Injuries to DNA that introduce deviations from its normal, intact structure and which may, if left unrepaired, result in a MUTATION or a block of DNA REPLICATION. These deviations may be caused by physical or chemical agents and occur by natural or unnatural, introduced circumstances. They include the introduction of illegitimate bases during replication or by deamination or other modification of bases; the loss of a base from the DNA backbone leaving an abasic site; single-strand breaks; double strand breaks; and intrastrand (PYRIMIDINE DIMERS) or interstrand crosslinking. Damage can often be repaired (DNA REPAIR). If the damage is extensive, it can induce APOPTOSIS.
The reconstruction of a continuous two-stranded DNA molecule without mismatch from a molecule which contained damaged regions. The major repair mechanisms are excision repair, in which defective regions in one strand are excised and resynthesized using the complementary base pairing information in the intact strand; photoreactivation repair, in which the lethal and mutagenic effects of ultraviolet light are eliminated; and post-replication repair, in which the primary lesions are not repaired, but the gaps in one daughter duplex are filled in by incorporation of portions of the other (undamaged) daughter duplex. Excision repair and post-replication repair are sometimes referred to as "dark repair" because they do not require light.
A genotoxicological technique for measuring DNA damage in an individual cell using single-cell gel electrophoresis. Cell DNA fragments assume a "comet with tail" formation on electrophoresis and are detected with an image analysis system. Alkaline assay conditions facilitate sensitive detection of single-strand damage.
A group of PROTEIN-SERINE-THREONINE KINASES which activate critical signaling cascades in double strand breaks, APOPTOSIS, and GENOTOXIC STRESS such as ionizing ultraviolet A light, thereby acting as a DNA damage sensor. These proteins play a role in a wide range of signaling mechanisms in cell cycle control.
Proteins that control the CELL DIVISION CYCLE. This family of proteins includes a wide variety of classes, including CYCLIN-DEPENDENT KINASES, mitogen-activated kinases, CYCLINS, and PHOSPHOPROTEIN PHOSPHATASES as well as their putative substrates such as chromatin-associated proteins, CYTOSKELETAL PROTEINS, and TRANSCRIPTION FACTORS.
That portion of the electromagnetic spectrum immediately below the visible range and extending into the x-ray frequencies. The longer wavelengths (near-UV or biotic or vital rays) are necessary for the endogenous synthesis of vitamin D and are also called antirachitic rays; the shorter, ionizing wavelengths (far-UV or abiotic or extravital rays) are viricidal, bactericidal, mutagenic, and carcinogenic and are used as disinfectants.
A nucleoside consisting of the base guanine and the sugar deoxyribose.
Enzyme activated in response to DNA DAMAGE involved in cell cycle arrest. The gene is located on the long (q) arm of chromosome 22 at position 12.1. In humans it is encoded by the CHEK2 gene.
Nuclear phosphoprotein encoded by the p53 gene (GENES, P53) whose normal function is to control CELL PROLIFERATION and APOPTOSIS. A mutant or absent p53 protein has been found in LEUKEMIA; OSTEOSARCOMA; LUNG CANCER; and COLORECTAL CANCER.
Proteins which bind to DNA. The family includes proteins which bind to both double- and single-stranded DNA and also includes specific DNA binding proteins in serum which can be used as markers for malignant diseases.
Interruptions in the sugar-phosphate backbone of DNA, across both strands adjacently.
A group of enzymes that catalyzes the phosphorylation of serine or threonine residues in proteins, with ATP or other nucleotides as phosphate donors.
A disturbance in the prooxidant-antioxidant balance in favor of the former, leading to potential damage. Indicators of oxidative stress include damaged DNA bases, protein oxidation products, and lipid peroxidation products (Sies, Oxidative Stress, 1991, pxv-xvi).
The complex series of phenomena, occurring between the end of one CELL DIVISION and the end of the next, by which cellular material is duplicated and then divided between two daughter cells. The cell cycle includes INTERPHASE, which includes G0 PHASE; G1 PHASE; S PHASE; and G2 PHASE, and CELL DIVISION PHASE.
Proteins that are normally involved in holding cellular growth in check. Deficiencies or abnormalities in these proteins may lead to unregulated cell growth and tumor development.
Penetrating, high-energy electromagnetic radiation emitted from atomic nuclei during NUCLEAR DECAY. The range of wavelengths of emitted radiation is between 0.1 - 100 pm which overlaps the shorter, more energetic hard X-RAYS wavelengths. The distinction between gamma rays and X-rays is based on their radiation source.
One of the mechanisms by which CELL DEATH occurs (compare with NECROSIS and AUTOPHAGOCYTOSIS). Apoptosis is the mechanism responsible for the physiological deletion of cells and appears to be intrinsically programmed. It is characterized by distinctive morphologic changes in the nucleus and cytoplasm, chromatin cleavage at regularly spaced sites, and the endonucleolytic cleavage of genomic DNA; (DNA FRAGMENTATION); at internucleosomal sites. This mode of cell death serves as a balance to mitosis in regulating the size of animal tissues and in mediating pathologic processes associated with tumor growth.
A deoxyribonucleotide polymer that is the primary genetic material of all cells. Eukaryotic and prokaryotic organisms normally contain DNA in a double-stranded state, yet several important biological processes transiently involve single-stranded regions. DNA, which consists of a polysugar-phosphate backbone possessing projections of purines (adenine and guanine) and pyrimidines (thymine and cytosine), forms a double helix that is held together by hydrogen bonds between these purines and pyrimidines (adenine to thymine and guanine to cytosine).
An alkylating agent in cancer therapy that may also act as a mutagen by interfering with and causing damage to DNA.
Small chromosomal proteins (approx 12-20 kD) possessing an open, unfolded structure and attached to the DNA in cell nuclei by ionic linkages. Classification into the various types (designated histone I, histone II, etc.) is based on the relative amounts of arginine and lysine in each.
Chemical agents that increase the rate of genetic mutation by interfering with the function of nucleic acids. A clastogen is a specific mutagen that causes breaks in chromosomes.
Proteins found in the nucleus of a cell. Do not confuse with NUCLEOPROTEINS which are proteins conjugated with nucleic acids, that are not necessarily present in the nucleus.
An increased tendency of the GENOME to acquire MUTATIONS when various processes involved in maintaining and replicating the genome are dysfunctional.
The period of the CELL CYCLE following DNA synthesis (S PHASE) and preceding M PHASE (cell division phase). The CHROMOSOMES are tetraploid in this point.
The process by which a DNA molecule is duplicated.
Any detectable and heritable change in the genetic material that causes a change in the GENOTYPE and which is transmitted to daughter cells and to succeeding generations.
The introduction of a phosphoryl group into a compound through the formation of an ester bond between the compound and a phosphorus moiety.
A cell line derived from cultured tumor cells.
Enzymes that are involved in the reconstruction of a continuous two-stranded DNA molecule without mismatch from a molecule, which contained damaged regions.
The relationship between the dose of administered radiation and the response of the organism or tissue to the radiation.
The products of chemical reactions that result in the addition of extraneous chemical groups to DNA.
Molecules or ions formed by the incomplete one-electron reduction of oxygen. These reactive oxygen intermediates include SINGLET OXYGEN; SUPEROXIDES; PEROXIDES; HYDROXYL RADICAL; and HYPOCHLOROUS ACID. They contribute to the microbicidal activity of PHAGOCYTES, regulation of signal transduction and gene expression, and the oxidative damage to NUCLEIC ACIDS; PROTEINS; and LIPIDS.
The span of viability of a cell characterized by the capacity to perform certain functions such as metabolism, growth, reproduction, some form of responsiveness, and adaptability.
Established cell cultures that have the potential to propagate indefinitely.
A strong oxidizing agent used in aqueous solution as a ripening agent, bleach, and topical anti-infective. It is relatively unstable and solutions deteriorate over time unless stabilized by the addition of acetanilide or similar organic materials.
Interruptions in the sugar-phosphate backbone of DNA.
The ability of some cells or tissues to survive lethal doses of IONIZING RADIATION. Tolerance depends on the species, cell type, and physical and chemical variables, including RADIATION-PROTECTIVE AGENTS and RADIATION-SENSITIZING AGENTS.
A family of enzymes that catalyze the conversion of ATP and a protein to ADP and a phosphoprotein.
Enzymes that catalyze the transfer of multiple ADP-RIBOSE groups from nicotinamide-adenine dinucleotide (NAD) onto protein targets, thus building up a linear or branched homopolymer of repeating ADP-ribose units i.e., POLY ADENOSINE DIPHOSPHATE RIBOSE.
Genes that code for proteins that regulate the CELL DIVISION CYCLE. These genes form a regulatory network that culminates in the onset of MITOSIS by activating the p34cdc2 protein (PROTEIN P34CDC2).
Regulatory signaling systems that control the progression through the CELL CYCLE. They ensure that the cell has completed, in the correct order and without mistakes, all the processes required to replicate the GENOME and CYTOPLASM, and divide them equally between two daughter cells. If cells sense they have not completed these processes or that the environment does not have the nutrients and growth hormones in place to proceed, then the cells are restrained (or "arrested") until the processes are completed and growth conditions are suitable.
Interruptions in one of the strands of the sugar-phosphate backbone of double-stranded DNA.
The intracellular transfer of information (biological activation/inhibition) through a signal pathway. In each signal transduction system, an activation/inhibition signal from a biologically active molecule (hormone, neurotransmitter) is mediated via the coupling of a receptor/enzyme to a second messenger system or to an ion channel. Signal transduction plays an important role in activating cellular functions, cell differentiation, and cell proliferation. Examples of signal transduction systems are the GAMMA-AMINOBUTYRIC ACID-postsynaptic receptor-calcium ion channel system, the receptor-mediated T-cell activation pathway, and the receptor-mediated activation of phospholipases. Those coupled to membrane depolarization or intracellular release of calcium include the receptor-mediated activation of cytotoxic functions in granulocytes and the synaptic potentiation of protein kinase activation. Some signal transduction pathways may be part of larger signal transduction pathways; for example, protein kinase activation is part of the platelet activation signal pathway.
A Rec A recombinase found in eukaryotes. Rad51 is involved in DNA REPAIR of double-strand breaks.
A family of DNA repair enzymes that recognize damaged nucleotide bases and remove them by hydrolyzing the N-glycosidic bond that attaches them to the sugar backbone of the DNA molecule. The process called BASE EXCISION REPAIR can be completed by a DNA-(APURINIC OR APYRIMIDINIC SITE) LYASE which excises the remaining RIBOSE sugar from the DNA.
The first continuously cultured human malignant CELL LINE, derived from the cervical carcinoma of Henrietta Lacks. These cells are used for VIRUS CULTIVATION and antitumor drug screening assays.
Phase of the CELL CYCLE following G1 and preceding G2 when the entire DNA content of the nucleus is replicated. It is achieved by bidirectional replication at multiple sites along each chromosome.
Proteins obtained from the species SACCHAROMYCES CEREVISIAE. The function of specific proteins from this organism are the subject of intense scientific interest and have been used to derive basic understanding of the functioning similar proteins in higher eukaryotes.
The phosphoprotein encoded by the BRCA1 gene (GENE, BRCA1). In normal cells the BRCA1 protein is localized in the nucleus, whereas in the majority of breast cancer cell lines and in malignant pleural effusions from breast cancer patients, it is localized mainly in the cytoplasm. (Science 1995;270(5237):713,789-91)
Cells propagated in vitro in special media conducive to their growth. Cultured cells are used to study developmental, morphologic, metabolic, physiologic, and genetic processes, among others.
Connective tissue cells which secrete an extracellular matrix rich in collagen and other macromolecules.
A type of CELL NUCLEUS division by means of which the two daughter nuclei normally receive identical complements of the number of CHROMOSOMES of the somatic cells of the species.
The decrease in the cell's ability to proliferate with the passing of time. Each cell is programmed for a certain number of cell divisions and at the end of that time proliferation halts. The cell enters a quiescent state after which it experiences CELL DEATH via the process of APOPTOSIS.
Induction and quantitative measurement of chromosomal damage leading to the formation of micronuclei (MICRONUCLEI, CHROMOSOME-DEFECTIVE) in cells which have been exposed to genotoxic agents or IONIZING RADIATION.
A species of the genus SACCHAROMYCES, family Saccharomycetaceae, order Saccharomycetales, known as "baker's" or "brewer's" yeast. The dried form is used as a dietary supplement.
Within a eukaryotic cell, a membrane-limited body which contains chromosomes and one or more nucleoli (CELL NUCLEOLUS). The nuclear membrane consists of a double unit-type membrane which is perforated by a number of pores; the outermost membrane is continuous with the ENDOPLASMIC RETICULUM. A cell may contain more than one nucleus. (From Singleton & Sainsbury, Dictionary of Microbiology and Molecular Biology, 2d ed)
An antineoplastic agent that inhibits DNA synthesis through the inhibition of ribonucleoside diphosphate reductase.
Naturally occurring or synthetic substances that inhibit or retard the oxidation of a substance to which it is added. They counteract the harmful and damaging effects of oxidation in animal tissues.
A serine-threonine protein kinase that, when activated by DNA, phosphorylates several DNA-binding protein substrates including the TUMOR SUPPRESSOR PROTEIN P53 and a variety of TRANSCRIPTION FACTORS.
CELL CYCLE regulatory signaling systems that are triggered by DNA DAMAGE or lack of nutrients during G2 PHASE. When triggered they restrain cells transitioning from G2 phase to M PHASE.
Proteins and peptides that are involved in SIGNAL TRANSDUCTION within the cell. Included here are peptides and proteins that regulate the activity of TRANSCRIPTION FACTORS and cellular processes in response to signals from CELL SURFACE RECEPTORS. Intracellular signaling peptide and proteins may be part of an enzymatic signaling cascade or act through binding to and modifying the action of other signaling factors.
The material of CHROMOSOMES. It is a complex of DNA; HISTONES; and nonhistone proteins (CHROMOSOMAL PROTEINS, NON-HISTONE) found within the nucleus of a cell.
Highly reactive chemicals that introduce alkyl radicals into biologically active molecules and thereby prevent their proper functioning. Many are used as antineoplastic agents, but most are very toxic, with carcinogenic, mutagenic, teratogenic, and immunosuppressant actions. They have also been used as components in poison gases.
Penetrating electromagnetic radiation emitted when the inner orbital electrons of an atom are excited and release radiant energy. X-ray wavelengths range from 1 pm to 10 nm. Hard X-rays are the higher energy, shorter wavelength X-rays. Soft x-rays or Grenz rays are less energetic and longer in wavelength. The short wavelength end of the X-ray spectrum overlaps the GAMMA RAYS wavelength range. The distinction between gamma rays and X-rays is based on their radiation source.
Elements of limited time intervals, contributing to particular results or situations.
A single-stranded DNA-binding protein that is found in EUKARYOTIC CELLS. It is required for DNA REPLICATION; DNA REPAIR; and GENETIC RECOMBINATION.
Repair of DNA DAMAGE by exchange of DNA between matching sequences, usually between the allelic DNA (ALLELES) of sister chromatids.
Theoretical representations that simulate the behavior or activity of biological processes or diseases. For disease models in living animals, DISEASE MODELS, ANIMAL is available. Biological models include the use of mathematical equations, computers, and other electronic equipment.
The process in which substances, either endogenous or exogenous, bind to proteins, peptides, enzymes, protein precursors, or allied compounds. Specific protein-binding measures are often used as assays in diagnostic assessments.
Proteins that catalyze the unwinding of duplex DNA during replication by binding cooperatively to single-stranded regions of DNA or to short regions of duplex DNA that are undergoing transient opening. In addition DNA helicases are DNA-dependent ATPases that harness the free energy of ATP hydrolysis to translocate DNA strands.
A chemical reaction in which an electron is transferred from one molecule to another. The electron-donating molecule is the reducing agent or reductant; the electron-accepting molecule is the oxidizing agent or oxidant. Reducing and oxidizing agents function as conjugate reductant-oxidant pairs or redox pairs (Lehninger, Principles of Biochemistry, 1982, p471).
A polynucleotide formed from the ADP-RIBOSE moiety of nicotinamide-adenine dinucleotide (NAD) by POLY(ADP-RIBOSE) POLYMERASES.
The termination of the cell's ability to carry out vital functions such as metabolism, growth, reproduction, responsiveness, and adaptability.
Small double-stranded, non-protein coding RNAs (21-31 nucleotides) involved in GENE SILENCING functions, especially RNA INTERFERENCE (RNAi). Endogenously, siRNAs are generated from dsRNAs (RNA, DOUBLE-STRANDED) by the same ribonuclease, Dicer, that generates miRNAs (MICRORNAS). The perfect match of the siRNAs' antisense strand to their target RNAs mediates RNAi by siRNA-guided RNA cleavage. siRNAs fall into different classes including trans-acting siRNA (tasiRNA), repeat-associated RNA (rasiRNA), small-scan RNA (scnRNA), and Piwi protein-interacting RNA (piRNA) and have different specific gene silencing functions.
Descriptions of specific amino acid, carbohydrate, or nucleotide sequences which have appeared in the published literature and/or are deposited in and maintained by databanks such as GENBANK, European Molecular Biology Laboratory (EMBL), National Biomedical Research Foundation (NBRF), or other sequence repositories.
An error-prone mechanism or set of functions for repairing damaged microbial DNA. SOS functions (a concept reputedly derived from the SOS of the international distress signal) are involved in DNA repair and mutagenesis, in cell division inhibition, in recovery of normal physiological conditions after DNA repair, and possibly in cell death when DNA damage is extensive.
An E3 UBIQUITIN LIGASE that interacts with and inhibits TUMOR SUPPRESSOR PROTEIN P53. Its ability to ubiquitinate p53 is regulated by TUMOR SUPPRESSOR PROTEIN P14ARF.
Production of new arrangements of DNA by various mechanisms such as assortment and segregation, CROSSING OVER; GENE CONVERSION; GENETIC TRANSFORMATION; GENETIC CONJUGATION; GENETIC TRANSDUCTION; or mixed infection of viruses.
Dimers found in DNA chains damaged by ULTRAVIOLET RAYS. They consist of two adjacent PYRIMIDINE NUCLEOTIDES, usually THYMINE nucleotides, in which the pyrimidine residues are covalently joined by a cyclobutane ring. These dimers block DNA REPLICATION.
A DNA repair enzyme that is an N-glycosyl hydrolase with specificity for DNA-containing ring-opened N(7)-methylguanine residues.
An antineoplastic antibiotic produced by Streptomyces caespitosus. It is one of the bi- or tri-functional ALKYLATING AGENTS causing cross-linking of DNA and inhibition of DNA synthesis.
Human COLORECTAL CARCINOMA cell line.
Defective nuclei produced during the TELOPHASE of MITOSIS or MEIOSIS by lagging CHROMOSOMES or chromosome fragments derived from spontaneous or experimentally induced chromosomal structural changes.
The act of ligating UBIQUITINS to PROTEINS to form ubiquitin-protein ligase complexes to label proteins for transport to the PROTEASOME ENDOPEPTIDASE COMPLEX where proteolysis occurs.
A cyclin-dependent kinase inhibitor that mediates TUMOR SUPPRESSOR PROTEIN P53-dependent CELL CYCLE arrest. p21 interacts with a range of CYCLIN-DEPENDENT KINASES and associates with PROLIFERATING CELL NUCLEAR ANTIGEN and CASPASE 3.
A terminal section of a chromosome which has a specialized structure and which is involved in chromosomal replication and stability. Its length is believed to be a few hundred base pairs.
Compounds that inhibit cell production of DNA or RNA.
The repair of DOUBLE-STRAND DNA BREAKS by rejoining the broken ends of DNA to each other directly.
Identification of proteins or peptides that have been electrophoretically separated by blot transferring from the electrophoresis gel to strips of nitrocellulose paper, followed by labeling with antibody probes.
Tests of chemical substances and physical agents for mutagenic potential. They include microbial, insect, mammalian cell, and whole animal tests.
Guanine is a purine nucleobase, one of the four nucleobases in the nucleic acid of DNA and RNA, involved in forming hydrogen bonds between complementary base pairs in double-stranded DNA molecules.
The relationship between the dose of an administered drug and the response of the organism to the drug.
A ZINC FINGER MOTIF protein that recognizes and interacts with damaged DNA. It is a DNA-binding protein that plays an essential role in NUCLEOTIDE EXCISION REPAIR. Mutations in this protein are associated with the most severe form of XERODERMA PIGMENTOSUM.
White blood cells formed in the body's lymphoid tissue. The nucleus is round or ovoid with coarse, irregularly clumped chromatin while the cytoplasm is typically pale blue with azurophilic (if any) granules. Most lymphocytes can be classified as either T or B (with subpopulations of each), or NATURAL KILLER CELLS.
Deoxyribonucleic acid that makes up the genetic material of fungi.
An autosomal recessive inherited disorder characterized by choreoathetosis beginning in childhood, progressive CEREBELLAR ATAXIA; TELANGIECTASIS of CONJUNCTIVA and SKIN; DYSARTHRIA; B- and T-cell immunodeficiency, and RADIOSENSITIVITY to IONIZING RADIATION. Affected individuals are prone to recurrent sinobronchopulmonary infections, lymphoreticular neoplasms, and other malignancies. Serum ALPHA-FETOPROTEINS are usually elevated. (Menkes, Textbook of Child Neurology, 5th ed, p688) The gene for this disorder (ATM) encodes a cell cycle checkpoint protein kinase and has been mapped to chromosome 11 (11q22-q23).
Electron-accepting molecules in chemical reactions in which electrons are transferred from one molecule to another (OXIDATION-REDUCTION).
A group of enzymes catalyzing the endonucleolytic cleavage of DNA. They include members of EC 3.1.21.-, EC 3.1.22.-, EC 3.1.23.- (DNA RESTRICTION ENZYMES), EC 3.1.24.- (DNA RESTRICTION ENZYMES), and EC 3.1.25.-.
A subclass of dual specificity phosphatases that play a role in the progression of the CELL CYCLE. They dephosphorylate and activate CYCLIN-DEPENDENT KINASES.
Nuclear antigen with a role in DNA synthesis, DNA repair, and cell cycle progression. PCNA is required for the coordinated synthesis of both leading and lagging strands at the replication fork during DNA replication. PCNA expression correlates with the proliferation activity of several malignant and non-malignant cell types.
Immunologically detectable substances found in the CELL NUCLEUS.
Enzymes that catalyze the hydrolysis of the internal bonds and thereby the formation of polynucleotides or oligonucleotides from ribo- or deoxyribonucleotide chains. EC 3.1.-.
A gene silencing phenomenon whereby specific dsRNAs (RNA, DOUBLE-STRANDED) trigger the degradation of homologous mRNA (RNA, MESSENGER). The specific dsRNAs are processed into SMALL INTERFERING RNA (siRNA) which serves as a guide for cleavage of the homologous mRNA in the RNA-INDUCED SILENCING COMPLEX. DNA METHYLATION may also be triggered during this process.
Strains of mice in which certain GENES of their GENOMES have been disrupted, or "knocked-out". To produce knockouts, using RECOMBINANT DNA technology, the normal DNA sequence of the gene being studied is altered to prevent synthesis of a normal gene product. Cloned cells in which this DNA alteration is successful are then injected into mouse EMBRYOS to produce chimeric mice. The chimeric mice are then bred to yield a strain in which all the cells of the mouse contain the disrupted gene. Knockout mice are used as EXPERIMENTAL ANIMAL MODELS for diseases (DISEASE MODELS, ANIMAL) and to clarify the functions of the genes.
Substances that increase the risk of NEOPLASMS in humans or animals. Both genotoxic chemicals, which affect DNA directly, and nongenotoxic chemicals, which induce neoplasms by other mechanism, are included.
An exchange of DNA between matching or similar sequences.
Tumor suppressor genes located on the short arm of human chromosome 17 and coding for the phosphoprotein p53.
Proteins obtained from the species Schizosaccharomyces pombe. The function of specific proteins from this organism are the subject of intense scientific interest and have been used to derive basic understanding of the functioning similar proteins in higher eukaryotes.
A condition characterized by long-standing brain dysfunction or damage, usually of three months duration or longer. Potential etiologies include BRAIN INFARCTION; certain NEURODEGENERATIVE DISORDERS; CRANIOCEREBRAL TRAUMA; ANOXIA, BRAIN; ENCEPHALITIS; certain NEUROTOXICITY SYNDROMES; metabolic disorders (see BRAIN DISEASES, METABOLIC); and other conditions.
Splitting the DNA into shorter pieces by endonucleolytic DNA CLEAVAGE at multiple sites. It includes the internucleosomal DNA fragmentation, which along with chromatin condensation, are considered to be the hallmarks of APOPTOSIS.
Peroxidase catalyzed oxidation of lipids using hydrogen peroxide as an electron acceptor.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
Substances that inhibit or prevent the proliferation of NEOPLASMS.
7,8,8a,9a-Tetrahydrobenzo(10,11)chryseno (3,4-b)oxirene-7,8-diol. A benzopyrene derivative with carcinogenic and mutagenic activity.
All of the processes involved in increasing CELL NUMBER including CELL DIVISION.
A genus of ascomycetous fungi of the family Schizosaccharomycetaceae, order Schizosaccharomycetales.
The biosynthesis of RNA carried out on a template of DNA. The biosynthesis of DNA from an RNA template is called REVERSE TRANSCRIPTION.
A semisynthetic derivative of PODOPHYLLOTOXIN that exhibits antitumor activity. Etoposide inhibits DNA synthesis by forming a complex with topoisomerase II and DNA. This complex induces breaks in double stranded DNA and prevents repair by topoisomerase II binding. Accumulated breaks in DNA prevent entry into the mitotic phase of cell division, and lead to cell death. Etoposide acts primarily in the G2 and S phases of the cell cycle.
Any of the processes by which nuclear, cytoplasmic, or intercellular factors influence the differential control (induction or repression) of gene action at the level of transcription or translation.
A family of enzymes that catalyze the exonucleolytic cleavage of DNA. It includes members of the class EC 3.1.11 that produce 5'-phosphomonoesters as cleavage products.
Inbred C57BL mice are a strain of laboratory mice that have been produced by many generations of brother-sister matings, resulting in a high degree of genetic uniformity and homozygosity, making them widely used for biomedical research, including studies on genetics, immunology, cancer, and neuroscience.
Rate of energy dissipation along the path of charged particles. In radiobiology and health physics, exposure is measured in kiloelectron volts per micrometer of tissue (keV/micrometer T).
A highly conserved 76-amino acid peptide universally found in eukaryotic cells that functions as a marker for intracellular PROTEIN TRANSPORT and degradation. Ubiquitin becomes activated through a series of complicated steps and forms an isopeptide bond to lysine residues of specific proteins within the cell. These "ubiquitinated" proteins can be recognized and degraded by proteosomes or be transported to specific compartments within the cell.
A diverse class of enzymes that interact with UBIQUITIN-CONJUGATING ENZYMES and ubiquitination-specific protein substrates. Each member of this enzyme group has its own distinct specificity for a substrate and ubiquitin-conjugating enzyme. Ubiquitin-protein ligases exist as both monomeric proteins multiprotein complexes.
Cells grown in vitro from neoplastic tissue. If they can be established as a TUMOR CELL LINE, they can be propagated in cell culture indefinitely.
Technique using an instrument system for making, processing, and displaying one or more measurements on individual cells obtained from a cell suspension. Cells are usually stained with one or more fluorescent dyes specific to cell components of interest, e.g., DNA, and fluorescence of each cell is measured as it rapidly transverses the excitation beam (laser or mercury arc lamp). Fluorescence provides a quantitative measure of various biochemical and biophysical properties of the cell, as well as a basis for cell sorting. Other measurable optical parameters include light absorption and light scattering, the latter being applicable to the measurement of cell size, shape, density, granularity, and stain uptake.
Nucleoproteins, which in contrast to HISTONES, are acid insoluble. They are involved in chromosomal functions; e.g. they bind selectively to DNA, stimulate transcription resulting in tissue-specific RNA synthesis and undergo specific changes in response to various hormones or phytomitogens.
Mature male germ cells derived from SPERMATIDS. As spermatids move toward the lumen of the SEMINIFEROUS TUBULES, they undergo extensive structural changes including the loss of cytoplasm, condensation of CHROMATIN into the SPERM HEAD, formation of the ACROSOME cap, the SPERM MIDPIECE and the SPERM TAIL that provides motility.
A potent mutagen and carcinogen. This compound and its metabolite 4-HYDROXYAMINOQUINOLINE-1-OXIDE bind to nucleic acids. It inactivates bacteria but not bacteriophage.
An in situ method for detecting areas of DNA which are nicked during APOPTOSIS. Terminal deoxynucleotidyl transferase is used to add labeled dUTP, in a template-independent manner, to the 3 prime OH ends of either single- or double-stranded DNA. The terminal deoxynucleotidyl transferase nick end labeling, or TUNEL, assay labels apoptosis on a single-cell level, making it more sensitive than agarose gel electrophoresis for analysis of DNA FRAGMENTATION.
An exchange of segments between the sister chromatids of a chromosome, either between the sister chromatids of a meiotic tetrad or between the sister chromatids of a duplicated somatic chromosome. Its frequency is increased by ultraviolet and ionizing radiation and other mutagenic agents and is particularly high in BLOOM SYNDROME.
A genetic rearrangement through loss of segments of DNA or RNA, bringing sequences which are normally separated into close proximity. This deletion may be detected using cytogenetic techniques and can also be inferred from the phenotype, indicating a deletion at one specific locus.
An enediyne that alkylates DNA and RNA like MITOMYCIN does, so it is cytotoxic.
A single chain of deoxyribonucleotides that occurs in some bacteria and viruses. It usually exists as a covalently closed circle.
Endogenous substances, usually proteins, which are effective in the initiation, stimulation, or termination of the genetic transcription process.
The order of amino acids as they occur in a polypeptide chain. This is referred to as the primary structure of proteins. It is of fundamental importance in determining PROTEIN CONFORMATION.
Process of generating a genetic MUTATION. It may occur spontaneously or be induced by MUTAGENS.
A family of structurally-related DNA helicases that play an essential role in the maintenance of genome integrity. RecQ helicases were originally discovered in E COLI and are highly conserved across both prokaryotic and eukaryotic organisms. Genetic mutations that result in loss of RecQ helicase activity gives rise to disorders that are associated with CANCER predisposition and premature aging.
A class of enzymes involved in the hydrolysis of the N-glycosidic bond of nitrogen-linked sugars.
A ubiquitously expressed telomere-binding protein that is present at TELOMERES throughout the cell cycle. It is a suppressor of telomere elongation and may be involved in stabilization of telomere length. It is structurally different from TELOMERIC REPEAT BINDING PROTEIN 1 in that it contains basic N-terminal amino acid residues.
DNA present in neoplastic tissue.
A DNA repair enzyme that catalyses the excision of ribose residues at apurinic and apyrimidinic DNA sites that can result from the action of DNA GLYCOSYLASES. The enzyme catalyzes a beta-elimination reaction in which the C-O-P bond 3' to the apurinic or apyrimidinic site in DNA is broken, leaving a 3'-terminal unsaturated sugar and a product with a terminal 5'-phosphate. This enzyme was previously listed under EC 3.1.25.2.
The level of protein structure in which combinations of secondary protein structures (alpha helices, beta sheets, loop regions, and motifs) pack together to form folded shapes called domains. Disulfide bridges between cysteines in two different parts of the polypeptide chain along with other interactions between the chains play a role in the formation and stabilization of tertiary structure. Small proteins usually consist of only one domain but larger proteins may contain a number of domains connected by segments of polypeptide chain which lack regular secondary structure.
The mechanisms effecting establishment, maintenance, and modification of that specific physical conformation of CHROMATIN determining the transcriptional accessibility or inaccessibility of the DNA.
Agents that reduce the frequency or rate of spontaneous or induced mutations independently of the mechanism involved.
Cell regulatory signaling system that controls progression through S PHASE and stabilizes the replication forks during conditions that could affect the fidelity of DNA REPLICATION, such as DNA DAMAGE or depletion of nucleotide pools.
The period of the CELL CYCLE preceding DNA REPLICATION in S PHASE. Subphases of G1 include "competence" (to respond to growth factors), G1a (entry into G1), G1b (progression), and G1c (assembly). Progression through the G1 subphases is effected by limiting growth factors, nutrients, or inhibitors.
An inorganic and water-soluble platinum complex. After undergoing hydrolysis, it reacts with DNA to produce both intra and interstrand crosslinks. These crosslinks appear to impair replication and transcription of DNA. The cytotoxicity of cisplatin correlates with cellular arrest in the G2 phase of the cell cycle.
Highly reactive molecules with an unsatisfied electron valence pair. Free radicals are produced in both normal and pathological processes. They are proven or suspected agents of tissue damage in a wide variety of circumstances including radiation, damage from environment chemicals, and aging. Natural and pharmacological prevention of free radical damage is being actively investigated.
A rare, pigmentary, and atrophic autosomal recessive disease. It is manifested as an extreme photosensitivity to ULTRAVIOLET RAYS as the result of a deficiency in the enzyme that permits excisional repair of ultraviolet-damaged DNA.
Abnormal number or structure of chromosomes. Chromosome aberrations may result in CHROMOSOME DISORDERS.
An oxidoreductase that catalyzes the reaction between superoxide anions and hydrogen to yield molecular oxygen and hydrogen peroxide. The enzyme protects the cell against dangerous levels of superoxide. EC 1.15.1.1.
Microscopy of specimens stained with fluorescent dye (usually fluorescein isothiocyanate) or of naturally fluorescent materials, which emit light when exposed to ultraviolet or blue light. Immunofluorescence microscopy utilizes antibodies that are labeled with fluorescent dye.
Conversion of an inactive form of an enzyme to one possessing metabolic activity. It includes 1, activation by ions (activators); 2, activation by cofactors (coenzymes); and 3, conversion of an enzyme precursor (proenzyme or zymogen) to an active enzyme.
DNA-dependent DNA polymerases found in bacteria, animal and plant cells. During the replication process, these enzymes catalyze the addition of deoxyribonucleotide residues to the end of a DNA strand in the presence of DNA as template-primer. They also possess exonuclease activity and therefore function in DNA repair.
Naturally occurring or experimentally induced animal diseases with pathological processes sufficiently similar to those of human diseases. They are used as study models for human diseases.
Antineoplastic antibiotic obtained from Streptomyces peucetius. It is a hydroxy derivative of DAUNORUBICIN.
An alkaloid isolated from the stem wood of the Chinese tree, Camptotheca acuminata. This compound selectively inhibits the nuclear enzyme DNA TOPOISOMERASES, TYPE I. Several semisynthetic analogs of camptothecin have demonstrated antitumor activity.
Substances that influence the course of a chemical reaction by ready combination with free radicals. Among other effects, this combining activity protects pancreatic islets against damage by cytokines and prevents myocardial and pulmonary perfusion injuries.
The uptake of naked or purified DNA by CELLS, usually meaning the process as it occurs in eukaryotic cells. It is analogous to bacterial transformation (TRANSFORMATION, BACTERIAL) and both are routinely employed in GENE TRANSFER TECHNIQUES.
Chemical substances, produced by microorganisms, inhibiting or preventing the proliferation of neoplasms.
New abnormal growth of tissue. Malignant neoplasms show a greater degree of anaplasia and have the properties of invasion and metastasis, compared to benign neoplasms.
Drugs used to protect against ionizing radiation. They are usually of interest for use in radiation therapy but have been considered for other, e.g. military, purposes.
A tripeptide with many roles in cells. It conjugates to drugs to make them more soluble for excretion, is a cofactor for some enzymes, is involved in protein disulfide bond rearrangement and reduces peroxides.
A Fanconi anemia complementation group protein that undergoes mono-ubiquitination by FANCL PROTEIN in response to DNA DAMAGE. Also, in response to IONIZING RADIATION it can undergo PHOSPHORYLATION by ataxia telangiectasia mutated protein. Modified FANCD2 interacts with BRCA2 PROTEIN in a stable complex with CHROMATIN, and it is involved in DNA REPAIR by homologous RECOMBINATION.
An oxidoreductase that catalyzes the conversion of HYDROGEN PEROXIDE to water and oxygen. It is present in many animal cells. A deficiency of this enzyme results in ACATALASIA.
Compounds or agents that combine with an enzyme in such a manner as to prevent the normal substrate-enzyme combination and the catalytic reaction.
The fission of a CELL. It includes CYTOKINESIS, when the CYTOPLASM of a cell is divided, and CELL NUCLEUS DIVISION.
Compounds that inhibit the activity of DNA TOPOISOMERASE I.
An increased tendency to acquire CHROMOSOME ABERRATIONS when various processes involved in chromosome replication, repair, or segregation are dysfunctional.
Formation of an acetyl derivative. (Stedman, 25th ed)
Double-stranded DNA of MITOCHONDRIA. In eukaryotes, the mitochondrial GENOME is circular and codes for ribosomal RNAs, transfer RNAs, and about 10 proteins.
Phosphoprotein with protein kinase activity that functions in the G2/M phase transition of the CELL CYCLE. It is the catalytic subunit of the MATURATION-PROMOTING FACTOR and complexes with both CYCLIN A and CYCLIN B in mammalian cells. The maximal activity of cyclin-dependent kinase 1 is achieved when it is fully dephosphorylated.
Changes in the organism associated with senescence, occurring at an accelerated rate.
RNA sequences that serve as templates for protein synthesis. Bacterial mRNAs are generally primary transcripts in that they do not require post-transcriptional processing. Eukaryotic mRNA is synthesized in the nucleus and must be exported to the cytoplasm for translation. Most eukaryotic mRNAs have a sequence of polyadenylic acid at the 3' end, referred to as the poly(A) tail. The function of this tail is not known for certain, but it may play a role in the export of mature mRNA from the nucleus as well as in helping stabilize some mRNA molecules by retarding their degradation in the cytoplasm.
A nitrosoguanidine derivative with potent mutagenic and carcinogenic properties.
Semiautonomous, self-reproducing organelles that occur in the cytoplasm of all cells of most, but not all, eukaryotes. Each mitochondrion is surrounded by a double limiting membrane. The inner membrane is highly invaginated, and its projections are called cristae. Mitochondria are the sites of the reactions of oxidative phosphorylation, which result in the formation of ATP. They contain distinctive RIBOSOMES, transfer RNAs (RNA, TRANSFER); AMINO ACYL T RNA SYNTHETASES; and elongation and termination factors. Mitochondria depend upon genes within the nucleus of the cells in which they reside for many essential messenger RNAs (RNA, MESSENGER). Mitochondria are believed to have arisen from aerobic bacteria that established a symbiotic relationship with primitive protoeukaryotes. (King & Stansfield, A Dictionary of Genetics, 4th ed)
A negative regulatory effect on physiological processes at the molecular, cellular, or systemic level. At the molecular level, the major regulatory sites include membrane receptors, genes (GENE EXPRESSION REGULATION), mRNAs (RNA, MESSENGER), and proteins.
DNA TOPOISOMERASES that catalyze ATP-independent breakage of one of the two strands of DNA, passage of the unbroken strand through the break, and rejoining of the broken strand. DNA Topoisomerases, Type I enzymes reduce the topological stress in the DNA structure by relaxing the superhelical turns and knotted rings in the DNA helix.
The artificial induction of GENE SILENCING by the use of RNA INTERFERENCE to reduce the expression of a specific gene. It includes the use of DOUBLE-STRANDED RNA, such as SMALL INTERFERING RNA and RNA containing HAIRPIN LOOP SEQUENCE, and ANTI-SENSE OLIGONUCLEOTIDES.
Proteins found in any species of fungus.
A non-essential amino acid occurring in natural form as the L-isomer. It is synthesized from GLYCINE or THREONINE. It is involved in the biosynthesis of PURINES; PYRIMIDINES; and other amino acids.
A DNA-binding protein that mediates DNA REPAIR of double strand breaks, and HOMOLOGOUS RECOMBINATION.
A cell line generated from human embryonic kidney cells that were transformed with human adenovirus type 5.
Transport proteins that carry specific substances in the blood or across cell membranes.
Any of various enzymatically catalyzed post-translational modifications of PEPTIDES or PROTEINS in the cell of origin. These modifications include carboxylation; HYDROXYLATION; ACETYLATION; PHOSPHORYLATION; METHYLATION; GLYCOSYLATION; ubiquitination; oxidation; proteolysis; and crosslinking and result in changes in molecular weight and electrophoretic motility.
Congenital disorder affecting all bone marrow elements, resulting in ANEMIA; LEUKOPENIA; and THROMBOPENIA, and associated with cardiac, renal, and limb malformations as well as dermal pigmentary changes. Spontaneous CHROMOSOME BREAKAGE is a feature of this disease along with predisposition to LEUKEMIA. There are at least 7 complementation groups in Fanconi anemia: FANCA, FANCB, FANCC, FANCD1, FANCD2, FANCE, FANCF, FANCG, and FANCL. (from Online Mendelian Inheritance in Man, http://www.ncbi.nlm.nih.gov/entrez/dispomim.cgi?id=227650, August 20, 2004)
Immunologic method used for detecting or quantifying immunoreactive substances. The substance is identified by first immobilizing it by blotting onto a membrane and then tagging it with labeled antibodies.
Compounds that inhibit the activity of DNA TOPOISOMERASES.
Extrachromosomal, usually CIRCULAR DNA molecules that are self-replicating and transferable from one organism to another. They are found in a variety of bacterial, archaeal, fungal, algal, and plant species. They are used in GENETIC ENGINEERING as CLONING VECTORS.
A large lobed glandular organ in the abdomen of vertebrates that is responsible for detoxification, metabolism, synthesis and storage of various substances.
Histochemical localization of immunoreactive substances using labeled antibodies as reagents.
Drugs used to potentiate the effectiveness of radiation therapy in destroying unwanted cells.
A strain of albino rat used widely for experimental purposes because of its calmness and ease of handling. It was developed by the Sprague-Dawley Animal Company.
The gradual irreversible changes in structure and function of an organism that occur as a result of the passage of time.
Measurable and quantifiable biological parameters (e.g., specific enzyme concentration, specific hormone concentration, specific gene phenotype distribution in a population, presence of biological substances) which serve as indices for health- and physiology-related assessments, such as disease risk, psychiatric disorders, environmental exposure and its effects, disease diagnosis, metabolic processes, substance abuse, pregnancy, cell line development, epidemiologic studies, etc.
A family of recombinases initially identified in BACTERIA. They catalyze the ATP-driven exchange of DNA strands in GENETIC RECOMBINATION. The product of the reaction consists of a duplex and a displaced single-stranded loop, which has the shape of the letter D and is therefore called a D-loop structure.
The outer covering of the body that protects it from the environment. It is composed of the DERMIS and the EPIDERMIS.
The covalent bonding of an alkyl group to an organic compound. It can occur by a simple addition reaction or by substitution of another functional group.
A subfamily in the family MURIDAE, comprising the hamsters. Four of the more common genera are Cricetus, CRICETULUS; MESOCRICETUS; and PHODOPUS.
An E2F transcription factor that interacts directly with RETINOBLASTOMA PROTEIN and CYCLIN A and activates GENETIC TRANSCRIPTION required for CELL CYCLE entry and DNA synthesis. E2F1 is involved in DNA REPAIR and APOPTOSIS.
Macromolecular complexes formed from the association of defined protein subunits.

Long-range oxidative damage to DNA: effects of distance and sequence. (1/17364)

INTRODUCTION: Oxidative damage to DNA in vivo can lead to mutations and cancer. DNA damage and repair studies have not yet revealed whether permanent oxidative lesions are generated by charges migrating over long distances. Both photoexcited *Rh(III) and ground-state Ru(III) intercalators were previously shown to oxidize guanine bases from a remote site in oligonucleotide duplexes by DNA-mediated electron transfer. Here we examine much longer charge-transport distances and explore the sensitivity of the reaction to intervening sequences. RESULTS: Oxidative damage was examined in a series of DNA duplexes containing a pendant intercalating photooxidant. These studies revealed a shallow dependence on distance and no dependence on the phasing orientation of the oxidant relative to the site of damage, 5'-GG-3'. The intervening DNA sequence has a significant effect on the yield of guanine oxidation, however. Oxidation through multiple 5'-TA-3' steps is substantially diminished compared to through other base steps. We observed intraduplex guanine oxidation by tethered *Rh(III) and Ru(III) over a distance of 200 A. The distribution of oxidized guanine varied as a function of temperature between 5 and 35 degrees C, with an increase in the proportion of long-range damage (> 100 A) occurring at higher temperatures. CONCLUSIONS: Guanines are oxidized as a result of DNA-mediated charge transport over significant distances (e.g. 200 A). Although long-range charge transfer is dependent on distance, it appears to be modulated by intervening sequence and sequence-dependent dynamics. These discoveries hold important implications with respect to DNA damage in vivo.  (+info)

Gadd45, a p53-responsive stress protein, modifies DNA accessibility on damaged chromatin. (2/17364)

This report demonstrates that Gadd45, a p53-responsive stress protein, can facilitate topoisomerase relaxing and cleavage activity in the presence of core histones. A correlation between reduced expression of Gadd45 and increased resistance to topoisomerase I and topoisomerase II inhibitors in a variety of human cell lines was also found. Gadd45 could potentially mediate this effect by destabilizing histone-DNA interactions since it was found to interact directly with the four core histones. To evaluate this possibility, we investigated the effect of Gadd45 on preassembled mononucleosomes. Our data indicate that Gadd45 directly associates with mononucleosomes that have been altered by histone acetylation or UV radiation. This interaction resulted in increased DNase I accessibility on hyperacetylated mononucleosomes and substantial reduction of T4 endonuclease V accessibility to cyclobutane pyrimidine dimers on UV-irradiated mononucleosomes but not on naked DNA. Both histone acetylation and UV radiation are thought to destabilize the nucleosomal structure. Hence, these results imply that Gadd45 can recognize an altered chromatin state and modulate DNA accessibility to cellular proteins.  (+info)

The Saccharomyces cerevisiae ETH1 gene, an inducible homolog of exonuclease III that provides resistance to DNA-damaging agents and limits spontaneous mutagenesis. (3/17364)

The recently sequenced Saccharomyces cerevisiae genome was searched for a gene with homology to the gene encoding the major human AP endonuclease, a component of the highly conserved DNA base excision repair pathway. An open reading frame was found to encode a putative protein (34% identical to the Schizosaccharomyces pombe eth1(+) [open reading frame SPBC3D6.10] gene product) with a 347-residue segment homologous to the exonuclease III family of AP endonucleases. Synthesis of mRNA from ETH1 in wild-type cells was induced sixfold relative to that in untreated cells after exposure to the alkylating agent methyl methanesulfonate (MMS). To investigate the function of ETH1, deletions of the open reading frame were made in a wild-type strain and a strain deficient in the known yeast AP endonuclease encoded by APN1. eth1 strains were not more sensitive to killing by MMS, hydrogen peroxide, or phleomycin D1, whereas apn1 strains were approximately 3-fold more sensitive to MMS and approximately 10-fold more sensitive to hydrogen peroxide than was the wild type. Double-mutant strains (apn1 eth1) were approximately 15-fold more sensitive to MMS and approximately 2- to 3-fold more sensitive to hydrogen peroxide and phleomycin D1 than were apn1 strains. Elimination of ETH1 in apn1 strains also increased spontaneous mutation rates 9- or 31-fold compared to the wild type as determined by reversion to adenine or lysine prototrophy, respectively. Transformation of apn1 eth1 cells with an expression vector containing ETH1 reversed the hypersensitivity to MMS and limited the rate of spontaneous mutagenesis. Expression of ETH1 in a dut-1 xthA3 Escherichia coli strain demonstrated that the gene product functionally complements the missing AP endonuclease activity. Thus, in apn1 cells where the major AP endonuclease activity is missing, ETH1 offers an alternate capacity for repair of spontaneous or induced damage to DNA that is normally repaired by Apn1 protein.  (+info)

Impaired translesion synthesis in xeroderma pigmentosum variant extracts. (4/17364)

Xeroderma pigmentosum variant (XPV) cells are characterized by a cellular defect in the ability to synthesize intact daughter DNA strands on damaged templates. Molecular mechanisms that facilitate replication fork progression on damaged DNA in normal cells are not well defined. In this study, we used single-stranded plasmid molecules containing a single N-2-acetylaminofluorene (AAF) adduct to analyze translesion synthesis (TLS) catalyzed by extracts of either normal or XPV primary skin fibroblasts. In one of the substrates, the single AAF adduct was located at the 3' end of a run of three guanines that was previously shown to induce deletion of one G by a slippage mechanism. Primer extension reactions performed by normal cellular extracts from four different individuals produced the same distinct pattern of TLS, with over 80% of the products resulting from the elongation of a slipped intermediate and the remaining 20% resulting from a nonslipped intermediate. In contrast, with cellular extracts from five different XPV patients, the TLS reaction was strongly reduced, yielding only low amounts of TLS via the nonslipped intermediate. With our second substrate, in which the AAF adduct was located at the first G in the run, thus preventing slippage from occurring, we confirmed that normal extracts were able to perform TLS 10-fold more efficiently than XPV extracts. These data demonstrate unequivocally that the defect in XPV cells resides in translesion synthesis independently of the slippage process.  (+info)

Postnatal growth failure, short life span, and early onset of cellular senescence and subsequent immortalization in mice lacking the xeroderma pigmentosum group G gene. (5/17364)

The xeroderma pigmentosum group G (XP-G) gene (XPG) encodes a structure-specific DNA endonuclease that functions in nucleotide excision repair (NER). XP-G patients show various symptoms, ranging from mild cutaneous abnormalities to severe dermatological impairments. In some cases, patients exhibit growth failure and life-shortening and neurological dysfunctions, which are characteristics of Cockayne syndrome (CS). The known XPG protein function as the 3' nuclease in NER, however, cannot explain the development of CS in certain XP-G patients. To gain an insight into the functions of the XPG protein, we have generated and examined mice lacking xpg (the mouse counterpart of the human XPG gene) alleles. The xpg-deficient mice exhibited postnatal growth failure and underwent premature death. Since XPA-deficient mice, which are totally defective in NER, do not show such symptoms, our data indicate that XPG performs an additional function(s) besides its role in NER. Our in vitro studies showed that primary embryonic fibroblasts isolated from the xpg-deficient mice underwent premature senescence and exhibited the early onset of immortalization and accumulation of p53.  (+info)

Analysis of genomic integrity and p53-dependent G1 checkpoint in telomerase-induced extended-life-span human fibroblasts. (6/17364)

Life span determination in normal human cells may be regulated by nucleoprotein structures called telomeres, the physical ends of eukaryotic chromosomes. Telomeres have been shown to be essential for chromosome stability and function and to shorten with each cell division in normal human cells in culture and with age in vivo. Reversal of telomere shortening by the forced expression of telomerase in normal cells has been shown to elongate telomeres and extend the replicative life span (H. Vaziri and S. Benchimol, Curr. Biol. 8:279-282, 1998; A. G. Bodnar et al., Science 279:349-352, 1998). Extension of the life span as a consequence of the functional inactivation of p53 is frequently associated with loss of genomic stability. Analysis of telomerase-induced extended-life-span fibroblast (TIELF) cells by G banding and spectral karyotyping indicated that forced extension of the life span by telomerase led to the transient formation of aberrant structures, which were subsequently resolved in higher passages. However, the p53-dependent G1 checkpoint was intact as assessed by functional activation of p53 protein in response to ionizing radiation and subsequent p53-mediated induction of p21(Waf1/Cip1/Sdi1). TIELF cells were not tumorigenic and had a normal DNA strand break rejoining activity and normal radiosensitivity in response to ionizing radiation.  (+info)

Phosphorylation of the DNA repair protein APE/REF-1 by CKII affects redox regulation of AP-1. (7/17364)

The DNA repair protein apurinic endonuclease (APE/Ref-1) exerts several physiological functions such as cleavage of apurinic/apyrimidinic sites and redox regulation of the transcription factor AP-1, whose activation is part of the cellular response to DNA damaging treatments. Here we demonstrate that APE/Ref-1 is phosphorylated by casein kinase II (CKII). This was shown for both the recombinant APE/Ref-1 protein (Km=0.55 mM) and for APE/Ref-1 expressed in COS cells. Phosphorylation of APE/Ref-1 did not alter the repair activity of the enzyme, whereas it stimulated its redox capability towards AP-1, thus promoting DNA binding activity of AP-1. Inhibition of CKII mediated phosphorylation of APE/Ref-1 blocked mutagen-stimulated increase in AP-1 binding. It also abrogated the induction of c-Jun protein and rendered cells more sensitive to induced DNA damage. Thus, phosphorylation of APE/Ref-1 appears to be involved in regulating the different physiological activities of the enzyme. CKII mediated phosphorylation of APE/Ref-1 and concomitant increase in AP-1 binding activity appears to be a novel mechanism of cellular stress response, forcing transcription of AP-1 target gene(s) the product(s) of which may exert protective function.  (+info)

Differential regulation of p21waf-1/cip-1 and Mdm2 by etoposide: etoposide inhibits the p53-Mdm2 autoregulatory feedback loop. (8/17364)

The Mdm2 protein is frequently overexpressed in human non-seminomatous germ cell tumours and transitional carcinoma of the bladder where it may contribute to tolerance of wtp53. Mdm2 forms an autoregulatory feedback loop with p53; the Mdm2 gene is responsive to transactivation by p53 and once synthesized the Mdm2 protein terminates the p53 response. We show here that the topoisomerase poison etoposide, like ultra violet irradiation, inhibits Mdm2 synthesis. Cytotoxic concentrations of etoposide (IC90 for > 3 h) result in inhibition of Mdm2 induction at both the RNA and protein level. Rapid apoptosis ensues. Global transcription is not inhibited: p21waf-1/cip1 and GADD45 expression increase in a dose dependent manner. Inhibition of Mdm2 synthesis depends on the continuous presence of etoposide, suggesting the DNA damage may prevent transcription. Downregulation of Mdm2 transcript occurs in cells expressing HPV16-E6 suggesting that inhibition of Mdm2 transcription is p53-independent. When cells are -treated with a pulse (1 h) of etoposide and reincubated in drug free medium, Mdm2 synthesis commences immediately after damage is repaired (3 h) and the p53 response is attenuated. Induction of apoptosis and loss of clonogenicity are 3-5-fold lower under pulse treatment conditions. This is the first observation of inhibition of Mdm2 transcription following treatment with topoisomerase (topo II) poisons, a feature that may be useful in tumour types where p53 is tolerated by overexpression of Mdm2.  (+info)

DNA damage refers to any alteration in the structure or composition of deoxyribonucleic acid (DNA), which is the genetic material present in cells. DNA damage can result from various internal and external factors, including environmental exposures such as ultraviolet radiation, tobacco smoke, and certain chemicals, as well as normal cellular processes such as replication and oxidative metabolism.

Examples of DNA damage include base modifications, base deletions or insertions, single-strand breaks, double-strand breaks, and crosslinks between the two strands of the DNA helix. These types of damage can lead to mutations, genomic instability, and chromosomal aberrations, which can contribute to the development of diseases such as cancer, neurodegenerative disorders, and aging-related conditions.

The body has several mechanisms for repairing DNA damage, including base excision repair, nucleotide excision repair, mismatch repair, and double-strand break repair. However, if the damage is too extensive or the repair mechanisms are impaired, the cell may undergo apoptosis (programmed cell death) to prevent the propagation of potentially harmful mutations.

DNA repair is the process by which cells identify and correct damage to the DNA molecules that encode their genome. DNA can be damaged by a variety of internal and external factors, such as radiation, chemicals, and metabolic byproducts. If left unrepaired, this damage can lead to mutations, which may in turn lead to cancer and other diseases.

There are several different mechanisms for repairing DNA damage, including:

1. Base excision repair (BER): This process repairs damage to a single base in the DNA molecule. An enzyme called a glycosylase removes the damaged base, leaving a gap that is then filled in by other enzymes.
2. Nucleotide excision repair (NER): This process repairs more severe damage, such as bulky adducts or crosslinks between the two strands of the DNA molecule. An enzyme cuts out a section of the damaged DNA, and the gap is then filled in by other enzymes.
3. Mismatch repair (MMR): This process repairs errors that occur during DNA replication, such as mismatched bases or small insertions or deletions. Specialized enzymes recognize the error and remove a section of the newly synthesized strand, which is then replaced by new nucleotides.
4. Double-strand break repair (DSBR): This process repairs breaks in both strands of the DNA molecule. There are two main pathways for DSBR: non-homologous end joining (NHEJ) and homologous recombination (HR). NHEJ directly rejoins the broken ends, while HR uses a template from a sister chromatid to repair the break.

Overall, DNA repair is a crucial process that helps maintain genome stability and prevent the development of diseases caused by genetic mutations.

The Comet Assay, also known as single-cell gel electrophoresis (SCGE), is a sensitive method used to detect and measure DNA damage at the level of individual cells. The assay gets its name from the comet-like shape that formed DNA fragments migrate towards the anode during electrophoresis, creating a "tail" that represents the damaged DNA.

In this assay, cells are embedded in low melting point agarose on a microscope slide and then lysed to remove the cell membranes and histones, leaving the DNA intact. The slides are then subjected to electrophoresis under neutral or alkaline conditions, which causes the negatively charged DNA fragments to migrate out of the nucleus towards the anode. After staining with a DNA-binding dye, the slides are visualized under a fluorescence microscope and the degree of DNA damage is quantified by measuring the length and intensity of the comet "tail."

The Comet Assay is widely used in genetic toxicology to assess the genotoxic potential of chemicals, drugs, and environmental pollutants. It can also be used to measure DNA repair capacity and oxidative DNA damage.

Ataxia telangiectasia mutated (ATM) proteins are a type of protein that play a crucial role in the maintenance and repair of DNA in cells. The ATM gene produces these proteins, which are involved in several important cellular processes such as:

1. DNA damage response: When DNA is damaged, ATM proteins help to detect and respond to the damage by activating various signaling pathways that lead to DNA repair or apoptosis (programmed cell death) if the damage is too severe.
2. Cell cycle regulation: ATM proteins regulate the cell cycle by controlling checkpoints that ensure proper DNA replication and division. This helps prevent the propagation of cells with damaged DNA.
3. Telomere maintenance: ATM proteins help maintain telomeres, which are the protective caps at the ends of chromosomes. Telomeres shorten as cells divide, and when they become too short, cells can no longer divide and enter a state of senescence or die.

Mutations in the ATM gene can lead to Ataxia-telangiectasia (A-T), a rare inherited disorder characterized by neurological problems, immune system dysfunction, increased risk of cancer, and sensitivity to ionizing radiation. People with A-T have defective ATM proteins that cannot properly respond to DNA damage, leading to genomic instability and increased susceptibility to disease.

Cell cycle proteins are a group of regulatory proteins that control the progression of the cell cycle, which is the series of events that take place in a eukaryotic cell leading to its division and duplication. These proteins can be classified into several categories based on their functions during different stages of the cell cycle.

The major groups of cell cycle proteins include:

1. Cyclin-dependent kinases (CDKs): CDKs are serine/threonine protein kinases that regulate key transitions in the cell cycle. They require binding to a regulatory subunit called cyclin to become active. Different CDK-cyclin complexes are activated at different stages of the cell cycle.
2. Cyclins: Cyclins are a family of regulatory proteins that bind and activate CDKs. Their levels fluctuate throughout the cell cycle, with specific cyclins expressed during particular phases. For example, cyclin D is important for the G1 to S phase transition, while cyclin B is required for the G2 to M phase transition.
3. CDK inhibitors (CKIs): CKIs are regulatory proteins that bind to and inhibit CDKs, thereby preventing their activation. CKIs can be divided into two main families: the INK4 family and the Cip/Kip family. INK4 family members specifically inhibit CDK4 and CDK6, while Cip/Kip family members inhibit a broader range of CDKs.
4. Anaphase-promoting complex/cyclosome (APC/C): APC/C is an E3 ubiquitin ligase that targets specific proteins for degradation by the 26S proteasome. During the cell cycle, APC/C regulates the metaphase to anaphase transition and the exit from mitosis by targeting securin and cyclin B for degradation.
5. Other regulatory proteins: Several other proteins play crucial roles in regulating the cell cycle, such as p53, a transcription factor that responds to DNA damage and arrests the cell cycle, and the polo-like kinases (PLKs), which are involved in various aspects of mitosis.

Overall, cell cycle proteins work together to ensure the proper progression of the cell cycle, maintain genomic stability, and prevent uncontrolled cell growth, which can lead to cancer.

According to the medical definition, ultraviolet (UV) rays are invisible radiations that fall in the range of the electromagnetic spectrum between 100-400 nanometers. UV rays are further divided into three categories: UVA (320-400 nm), UVB (280-320 nm), and UVC (100-280 nm).

UV rays have various sources, including the sun and artificial sources like tanning beds. Prolonged exposure to UV rays can cause damage to the skin, leading to premature aging, eye damage, and an increased risk of skin cancer. UVA rays penetrate deeper into the skin and are associated with skin aging, while UVB rays primarily affect the outer layer of the skin and are linked to sunburns and skin cancer. UVC rays are the most harmful but fortunately, they are absorbed by the Earth's atmosphere and do not reach the surface.

Healthcare professionals recommend limiting exposure to UV rays, wearing protective clothing, using broad-spectrum sunscreen with an SPF of at least 30, and avoiding tanning beds to reduce the risk of UV-related health problems.

Deoxyguanosine is a chemical compound that is a component of DNA (deoxyribonucleic acid), one of the nucleic acids. It is a nucleoside, which is a molecule consisting of a sugar (in this case, deoxyribose) and a nitrogenous base (in this case, guanine). Deoxyguanosine plays a crucial role in the structure and function of DNA, as it pairs with deoxycytidine through hydrogen bonding to form a rung in the DNA double helix. It is involved in the storage and transmission of genetic information.

Checkpoint Kinase 2 (Chk2) is a serine/threonine protein kinase that plays a crucial role in the DNA damage response and the regulation of the cell cycle. It is activated by various types of DNA damage, including double-strand breaks, and phosphorylates several downstream targets involved in cell cycle arrest, DNA repair, and apoptosis. Chk2 is a key player in the G2/M checkpoint, which prevents cells with damaged DNA from entering mitosis and dividing. Mutations in the Chk2 gene have been associated with increased risk of cancer.

Tumor suppressor protein p53, also known as p53 or tumor protein p53, is a nuclear phosphoprotein that plays a crucial role in preventing cancer development and maintaining genomic stability. It does so by regulating the cell cycle and acting as a transcription factor for various genes involved in apoptosis (programmed cell death), DNA repair, and cell senescence (permanent cell growth arrest).

In response to cellular stress, such as DNA damage or oncogene activation, p53 becomes activated and accumulates in the nucleus. Activated p53 can then bind to specific DNA sequences and promote the transcription of target genes that help prevent the proliferation of potentially cancerous cells. These targets include genes involved in cell cycle arrest (e.g., CDKN1A/p21), apoptosis (e.g., BAX, PUMA), and DNA repair (e.g., GADD45).

Mutations in the TP53 gene, which encodes p53, are among the most common genetic alterations found in human cancers. These mutations often lead to a loss or reduction of p53's tumor suppressive functions, allowing cancer cells to proliferate uncontrollably and evade apoptosis. As a result, p53 has been referred to as "the guardian of the genome" due to its essential role in preventing tumorigenesis.

DNA-binding proteins are a type of protein that have the ability to bind to DNA (deoxyribonucleic acid), the genetic material of organisms. These proteins play crucial roles in various biological processes, such as regulation of gene expression, DNA replication, repair and recombination.

The binding of DNA-binding proteins to specific DNA sequences is mediated by non-covalent interactions, including electrostatic, hydrogen bonding, and van der Waals forces. The specificity of binding is determined by the recognition of particular nucleotide sequences or structural features of the DNA molecule.

DNA-binding proteins can be classified into several categories based on their structure and function, such as transcription factors, histones, and restriction enzymes. Transcription factors are a major class of DNA-binding proteins that regulate gene expression by binding to specific DNA sequences in the promoter region of genes and recruiting other proteins to modulate transcription. Histones are DNA-binding proteins that package DNA into nucleosomes, the basic unit of chromatin structure. Restriction enzymes are DNA-binding proteins that recognize and cleave specific DNA sequences, and are widely used in molecular biology research and biotechnology applications.

Double-stranded DNA breaks (DSBs) refer to a type of damage that occurs in the DNA molecule when both strands of the double helix are severed or broken at the same location. This kind of damage is particularly harmful to cells because it can disrupt the integrity and continuity of the genetic material, potentially leading to genomic instability, mutations, and cell death if not properly repaired.

DSBs can arise from various sources, including exposure to ionizing radiation, chemical agents, free radicals, reactive oxygen species (ROS), and errors during DNA replication or repair processes. Unrepaired or incorrectly repaired DSBs have been implicated in numerous human diseases, such as cancer, neurodegenerative disorders, and premature aging.

Cells possess several mechanisms to repair double-stranded DNA breaks, including homologous recombination (HR) and non-homologous end joining (NHEJ). HR is a more accurate repair pathway that uses a homologous template, typically the sister chromatid, to restore the original DNA sequence. NHEJ, on the other hand, directly ligates the broken ends together, often resulting in small deletions or insertions at the break site and increased risk of errors. The choice between these two pathways depends on various factors, such as the cell cycle stage, the presence of nearby breaks, and the availability of repair proteins.

In summary, double-stranded DNA breaks are severe forms of DNA damage that can have detrimental consequences for cells if not properly repaired. Cells employ multiple mechanisms to address DSBs, with homologous recombination and non-homologous end joining being the primary repair pathways.

Protein-Serine-Threonine Kinases (PSTKs) are a type of protein kinase that catalyzes the transfer of a phosphate group from ATP to the hydroxyl side chains of serine or threonine residues on target proteins. This phosphorylation process plays a crucial role in various cellular signaling pathways, including regulation of metabolism, gene expression, cell cycle progression, and apoptosis. PSTKs are involved in many physiological and pathological processes, and their dysregulation has been implicated in several diseases, such as cancer, diabetes, and neurodegenerative disorders.

Oxidative stress is defined as an imbalance between the production of reactive oxygen species (free radicals) and the body's ability to detoxify them or repair the damage they cause. This imbalance can lead to cellular damage, oxidation of proteins, lipids, and DNA, disruption of cellular functions, and activation of inflammatory responses. Prolonged or excessive oxidative stress has been linked to various health conditions, including cancer, cardiovascular diseases, neurodegenerative disorders, and aging-related diseases.

The cell cycle is a series of events that take place in a cell leading to its division and duplication. It consists of four main phases: G1 phase, S phase, G2 phase, and M phase.

During the G1 phase, the cell grows in size and synthesizes mRNA and proteins in preparation for DNA replication. In the S phase, the cell's DNA is copied, resulting in two complete sets of chromosomes. During the G2 phase, the cell continues to grow and produces more proteins and organelles necessary for cell division.

The M phase is the final stage of the cell cycle and consists of mitosis (nuclear division) and cytokinesis (cytoplasmic division). Mitosis results in two genetically identical daughter nuclei, while cytokinesis divides the cytoplasm and creates two separate daughter cells.

The cell cycle is regulated by various checkpoints that ensure the proper completion of each phase before progressing to the next. These checkpoints help prevent errors in DNA replication and division, which can lead to mutations and cancer.

Tumor suppressor proteins are a type of regulatory protein that helps control the cell cycle and prevent cells from dividing and growing in an uncontrolled manner. They work to inhibit tumor growth by preventing the formation of tumors or slowing down their progression. These proteins can repair damaged DNA, regulate gene expression, and initiate programmed cell death (apoptosis) if the damage is too severe for repair.

Mutations in tumor suppressor genes, which provide the code for these proteins, can lead to a decrease or loss of function in the resulting protein. This can result in uncontrolled cell growth and division, leading to the formation of tumors and cancer. Examples of tumor suppressor proteins include p53, Rb (retinoblastoma), and BRCA1/2.

Gamma rays are a type of ionizing radiation that is released from the nucleus of an atom during radioactive decay. They are high-energy photons, with wavelengths shorter than 0.01 nanometers and frequencies greater than 3 x 10^19 Hz. Gamma rays are electromagnetic radiation, similar to X-rays, but with higher energy levels and the ability to penetrate matter more deeply. They can cause damage to living tissue and are used in medical imaging and cancer treatment.

Apoptosis is a programmed and controlled cell death process that occurs in multicellular organisms. It is a natural process that helps maintain tissue homeostasis by eliminating damaged, infected, or unwanted cells. During apoptosis, the cell undergoes a series of morphological changes, including cell shrinkage, chromatin condensation, and fragmentation into membrane-bound vesicles called apoptotic bodies. These bodies are then recognized and engulfed by neighboring cells or phagocytic cells, preventing an inflammatory response. Apoptosis is regulated by a complex network of intracellular signaling pathways that involve proteins such as caspases, Bcl-2 family members, and inhibitors of apoptosis (IAPs).

Deoxyribonucleic acid (DNA) is the genetic material present in the cells of organisms where it is responsible for the storage and transmission of hereditary information. DNA is a long molecule that consists of two strands coiled together to form a double helix. Each strand is made up of a series of four nucleotide bases - adenine (A), guanine (G), cytosine (C), and thymine (T) - that are linked together by phosphate and sugar groups. The sequence of these bases along the length of the molecule encodes genetic information, with A always pairing with T and C always pairing with G. This base-pairing allows for the replication and transcription of DNA, which are essential processes in the functioning and reproduction of all living organisms.

Methyl methanesulfonate (MMS) is not a medication, but rather a chemical compound with the formula CH3SO3CH3. It's an alkylating agent that is used in laboratory settings for various research purposes, including as a methylating agent in biochemical and genetic studies.

MMS works by transferring its methyl group (CH3) to other molecules, which can result in the modification of DNA and other biological macromolecules. This property makes it useful in laboratory research, but it also means that MMS is highly reactive and toxic. Therefore, it must be handled with care and appropriate safety precautions.

It's important to note that MMS is not used as a therapeutic agent in medicine due to its high toxicity and potential to cause serious harm if mishandled or misused.

Histones are highly alkaline proteins found in the chromatin of eukaryotic cells. They are rich in basic amino acid residues, such as arginine and lysine, which give them their positive charge. Histones play a crucial role in packaging DNA into a more compact structure within the nucleus by forming a complex with it called a nucleosome. Each nucleosome contains about 146 base pairs of DNA wrapped around an octamer of eight histone proteins (two each of H2A, H2B, H3, and H4). The N-terminal tails of these histones are subject to various post-translational modifications, such as methylation, acetylation, and phosphorylation, which can influence chromatin structure and gene expression. Histone variants also exist, which can contribute to the regulation of specific genes and other nuclear processes.

Mutagens are physical or chemical agents that can cause permanent changes in the structure of genetic material, including DNA and chromosomes, leading to mutations. These mutations can be passed down to future generations and may increase the risk of cancer and other diseases. Examples of mutagens include ultraviolet (UV) radiation, tobacco smoke, and certain chemicals found in industrial settings. It is important to note that not all mutations are harmful, but some can have negative effects on health and development.

Nuclear proteins are a category of proteins that are primarily found in the nucleus of a eukaryotic cell. They play crucial roles in various nuclear functions, such as DNA replication, transcription, repair, and RNA processing. This group includes structural proteins like lamins, which form the nuclear lamina, and regulatory proteins, such as histones and transcription factors, that are involved in gene expression. Nuclear localization signals (NLS) often help target these proteins to the nucleus by interacting with importin proteins during active transport across the nuclear membrane.

Genomic instability is a term used in genetics and molecular biology to describe a state of increased susceptibility to genetic changes or mutations in the genome. It can be defined as a condition where the integrity and stability of the genome are compromised, leading to an increased rate of DNA alterations such as point mutations, insertions, deletions, and chromosomal rearrangements.

Genomic instability is a hallmark of cancer cells and can also be observed in various other diseases, including genetic disorders and aging. It can arise due to defects in the DNA repair mechanisms, telomere maintenance, epigenetic regulation, or chromosome segregation during cell division. These defects can result from inherited genetic mutations, acquired somatic mutations, exposure to environmental mutagens, or age-related degenerative changes.

Genomic instability is a significant factor in the development and progression of cancer as it promotes the accumulation of oncogenic mutations that contribute to tumor initiation, growth, and metastasis. Therefore, understanding the mechanisms underlying genomic instability is crucial for developing effective strategies for cancer prevention, diagnosis, and treatment.

The G2 phase, also known as the "gap 2 phase," is a stage in the cell cycle that occurs after DNA replication (S phase) and before cell division (mitosis). During this phase, the cell prepares for mitosis by completing the synthesis of proteins and organelles needed for chromosome separation. The cell also checks for any errors or damage to the DNA before entering mitosis. This phase is a critical point in the cell cycle where proper regulation ensures the faithful transmission of genetic information from one generation of cells to the next. If significant DNA damage is detected during G2, the cell may undergo programmed cell death (apoptosis) instead of dividing.

DNA replication is the biological process by which DNA makes an identical copy of itself during cell division. It is a fundamental mechanism that allows genetic information to be passed down from one generation of cells to the next. During DNA replication, each strand of the double helix serves as a template for the synthesis of a new complementary strand. This results in the creation of two identical DNA molecules. The enzymes responsible for DNA replication include helicase, which unwinds the double helix, and polymerase, which adds nucleotides to the growing strands.

A mutation is a permanent change in the DNA sequence of an organism's genome. Mutations can occur spontaneously or be caused by environmental factors such as exposure to radiation, chemicals, or viruses. They may have various effects on the organism, ranging from benign to harmful, depending on where they occur and whether they alter the function of essential proteins. In some cases, mutations can increase an individual's susceptibility to certain diseases or disorders, while in others, they may confer a survival advantage. Mutations are the driving force behind evolution, as they introduce new genetic variability into populations, which can then be acted upon by natural selection.

Phosphorylation is the process of adding a phosphate group (a molecule consisting of one phosphorus atom and four oxygen atoms) to a protein or other organic molecule, which is usually done by enzymes called kinases. This post-translational modification can change the function, localization, or activity of the target molecule, playing a crucial role in various cellular processes such as signal transduction, metabolism, and regulation of gene expression. Phosphorylation is reversible, and the removal of the phosphate group is facilitated by enzymes called phosphatases.

A cell line that is derived from tumor cells and has been adapted to grow in culture. These cell lines are often used in research to study the characteristics of cancer cells, including their growth patterns, genetic changes, and responses to various treatments. They can be established from many different types of tumors, such as carcinomas, sarcomas, and leukemias. Once established, these cell lines can be grown and maintained indefinitely in the laboratory, allowing researchers to conduct experiments and studies that would not be feasible using primary tumor cells. It is important to note that tumor cell lines may not always accurately represent the behavior of the original tumor, as they can undergo genetic changes during their time in culture.

DNA repair enzymes are a group of enzymes that are responsible for identifying and correcting damage to the DNA molecule. These enzymes play a critical role in maintaining the integrity of an organism's genetic material, as they help to ensure that the information stored in DNA is accurately transmitted during cell division and reproduction.

There are several different types of DNA repair enzymes, each responsible for correcting specific types of damage. For example, base excision repair enzymes remove and replace damaged or incorrect bases, while nucleotide excision repair enzymes remove larger sections of damaged DNA and replace them with new nucleotides. Other types of DNA repair enzymes include mismatch repair enzymes, which correct errors that occur during DNA replication, and double-strand break repair enzymes, which are responsible for fixing breaks in both strands of the DNA molecule.

Defects in DNA repair enzymes have been linked to a variety of diseases, including cancer, neurological disorders, and premature aging. For example, individuals with xeroderma pigmentosum, a rare genetic disorder characterized by an increased risk of skin cancer, have mutations in genes that encode nucleotide excision repair enzymes. Similarly, defects in mismatch repair enzymes have been linked to hereditary nonpolyposis colorectal cancer, a type of colon cancer that is inherited and tends to occur at a younger age than sporadic colon cancer.

Overall, DNA repair enzymes play a critical role in maintaining the stability and integrity of an organism's genetic material, and defects in these enzymes can have serious consequences for human health.

A dose-response relationship in radiation refers to the correlation between the amount of radiation exposure (dose) and the biological response or adverse health effects observed in exposed individuals. As the level of radiation dose increases, the severity and frequency of the adverse health effects also tend to increase. This relationship is crucial in understanding the risks associated with various levels of radiation exposure and helps inform radiation protection standards and guidelines.

The effects of ionizing radiation can be categorized into two types: deterministic and stochastic. Deterministic effects have a threshold dose below which no effect is observed, and above this threshold, the severity of the effect increases with higher doses. Examples include radiation-induced cataracts or radiation dermatitis. Stochastic effects, on the other hand, do not have a clear threshold and are based on probability; as the dose increases, so does the likelihood of the adverse health effect occurring, such as an increased risk of cancer.

Understanding the dose-response relationship in radiation exposure is essential for setting limits on occupational and public exposure to ionizing radiation, optimizing radiation protection practices, and developing effective medical countermeasures in case of radiation emergencies.

DNA adducts are chemical modifications or alterations that occur when DNA molecules become attached to or bound with certain harmful substances, such as toxic chemicals or carcinogens. These attachments can disrupt the normal structure and function of the DNA, potentially leading to mutations, genetic damage, and an increased risk of cancer and other diseases.

DNA adducts are formed when a reactive molecule from a chemical agent binds covalently to a base in the DNA molecule. This process can occur either spontaneously or as a result of exposure to environmental toxins, such as those found in tobacco smoke, certain industrial chemicals, and some medications.

The formation of DNA adducts is often used as a biomarker for exposure to harmful substances, as well as an indicator of potential health risks associated with that exposure. Researchers can measure the levels of specific DNA adducts in biological samples, such as blood or urine, to assess the extent and duration of exposure to certain chemicals or toxins.

It's important to note that not all DNA adducts are necessarily harmful, and some may even play a role in normal cellular processes. However, high levels of certain DNA adducts have been linked to an increased risk of cancer and other diseases, making them a focus of ongoing research and investigation.

Reactive Oxygen Species (ROS) are highly reactive molecules containing oxygen, including peroxides, superoxide, hydroxyl radical, and singlet oxygen. They are naturally produced as byproducts of normal cellular metabolism in the mitochondria, and can also be generated by external sources such as ionizing radiation, tobacco smoke, and air pollutants. At low or moderate concentrations, ROS play important roles in cell signaling and homeostasis, but at high concentrations, they can cause significant damage to cell structures, including lipids, proteins, and DNA, leading to oxidative stress and potential cell death.

Cell survival refers to the ability of a cell to continue living and functioning normally, despite being exposed to potentially harmful conditions or treatments. This can include exposure to toxins, radiation, chemotherapeutic drugs, or other stressors that can damage cells or interfere with their normal processes.

In scientific research, measures of cell survival are often used to evaluate the effectiveness of various therapies or treatments. For example, researchers may expose cells to a particular drug or treatment and then measure the percentage of cells that survive to assess its potential therapeutic value. Similarly, in toxicology studies, measures of cell survival can help to determine the safety of various chemicals or substances.

It's important to note that cell survival is not the same as cell proliferation, which refers to the ability of cells to divide and multiply. While some treatments may promote cell survival, they may also inhibit cell proliferation, making them useful for treating diseases such as cancer. Conversely, other treatments may be designed to specifically target and kill cancer cells, even if it means sacrificing some healthy cells in the process.

A cell line is a culture of cells that are grown in a laboratory for use in research. These cells are usually taken from a single cell or group of cells, and they are able to divide and grow continuously in the lab. Cell lines can come from many different sources, including animals, plants, and humans. They are often used in scientific research to study cellular processes, disease mechanisms, and to test new drugs or treatments. Some common types of human cell lines include HeLa cells (which come from a cancer patient named Henrietta Lacks), HEK293 cells (which come from embryonic kidney cells), and HUVEC cells (which come from umbilical vein endothelial cells). It is important to note that cell lines are not the same as primary cells, which are cells that are taken directly from a living organism and have not been grown in the lab.

Hydrogen peroxide (H2O2) is a colorless, odorless, clear liquid with a slightly sweet taste, although drinking it is harmful and can cause poisoning. It is a weak oxidizing agent and is used as an antiseptic and a bleaching agent. In diluted form, it is used to disinfect wounds and kill bacteria and viruses on the skin; in higher concentrations, it can be used to bleach hair or remove stains from clothing. It is also used as a propellant in rocketry and in certain industrial processes. Chemically, hydrogen peroxide is composed of two hydrogen atoms and two oxygen atoms, and it is structurally similar to water (H2O), with an extra oxygen atom. This gives it its oxidizing properties, as the additional oxygen can be released and used to react with other substances.

DNA breaks refer to any damage or disruption in the DNA molecule that results in a separation of the double helix strands. There are two types of DNA breaks: single-strand breaks (SSBs) and double-strand breaks (DSBs).

Single-strand breaks occur when one of the two strands in the DNA duplex is cleaved, leaving the other strand intact. These breaks are usually repaired quickly and efficiently by enzymes that can recognize and repair the damage.

Double-strand breaks, on the other hand, are more serious forms of DNA damage because they result in a complete separation of both strands of the DNA duplex. DSBs can lead to genomic instability, chromosomal aberrations, and cell death if not repaired promptly and accurately.

DSBs can be caused by various factors, including ionizing radiation, chemotherapeutic agents, oxidative stress, and errors during DNA replication or repair. The body has several mechanisms to repair DSBs, including non-homologous end joining (NHEJ) and homologous recombination (HR). However, if these repair pathways are impaired or overwhelmed, DSBs can lead to mutations, cancer, and other diseases.

Radiation tolerance, in the context of medicine and particularly radiation oncology, refers to the ability of tissues or organs to withstand and recover from exposure to ionizing radiation without experiencing significant damage or loss of function. It is often used to describe the maximum dose of radiation that can be safely delivered to a specific area of the body during radiotherapy treatments.

Radiation tolerance varies depending on the type and location of the tissue or organ. For example, some tissues such as the brain, spinal cord, and lungs have lower radiation tolerance than others like the skin or bone. Factors that can affect radiation tolerance include the total dose of radiation, the fractionation schedule (the number and size of radiation doses), the volume of tissue treated, and the individual patient's overall health and genetic factors.

Assessing radiation tolerance is critical in designing safe and effective radiotherapy plans for cancer patients, as excessive radiation exposure can lead to serious side effects such as radiation-induced injury, fibrosis, or even secondary malignancies.

Protein kinases are a group of enzymes that play a crucial role in many cellular processes by adding phosphate groups to other proteins, a process known as phosphorylation. This modification can activate or deactivate the target protein's function, thereby regulating various signaling pathways within the cell. Protein kinases are essential for numerous biological functions, including metabolism, signal transduction, cell cycle progression, and apoptosis (programmed cell death). Abnormal regulation of protein kinases has been implicated in several diseases, such as cancer, diabetes, and neurological disorders.

I'm happy to help! However, I believe there may be a slight mistake in your question. The abbreviation "cdc" is not typically associated with genetics or genes in the context of medical definitions.

If you meant to ask for a definition of "genes," here it is:

Genes are segments of DNA (deoxyribonucleic acid) that contain the instructions for the development, function, and reproduction of all living organisms. They are the basic units of heredity, passed down from one generation to the next. Genes encode specific proteins or RNA molecules that play critical roles in the structure, function, and regulation of the body's cells, tissues, and organs.

If you had a different term in mind, please let me know, and I will be happy to provide a definition for it!

Cell cycle checkpoints are control mechanisms that regulate the cell cycle and ensure the accurate and timely progression through different phases of the cell cycle. These checkpoints monitor specific cellular events, such as DNA replication and damage, chromosome separation, and proper attachment of the mitotic spindle to the chromosomes. If any of these events fail to occur properly or are delayed, the cell cycle checkpoints trigger a response that can halt the cell cycle until the problem is resolved. This helps to prevent cells with damaged or incomplete genomes from dividing and potentially becoming cancerous.

There are three main types of cell cycle checkpoints:

1. G1 Checkpoint: Also known as the restriction point, this checkpoint controls the transition from the G1 phase to the S phase of the cell cycle. It monitors the availability of nutrients, growth factors, and the integrity of the genome before allowing the cell to proceed into DNA replication.
2. G2 Checkpoint: This checkpoint regulates the transition from the G2 phase to the M phase of the cell cycle. It checks for completion of DNA replication and absence of DNA damage before allowing the cell to enter mitosis.
3. Mitotic (M) Checkpoint: Also known as the spindle assembly checkpoint, this checkpoint ensures that all chromosomes are properly attached to the mitotic spindle before anaphase begins. It prevents the separation of sister chromatids until all kinetochores are correctly attached and tension is established between them.

Cell cycle checkpoints play a crucial role in maintaining genomic stability, preventing tumorigenesis, and ensuring proper cell division. Dysregulation of these checkpoints can lead to various diseases, including cancer.

Single-stranded DNA breaks (SSBs) refer to a type of DNA damage in which one strand of the double-helix structure is cleaved or broken. This kind of damage can occur spontaneously due to cellular metabolism or can be induced by various genotoxic agents, such as ionizing radiation and certain chemicals.

SSBs are typically repaired rapidly and efficiently by enzymes known as DNA repair proteins. However, if left unrepaired or misrepaired, they can lead to mutations, genomic instability, and increased risk of diseases, including cancer. In some cases, single-stranded breaks may also precede the formation of more severe double-stranded DNA breaks (DSBs).

It is important to note that while SSBs are less catastrophic than DSBs, they still play a significant role in genome maintenance and cellular health.

Signal transduction is the process by which a cell converts an extracellular signal, such as a hormone or neurotransmitter, into an intracellular response. This involves a series of molecular events that transmit the signal from the cell surface to the interior of the cell, ultimately resulting in changes in gene expression, protein activity, or metabolism.

The process typically begins with the binding of the extracellular signal to a receptor located on the cell membrane. This binding event activates the receptor, which then triggers a cascade of intracellular signaling molecules, such as second messengers, protein kinases, and ion channels. These molecules amplify and propagate the signal, ultimately leading to the activation or inhibition of specific cellular responses.

Signal transduction pathways are highly regulated and can be modulated by various factors, including other signaling molecules, post-translational modifications, and feedback mechanisms. Dysregulation of these pathways has been implicated in a variety of diseases, including cancer, diabetes, and neurological disorders.

Rad51 recombinase is a protein involved in the repair of double-stranded DNA breaks through homologous recombination, a process that helps maintain genomic stability. This protein forms a nucleoprotein filament on single-stranded DNA, facilitating the search for and invasion of homologous sequences in double-stranded DNA. Rad51 recombinase is highly conserved across various species, including humans, and plays a crucial role in preventing genetic disorders, cancer, and aging caused by DNA damage.

DNA glycosylases are a group of enzymes that play a crucial role in the maintenance of genetic material. They are responsible for initiating the base excision repair (BER) pathway, which is one of the major DNA repair mechanisms in cells.

The function of DNA glycosylases is to remove damaged or mismatched bases from DNA molecules. These enzymes recognize and bind to specific types of damaged or incorrect bases, and then cleave the N-glycosidic bond between the base and the deoxyribose sugar in the DNA backbone. This results in the formation of an apurinic/apyrimidinic (AP) site, which is subsequently processed by other enzymes in the BER pathway.

There are several different types of DNA glycosylases that recognize and remove specific types of damaged or incorrect bases. For example, some DNA glycosylases specialize in removing oxidized bases, while others are responsible for removing mismatched bases or those that have been alkylated or methylated.

Overall, the proper functioning of DNA glycosylases is essential for maintaining genomic stability and preventing the accumulation of mutations that can lead to diseases such as cancer.

HeLa cells are a type of immortalized cell line used in scientific research. They are derived from a cancer that developed in the cervical tissue of Henrietta Lacks, an African-American woman, in 1951. After her death, cells taken from her tumor were found to be capable of continuous division and growth in a laboratory setting, making them an invaluable resource for medical research.

HeLa cells have been used in a wide range of scientific studies, including research on cancer, viruses, genetics, and drug development. They were the first human cell line to be successfully cloned and are able to grow rapidly in culture, doubling their population every 20-24 hours. This has made them an essential tool for many areas of biomedical research.

It is important to note that while HeLa cells have been instrumental in numerous scientific breakthroughs, the story of their origin raises ethical questions about informed consent and the use of human tissue in research.

In the context of cell biology, "S phase" refers to the part of the cell cycle during which DNA replication occurs. The "S" stands for synthesis, reflecting the active DNA synthesis that takes place during this phase. It is preceded by G1 phase (gap 1) and followed by G2 phase (gap 2), with mitosis (M phase) being the final stage of the cell cycle.

During S phase, the cell's DNA content effectively doubles as each chromosome is replicated to ensure that the two resulting daughter cells will have the same genetic material as the parent cell. This process is carefully regulated and coordinated with other events in the cell cycle to maintain genomic stability.

Saccharomyces cerevisiae proteins are the proteins that are produced by the budding yeast, Saccharomyces cerevisiae. This organism is a single-celled eukaryote that has been widely used as a model organism in scientific research for many years due to its relatively simple genetic makeup and its similarity to higher eukaryotic cells.

The genome of Saccharomyces cerevisiae has been fully sequenced, and it is estimated to contain approximately 6,000 genes that encode proteins. These proteins play a wide variety of roles in the cell, including catalyzing metabolic reactions, regulating gene expression, maintaining the structure of the cell, and responding to environmental stimuli.

Many Saccharomyces cerevisiae proteins have human homologs and are involved in similar biological processes, making this organism a valuable tool for studying human disease. For example, many of the proteins involved in DNA replication, repair, and recombination in yeast have human counterparts that are associated with cancer and other diseases. By studying these proteins in yeast, researchers can gain insights into their function and regulation in humans, which may lead to new treatments for disease.

BRCA1 protein is a tumor suppressor protein that plays a crucial role in repairing damaged DNA and maintaining genomic stability. The BRCA1 gene provides instructions for making this protein. Mutations in the BRCA1 gene can lead to impaired function of the BRCA1 protein, significantly increasing the risk of developing breast, ovarian, and other types of cancer.

The BRCA1 protein forms complexes with several other proteins to participate in various cellular processes, such as:

1. DNA damage response and repair: BRCA1 helps recognize and repair double-strand DNA breaks through homologous recombination, a precise error-free repair mechanism.
2. Cell cycle checkpoints: BRCA1 is involved in regulating the G1/S and G2/M cell cycle checkpoints to ensure proper DNA replication and cell division.
3. Transcription regulation: BRCA1 can act as a transcriptional co-regulator, influencing the expression of genes involved in various cellular processes, including DNA repair and cell cycle control.
4. Apoptosis: In cases of severe or irreparable DNA damage, BRCA1 helps trigger programmed cell death (apoptosis) to eliminate potentially cancerous cells.

Individuals with inherited mutations in the BRCA1 gene have a higher risk of developing breast and ovarian cancers compared to the general population. Genetic testing for BRCA1 mutations is available for individuals with a family history of these cancers or those who meet specific clinical criteria. Identifying carriers of BRCA1 mutations allows for enhanced cancer surveillance, risk reduction strategies, and potential targeted therapies.

"Cells, cultured" is a medical term that refers to cells that have been removed from an organism and grown in controlled laboratory conditions outside of the body. This process is called cell culture and it allows scientists to study cells in a more controlled and accessible environment than they would have inside the body. Cultured cells can be derived from a variety of sources, including tissues, organs, or fluids from humans, animals, or cell lines that have been previously established in the laboratory.

Cell culture involves several steps, including isolation of the cells from the tissue, purification and characterization of the cells, and maintenance of the cells in appropriate growth conditions. The cells are typically grown in specialized media that contain nutrients, growth factors, and other components necessary for their survival and proliferation. Cultured cells can be used for a variety of purposes, including basic research, drug development and testing, and production of biological products such as vaccines and gene therapies.

It is important to note that cultured cells may behave differently than they do in the body, and results obtained from cell culture studies may not always translate directly to human physiology or disease. Therefore, it is essential to validate findings from cell culture experiments using additional models and ultimately in clinical trials involving human subjects.

Fibroblasts are specialized cells that play a critical role in the body's immune response and wound healing process. They are responsible for producing and maintaining the extracellular matrix (ECM), which is the non-cellular component present within all tissues and organs, providing structural support and biochemical signals for surrounding cells.

Fibroblasts produce various ECM proteins such as collagens, elastin, fibronectin, and laminins, forming a complex network of fibers that give tissues their strength and flexibility. They also help in the regulation of tissue homeostasis by controlling the turnover of ECM components through the process of remodeling.

In response to injury or infection, fibroblasts become activated and start to proliferate rapidly, migrating towards the site of damage. Here, they participate in the inflammatory response, releasing cytokines and chemokines that attract immune cells to the area. Additionally, they deposit new ECM components to help repair the damaged tissue and restore its functionality.

Dysregulation of fibroblast activity has been implicated in several pathological conditions, including fibrosis (excessive scarring), cancer (where they can contribute to tumor growth and progression), and autoimmune diseases (such as rheumatoid arthritis).

Mitosis is a type of cell division in which the genetic material of a single cell, called the mother cell, is equally distributed into two identical daughter cells. It's a fundamental process that occurs in multicellular organisms for growth, maintenance, and repair, as well as in unicellular organisms for reproduction.

The process of mitosis can be broken down into several stages: prophase, prometaphase, metaphase, anaphase, and telophase. During prophase, the chromosomes condense and become visible, and the nuclear envelope breaks down. In prometaphase, the nuclear membrane is completely disassembled, and the mitotic spindle fibers attach to the chromosomes at their centromeres.

During metaphase, the chromosomes align at the metaphase plate, an imaginary line equidistant from the two spindle poles. In anaphase, sister chromatids are pulled apart by the spindle fibers and move toward opposite poles of the cell. Finally, in telophase, new nuclear envelopes form around each set of chromosomes, and the chromosomes decondense and become less visible.

Mitosis is followed by cytokinesis, a process that divides the cytoplasm of the mother cell into two separate daughter cells. The result of mitosis and cytokinesis is two genetically identical cells, each with the same number and kind of chromosomes as the original parent cell.

Cellular aging, also known as cellular senescence, is a natural process that occurs as cells divide and grow older. Over time, cells accumulate damage to their DNA, proteins, and lipids due to various factors such as genetic mutations, oxidative stress, and epigenetic changes. This damage can impair the cell's ability to function properly and can lead to changes associated with aging, such as decreased tissue repair and regeneration, increased inflammation, and increased risk of age-related diseases.

Cellular aging is characterized by several features, including:

1. Shortened telomeres: Telomeres are the protective caps on the ends of chromosomes that shorten each time a cell divides. When telomeres become too short, the cell can no longer divide and becomes senescent or dies.
2. Epigenetic changes: Epigenetic modifications refer to chemical changes to DNA and histone proteins that affect gene expression without changing the underlying genetic code. As cells age, they accumulate epigenetic changes that can alter gene expression and contribute to cellular aging.
3. Oxidative stress: Reactive oxygen species (ROS) are byproducts of cellular metabolism that can damage DNA, proteins, and lipids. Accumulated ROS over time can lead to oxidative stress, which is associated with cellular aging.
4. Inflammation: Senescent cells produce pro-inflammatory cytokines, chemokines, and matrix metalloproteinases that contribute to a low-grade inflammation known as inflammaging. This chronic inflammation can lead to tissue damage and increase the risk of age-related diseases.
5. Genomic instability: DNA damage accumulates with age, leading to genomic instability and an increased risk of mutations and cancer.

Understanding cellular aging is crucial for developing interventions that can delay or prevent age-related diseases and improve healthy lifespan.

A micronucleus test is a type of genetic toxicology assay used to detect the presence of micronuclei in cells, which are small chromosomal fragments or whole chromosomes that have been missegregated during cell division. The test measures the frequency of micronuclei in cells exposed to a potential genotoxic agent, such as a chemical or radiation, and compares it to the frequency in untreated control cells.

The assay is typically performed on cultured mammalian cells, such as human lymphocytes or Chinese hamster ovary (CHO) cells, and involves exposing the cells to the test agent for a specific period of time, followed by staining and examination of the cells under a microscope. The micronuclei are identified based on their size, shape, and staining characteristics, and the frequency of micronucleated cells is calculated as a measure of genotoxic potential.

Micronucleus tests are widely used in regulatory toxicology to assess the genetic safety of chemicals, drugs, and other substances, and can provide valuable information on potential risks to human health. The test is also used in basic research to study the mechanisms of genotoxicity and chromosomal instability.

"Saccharomyces cerevisiae" is not typically considered a medical term, but it is a scientific name used in the field of microbiology. It refers to a species of yeast that is commonly used in various industrial processes, such as baking and brewing. It's also widely used in scientific research due to its genetic tractability and eukaryotic cellular organization.

However, it does have some relevance to medical fields like medicine and nutrition. For example, certain strains of S. cerevisiae are used as probiotics, which can provide health benefits when consumed. They may help support gut health, enhance the immune system, and even assist in the digestion of certain nutrients.

In summary, "Saccharomyces cerevisiae" is a species of yeast with various industrial and potential medical applications.

The cell nucleus is a membrane-bound organelle found in the eukaryotic cells (cells with a true nucleus). It contains most of the cell's genetic material, organized as DNA molecules in complex with proteins, RNA molecules, and histones to form chromosomes.

The primary function of the cell nucleus is to regulate and control the activities of the cell, including growth, metabolism, protein synthesis, and reproduction. It also plays a crucial role in the process of mitosis (cell division) by separating and protecting the genetic material during this process. The nuclear membrane, or nuclear envelope, surrounding the nucleus is composed of two lipid bilayers with numerous pores that allow for the selective transport of molecules between the nucleoplasm (nucleus interior) and the cytoplasm (cell exterior).

The cell nucleus is a vital structure in eukaryotic cells, and its dysfunction can lead to various diseases, including cancer and genetic disorders.

Hydroxyurea is an antimetabolite drug that is primarily used in the treatment of myeloproliferative disorders such as chronic myelogenous leukemia (CML), essential thrombocythemia, and polycythemia vera. It works by interfering with the synthesis of DNA, which inhibits the growth of cancer cells.

In addition to its use in cancer therapy, hydroxyurea is also used off-label for the management of sickle cell disease. In this context, it helps to reduce the frequency and severity of painful vaso-occlusive crises by increasing the production of fetal hemoglobin (HbF), which decreases the formation of sickled red blood cells.

The medical definition of hydroxyurea is:

A hydantoin derivative and antimetabolite that inhibits ribonucleoside diphosphate reductase, thereby interfering with DNA synthesis. It has been used as an antineoplastic agent, particularly in the treatment of myeloproliferative disorders, and more recently for the management of sickle cell disease to reduce the frequency and severity of painful vaso-occlusive crises by increasing fetal hemoglobin production.

Antioxidants are substances that can prevent or slow damage to cells caused by free radicals, which are unstable molecules that the body produces as a reaction to environmental and other pressures. Antioxidants are able to neutralize free radicals by donating an electron to them, thus stabilizing them and preventing them from causing further damage to the cells.

Antioxidants can be found in a variety of foods, including fruits, vegetables, nuts, and grains. Some common antioxidants include vitamins C and E, beta-carotene, and selenium. Antioxidants are also available as dietary supplements.

In addition to their role in protecting cells from damage, antioxidants have been studied for their potential to prevent or treat a number of health conditions, including cancer, heart disease, and age-related macular degeneration. However, more research is needed to fully understand the potential benefits and risks of using antioxidant supplements.

DNA-activated protein kinase (DNA-PK) is a type of serine/threonine protein kinase that plays a crucial role in the DNA damage response and repair processes in cells. It is composed of a catalytic subunit, DNA-PKcs, and a regulatory subunit, Ku, which binds to double-stranded DNA breaks and recruits DNA-PKcs to the site of damage.

Once activated by DNA damage, DNA-PK phosphorylates various downstream targets involved in DNA repair, including proteins involved in non-homologous end joining (NHEJ) and homologous recombination (HR). NHEJ is a major pathway for the repair of double-stranded DNA breaks, while HR is a more accurate but slower process that requires a template for repair.

Dysregulation of DNA-PK has been implicated in various human diseases, including cancer and neurological disorders. Inhibitors of DNA-PK are being investigated as potential therapeutic agents for the treatment of cancer, particularly in combination with other DNA damage response inhibitors or radiation therapy.

The G2 phase cell cycle checkpoint is a point in the cell cycle, specifically in the G2 phase, where the cell checks for any DNA damage or other issues that may have occurred during the DNA synthesis phase (S phase) before proceeding to mitosis. This checkpoint serves as a quality control mechanism to ensure that the genetic material is accurately and completely replicated and that the cell is ready to divide. If DNA damage or other problems are detected, the cell cycle is halted at the G2 checkpoint until the issues can be resolved. If the damage is too severe or cannot be repaired, the cell may undergo programmed cell death (apoptosis) to prevent the propagation of potentially harmful mutations.

Intracellular signaling peptides and proteins are molecules that play a crucial role in transmitting signals within cells, which ultimately lead to changes in cell behavior or function. These signals can originate from outside the cell (extracellular) or within the cell itself. Intracellular signaling molecules include various types of peptides and proteins, such as:

1. G-protein coupled receptors (GPCRs): These are seven-transmembrane domain receptors that bind to extracellular signaling molecules like hormones, neurotransmitters, or chemokines. Upon activation, they initiate a cascade of intracellular signals through G proteins and secondary messengers.
2. Receptor tyrosine kinases (RTKs): These are transmembrane receptors that bind to growth factors, cytokines, or hormones. Activation of RTKs leads to autophosphorylation of specific tyrosine residues, creating binding sites for intracellular signaling proteins such as adapter proteins, phosphatases, and enzymes like Ras, PI3K, and Src family kinases.
3. Second messenger systems: Intracellular second messengers are small molecules that amplify and propagate signals within the cell. Examples include cyclic adenosine monophosphate (cAMP), cyclic guanosine monophosphate (cGMP), diacylglycerol (DAG), inositol triphosphate (IP3), calcium ions (Ca2+), and nitric oxide (NO). These second messengers activate or inhibit various downstream effectors, leading to changes in cellular responses.
4. Signal transduction cascades: Intracellular signaling proteins often form complex networks of interacting molecules that relay signals from the plasma membrane to the nucleus. These cascades involve kinases (protein kinases A, B, C, etc.), phosphatases, and adapter proteins, which ultimately regulate gene expression, cell cycle progression, metabolism, and other cellular processes.
5. Ubiquitination and proteasome degradation: Intracellular signaling pathways can also control protein stability by modulating ubiquitin-proteasome degradation. E3 ubiquitin ligases recognize specific substrates and conjugate them with ubiquitin molecules, targeting them for proteasomal degradation. This process regulates the abundance of key signaling proteins and contributes to signal termination or amplification.

In summary, intracellular signaling pathways involve a complex network of interacting proteins that relay signals from the plasma membrane to various cellular compartments, ultimately regulating gene expression, metabolism, and other cellular processes. Dysregulation of these pathways can contribute to disease development and progression, making them attractive targets for therapeutic intervention.

Chromatin is the complex of DNA, RNA, and proteins that make up the chromosomes in the nucleus of a cell. It is responsible for packaging the long DNA molecules into a more compact form that fits within the nucleus. Chromatin is made up of repeating units called nucleosomes, which consist of a histone protein octamer wrapped tightly by DNA. The structure of chromatin can be altered through chemical modifications to the histone proteins and DNA, which can influence gene expression and other cellular processes.

Alkylating agents are a class of chemotherapy drugs that work by alkylating, or adding an alkyl group to, DNA molecules. This process can damage the DNA and prevent cancer cells from dividing and growing. Alkylating agents are often used to treat various types of cancer, including Hodgkin's lymphoma, non-Hodgkin's lymphoma, multiple myeloma, and solid tumors. Examples of alkylating agents include cyclophosphamide, melphalan, and chlorambucil. These drugs can have significant side effects, including nausea, vomiting, hair loss, and an increased risk of infection. They can also cause long-term damage to the heart, lungs, and reproductive system.

X-rays, also known as radiographs, are a type of electromagnetic radiation with higher energy and shorter wavelength than visible light. In medical imaging, X-rays are used to produce images of the body's internal structures, such as bones and organs, by passing the X-rays through the body and capturing the resulting shadows or patterns on a specialized film or digital detector.

The amount of X-ray radiation used is carefully controlled to minimize exposure and ensure patient safety. Different parts of the body absorb X-rays at different rates, allowing for contrast between soft tissues and denser structures like bone. This property makes X-rays an essential tool in diagnosing and monitoring a wide range of medical conditions, including fractures, tumors, infections, and foreign objects within the body.

In the field of medicine, "time factors" refer to the duration of symptoms or time elapsed since the onset of a medical condition, which can have significant implications for diagnosis and treatment. Understanding time factors is crucial in determining the progression of a disease, evaluating the effectiveness of treatments, and making critical decisions regarding patient care.

For example, in stroke management, "time is brain," meaning that rapid intervention within a specific time frame (usually within 4.5 hours) is essential to administering tissue plasminogen activator (tPA), a clot-busting drug that can minimize brain damage and improve patient outcomes. Similarly, in trauma care, the "golden hour" concept emphasizes the importance of providing definitive care within the first 60 minutes after injury to increase survival rates and reduce morbidity.

Time factors also play a role in monitoring the progression of chronic conditions like diabetes or heart disease, where regular follow-ups and assessments help determine appropriate treatment adjustments and prevent complications. In infectious diseases, time factors are crucial for initiating antibiotic therapy and identifying potential outbreaks to control their spread.

Overall, "time factors" encompass the significance of recognizing and acting promptly in various medical scenarios to optimize patient outcomes and provide effective care.

Replication Protein A (RPA) is a single-stranded DNA binding protein complex that plays a crucial role in the process of DNA replication, repair, and recombination. In eukaryotic cells, RPA is composed of three subunits: RPA70, RPA32, and RPA14. The primary function of RPA is to coat single-stranded DNA (ssDNA) generated during these processes, protecting it from degradation, preventing the formation of secondary structures, and promoting the recruitment of other proteins involved in DNA metabolism.

RPA binds ssDNA with high affinity and specificity, forming a stable complex that protects the DNA from nucleases, chemical modifications, and other damaging agents. The protein also participates in the regulation of various enzymatic activities, such as helicase loading and activation, end processing, and polymerase processivity.

During DNA replication, RPA is essential for the initiation and elongation phases. It facilitates the assembly of the pre-replicative complex (pre-RC) at origins of replication, aids in the recruitment and activation of helicases, and promotes the switch from MCM2-7 helicase to polymerase processivity during DNA synthesis.

In addition to its role in DNA replication, RPA is involved in various DNA repair pathways, including nucleotide excision repair (NER), base excision repair (BER), mismatch repair (MMR), and double-strand break repair (DSBR). It also plays a critical role in meiotic recombination during sexual reproduction.

In summary, Replication Protein A (RPA) is a eukaryotic single-stranded DNA binding protein complex that protects, stabilizes, and regulates ssDNA during DNA replication, repair, and recombination processes.

Recombinational DNA repair is a biological process that takes place in cells to correct damage to the DNA molecule. This type of repair is particularly important in maintaining the stability and integrity of the genetic code, especially in response to double-strand breaks (DSBs) in the DNA.

In recombinational DNA repair, the cell uses a template from a homologous DNA sequence, typically a sister chromatid, to restore the damaged region. The process involves several steps:

1. Resection: The broken ends of the DNA molecule are processed by enzymes that remove nucleotides and create 3' single-stranded overhangs.
2. Recombination: The single-stranded overhangs invade a homologous DNA sequence, forming a displacement loop (D-loop) structure. This invasion is facilitated by recombinase proteins such as Rad51 and Dmc1.
3. Strand exchange: The invading 3' end of the single strand pairs with the complementary sequence in the template DNA, and DNA synthesis occurs using the template to restore the missing genetic information.
4. Resolution: The recombination intermediate is resolved, and the repaired DNA molecule is ligated together. This step can result in different outcomes, including crossover or non-crossover events, depending on the specific mechanisms involved.

Recombinational DNA repair plays a crucial role in maintaining genome stability and preventing mutations that could lead to diseases such as cancer. Additionally, this process is exploited in genetic engineering techniques like homologous recombination-mediated gene targeting and CRISPR-Cas9 genome editing.

Biological models, also known as physiological models or organismal models, are simplified representations of biological systems, processes, or mechanisms that are used to understand and explain the underlying principles and relationships. These models can be theoretical (conceptual or mathematical) or physical (such as anatomical models, cell cultures, or animal models). They are widely used in biomedical research to study various phenomena, including disease pathophysiology, drug action, and therapeutic interventions.

Examples of biological models include:

1. Mathematical models: These use mathematical equations and formulas to describe complex biological systems or processes, such as population dynamics, metabolic pathways, or gene regulation networks. They can help predict the behavior of these systems under different conditions and test hypotheses about their underlying mechanisms.
2. Cell cultures: These are collections of cells grown in a controlled environment, typically in a laboratory dish or flask. They can be used to study cellular processes, such as signal transduction, gene expression, or metabolism, and to test the effects of drugs or other treatments on these processes.
3. Animal models: These are living organisms, usually vertebrates like mice, rats, or non-human primates, that are used to study various aspects of human biology and disease. They can provide valuable insights into the pathophysiology of diseases, the mechanisms of drug action, and the safety and efficacy of new therapies.
4. Anatomical models: These are physical representations of biological structures or systems, such as plastic models of organs or tissues, that can be used for educational purposes or to plan surgical procedures. They can also serve as a basis for developing more sophisticated models, such as computer simulations or 3D-printed replicas.

Overall, biological models play a crucial role in advancing our understanding of biology and medicine, helping to identify new targets for therapeutic intervention, develop novel drugs and treatments, and improve human health.

Protein binding, in the context of medical and biological sciences, refers to the interaction between a protein and another molecule (known as the ligand) that results in a stable complex. This process is often reversible and can be influenced by various factors such as pH, temperature, and concentration of the involved molecules.

In clinical chemistry, protein binding is particularly important when it comes to drugs, as many of them bind to proteins (especially albumin) in the bloodstream. The degree of protein binding can affect a drug's distribution, metabolism, and excretion, which in turn influence its therapeutic effectiveness and potential side effects.

Protein-bound drugs may be less available for interaction with their target tissues, as only the unbound or "free" fraction of the drug is active. Therefore, understanding protein binding can help optimize dosing regimens and minimize adverse reactions.

DNA helicases are a group of enzymes that are responsible for separating the two strands of DNA during processes such as replication and transcription. They do this by unwinding the double helix structure of DNA, using energy from ATP to break the hydrogen bonds between the base pairs. This allows other proteins to access the individual strands of DNA and carry out functions such as copying the genetic code or transcribing it into RNA.

During replication, DNA helicases help to create a replication fork, where the two strands of DNA are separated and new complementary strands are synthesized. In transcription, DNA helicases help to unwind the DNA double helix at the promoter region, allowing the RNA polymerase enzyme to bind and begin transcribing the DNA into RNA.

DNA helicases play a crucial role in maintaining the integrity of the genetic code and are essential for the normal functioning of cells. Defects in DNA helicases have been linked to various diseases, including cancer and neurological disorders.

Oxidation-Reduction (redox) reactions are a type of chemical reaction involving a transfer of electrons between two species. The substance that loses electrons in the reaction is oxidized, and the substance that gains electrons is reduced. Oxidation and reduction always occur together in a redox reaction, hence the term "oxidation-reduction."

In biological systems, redox reactions play a crucial role in many cellular processes, including energy production, metabolism, and signaling. The transfer of electrons in these reactions is often facilitated by specialized molecules called electron carriers, such as nicotinamide adenine dinucleotide (NAD+/NADH) and flavin adenine dinucleotide (FAD/FADH2).

The oxidation state of an element in a compound is a measure of the number of electrons that have been gained or lost relative to its neutral state. In redox reactions, the oxidation state of one or more elements changes as they gain or lose electrons. The substance that is oxidized has a higher oxidation state, while the substance that is reduced has a lower oxidation state.

Overall, oxidation-reduction reactions are fundamental to the functioning of living organisms and are involved in many important biological processes.

Poly(ADP-ribose) (PAR) is not strictly referred to as "Poly Adenosine Diphosphate Ribose" in the medical or biochemical context, although the term ADP-ribose is a component of it. Poly(ADP-ribose) is a polymer of ADP-ribose units that are synthesized by enzymes called poly(ADP-ribose) polymerases (PARPs).

Poly(ADP-ribosyl)ation, the process of adding PAR polymers to target proteins, plays a crucial role in various cellular processes such as DNA repair, genomic stability, and cell death. In medical research, alterations in PAR metabolism have been implicated in several diseases, including cancer and neurodegenerative disorders. Therefore, understanding the function and regulation of poly(ADP-ribose) is of significant interest in biomedical sciences.

Cell death is the process by which cells cease to function and eventually die. There are several ways that cells can die, but the two most well-known and well-studied forms of cell death are apoptosis and necrosis.

Apoptosis is a programmed form of cell death that occurs as a normal and necessary process in the development and maintenance of healthy tissues. During apoptosis, the cell's DNA is broken down into small fragments, the cell shrinks, and the membrane around the cell becomes fragmented, allowing the cell to be easily removed by phagocytic cells without causing an inflammatory response.

Necrosis, on the other hand, is a form of cell death that occurs as a result of acute tissue injury or overwhelming stress. During necrosis, the cell's membrane becomes damaged and the contents of the cell are released into the surrounding tissue, causing an inflammatory response.

There are also other forms of cell death, such as autophagy, which is a process by which cells break down their own organelles and proteins to recycle nutrients and maintain energy homeostasis, and pyroptosis, which is a form of programmed cell death that occurs in response to infection and involves the activation of inflammatory caspases.

Cell death is an important process in many physiological and pathological processes, including development, tissue homeostasis, and disease. Dysregulation of cell death can contribute to the development of various diseases, including cancer, neurodegenerative disorders, and autoimmune diseases.

Small interfering RNA (siRNA) is a type of short, double-stranded RNA molecule that plays a role in the RNA interference (RNAi) pathway. The RNAi pathway is a natural cellular process that regulates gene expression by targeting and destroying specific messenger RNA (mRNA) molecules, thereby preventing the translation of those mRNAs into proteins.

SiRNAs are typically 20-25 base pairs in length and are generated from longer double-stranded RNA precursors called hairpin RNAs or dsRNAs by an enzyme called Dicer. Once generated, siRNAs associate with a protein complex called the RNA-induced silencing complex (RISC), which uses one strand of the siRNA (the guide strand) to recognize and bind to complementary sequences in the target mRNA. The RISC then cleaves the target mRNA, leading to its degradation and the inhibition of protein synthesis.

SiRNAs have emerged as a powerful tool for studying gene function and have shown promise as therapeutic agents for a variety of diseases, including viral infections, cancer, and genetic disorders. However, their use as therapeutics is still in the early stages of development, and there are challenges associated with delivering siRNAs to specific cells and tissues in the body.

Molecular sequence data refers to the specific arrangement of molecules, most commonly nucleotides in DNA or RNA, or amino acids in proteins, that make up a biological macromolecule. This data is generated through laboratory techniques such as sequencing, and provides information about the exact order of the constituent molecules. This data is crucial in various fields of biology, including genetics, evolution, and molecular biology, allowing for comparisons between different organisms, identification of genetic variations, and studies of gene function and regulation.

Proto-oncogene proteins, such as c-MDM2, are normal cellular proteins that play crucial roles in regulating various cellular processes, including cell growth, differentiation, and apoptosis (programmed cell death). When these genes undergo mutations or are overexpressed, they can become oncogenes, which contribute to the development of cancer.

The c-MDM2 protein is a key regulator of the cell cycle and is involved in the negative regulation of the tumor suppressor protein p53. Under normal conditions, p53 helps prevent the formation of tumors by inducing cell cycle arrest or apoptosis in response to DNA damage or other stress signals. However, when c-MDM2 is overexpressed or mutated, it can bind and inhibit p53, leading to uncontrolled cell growth and increased risk of cancer development.

In summary, proto-oncogene proteins like c-MDM2 are important regulators of normal cellular processes, but when they become dysregulated through mutations or overexpression, they can contribute to the formation of tumors and cancer progression.

Genetic recombination is the process by which genetic material is exchanged between two similar or identical molecules of DNA during meiosis, resulting in new combinations of genes on each chromosome. This exchange occurs during crossover, where segments of DNA are swapped between non-sister homologous chromatids, creating genetic diversity among the offspring. It is a crucial mechanism for generating genetic variability and facilitating evolutionary change within populations. Additionally, recombination also plays an essential role in DNA repair processes through mechanisms such as homologous recombinational repair (HRR) and non-homologous end joining (NHEJ).

Pyrimidine dimers are a type of DNA lesion that form when two adjacent pyrimidine bases on the same strand of DNA become covalently linked, usually as a result of exposure to ultraviolet (UV) light. The most common type of pyrimidine dimer is the cyclobutane pyrimidine dimer (CPD), which forms when two thymine bases are linked together in a cyclobutane ring structure.

Pyrimidine dimers can distort the DNA helix and interfere with normal replication and transcription processes, leading to mutations and potentially cancer. The formation of pyrimidine dimers is a major mechanism by which UV radiation causes skin damage and increases the risk of skin cancer.

The body has several mechanisms for repairing pyrimidine dimers, including nucleotide excision repair (NER) and base excision repair (BER). However, if these repair mechanisms are impaired or overwhelmed, pyrimidine dimers can persist and contribute to the development of cancer.

DNA-Formamidopyrimidine Glycosylase (Fpg) is an enzyme that plays a crucial role in the repair of DNA damage. It is involved in the base excision repair pathway, which is responsible for correcting damaged or mismatched bases in the DNA molecule.

The Fpg protein specifically recognizes and removes formamidopyrimidines, which are damaged bases that can arise from oxidative stress or exposure to certain chemicals or radiation. Formamidopyrimidines include two types of lesions: formamidopyrimidine (Fapy) adenine and Fapy guanine. These lesions can distort the structure of the DNA molecule, leading to mutations and genomic instability if not repaired.

By removing the damaged bases, Fpg allows for the insertion of a correct base during DNA replication, preventing the transmission of mutations to subsequent generations of cells. This enzyme is highly conserved across different species, indicating its importance in maintaining genome stability and preventing the development of diseases such as cancer.

Mitomycin is an antineoplastic antibiotic derived from Streptomyces caespitosus. It is primarily used in cancer chemotherapy, particularly in the treatment of various carcinomas including gastrointestinal tract malignancies and breast cancer. Mitomycin works by forming cross-links in DNA, thereby inhibiting its replication and transcription, which ultimately leads to cell death.

In addition to its systemic use, mitomycin is also used topically in ophthalmology for the treatment of certain eye conditions such as glaucoma and various ocular surface disorders. The topical application of mitomycin can help reduce scarring and fibrosis by inhibiting the proliferation of fibroblasts.

It's important to note that mitomycin has a narrow therapeutic index, meaning there is only a small range between an effective dose and a toxic one. Therefore, its use should be closely monitored to minimize side effects, which can include myelosuppression, mucositis, alopecia, and potential secondary malignancies.

HCT116 cells are a type of human colon cancer cell line that is widely used in scientific research. They were originally established in the early 1980s from a primary colon tumor that had metastasized to the liver. HCT116 cells are known for their stability, robust growth, and susceptibility to various genetic manipulations, making them a popular choice for studying cancer biology, drug discovery, and gene function.

These cells have several important features that make them useful in research. For example, they harbor mutations in key genes involved in colorectal cancer development, such as the adenomatous polyposis coli (APC) gene and the KRAS oncogene. Additionally, HCT116 cells can be easily cultured in the lab and are amenable to a variety of experimental techniques, including genetic modification, drug screening, and protein analysis.

It is important to note that while HCT116 cells provide valuable insights into colon cancer biology, they represent only one type of cancer cell line, and their behavior may not necessarily reflect the complexity of human tumors in vivo. Therefore, researchers must exercise caution when interpreting results obtained from these cells and consider other complementary approaches to validate their findings.

Micronuclei, chromosome-defective, refer to small additional nuclei that form during cell division when the genetic material is not properly divided between the two resulting daughter cells. These micronuclei can contain whole chromosomes or fragments of chromosomes that were not incorporated into either of the main nuclei during cell division. Chromosome-defective micronuclei are often associated with genomic instability, DNA damage, and chromosomal aberrations, which can lead to various health issues, including cancer and developmental defects. They can be used as a biomarker for genetic damage in cells and are commonly observed in response to exposure to mutagenic agents such as radiation or chemicals.

Ubiquitination is a post-translational modification process in which a ubiquitin protein is covalently attached to a target protein. This process plays a crucial role in regulating various cellular functions, including protein degradation, DNA repair, and signal transduction. The addition of ubiquitin can lead to different outcomes depending on the number and location of ubiquitin molecules attached to the target protein. Monoubiquitination (the attachment of a single ubiquitin molecule) or multiubiquitination (the attachment of multiple ubiquitin molecules) can mark proteins for degradation by the 26S proteasome, while specific types of ubiquitination (e.g., K63-linked polyubiquitination) can serve as a signal for nonproteolytic functions such as endocytosis, autophagy, or DNA repair. Ubiquitination is a highly regulated process that involves the coordinated action of three enzymes: E1 ubiquitin-activating enzyme, E2 ubiquitin-conjugating enzyme, and E3 ubiquitin ligase. Dysregulation of ubiquitination has been implicated in various diseases, including cancer, neurodegenerative disorders, and inflammatory conditions.

Cyclin-dependent kinase inhibitor p21, also known as CDKN1A or p21/WAF1/CIP1, is a protein that regulates the cell cycle. It inhibits the activity of cyclin-dependent kinases (CDKs), which are enzymes that play crucial roles in controlling the progression of the cell cycle.

The binding of p21 to CDKs prevents the phosphorylation and activation of downstream targets, leading to cell cycle arrest. This protein is transcriptionally activated by tumor suppressor protein p53 in response to DNA damage or other stress signals, and it functions as an important mediator of p53-dependent growth arrest.

By inhibiting CDKs, p21 helps to ensure that cells do not proceed through the cell cycle until damaged DNA has been repaired, thereby preventing the propagation of potentially harmful mutations. Additionally, p21 has been implicated in other cellular processes such as apoptosis, differentiation, and senescence. Dysregulation of p21 has been associated with various human diseases, including cancer.

A telomere is a region of repetitive DNA sequences found at the end of chromosomes, which protects the genetic data from damage and degradation during cell division. Telomeres naturally shorten as cells divide, and when they become too short, the cell can no longer divide and becomes senescent or dies. This natural process is associated with aging and various age-related diseases. The length of telomeres can also be influenced by various genetic and environmental factors, including stress, diet, and lifestyle.

Nucleic acid synthesis inhibitors are a class of antimicrobial, antiviral, or antitumor agents that block the synthesis of nucleic acids (DNA or RNA) by interfering with enzymes involved in their replication. These drugs can target various stages of nucleic acid synthesis, including DNA transcription, replication, and repair, as well as RNA transcription and processing.

Examples of nucleic acid synthesis inhibitors include:

1. Antibiotics like quinolones (e.g., ciprofloxacin), rifamycins (e.g., rifampin), and trimethoprim, which target bacterial DNA gyrase, RNA polymerase, or dihydrofolate reductase, respectively.
2. Antiviral drugs like reverse transcriptase inhibitors (e.g., zidovudine, lamivudine) and integrase strand transfer inhibitors (e.g., raltegravir), which target HIV replication by interfering with viral enzymes required for DNA synthesis.
3. Antitumor drugs like antimetabolites (e.g., methotrexate, 5-fluorouracil) and topoisomerase inhibitors (e.g., etoposide, doxorubicin), which interfere with DNA replication and repair in cancer cells.

These drugs have been widely used for treating various bacterial and viral infections, as well as cancers, due to their ability to selectively inhibit the growth of target cells without affecting normal cellular functions significantly. However, they may also cause side effects related to their mechanism of action or off-target effects on non-target cells.

DNA end-joining repair, also known as non-homologous end joining (NHEJ), is a primary mechanism for repairing double-stranded breaks in DNA. This pathway involves the direct rejoining of broken ends, often with some degree of imprecision, and it can result in small deletions or insertions at the site of the break. NHEJ plays a crucial role in maintaining genomic stability and is an important process for the repair of DNA damage that can occur as a result of ionizing radiation, chemotherapeutic agents, and other sources of genotoxic stress. The key proteins involved in NHEJ include the Ku heterodimer, DNA-dependent protein kinase (DNA-PK), XRCC4, XLF, and DNA ligase IV.

Western blotting is a laboratory technique used in molecular biology to detect and quantify specific proteins in a mixture of many different proteins. This technique is commonly used to confirm the expression of a protein of interest, determine its size, and investigate its post-translational modifications. The name "Western" blotting distinguishes this technique from Southern blotting (for DNA) and Northern blotting (for RNA).

The Western blotting procedure involves several steps:

1. Protein extraction: The sample containing the proteins of interest is first extracted, often by breaking open cells or tissues and using a buffer to extract the proteins.
2. Separation of proteins by electrophoresis: The extracted proteins are then separated based on their size by loading them onto a polyacrylamide gel and running an electric current through the gel (a process called sodium dodecyl sulfate-polyacrylamide gel electrophoresis or SDS-PAGE). This separates the proteins according to their molecular weight, with smaller proteins migrating faster than larger ones.
3. Transfer of proteins to a membrane: After separation, the proteins are transferred from the gel onto a nitrocellulose or polyvinylidene fluoride (PVDF) membrane using an electric current in a process called blotting. This creates a replica of the protein pattern on the gel but now immobilized on the membrane for further analysis.
4. Blocking: The membrane is then blocked with a blocking agent, such as non-fat dry milk or bovine serum albumin (BSA), to prevent non-specific binding of antibodies in subsequent steps.
5. Primary antibody incubation: A primary antibody that specifically recognizes the protein of interest is added and allowed to bind to its target protein on the membrane. This step may be performed at room temperature or 4°C overnight, depending on the antibody's properties.
6. Washing: The membrane is washed with a buffer to remove unbound primary antibodies.
7. Secondary antibody incubation: A secondary antibody that recognizes the primary antibody (often coupled to an enzyme or fluorophore) is added and allowed to bind to the primary antibody. This step may involve using a horseradish peroxidase (HRP)-conjugated or alkaline phosphatase (AP)-conjugated secondary antibody, depending on the detection method used later.
8. Washing: The membrane is washed again to remove unbound secondary antibodies.
9. Detection: A detection reagent is added to visualize the protein of interest by detecting the signal generated from the enzyme-conjugated or fluorophore-conjugated secondary antibody. This can be done using chemiluminescent, colorimetric, or fluorescent methods.
10. Analysis: The resulting image is analyzed to determine the presence and quantity of the protein of interest in the sample.

Western blotting is a powerful technique for identifying and quantifying specific proteins within complex mixtures. It can be used to study protein expression, post-translational modifications, protein-protein interactions, and more. However, it requires careful optimization and validation to ensure accurate and reproducible results.

Mutagenicity tests are a type of laboratory assays used to identify agents that can cause genetic mutations. These tests detect changes in the DNA of organisms, such as bacteria, yeast, or mammalian cells, after exposure to potential mutagens. The most commonly used mutagenicity test is the Ames test, which uses a strain of Salmonella bacteria that is sensitive to mutagens. If a chemical causes an increase in the number of revertants (reversion to the wild type) in the bacterial population, it is considered to be a mutagen. Other tests include the mouse lymphoma assay and the chromosomal aberration test. These tests are used to evaluate the potential genotoxicity of chemicals and are an important part of the safety evaluation process for new drugs, chemicals, and other substances.

Guanine is not a medical term per se, but it is a biological molecule that plays a crucial role in the body. Guanine is one of the four nucleobases found in the nucleic acids DNA and RNA, along with adenine, cytosine, and thymine (in DNA) or uracil (in RNA). Specifically, guanine pairs with cytosine via hydrogen bonds to form a base pair.

Guanine is a purine derivative, which means it has a double-ring structure. It is formed through the synthesis of simpler molecules in the body and is an essential component of genetic material. Guanine's chemical formula is C5H5N5O.

While guanine itself is not a medical term, abnormalities or mutations in genes that contain guanine nucleotides can lead to various medical conditions, including genetic disorders and cancer.

A dose-response relationship in the context of drugs refers to the changes in the effects or symptoms that occur as the dose of a drug is increased or decreased. Generally, as the dose of a drug is increased, the severity or intensity of its effects also increases. Conversely, as the dose is decreased, the effects of the drug become less severe or may disappear altogether.

The dose-response relationship is an important concept in pharmacology and toxicology because it helps to establish the safe and effective dosage range for a drug. By understanding how changes in the dose of a drug affect its therapeutic and adverse effects, healthcare providers can optimize treatment plans for their patients while minimizing the risk of harm.

The dose-response relationship is typically depicted as a curve that shows the relationship between the dose of a drug and its effect. The shape of the curve may vary depending on the drug and the specific effect being measured. Some drugs may have a steep dose-response curve, meaning that small changes in the dose can result in large differences in the effect. Other drugs may have a more gradual dose-response curve, where larger changes in the dose are needed to produce significant effects.

In addition to helping establish safe and effective dosages, the dose-response relationship is also used to evaluate the potential therapeutic benefits and risks of new drugs during clinical trials. By systematically testing different doses of a drug in controlled studies, researchers can identify the optimal dosage range for the drug and assess its safety and efficacy.

Xeroderma Pigmentosum Group A Protein, also known as XPA protein, is a crucial component of the nucleotide excision repair (NER) pathway in humans. The NER pathway is responsible for repairing damaged DNA, including DNA that has been damaged by ultraviolet (UV) light.

XPA protein plays a central role in the NER process by serving as a scaffold that helps to coordinate and regulate the activity of other NER proteins. XPA protein binds directly to the damaged DNA site, helping to recruit and position other NER proteins for efficient repair.

Mutations in the XPA gene can lead to Xeroderma Pigmentosum (XP), a rare genetic disorder characterized by extreme sensitivity to UV light. Individuals with XP are prone to developing skin cancer and other forms of cancer at an early age due to their inability to repair DNA damage caused by UV light.

Lymphocytes are a type of white blood cell that is an essential part of the immune system. They are responsible for recognizing and responding to potentially harmful substances such as viruses, bacteria, and other foreign invaders. There are two main types of lymphocytes: B-lymphocytes (B-cells) and T-lymphocytes (T-cells).

B-lymphocytes produce antibodies, which are proteins that help to neutralize or destroy foreign substances. When a B-cell encounters a foreign substance, it becomes activated and begins to divide and differentiate into plasma cells, which produce and secrete large amounts of antibodies. These antibodies bind to the foreign substance, marking it for destruction by other immune cells.

T-lymphocytes, on the other hand, are involved in cell-mediated immunity. They directly attack and destroy infected cells or cancerous cells. T-cells can also help to regulate the immune response by producing chemical signals that activate or inhibit other immune cells.

Lymphocytes are produced in the bone marrow and mature in either the bone marrow (B-cells) or the thymus gland (T-cells). They circulate throughout the body in the blood and lymphatic system, where they can be found in high concentrations in lymph nodes, the spleen, and other lymphoid organs.

Abnormalities in the number or function of lymphocytes can lead to a variety of immune-related disorders, including immunodeficiency diseases, autoimmune disorders, and cancer.

Fungal DNA refers to the genetic material present in fungi, which are a group of eukaryotic organisms that include microorganisms such as yeasts and molds, as well as larger organisms like mushrooms. The DNA of fungi, like that of all living organisms, is made up of nucleotides that are arranged in a double helix structure.

Fungal DNA contains the genetic information necessary for the growth, development, and reproduction of fungi. This includes the instructions for making proteins, which are essential for the structure and function of cells, as well as other important molecules such as enzymes and nucleic acids.

Studying fungal DNA can provide valuable insights into the biology and evolution of fungi, as well as their potential uses in medicine, agriculture, and industry. For example, researchers have used genetic engineering techniques to modify the DNA of fungi to produce drugs, biofuels, and other useful products. Additionally, understanding the genetic makeup of pathogenic fungi can help scientists develop new strategies for preventing and treating fungal infections.

Ataxia telangiectasia is a rare, inherited genetic disorder that affects the nervous system, immune system, and overall development. The condition is characterized by progressive difficulty with coordination and balance (ataxia), as well as the development of small, dilated blood vessels (telangiectasias) on the skin and eyes.

The underlying cause of ataxia telangiectasia is a mutation in the ATM gene, which provides instructions for making a protein that plays a critical role in DNA repair and maintaining genetic stability. When this gene is mutated, cells are unable to properly repair damaged DNA, leading to an increased risk of cancer and other health problems.

Individuals with ataxia telangiectasia typically begin to show symptoms during early childhood, with progressive difficulties in coordination and balance, slurred speech, and recurrent respiratory infections due to weakened immune function. Over time, these symptoms can worsen, leading to significant disability and reduced life expectancy.

There is currently no cure for ataxia telangiectasia, and treatment is focused on managing the symptoms and complications of the condition. This may include physical therapy, speech therapy, and medications to help control infections and other health problems.

Medical definitions of "oxidants" refer to them as oxidizing agents or substances that can gain electrons and be reduced. They are capable of accepting electrons from other molecules in chemical reactions, leading to the production of oxidation products. In biological systems, oxidants play a crucial role in various cellular processes such as energy production and immune responses. However, an imbalance between oxidant and antioxidant levels can lead to a state of oxidative stress, which has been linked to several diseases, including cancer, cardiovascular disease, and neurodegenerative disorders. Examples of oxidants include reactive oxygen species (ROS), such as superoxide anion, hydrogen peroxide, and hydroxyl radical, as well as reactive nitrogen species (RNS), such as nitric oxide and peroxynitrite.

Endodeoxyribonucleases are a type of enzyme that cleave, or cut, phosphodiester bonds within the backbone of DNA molecules. These enzymes are also known as restriction endonucleases or simply restriction enzymes. They are called "restriction" enzymes because they were first discovered in bacteria, where they function to protect the organism from foreign DNA by cleaving and destroying invading viral DNA.

Endodeoxyribonucleases recognize specific sequences of nucleotides within the DNA molecule, known as recognition sites or restriction sites, and cut the phosphodiester bonds at specific locations within these sites. The cuts made by endodeoxyribonucleases can be either "sticky" or "blunt," depending on whether the enzyme leaves single-stranded overhangs or creates blunt ends at the site of cleavage, respectively.

Endodeoxyribonucleases are widely used in molecular biology research for various applications, including DNA cloning, genome mapping, and genetic engineering. They allow researchers to cut DNA molecules at specific sites, creating defined fragments that can be manipulated and recombined in a variety of ways.

CDC25 phosphatases are a group of enzymes that play crucial roles in the regulation of the cell cycle, which is the series of events that cells undergo as they grow and divide. Specifically, CDC25 phosphatases function to remove inhibitory phosphates from certain cyclin-dependent kinases (CDKs), thereby activating them and allowing the cell cycle to progress.

There are three main types of CDC25 phosphatases in humans, known as CDC25A, CDC25B, and CDC25C. These enzymes are named after the original yeast homolog, called Cdc25, which was discovered to be essential for cell cycle progression.

CDC25 phosphatases are tightly regulated during the cell cycle, with their activity being controlled by various mechanisms such as phosphorylation, protein-protein interactions, and subcellular localization. Dysregulation of CDC25 phosphatases has been implicated in several human diseases, including cancer, where they can contribute to uncontrolled cell growth and division. Therefore, understanding the functions and regulation of CDC25 phosphatases is an important area of research in molecular biology and medicine.

Proliferating Cell Nuclear Antigen (PCNA) is a protein that plays an essential role in the process of DNA replication and repair in eukaryotic cells. It functions as a cofactor for DNA polymerase delta, enhancing its activity during DNA synthesis. PCNA forms a sliding clamp around DNA, allowing it to move along the template and coordinate the actions of various enzymes involved in DNA metabolism.

PCNA is often used as a marker for cell proliferation because its levels increase in cells that are actively dividing or have been stimulated to enter the cell cycle. Immunostaining techniques can be used to detect PCNA and determine the proliferative status of tissues or cultures. In this context, 'proliferating' refers to the rapid multiplication of cells through cell division.

Nuclear antigens are proteins or other molecules found in the nucleus of a cell that can stimulate an immune response and produce antibodies when they are recognized as foreign by the body's immune system. These antigens are normally located inside the cell and are not typically exposed to the immune system, but under certain circumstances, such as during cell death or damage, they may be released and become targets of the immune system.

Nuclear antigens can play a role in the development of some autoimmune diseases, such as systemic lupus erythematosus (SLE), where the body's immune system mistakenly attacks its own cells and tissues. In SLE, nuclear antigens such as double-stranded DNA and nucleoproteins are common targets of the abnormal immune response.

Testing for nuclear antigens is often used in the diagnosis and monitoring of autoimmune diseases. For example, a positive test for anti-double-stranded DNA antibodies is a specific indicator of SLE and can help confirm the diagnosis. However, it's important to note that not all people with SLE will have positive nuclear antigen tests, and other factors must also be considered in making a diagnosis.

Endonucleases are enzymes that cleave, or cut, phosphodiester bonds within a polynucleotide chain, specifically within the same molecule of DNA or RNA. They can be found in all living organisms and play crucial roles in various biological processes, such as DNA replication, repair, and recombination.

Endonucleases can recognize specific nucleotide sequences (sequence-specific endonucleases) or have no sequence preference (non-specific endonucleases). Some endonucleases generate sticky ends, overhangs of single-stranded DNA after cleavage, while others produce blunt ends without any overhang.

These enzymes are widely used in molecular biology techniques, such as restriction digestion, cloning, and genome editing (e.g., CRISPR-Cas9 system). Restriction endonucleases recognize specific DNA sequences called restriction sites and cleave the phosphodiester bonds at or near these sites, generating defined fragment sizes that can be separated by agarose gel electrophoresis. This property is essential for various applications in genetic engineering and biotechnology.

RNA interference (RNAi) is a biological process in which RNA molecules inhibit the expression of specific genes. This process is mediated by small RNA molecules, including microRNAs (miRNAs) and small interfering RNAs (siRNAs), that bind to complementary sequences on messenger RNA (mRNA) molecules, leading to their degradation or translation inhibition.

RNAi plays a crucial role in regulating gene expression and defending against foreign genetic elements, such as viruses and transposons. It has also emerged as an important tool for studying gene function and developing therapeutic strategies for various diseases, including cancer and viral infections.

A "knockout" mouse is a genetically engineered mouse in which one or more genes have been deleted or "knocked out" using molecular biology techniques. This allows researchers to study the function of specific genes and their role in various biological processes, as well as potential associations with human diseases. The mice are generated by introducing targeted DNA modifications into embryonic stem cells, which are then used to create a live animal. Knockout mice have been widely used in biomedical research to investigate gene function, disease mechanisms, and potential therapeutic targets.

Carcinogens are agents (substances or mixtures of substances) that can cause cancer. They may be naturally occurring or man-made. Carcinogens can increase the risk of cancer by altering cellular DNA, disrupting cellular function, or promoting cell growth. Examples of carcinogens include certain chemicals found in tobacco smoke, asbestos, UV radiation from the sun, and some viruses.

It's important to note that not all exposures to carcinogens will result in cancer, and the risk typically depends on factors such as the level and duration of exposure, individual genetic susceptibility, and lifestyle choices. The International Agency for Research on Cancer (IARC) classifies carcinogens into different groups based on the strength of evidence linking them to cancer:

Group 1: Carcinogenic to humans
Group 2A: Probably carcinogenic to humans
Group 2B: Possibly carcinogenic to humans
Group 3: Not classifiable as to its carcinogenicity to humans
Group 4: Probably not carcinogenic to humans

This information is based on medical research and may be subject to change as new studies become available. Always consult a healthcare professional for medical advice.

Homologous recombination is a type of genetic recombination that occurs between two similar or identical (homologous) segments of DNA. It is a natural process that helps to maintain the stability of an organism's genome and plays a crucial role in DNA repair, particularly the repair of double-strand breaks.

In homologous recombination, the two DNA molecules exchange genetic information through a series of steps, including the formation of Holliday junctions, where the strands cross over and exchange partners. This process can result in new combinations of genetic material, which can increase genetic diversity and contribute to evolution.

Homologous recombination is also used in biotechnology and genetic engineering to introduce specific changes into DNA sequences or to create genetically modified organisms.

p53 is a tumor suppressor gene that encodes a protein responsible for controlling cell growth and division. The p53 protein plays a crucial role in preventing the development of cancer by regulating the cell cycle and activating DNA repair processes when genetic damage is detected. If the damage is too severe to be repaired, p53 can trigger apoptosis, or programmed cell death, to prevent the propagation of potentially cancerous cells. Mutations in the TP53 gene, which encodes the p53 protein, are among the most common genetic alterations found in human cancers and are often associated with a poor prognosis.

I'm sorry for any confusion, but "Schizosaccharomyces pombe proteins" is not a medical term or concept. Schizosaccharomyces pombe is a type of single-celled microorganism called a yeast, which is often used as a model organism in scientific research. Proteins are complex molecules that do most of the work in cells and are necessary for the structure, function, and regulation of the body's tissues and organs.

In the context of scientific research, "Schizosaccharomyces pombe proteins" would refer to the specific proteins found in or studied using this particular type of yeast. These proteins may have similarities to human proteins and can be used to help understand basic biological processes, as well as diseases that occur in humans. However, it is important to note that while research using model organisms like Schizosaccharomyces pombe has led to many important discoveries, the findings may not always translate directly to humans.

Chronic brain damage is a condition characterized by long-term, persistent injury to the brain that results in cognitive, physical, and behavioral impairments. It can be caused by various factors such as trauma, hypoxia (lack of oxygen), infection, toxic exposure, or degenerative diseases. The effects of chronic brain damage may not be immediately apparent and can worsen over time, leading to significant disability and reduced quality of life.

The symptoms of chronic brain damage can vary widely depending on the severity and location of the injury. They may include:

* Cognitive impairments such as memory loss, difficulty concentrating, trouble with problem-solving and decision-making, and decreased learning ability
* Motor impairments such as weakness, tremors, poor coordination, and balance problems
* Sensory impairments such as hearing or vision loss, numbness, tingling, or altered sense of touch
* Speech and language difficulties such as aphasia (problems with understanding or producing speech) or dysarthria (slurred or slow speech)
* Behavioral changes such as irritability, mood swings, depression, anxiety, and personality changes

Chronic brain damage can be diagnosed through a combination of medical history, physical examination, neurological evaluation, and imaging studies such as MRI or CT scans. Treatment typically focuses on managing symptoms and maximizing function through rehabilitation therapies such as occupational therapy, speech therapy, and physical therapy. In some cases, medication or surgery may be necessary to address specific symptoms or underlying causes of the brain damage.

DNA fragmentation is the breaking of DNA strands into smaller pieces. This process can occur naturally during apoptosis, or programmed cell death, where the DNA is broken down and packaged into apoptotic bodies to be safely eliminated from the body. However, excessive or abnormal DNA fragmentation can also occur due to various factors such as oxidative stress, exposure to genotoxic agents, or certain medical conditions. This can lead to genetic instability, cellular dysfunction, and increased risk of diseases such as cancer. In the context of reproductive medicine, high levels of DNA fragmentation in sperm cells have been linked to male infertility and poor assisted reproductive technology outcomes.

Lipid peroxidation is a process in which free radicals, such as reactive oxygen species (ROS), steal electrons from lipids containing carbon-carbon double bonds, particularly polyunsaturated fatty acids (PUFAs). This results in the formation of lipid hydroperoxides, which can decompose to form a variety of compounds including reactive carbonyl compounds, aldehydes, and ketones.

Malondialdehyde (MDA) is one such compound that is commonly used as a marker for lipid peroxidation. Lipid peroxidation can cause damage to cell membranes, leading to changes in their fluidity and permeability, and can also result in the modification of proteins and DNA, contributing to cellular dysfunction and ultimately cell death. It is associated with various pathological conditions such as atherosclerosis, neurodegenerative diseases, and cancer.

A base sequence in the context of molecular biology refers to the specific order of nucleotides in a DNA or RNA molecule. In DNA, these nucleotides are adenine (A), guanine (G), cytosine (C), and thymine (T). In RNA, uracil (U) takes the place of thymine. The base sequence contains genetic information that is transcribed into RNA and ultimately translated into proteins. It is the exact order of these bases that determines the genetic code and thus the function of the DNA or RNA molecule.

Antineoplastic agents are a class of drugs used to treat malignant neoplasms or cancer. These agents work by inhibiting the growth and proliferation of cancer cells, either by killing them or preventing their division and replication. Antineoplastic agents can be classified based on their mechanism of action, such as alkylating agents, antimetabolites, topoisomerase inhibitors, mitotic inhibitors, and targeted therapy agents.

Alkylating agents work by adding alkyl groups to DNA, which can cause cross-linking of DNA strands and ultimately lead to cell death. Antimetabolites interfere with the metabolic processes necessary for DNA synthesis and replication, while topoisomerase inhibitors prevent the relaxation of supercoiled DNA during replication. Mitotic inhibitors disrupt the normal functioning of the mitotic spindle, which is essential for cell division. Targeted therapy agents are designed to target specific molecular abnormalities in cancer cells, such as mutated oncogenes or dysregulated signaling pathways.

It's important to note that antineoplastic agents can also affect normal cells and tissues, leading to various side effects such as nausea, vomiting, hair loss, and myelosuppression (suppression of bone marrow function). Therefore, the use of these drugs requires careful monitoring and management of their potential adverse effects.

Cell proliferation is the process by which cells increase in number, typically through the process of cell division. In the context of biology and medicine, it refers to the reproduction of cells that makes up living tissue, allowing growth, maintenance, and repair. It involves several stages including the transition from a phase of quiescence (G0 phase) to an active phase (G1 phase), DNA replication in the S phase, and mitosis or M phase, where the cell divides into two daughter cells.

Abnormal or uncontrolled cell proliferation is a characteristic feature of many diseases, including cancer, where deregulated cell cycle control leads to excessive and unregulated growth of cells, forming tumors that can invade surrounding tissues and metastasize to distant sites in the body.

I apologize for any confusion, but "Schizosaccharomyces" is not a medical term. It is a genus name in the field of microbiology and genetics, referring to a group of budding, tear-shaped yeasts that are widely used as model organisms in scientific research. The most well-known species within this genus is Schizosaccharomyces pombe, which has been extensively studied for its cell cycle regulation, DNA repair mechanisms, and other fundamental biological processes.

If you have any questions related to medical terminology or health-related topics, I would be happy to help.

Genetic transcription is the process by which the information in a strand of DNA is used to create a complementary RNA molecule. This process is the first step in gene expression, where the genetic code in DNA is converted into a form that can be used to produce proteins or functional RNAs.

During transcription, an enzyme called RNA polymerase binds to the DNA template strand and reads the sequence of nucleotide bases. As it moves along the template, it adds complementary RNA nucleotides to the growing RNA chain, creating a single-stranded RNA molecule that is complementary to the DNA template strand. Once transcription is complete, the RNA molecule may undergo further processing before it can be translated into protein or perform its functional role in the cell.

Transcription can be either "constitutive" or "regulated." Constitutive transcription occurs at a relatively constant rate and produces essential proteins that are required for basic cellular functions. Regulated transcription, on the other hand, is subject to control by various intracellular and extracellular signals, allowing cells to respond to changing environmental conditions or developmental cues.

Etoposide is a chemotherapy medication used to treat various types of cancer, including lung cancer, testicular cancer, and certain types of leukemia. It works by inhibiting the activity of an enzyme called topoisomerase II, which is involved in DNA replication and transcription. By doing so, etoposide can interfere with the growth and multiplication of cancer cells.

Etoposide is often administered intravenously in a hospital or clinic setting, although it may also be given orally in some cases. The medication can cause a range of side effects, including nausea, vomiting, hair loss, and an increased risk of infection. It can also have more serious side effects, such as bone marrow suppression, which can lead to anemia, bleeding, and a weakened immune system.

Like all chemotherapy drugs, etoposide is not without risks and should only be used under the close supervision of a qualified healthcare provider. It is important for patients to discuss the potential benefits and risks of this medication with their doctor before starting treatment.

'Gene expression regulation' refers to the processes that control whether, when, and where a particular gene is expressed, meaning the production of a specific protein or functional RNA encoded by that gene. This complex mechanism can be influenced by various factors such as transcription factors, chromatin remodeling, DNA methylation, non-coding RNAs, and post-transcriptional modifications, among others. Proper regulation of gene expression is crucial for normal cellular function, development, and maintaining homeostasis in living organisms. Dysregulation of gene expression can lead to various diseases, including cancer and genetic disorders.

Exodeoxyribonucleases are a type of enzyme that cleave (break) nucleotides from the ends of DNA molecules. They are further classified into 5' exodeoxyribonucleases and 3' exodeoxyribonucleases based on the end of the DNA molecule they act upon.

5' Exodeoxyribonucleases remove nucleotides from the 5' end (phosphate group) of a DNA strand, while 3' exodeoxyribonucleases remove nucleotides from the 3' end (hydroxyl group) of a DNA strand.

These enzymes play important roles in various biological processes such as DNA replication, repair, and degradation. They are also used in molecular biology research for various applications such as DNA sequencing, cloning, and genetic engineering.

C57BL/6 (C57 Black 6) is an inbred strain of laboratory mouse that is widely used in biomedical research. The term "inbred" refers to a strain of animals where matings have been carried out between siblings or other closely related individuals for many generations, resulting in a population that is highly homozygous at most genetic loci.

The C57BL/6 strain was established in 1920 by crossing a female mouse from the dilute brown (DBA) strain with a male mouse from the black strain. The resulting offspring were then interbred for many generations to create the inbred C57BL/6 strain.

C57BL/6 mice are known for their robust health, longevity, and ease of handling, making them a popular choice for researchers. They have been used in a wide range of biomedical research areas, including studies of cancer, immunology, neuroscience, cardiovascular disease, and metabolism.

One of the most notable features of the C57BL/6 strain is its sensitivity to certain genetic modifications, such as the introduction of mutations that lead to obesity or impaired glucose tolerance. This has made it a valuable tool for studying the genetic basis of complex diseases and traits.

Overall, the C57BL/6 inbred mouse strain is an important model organism in biomedical research, providing a valuable resource for understanding the genetic and molecular mechanisms underlying human health and disease.

Linear Energy Transfer (LET) is a concept in radiation physics that describes the amount of energy that is transferred from an ionizing particle to a medium per unit length along its path. It is usually expressed in units of keV/μm (kiloelectron volts per micrometer). High-LET radiations, such as alpha particles and heavy ions, transfer more energy to the medium per unit length than low-LET radiations, such as X-rays and gamma rays. This results in a higher probability of producing dense ionizations and causing biological damage along the particle's path. Therefore, LET is an important factor in determining the relative biological effectiveness (RBE) of different types of radiation.

Ubiquitin is a small protein that is present in all eukaryotic cells and plays a crucial role in the regulation of various cellular processes, such as protein degradation, DNA repair, and stress response. It is involved in marking proteins for destruction by attaching to them, a process known as ubiquitination. This modification can target proteins for degradation by the proteasome, a large protein complex that breaks down unneeded or damaged proteins in the cell. Ubiquitin also has other functions, such as regulating the localization and activity of certain proteins. The ability of ubiquitin to modify many different proteins and play a role in multiple cellular processes makes it an essential player in maintaining cellular homeostasis.

Ubiquitin-protein ligases, also known as E3 ubiquitin ligases, are a group of enzymes that play a crucial role in the ubiquitination process. Ubiquitination is a post-translational modification where ubiquitin molecules are attached to specific target proteins, marking them for degradation by the proteasome or for other regulatory functions.

Ubiquitin-protein ligases catalyze the final step in this process by binding to both the ubiquitin protein and the target protein, facilitating the transfer of ubiquitin from an E2 ubiquitin-conjugating enzyme to the target protein. There are several different types of ubiquitin-protein ligases, each with their own specificity for particular target proteins and regulatory functions.

Ubiquitin-protein ligases have been implicated in various cellular processes such as protein degradation, DNA repair, signal transduction, and regulation of the cell cycle. Dysregulation of ubiquitination has been associated with several diseases, including cancer, neurodegenerative disorders, and inflammatory responses. Therefore, understanding the function and regulation of ubiquitin-protein ligases is an important area of research in biology and medicine.

'Tumor cells, cultured' refers to the process of removing cancerous cells from a tumor and growing them in controlled laboratory conditions. This is typically done by isolating the tumor cells from a patient's tissue sample, then placing them in a nutrient-rich environment that promotes their growth and multiplication.

The resulting cultured tumor cells can be used for various research purposes, including the study of cancer biology, drug development, and toxicity testing. They provide a valuable tool for researchers to better understand the behavior and characteristics of cancer cells outside of the human body, which can lead to the development of more effective cancer treatments.

It is important to note that cultured tumor cells may not always behave exactly the same way as they do in the human body, so findings from cell culture studies must be validated through further research, such as animal models or clinical trials.

Flow cytometry is a medical and research technique used to measure physical and chemical characteristics of cells or particles, one cell at a time, as they flow in a fluid stream through a beam of light. The properties measured include:

* Cell size (light scatter)
* Cell internal complexity (granularity, also light scatter)
* Presence or absence of specific proteins or other molecules on the cell surface or inside the cell (using fluorescent antibodies or other fluorescent probes)

The technique is widely used in cell counting, cell sorting, protein engineering, biomarker discovery and monitoring disease progression, particularly in hematology, immunology, and cancer research.

Chromosomal proteins, non-histone, are a diverse group of proteins that are associated with chromatin, the complex of DNA and histone proteins, but do not have the characteristic structure of histones. These proteins play important roles in various nuclear processes such as DNA replication, transcription, repair, recombination, and chromosome condensation and segregation during cell division. They can be broadly classified into several categories based on their functions, including architectural proteins, enzymes, transcription factors, and structural proteins. Examples of non-histone chromosomal proteins include high mobility group (HMG) proteins, poly(ADP-ribose) polymerases (PARPs), and condensins.

Spermatozoa are the male reproductive cells, or gametes, that are produced in the testes. They are microscopic, flagellated (tail-equipped) cells that are highly specialized for fertilization. A spermatozoon consists of a head, neck, and tail. The head contains the genetic material within the nucleus, covered by a cap-like structure called the acrosome which contains enzymes to help the sperm penetrate the female's egg (ovum). The long, thin tail propels the sperm forward through fluid, such as semen, enabling its journey towards the egg for fertilization.

4-Nitroquinoline-1-oxide is a chemical compound that is often used in laboratory research as a carcinogenic agent. Its molecular formula is C6H4N2O3, and it is known to cause DNA damage and mutations, which can lead to the development of cancer. It is primarily used in scientific research to study the mechanisms of carcinogenesis and to test the effectiveness of potential cancer treatments.

It is important to note that 4-Nitroquinoline-1-oxide is not a medication or a treatment for any medical condition, and it should only be handled by trained professionals in a controlled laboratory setting.

In situ nick-end labeling (ISEL, also known as TUNEL) is a technique used in pathology and molecular biology to detect DNA fragmentation, which is a characteristic of apoptotic cells (cells undergoing programmed cell death). The method involves labeling the 3'-hydroxyl termini of double or single stranded DNA breaks in situ (within tissue sections or individual cells) using modified nucleotides that are coupled to a detectable marker, such as a fluorophore or an enzyme. This technique allows for the direct visualization and quantification of apoptotic cells within complex tissues or cell populations.

Sister chromatid exchange (SCE) is a type of genetic recombination that takes place between two identical sister chromatids during the DNA repair process in meiosis or mitosis. It results in an exchange of genetic material between the two chromatids, creating a new combination of genes on each chromatid. This event is a normal part of cell division and helps to increase genetic variability within a population. However, an increased rate of SCEs can also be indicative of exposure to certain genotoxic agents or conditions that cause DNA damage.

Gene deletion is a type of mutation where a segment of DNA, containing one or more genes, is permanently lost or removed from a chromosome. This can occur due to various genetic mechanisms such as homologous recombination, non-homologous end joining, or other types of genomic rearrangements.

The deletion of a gene can have varying effects on the organism, depending on the function of the deleted gene and its importance for normal physiological processes. If the deleted gene is essential for survival, the deletion may result in embryonic lethality or developmental abnormalities. However, if the gene is non-essential or has redundant functions, the deletion may not have any noticeable effects on the organism's phenotype.

Gene deletions can also be used as a tool in genetic research to study the function of specific genes and their role in various biological processes. For example, researchers may use gene deletion techniques to create genetically modified animal models to investigate the impact of gene deletion on disease progression or development.

Zinostatin is not a widely recognized or commonly used term in medicine. However, it appears to be a brand name for a formulation of the anti-cancer drug Neocarzinostatin (NCS). Neocarzinostatin is a protein produced by the bacterium Streptomyces carzinostaticus and has been studied for its potential to inhibit the growth of various types of cancer cells.

Zinostatin is specifically used in the treatment of hepatocellular carcinoma (HCC), which is a type of liver cancer. It is administered via arterial infusion, where the drug is delivered directly into the hepatic artery that supplies blood to the liver. This method allows for higher concentrations of the drug to reach the tumor site while minimizing systemic exposure and potential side effects.

It's important to note that medical terminology can vary by region and context, so it's possible that "Zinostatin" may not be a term used in all medical communities or for all purposes. Always consult with a healthcare professional or trusted medical source for accurate information.

Single-stranded DNA (ssDNA) is a form of DNA that consists of a single polynucleotide chain. In contrast, double-stranded DNA (dsDNA) consists of two complementary polynucleotide chains that are held together by hydrogen bonds.

In the double-helix structure of dsDNA, each nucleotide base on one strand pairs with a specific base on the other strand through hydrogen bonding: adenine (A) with thymine (T), and guanine (G) with cytosine (C). This base pairing provides stability to the double-stranded structure.

Single-stranded DNA, on the other hand, lacks this complementary base pairing and is therefore less stable than dsDNA. However, ssDNA can still form secondary structures through intrastrand base pairing, such as hairpin loops or cruciform structures.

Single-stranded DNA is found in various biological contexts, including viral genomes, transcription bubbles during gene expression, and in certain types of genetic recombination. It also plays a critical role in some laboratory techniques, such as polymerase chain reaction (PCR) and DNA sequencing.

Transcription factors are proteins that play a crucial role in regulating gene expression by controlling the transcription of DNA to messenger RNA (mRNA). They function by binding to specific DNA sequences, known as response elements, located in the promoter region or enhancer regions of target genes. This binding can either activate or repress the initiation of transcription, depending on the properties and interactions of the particular transcription factor. Transcription factors often act as part of a complex network of regulatory proteins that determine the precise spatiotemporal patterns of gene expression during development, differentiation, and homeostasis in an organism.

An amino acid sequence is the specific order of amino acids in a protein or peptide molecule, formed by the linking of the amino group (-NH2) of one amino acid to the carboxyl group (-COOH) of another amino acid through a peptide bond. The sequence is determined by the genetic code and is unique to each type of protein or peptide. It plays a crucial role in determining the three-dimensional structure and function of proteins.

Mutagenesis is the process by which the genetic material (DNA or RNA) of an organism is changed in a way that can alter its phenotype, or observable traits. These changes, known as mutations, can be caused by various factors such as chemicals, radiation, or viruses. Some mutations may have no effect on the organism, while others can cause harm, including diseases and cancer. Mutagenesis is a crucial area of study in genetics and molecular biology, with implications for understanding evolution, genetic disorders, and the development of new medical treatments.

RecQ helicases are a group of enzymes that belong to the RecQ family, which are named after the E. coli RecQ protein. These helicases play crucial roles in maintaining genomic stability by participating in various DNA metabolic processes such as DNA replication, repair, recombination, and transcription. They are highly conserved across different species, including bacteria, yeast, plants, and mammals.

In humans, there are five RecQ helicases: RECQL1, RECQL4, RECQL5, BLM (RecQ-like helicase), and WRN (Werner syndrome ATP-dependent helicase). Defects in these proteins have been linked to various genetic disorders. For instance, mutations in the BLM gene cause Bloom's syndrome, while mutations in the WRN gene lead to Werner syndrome, both of which are characterized by genomic instability and increased cancer predisposition.

RecQ helicases possess 3'-5' DNA helicase activity, unwinding double-stranded DNA into single strands, and can also perform other functions like branch migration, strand annealing, and removal of protein-DNA crosslinks. Their roles in DNA metabolism help prevent and resolve DNA damage, maintain proper chromosome segregation during cell division, and ensure the integrity of the genome.

N-Glycosyl hydrolases (or N-glycanases) are a class of enzymes that catalyze the hydrolysis of the glycosidic bond between an N-glycosyl group and an aglycon, which is typically another part of a larger molecule such as a protein or lipid. N-Glycosyl groups refer to carbohydrate moieties attached to an nitrogen atom, usually in the side chain of an amino acid such as asparagine (Asn) in proteins.

N-Glycosyl hydrolases play important roles in various biological processes, including the degradation and processing of glycoproteins, the modification of glycolipids, and the breakdown of complex carbohydrates. These enzymes are widely distributed in nature and have been found in many organisms, from bacteria to humans.

The classification and nomenclature of N-Glycosyl hydrolases are based on the type of glycosidic bond they cleave and the stereochemistry of the reaction they catalyze. They are grouped into different families in the Carbohydrate-Active enZymes (CAZy) database, which provides a comprehensive resource for the study of carbohydrate-active enzymes.

It is worth noting that N-Glycosyl hydrolases can have both beneficial and detrimental effects on human health. For example, they are involved in the normal turnover and degradation of glycoproteins in the body, but they can also contribute to the pathogenesis of certain diseases, such as lysosomal storage disorders, where mutations in N-Glycosyl hydrolases lead to the accumulation of undigested glycoconjugates and cellular damage.

Telomeric Repeat Binding Protein 2 (TRF2) is a protein that binds to the telomeres, which are the repetitive DNA sequences found at the ends of chromosomes. TRF2 plays a crucial role in protecting the telomeres from being recognized as damaged or broken DNA, which could otherwise lead to chromosomal instability and cellular senescence or apoptosis.

TRF2 is a member of the shelterin complex, a group of proteins that bind to and protect telomeres. TRF2 specifically binds to double-stranded TTAGGG repeats in the telomeric DNA through its N-terminal Myb-like DNA binding domain. By binding to the telomeres, TRF2 helps to prevent the activation of the DNA damage response (DDR) pathway and the subsequent activation of p53-dependent cell cycle checkpoints or apoptosis.

TRF2 has also been shown to play a role in regulating the length of telomeres. It can inhibit the activity of telomerase, an enzyme that adds repetitive DNA sequences to the ends of chromosomes, thereby limiting the extension of telomeres. TRF2 can also promote the formation of t-loops, a higher-order structure in which the 3' overhang of the telomere invades the double-stranded telomeric DNA, forming a displacement loop (D-loop). This helps to protect the telomere from being recognized as a double-strand break and degraded by nucleases.

Mutations in TRF2 have been associated with several human diseases, including premature aging disorders such as dyskeratosis congenita and Hoyeraal-Hreidarsson syndrome, as well as cancer.

The term "DNA, neoplasm" is not a standard medical term or concept. DNA refers to deoxyribonucleic acid, which is the genetic material present in the cells of living organisms. A neoplasm, on the other hand, is a tumor or growth of abnormal tissue that can be benign (non-cancerous) or malignant (cancerous).

In some contexts, "DNA, neoplasm" may refer to genetic alterations found in cancer cells. These genetic changes can include mutations, amplifications, deletions, or rearrangements of DNA sequences that contribute to the development and progression of cancer. Identifying these genetic abnormalities can help doctors diagnose and treat certain types of cancer more effectively.

However, it's important to note that "DNA, neoplasm" is not a term that would typically be used in medical reports or research papers without further clarification. If you have any specific questions about DNA changes in cancer cells or neoplasms, I would recommend consulting with a healthcare professional or conducting further research on the topic.

Tertiary protein structure refers to the three-dimensional arrangement of all the elements (polypeptide chains) of a single protein molecule. It is the highest level of structural organization and results from interactions between various side chains (R groups) of the amino acids that make up the protein. These interactions, which include hydrogen bonds, ionic bonds, van der Waals forces, and disulfide bridges, give the protein its unique shape and stability, which in turn determines its function. The tertiary structure of a protein can be stabilized by various factors such as temperature, pH, and the presence of certain ions. Any changes in these factors can lead to denaturation, where the protein loses its tertiary structure and thus its function.

Chromatin assembly and disassembly refer to the processes by which chromatin, the complex of DNA, histone proteins, and other molecules that make up chromosomes, is organized within the nucleus of a eukaryotic cell.

Chromatin assembly refers to the process by which DNA wraps around histone proteins to form nucleosomes, which are then packed together to form higher-order structures. This process is essential for compacting the vast amount of genetic material contained within the cell nucleus and for regulating gene expression. Chromatin assembly is mediated by a variety of protein complexes, including the histone chaperones and ATP-dependent chromatin remodeling enzymes.

Chromatin disassembly, on the other hand, refers to the process by which these higher-order structures are disassembled during cell division, allowing for the equal distribution of genetic material to daughter cells. This process is mediated by phosphorylation of histone proteins by kinases, which leads to the dissociation of nucleosomes and the decondensation of chromatin.

Both Chromatin assembly and disassembly are dynamic and highly regulated processes that play crucial roles in the maintenance of genome stability and the regulation of gene expression.

Antimutagenic agents are substances that prevent or reduce the frequency of mutations in DNA, which can be caused by various factors such as radiation, chemicals, and free radicals. These agents work by preventing the formation of mutations or by repairing the damage already done to the DNA. They can be found naturally in foods, such as antioxidants, or they can be synthesized in a laboratory. Antimutagenic agents have potential use in cancer prevention and treatment, as well as in reducing the negative effects of environmental mutagens.

The S phase cell cycle checkpoints are mechanisms that ensure the accurate and timely progression through the DNA synthesis (S) phase of the eukaryotic cell cycle. These checkpoints monitor the completion of DNA replication and the proper repair of any DNA damage before the cell moves on to the next phase, namely the mitosis (M) phase.

The S phase checkpoint is primarily focused on detecting and responding to DNA damage that may occur during the replication process. When DNA damage is detected, the checkpoint machinery triggers a series of events that lead to the activation of cell cycle arrest, DNA repair pathways, and/or apoptosis (programmed cell death) if the damage is too severe or cannot be repaired.

The primary components of the S phase checkpoint include sensors, transducers, and effectors. The sensors detect DNA damage or stalled replication forks, while the transducers transmit and amplify the signal to activate the effectors. The effectors then bring about cell cycle arrest, allowing time for repair or initiating apoptosis if necessary.

Overall, the S phase cell cycle checkpoints play a crucial role in maintaining genomic stability and preventing the propagation of cells with damaged DNA, which can lead to tumorigenesis and other diseases.

The G1 phase, or Gap 1 phase, is the first phase of the cell cycle, during which the cell grows in size and synthesizes mRNA and proteins in preparation for subsequent steps leading to mitosis. During this phase, the cell also checks its growth and makes sure that it is large enough to proceed through the cell cycle. If the cell is not large enough, it will arrest in the G1 phase until it has grown sufficiently. The G1 phase is followed by the S phase, during which DNA replication occurs.

Cisplatin is a chemotherapeutic agent used to treat various types of cancers, including testicular, ovarian, bladder, head and neck, lung, and cervical cancers. It is an inorganic platinum compound that contains a central platinum atom surrounded by two chloride atoms and two ammonia molecules in a cis configuration.

Cisplatin works by forming crosslinks between DNA strands, which disrupts the structure of DNA and prevents cancer cells from replicating. This ultimately leads to cell death and slows down or stops the growth of tumors. However, cisplatin can also cause damage to normal cells, leading to side effects such as nausea, vomiting, hearing loss, and kidney damage. Therefore, it is essential to monitor patients closely during treatment and manage any adverse effects promptly.

Free radicals are molecules or atoms that have one or more unpaired electrons in their outermost shell, making them highly reactive. They can be formed naturally in the body through processes such as metabolism and exercise, or they can come from external sources like pollution, radiation, and certain chemicals. Free radicals can cause damage to cells and contribute to the development of various diseases, including cancer, cardiovascular disease, and neurodegenerative disorders. Antioxidants are substances that can neutralize free radicals and help protect against their harmful effects.

Xeroderma Pigmentosum (XP) is a rare, genetic disorder that affects the body's ability to repair damage to DNA caused by ultraviolet (UV) radiation from sunlight. The condition results in extreme sensitivity to UV light. People with XP develop freckles and moles on sun-exposed skin at an early age, and are prone to developing various forms of skin cancer. In severe cases, the disease may also affect the eyes and nervous system.

The disorder is caused by mutations in genes that are responsible for repairing damaged DNA. If not diagnosed and managed properly, XP can lead to serious health complications, including disability and death. Treatment typically involves strict sun protection measures, such as avoiding sunlight, using sunscreen, wearing protective clothing, and in some cases, medication or surgery.

Chromosome aberrations refer to structural and numerical changes in the chromosomes that can occur spontaneously or as a result of exposure to mutagenic agents. These changes can affect the genetic material encoded in the chromosomes, leading to various consequences such as developmental abnormalities, cancer, or infertility.

Structural aberrations include deletions, duplications, inversions, translocations, and rings, which result from breaks and rearrangements of chromosome segments. Numerical aberrations involve changes in the number of chromosomes, such as aneuploidy (extra or missing chromosomes) or polyploidy (multiples of a complete set of chromosomes).

Chromosome aberrations can be detected and analyzed using various cytogenetic techniques, including karyotyping, fluorescence in situ hybridization (FISH), and comparative genomic hybridization (CGH). These methods allow for the identification and characterization of chromosomal changes at the molecular level, providing valuable information for genetic counseling, diagnosis, and research.

Medical Definition:

Superoxide dismutase (SOD) is an enzyme that catalyzes the dismutation of superoxide radicals (O2-) into oxygen (O2) and hydrogen peroxide (H2O2). This essential antioxidant defense mechanism helps protect the body's cells from damage caused by reactive oxygen species (ROS), which are produced during normal metabolic processes and can lead to oxidative stress when their levels become too high.

There are three main types of superoxide dismutase found in different cellular locations:
1. Copper-zinc superoxide dismutase (CuZnSOD or SOD1) - Present mainly in the cytoplasm of cells.
2. Manganese superoxide dismutase (MnSOD or SOD2) - Located within the mitochondrial matrix.
3. Extracellular superoxide dismutase (EcSOD or SOD3) - Found in the extracellular spaces, such as blood vessels and connective tissues.

Imbalances in SOD levels or activity have been linked to various pathological conditions, including neurodegenerative diseases, cancer, and aging-related disorders.

Fluorescence microscopy is a type of microscopy that uses fluorescent dyes or proteins to highlight and visualize specific components within a sample. In this technique, the sample is illuminated with high-energy light, typically ultraviolet (UV) or blue light, which excites the fluorescent molecules causing them to emit lower-energy, longer-wavelength light, usually visible light in the form of various colors. This emitted light is then collected by the microscope and detected to produce an image.

Fluorescence microscopy has several advantages over traditional brightfield microscopy, including the ability to visualize specific structures or molecules within a complex sample, increased sensitivity, and the potential for quantitative analysis. It is widely used in various fields of biology and medicine, such as cell biology, neuroscience, and pathology, to study the structure, function, and interactions of cells and proteins.

There are several types of fluorescence microscopy techniques, including widefield fluorescence microscopy, confocal microscopy, two-photon microscopy, and total internal reflection fluorescence (TIRF) microscopy, each with its own strengths and limitations. These techniques can provide valuable insights into the behavior of cells and proteins in health and disease.

Enzyme activation refers to the process by which an enzyme becomes biologically active and capable of carrying out its specific chemical or biological reaction. This is often achieved through various post-translational modifications, such as proteolytic cleavage, phosphorylation, or addition of cofactors or prosthetic groups to the enzyme molecule. These modifications can change the conformation or structure of the enzyme, exposing or creating a binding site for the substrate and allowing the enzymatic reaction to occur.

For example, in the case of proteolytic cleavage, an inactive precursor enzyme, known as a zymogen, is cleaved into its active form by a specific protease. This is seen in enzymes such as trypsin and chymotrypsin, which are initially produced in the pancreas as inactive precursors called trypsinogen and chymotrypsinogen, respectively. Once they reach the small intestine, they are activated by enteropeptidase, a protease that cleaves a specific peptide bond, releasing the active enzyme.

Phosphorylation is another common mechanism of enzyme activation, where a phosphate group is added to a specific serine, threonine, or tyrosine residue on the enzyme by a protein kinase. This modification can alter the conformation of the enzyme and create a binding site for the substrate, allowing the enzymatic reaction to occur.

Enzyme activation is a crucial process in many biological pathways, as it allows for precise control over when and where specific reactions take place. It also provides a mechanism for regulating enzyme activity in response to various signals and stimuli, such as hormones, neurotransmitters, or changes in the intracellular environment.

DNA-directed DNA polymerase is a type of enzyme that synthesizes new strands of DNA by adding nucleotides to an existing DNA template in a 5' to 3' direction. These enzymes are essential for DNA replication, repair, and recombination. They require a single-stranded DNA template, a primer with a free 3' hydroxyl group, and the four deoxyribonucleoside triphosphates (dNTPs) as substrates to carry out the polymerization reaction.

DNA polymerases also have proofreading activity, which allows them to correct errors that occur during DNA replication by removing mismatched nucleotides and replacing them with the correct ones. This helps ensure the fidelity of the genetic information passed from one generation to the next.

There are several different types of DNA polymerases, each with specific functions and characteristics. For example, DNA polymerase I is involved in both DNA replication and repair, while DNA polymerase III is the primary enzyme responsible for DNA replication in bacteria. In eukaryotic cells, DNA polymerase alpha, beta, gamma, delta, and epsilon have distinct roles in DNA replication, repair, and maintenance.

Animal disease models are specialized animals, typically rodents such as mice or rats, that have been genetically engineered or exposed to certain conditions to develop symptoms and physiological changes similar to those seen in human diseases. These models are used in medical research to study the pathophysiology of diseases, identify potential therapeutic targets, test drug efficacy and safety, and understand disease mechanisms.

The genetic modifications can include knockout or knock-in mutations, transgenic expression of specific genes, or RNA interference techniques. The animals may also be exposed to environmental factors such as chemicals, radiation, or infectious agents to induce the disease state.

Examples of animal disease models include:

1. Mouse models of cancer: Genetically engineered mice that develop various types of tumors, allowing researchers to study cancer initiation, progression, and metastasis.
2. Alzheimer's disease models: Transgenic mice expressing mutant human genes associated with Alzheimer's disease, which exhibit amyloid plaque formation and cognitive decline.
3. Diabetes models: Obese and diabetic mouse strains like the NOD (non-obese diabetic) or db/db mice, used to study the development of type 1 and type 2 diabetes, respectively.
4. Cardiovascular disease models: Atherosclerosis-prone mice, such as ApoE-deficient or LDLR-deficient mice, that develop plaque buildup in their arteries when fed a high-fat diet.
5. Inflammatory bowel disease models: Mice with genetic mutations affecting intestinal barrier function and immune response, such as IL-10 knockout or SAMP1/YitFc mice, which develop colitis.

Animal disease models are essential tools in preclinical research, but it is important to recognize their limitations. Differences between species can affect the translatability of results from animal studies to human patients. Therefore, researchers must carefully consider the choice of model and interpret findings cautiously when applying them to human diseases.

Doxorubicin is a type of chemotherapy medication known as an anthracycline. It works by interfering with the DNA in cancer cells, which prevents them from growing and multiplying. Doxorubicin is used to treat a wide variety of cancers, including leukemia, lymphoma, breast cancer, lung cancer, ovarian cancer, and many others. It may be given alone or in combination with other chemotherapy drugs.

Doxorubicin is usually administered through a vein (intravenously) and can cause side effects such as nausea, vomiting, hair loss, mouth sores, and increased risk of infection. It can also cause damage to the heart muscle, which can lead to heart failure in some cases. For this reason, doctors may monitor patients' heart function closely while they are receiving doxorubicin treatment.

It is important for patients to discuss the potential risks and benefits of doxorubicin therapy with their healthcare provider before starting treatment.

Camptothecin is a topoisomerase I inhibitor, which is a type of chemotherapeutic agent used in cancer treatment. It works by interfering with the function of an enzyme called topoisomerase I, which helps to uncoil DNA during cell division. By inhibiting this enzyme, camptothecin prevents the cancer cells from dividing and growing, ultimately leading to their death.

Camptothecin is found naturally in the bark and stem of the Camptotheca acuminata tree, also known as the "happy tree," which is native to China. It was first isolated in 1966 and has since been developed into several synthetic derivatives, including irinotecan and topotecan, which are used clinically to treat various types of cancer, such as colon, lung, and ovarian cancers.

Like other chemotherapeutic agents, camptothecin can have significant side effects, including nausea, vomiting, diarrhea, and myelosuppression (suppression of bone marrow function). It is important for patients receiving camptothecin-based therapies to be closely monitored by their healthcare team to manage these side effects effectively.

Free radical scavengers, also known as antioxidants, are substances that neutralize or stabilize free radicals. Free radicals are highly reactive atoms or molecules with unpaired electrons, capable of causing damage to cells and tissues in the body through a process called oxidative stress. Antioxidants donate an electron to the free radical, thereby neutralizing it and preventing it from causing further damage. They can be found naturally in foods such as fruits, vegetables, and nuts, or they can be synthesized and used as dietary supplements. Examples of antioxidants include vitamins C and E, beta-carotene, and selenium.

Transfection is a term used in molecular biology that refers to the process of deliberately introducing foreign genetic material (DNA, RNA or artificial gene constructs) into cells. This is typically done using chemical or physical methods, such as lipofection or electroporation. Transfection is widely used in research and medical settings for various purposes, including studying gene function, producing proteins, developing gene therapies, and creating genetically modified organisms. It's important to note that transfection is different from transduction, which is the process of introducing genetic material into cells using viruses as vectors.

Antibiotics are a type of medication used to treat infections caused by bacteria. They work by either killing the bacteria or inhibiting their growth.

Antineoplastics, also known as chemotherapeutic agents, are a class of drugs used to treat cancer. These medications target and destroy rapidly dividing cells, such as cancer cells, although they can also affect other quickly dividing cells in the body, such as those in the hair follicles or digestive tract, which can lead to side effects.

Antibiotics and antineoplastics are two different classes of drugs with distinct mechanisms of action and uses. It is important to use them appropriately and under the guidance of a healthcare professional.

Neoplasms are abnormal growths of cells or tissues in the body that serve no physiological function. They can be benign (non-cancerous) or malignant (cancerous). Benign neoplasms are typically slow growing and do not spread to other parts of the body, while malignant neoplasms are aggressive, invasive, and can metastasize to distant sites.

Neoplasms occur when there is a dysregulation in the normal process of cell division and differentiation, leading to uncontrolled growth and accumulation of cells. This can result from genetic mutations or other factors such as viral infections, environmental exposures, or hormonal imbalances.

Neoplasms can develop in any organ or tissue of the body and can cause various symptoms depending on their size, location, and type. Treatment options for neoplasms include surgery, radiation therapy, chemotherapy, immunotherapy, and targeted therapy, among others.

Radiation-protective agents, also known as radioprotectors, are substances that help in providing protection against the harmful effects of ionizing radiation. They can be used to prevent or reduce damage to biological tissues, including DNA, caused by exposure to radiation. These agents work through various mechanisms such as scavenging free radicals, modulating cellular responses to radiation-induced damage, and enhancing DNA repair processes.

Radiation-protective agents can be categorized into two main groups:

1. Radiosensitizers: These are substances that make cancer cells more sensitive to the effects of radiation therapy, increasing their susceptibility to damage and potentially improving treatment outcomes. However, radiosensitizers do not provide protection to normal tissues against radiation exposure.

2. Radioprotectors: These agents protect both normal and cancerous cells from radiation-induced damage. They can be further divided into two categories: direct and indirect radioprotectors. Direct radioprotectors interact directly with radiation, absorbing or scattering it away from sensitive tissues. Indirect radioprotectors work by neutralizing free radicals and reactive oxygen species generated during radiation exposure, which would otherwise cause damage to cellular structures and DNA.

Examples of radiation-protective agents include antioxidants like vitamins C and E, chemical compounds such as amifostine and cysteamine, and various natural substances found in plants and foods. It is important to note that while some radiation-protective agents have shown promise in preclinical studies, their efficacy and safety in humans require further investigation before they can be widely used in clinical settings.

Glutathione is a tripeptide composed of three amino acids: cysteine, glutamic acid, and glycine. It is a vital antioxidant that plays an essential role in maintaining cellular health and function. Glutathione helps protect cells from oxidative stress by neutralizing free radicals, which are unstable molecules that can damage cells and contribute to aging and diseases such as cancer, heart disease, and dementia. It also supports the immune system, detoxifies harmful substances, and regulates various cellular processes, including DNA synthesis and repair.

Glutathione is found in every cell of the body, with particularly high concentrations in the liver, lungs, and eyes. The body can produce its own glutathione, but levels may decline with age, illness, or exposure to toxins. As such, maintaining optimal glutathione levels through diet, supplementation, or other means is essential for overall health and well-being.

Fanconi Anemia Complementation Group D2 Protein, also known as FANCD2 protein, is a key player in the Fanconi anemia (FA) pathway, which is a DNA repair pathway that helps to maintain genomic stability. The FA pathway is responsible for the repair of DNA interstrand cross-links (ICLs), which are harmful lesions that can lead to genomic instability and cancer.

FANCD2 protein is part of the E3 ubiquitin ligase complex that monoubiquitinates FANCI protein, forming a heterodimeric complex known as ID2. The monoubiquitination of FANCD2/FANCI is a critical step in the FA pathway and is required for the recruitment of downstream repair factors to the site of DNA damage.

Mutations in the gene that encodes FANCD2 protein can lead to Fanconi anemia, a rare genetic disorder characterized by bone marrow failure, congenital abnormalities, and an increased risk of cancer. The disease is typically inherited in an autosomal recessive manner, meaning that an individual must inherit two copies of the mutated gene (one from each parent) to develop the condition.

Catalase is a type of enzyme that is found in many living organisms, including humans. Its primary function is to catalyze the decomposition of hydrogen peroxide (H2O2) into water (H2O) and oxygen (O2). This reaction helps protect cells from the harmful effects of hydrogen peroxide, which can be toxic at high concentrations.

The chemical reaction catalyzed by catalase can be represented as follows:

H2O2 + Catalase → H2O + O2 + Catalase

Catalase is a powerful antioxidant enzyme that plays an important role in protecting cells from oxidative damage. It is found in high concentrations in tissues that produce or are exposed to hydrogen peroxide, such as the liver, kidneys, and erythrocytes (red blood cells).

Deficiency in catalase activity has been linked to several diseases, including cancer, neurodegenerative disorders, and aging. On the other hand, overexpression of catalase has been shown to have potential therapeutic benefits in various disease models, such as reducing inflammation and oxidative stress.

Enzyme inhibitors are substances that bind to an enzyme and decrease its activity, preventing it from catalyzing a chemical reaction in the body. They can work by several mechanisms, including blocking the active site where the substrate binds, or binding to another site on the enzyme to change its shape and prevent substrate binding. Enzyme inhibitors are often used as drugs to treat various medical conditions, such as high blood pressure, abnormal heart rhythms, and bacterial infections. They can also be found naturally in some foods and plants, and can be used in research to understand enzyme function and regulation.

Cell division is the process by which a single eukaryotic cell (a cell with a true nucleus) divides into two identical daughter cells. This complex process involves several stages, including replication of DNA, separation of chromosomes, and division of the cytoplasm. There are two main types of cell division: mitosis and meiosis.

Mitosis is the type of cell division that results in two genetically identical daughter cells. It is a fundamental process for growth, development, and tissue repair in multicellular organisms. The stages of mitosis include prophase, prometaphase, metaphase, anaphase, and telophase, followed by cytokinesis, which divides the cytoplasm.

Meiosis, on the other hand, is a type of cell division that occurs in the gonads (ovaries and testes) during the production of gametes (sex cells). Meiosis results in four genetically unique daughter cells, each with half the number of chromosomes as the parent cell. This process is essential for sexual reproduction and genetic diversity. The stages of meiosis include meiosis I and meiosis II, which are further divided into prophase, prometaphase, metaphase, anaphase, and telophase.

In summary, cell division is the process by which a single cell divides into two daughter cells, either through mitosis or meiosis. This process is critical for growth, development, tissue repair, and sexual reproduction in multicellular organisms.

Topoisomerase I inhibitors are a class of anticancer drugs that work by inhibiting the function of topoisomerase I, an enzyme that plays a crucial role in the relaxation and replication of DNA. By inhibiting this enzyme's activity, these drugs interfere with the normal unwinding and separation of DNA strands, leading to DNA damage and ultimately cell death. Topoisomerase I inhibitors are used in the treatment of various types of cancer, including colon, small cell lung, ovarian, and cervical cancers. Examples of topoisomerase I inhibitors include camptothecin, irinotecan, and topotecan.

Chromosomal instability is a term used in genetics to describe a type of genetic alteration where there are abnormalities in the number or structure of chromosomes within cells. Chromosomes are thread-like structures that contain our genetic material, and they usually exist in pairs in the nucleus of a cell.

Chromosomal instability can arise due to various factors, including errors in DNA replication or repair, problems during cell division, or exposure to environmental mutagens. This instability can lead to an increased frequency of chromosomal abnormalities, such as deletions, duplications, translocations, or changes in the number of chromosomes.

Chromosomal instability is associated with several human diseases, including cancer. In cancer cells, chromosomal instability can contribute to tumor heterogeneity, drug resistance, and disease progression. It is also observed in certain genetic disorders, such as Down syndrome, where an extra copy of chromosome 21 is present, and in some rare inherited syndromes, such as Bloom syndrome and Fanconi anemia, which are characterized by a high risk of cancer and other health problems.

Acetylation is a chemical process that involves the addition of an acetyl group (-COCH3) to a molecule. In the context of medical biochemistry, acetylation often refers to the post-translational modification of proteins, where an acetyl group is added to the amino group of a lysine residue in a protein by an enzyme called acetyltransferase. This modification can alter the function or stability of the protein and plays a crucial role in regulating various cellular processes such as gene expression, DNA repair, and cell signaling. Acetylation can also occur on other types of molecules, including lipids and carbohydrates, and has important implications for drug metabolism and toxicity.

Mitochondrial DNA (mtDNA) is the genetic material present in the mitochondria, which are specialized structures within cells that generate energy. Unlike nuclear DNA, which is present in the cell nucleus and inherited from both parents, mtDNA is inherited solely from the mother.

MtDNA is a circular molecule that contains 37 genes, including 13 genes that encode for proteins involved in oxidative phosphorylation, a process that generates energy in the form of ATP. The remaining genes encode for rRNAs and tRNAs, which are necessary for protein synthesis within the mitochondria.

Mutations in mtDNA can lead to a variety of genetic disorders, including mitochondrial diseases, which can affect any organ system in the body. These mutations can also be used in forensic science to identify individuals and establish biological relationships.

CDC2 protein kinase, also known as cell division cycle 2 or CDK1, is a type of enzyme that plays a crucial role in the regulation of the cell cycle. The cell cycle is the series of events that cells undergo as they grow, replicate their DNA, and divide into two daughter cells.

CDC2 protein kinase is a member of the cyclin-dependent kinase (CDK) family, which are serine/threonine protein kinases that are activated by binding to regulatory subunits called cyclins. CDC2 protein kinase is primarily associated with the regulation of the G2 phase and the entry into mitosis, the stage of the cell cycle where nuclear and cytoplasmic division occur.

CDC2 protein kinase functions by phosphorylating various target proteins, which alters their activity and contributes to the coordination of the different events that occur during the cell cycle. The activity of CDC2 protein kinase is tightly regulated through a variety of mechanisms, including phosphorylation and dephosphorylation, as well as the binding and destruction of cyclin subunits.

Dysregulation of CDC2 protein kinase has been implicated in various human diseases, including cancer, where uncontrolled cell division can lead to the formation of tumors. Therefore, understanding the regulation and function of CDC2 protein kinase is an important area of research in molecular biology and medicine.

Premature aging, also known as "accelerated aging" or "early aging," refers to the physiological process in which the body shows signs of aging at an earlier age than typically expected. This can include various symptoms such as wrinkles, graying hair, decreased energy and mobility, cognitive decline, and increased risk of chronic diseases.

The medical definition of premature aging is not well-established, as aging is a complex process influenced by a variety of genetic and environmental factors. However, certain conditions and syndromes are associated with premature aging, such as Hutchinson-Gilford progeria syndrome, Werner syndrome, and Down syndrome.

In general, the signs of premature aging may be caused by a combination of genetic predisposition, lifestyle factors (such as smoking, alcohol consumption, and poor diet), exposure to environmental toxins, and chronic stress. While some aspects of aging are inevitable, maintaining a healthy lifestyle and reducing exposure to harmful factors can help slow down the aging process and improve overall quality of life.

Messenger RNA (mRNA) is a type of RNA (ribonucleic acid) that carries genetic information copied from DNA in the form of a series of three-base code "words," each of which specifies a particular amino acid. This information is used by the cell's machinery to construct proteins, a process known as translation. After being transcribed from DNA, mRNA travels out of the nucleus to the ribosomes in the cytoplasm where protein synthesis occurs. Once the protein has been synthesized, the mRNA may be degraded and recycled. Post-transcriptional modifications can also occur to mRNA, such as alternative splicing and addition of a 5' cap and a poly(A) tail, which can affect its stability, localization, and translation efficiency.

Methylnitronitrosoguanidine (MNNG) is not typically referred to as a medical term, but it is a chemical compound with potential implications in medical research and toxicology. Therefore, I will provide you with a general definition of this compound.

Methylnitronitrosoguanidine (C2H6N4O2), also known as MNNG or nitroso-guanidine, is a nitrosamine compound used primarily in laboratory research. It is an alkylating agent, which means it can introduce alkyl groups into other molecules through chemical reactions. In this case, MNNG is particularly reactive towards DNA and RNA, making it a potent mutagen and carcinogen.

MNNG has been used in research to study the mechanisms of carcinogenesis (the development of cancer) and mutations at the molecular level. However, due to its high toxicity and potential for causing damage to genetic material, its use is strictly regulated and typically limited to laboratory settings.

Mitochondria are specialized structures located inside cells that convert the energy from food into ATP (adenosine triphosphate), which is the primary form of energy used by cells. They are often referred to as the "powerhouses" of the cell because they generate most of the cell's supply of chemical energy. Mitochondria are also involved in various other cellular processes, such as signaling, differentiation, and apoptosis (programmed cell death).

Mitochondria have their own DNA, known as mitochondrial DNA (mtDNA), which is inherited maternally. This means that mtDNA is passed down from the mother to her offspring through the egg cells. Mitochondrial dysfunction has been linked to a variety of diseases and conditions, including neurodegenerative disorders, diabetes, and aging.

Down-regulation is a process that occurs in response to various stimuli, where the number or sensitivity of cell surface receptors or the expression of specific genes is decreased. This process helps maintain homeostasis within cells and tissues by reducing the ability of cells to respond to certain signals or molecules.

In the context of cell surface receptors, down-regulation can occur through several mechanisms:

1. Receptor internalization: After binding to their ligands, receptors can be internalized into the cell through endocytosis. Once inside the cell, these receptors may be degraded or recycled back to the cell surface in smaller numbers.
2. Reduced receptor synthesis: Down-regulation can also occur at the transcriptional level, where the expression of genes encoding for specific receptors is decreased, leading to fewer receptors being produced.
3. Receptor desensitization: Prolonged exposure to a ligand can lead to a decrease in receptor sensitivity or affinity, making it more difficult for the cell to respond to the signal.

In the context of gene expression, down-regulation refers to the decreased transcription and/or stability of specific mRNAs, leading to reduced protein levels. This process can be induced by various factors, including microRNA (miRNA)-mediated regulation, histone modification, or DNA methylation.

Down-regulation is an essential mechanism in many physiological processes and can also contribute to the development of several diseases, such as cancer and neurodegenerative disorders.

DNA topoisomerases are enzymes that modify the topological structure of DNA by regulating the number of twists or supercoils in the double helix. There are two main types of DNA topoisomerases: type I and type II.

Type I DNA topoisomerases function by cutting one strand of the DNA duplex, allowing the uncut strand to rotate around the break, and then resealing the break. This process can relieve both positive and negative supercoiling in DNA, as well as introduce single-stranded breaks into the DNA molecule.

Type I topoisomerases are further divided into three subtypes: type IA, type IB, and type IC. These subtypes differ in their mechanism of action and the structure of the active site tyrosine residue that makes the transient break in the DNA strand.

Overall, DNA topoisomerases play a crucial role in many cellular processes involving DNA, including replication, transcription, recombination, and chromosome segregation. Dysregulation of these enzymes has been implicated in various human diseases, including cancer and genetic disorders.

Gene knockdown techniques are methods used to reduce the expression or function of specific genes in order to study their role in biological processes. These techniques typically involve the use of small RNA molecules, such as siRNAs (small interfering RNAs) or shRNAs (short hairpin RNAs), which bind to and promote the degradation of complementary mRNA transcripts. This results in a decrease in the production of the protein encoded by the targeted gene.

Gene knockdown techniques are often used as an alternative to traditional gene knockout methods, which involve completely removing or disrupting the function of a gene. Knockdown techniques allow for more subtle and reversible manipulation of gene expression, making them useful for studying genes that are essential for cell survival or have redundant functions.

These techniques are widely used in molecular biology research to investigate gene function, genetic interactions, and disease mechanisms. However, it is important to note that gene knockdown can have off-target effects and may not completely eliminate the expression of the targeted gene, so results should be interpreted with caution.

Fungal proteins are a type of protein that is specifically produced and present in fungi, which are a group of eukaryotic organisms that include microorganisms such as yeasts and molds. These proteins play various roles in the growth, development, and survival of fungi. They can be involved in the structure and function of fungal cells, metabolism, pathogenesis, and other cellular processes. Some fungal proteins can also have important implications for human health, both in terms of their potential use as therapeutic targets and as allergens or toxins that can cause disease.

Fungal proteins can be classified into different categories based on their functions, such as enzymes, structural proteins, signaling proteins, and toxins. Enzymes are proteins that catalyze chemical reactions in fungal cells, while structural proteins provide support and protection for the cell. Signaling proteins are involved in communication between cells and regulation of various cellular processes, and toxins are proteins that can cause harm to other organisms, including humans.

Understanding the structure and function of fungal proteins is important for developing new treatments for fungal infections, as well as for understanding the basic biology of fungi. Research on fungal proteins has led to the development of several antifungal drugs that target specific fungal enzymes or other proteins, providing effective treatment options for a range of fungal diseases. Additionally, further study of fungal proteins may reveal new targets for drug development and help improve our ability to diagnose and treat fungal infections.

Serine is an amino acid, which is a building block of proteins. More specifically, it is a non-essential amino acid, meaning that the body can produce it from other compounds, and it does not need to be obtained through diet. Serine plays important roles in the body, such as contributing to the formation of the protective covering of nerve fibers (myelin sheath), helping to synthesize another amino acid called tryptophan, and taking part in the metabolism of fatty acids. It is also involved in the production of muscle tissues, the immune system, and the forming of cell structures. Serine can be found in various foods such as soy, eggs, cheese, meat, peanuts, lentils, and many others.

Rad52 is a DNA repair and recombination protein that plays a crucial role in the maintenance of genomic stability in cells. It is highly conserved across various species, including yeast, humans, and other mammals. The primary function of Rad52 is to facilitate the process of homologous recombination (HR), which is a critical DNA repair mechanism that helps to maintain the integrity of the genetic material in the event of double-strand breaks (DSBs) or other types of DNA damage.

Rad52 has several essential roles in HR:

1. Rad52 promotes the formation of ssDNA-Rad51 nucleoprotein filaments: Rad52 interacts with single-stranded DNA (ssDNA) generated during resection of DSBs, facilitating the recruitment and loading of the Rad51 recombinase onto the ssDNA. This Rad51-ssDNA nucleoprotein filament formation is a key step in HR, as it enables the search for homologous sequences and subsequent strand invasion.

2. Rad52 mediates DNA annealing: Rad52 can catalyze the annealing of complementary ssDNA molecules, promoting the reannealing of invaded strands during HR or facilitating the pairing of RPA-coated ssDNA with homologous duplex DNA.

3. Rad52 stimulates D-loop formation and extension: Rad52 can stimulate the extension of D-loops, which are three-stranded structures formed when a single-stranded DNA invades a double-stranded DNA molecule during HR. This process is essential for the subsequent steps of homology search and strand exchange.

4. Rad52 facilitates RPA displacement: Rad52 can displace replication protein A (RPA) from ssDNA, allowing Rad51 to bind and form nucleoprotein filaments. This is a critical step in HR, as RPA inhibits Rad51 binding to ssDNA.

5. Rad52 interacts with other DNA repair proteins: Rad52 interacts with various DNA repair proteins, including BRCA1, BRCA2, and the single-strand binding protein RPA, to coordinate HR and other DNA repair pathways.

In summary, Rad52 is a crucial player in homologous recombination (HR) and DNA damage response. It functions as a mediator of DNA annealing, D-loop formation, and RPA displacement, promoting efficient HR and maintaining genome stability.

HEK293 cells, also known as human embryonic kidney 293 cells, are a line of cells used in scientific research. They were originally derived from human embryonic kidney cells and have been adapted to grow in a lab setting. HEK293 cells are widely used in molecular biology and biochemistry because they can be easily transfected (a process by which DNA is introduced into cells) and highly express foreign genes. As a result, they are often used to produce proteins for structural and functional studies. It's important to note that while HEK293 cells are derived from human tissue, they have been grown in the lab for many generations and do not retain the characteristics of the original embryonic kidney cells.

Carrier proteins, also known as transport proteins, are a type of protein that facilitates the movement of molecules across cell membranes. They are responsible for the selective and active transport of ions, sugars, amino acids, and other molecules from one side of the membrane to the other, against their concentration gradient. This process requires energy, usually in the form of ATP (adenosine triphosphate).

Carrier proteins have a specific binding site for the molecule they transport, and undergo conformational changes upon binding, which allows them to move the molecule across the membrane. Once the molecule has been transported, the carrier protein returns to its original conformation, ready to bind and transport another molecule.

Carrier proteins play a crucial role in maintaining the balance of ions and other molecules inside and outside of cells, and are essential for many physiological processes, including nerve impulse transmission, muscle contraction, and nutrient uptake.

Post-translational protein processing refers to the modifications and changes that proteins undergo after their synthesis on ribosomes, which are complex molecular machines responsible for protein synthesis. These modifications occur through various biochemical processes and play a crucial role in determining the final structure, function, and stability of the protein.

The process begins with the translation of messenger RNA (mRNA) into a linear polypeptide chain, which is then subjected to several post-translational modifications. These modifications can include:

1. Proteolytic cleavage: The removal of specific segments or domains from the polypeptide chain by proteases, resulting in the formation of mature, functional protein subunits.
2. Chemical modifications: Addition or modification of chemical groups to the side chains of amino acids, such as phosphorylation (addition of a phosphate group), glycosylation (addition of sugar moieties), methylation (addition of a methyl group), acetylation (addition of an acetyl group), and ubiquitination (addition of a ubiquitin protein).
3. Disulfide bond formation: The oxidation of specific cysteine residues within the polypeptide chain, leading to the formation of disulfide bonds between them. This process helps stabilize the three-dimensional structure of proteins, particularly in extracellular environments.
4. Folding and assembly: The acquisition of a specific three-dimensional conformation by the polypeptide chain, which is essential for its function. Chaperone proteins assist in this process to ensure proper folding and prevent aggregation.
5. Protein targeting: The directed transport of proteins to their appropriate cellular locations, such as the nucleus, mitochondria, endoplasmic reticulum, or plasma membrane. This is often facilitated by specific signal sequences within the protein that are recognized and bound by transport machinery.

Collectively, these post-translational modifications contribute to the functional diversity of proteins in living organisms, allowing them to perform a wide range of cellular processes, including signaling, catalysis, regulation, and structural support.

Fanconi anemia is a rare, inherited disorder that affects the body's ability to produce healthy blood cells. It is characterized by bone marrow failure, congenital abnormalities, and an increased risk of developing certain types of cancer. The condition is caused by mutations in genes responsible for repairing damaged DNA, leading to chromosomal instability and cell death.

The classic form of Fanconi anemia (type A) is typically diagnosed in childhood and is associated with various physical abnormalities such as short stature, skin pigmentation changes, thumb and radial ray anomalies, kidney and genitourinary malformations, and developmental delays. Other types of Fanconi anemia (B-G) may have different clinical presentations but share the common feature of bone marrow failure and cancer predisposition.

Bone marrow failure in Fanconi anemia results in decreased production of all three types of blood cells: red blood cells, white blood cells, and platelets. This can lead to anemia (low red blood cell count), neutropenia (low white blood cell count), and thrombocytopenia (low platelet count). These conditions increase the risk of infections, fatigue, and bleeding.

Individuals with Fanconi anemia have a significantly higher risk of developing various types of cancer, particularly acute myeloid leukemia (AML) and solid tumors such as squamous cell carcinomas of the head, neck, esophagus, and anogenital region.

Treatment for Fanconi anemia typically involves managing symptoms related to bone marrow failure, such as transfusions, growth factors, and antibiotics. Hematopoietic stem cell transplantation (HSCT) is the only curative treatment option for bone marrow failure but carries risks of its own, including graft-versus-host disease and transplant-related mortality. Regular cancer surveillance is essential due to the increased risk of malignancies in these patients.

Immunoblotting, also known as western blotting, is a laboratory technique used in molecular biology and immunogenetics to detect and quantify specific proteins in a complex mixture. This technique combines the electrophoretic separation of proteins by gel electrophoresis with their detection using antibodies that recognize specific epitopes (protein fragments) on the target protein.

The process involves several steps: first, the protein sample is separated based on size through sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Next, the separated proteins are transferred onto a nitrocellulose or polyvinylidene fluoride (PVDF) membrane using an electric field. The membrane is then blocked with a blocking agent to prevent non-specific binding of antibodies.

After blocking, the membrane is incubated with a primary antibody that specifically recognizes the target protein. Following this, the membrane is washed to remove unbound primary antibodies and then incubated with a secondary antibody conjugated to an enzyme such as horseradish peroxidase (HRP) or alkaline phosphatase (AP). The enzyme catalyzes a colorimetric or chemiluminescent reaction that allows for the detection of the target protein.

Immunoblotting is widely used in research and clinical settings to study protein expression, post-translational modifications, protein-protein interactions, and disease biomarkers. It provides high specificity and sensitivity, making it a valuable tool for identifying and quantifying proteins in various biological samples.

Topoisomerase inhibitors are a class of anticancer drugs that work by interfering with the function of topoisomerases, which are enzymes responsible for relaxing supercoiled DNA during processes such as replication and transcription. Topoisomerase I inhibitors selectively bind to and stabilize the cleavage complex formed between topoisomerase I and DNA, preventing the relegation of the broken DNA strand and resulting in DNA damage and cell death. Examples include irinotecan and topotecan. Topoisomerase II inhibitors, on the other hand, bind to and stabilize the cleavage complex formed between topoisomerase II and DNA, leading to double-stranded DNA breaks and cell death. Examples include doxorubicin, etoposide, and mitoxantrone. These drugs are used in the treatment of various types of cancer.

A plasmid is a small, circular, double-stranded DNA molecule that is separate from the chromosomal DNA of a bacterium or other organism. Plasmids are typically not essential for the survival of the organism, but they can confer beneficial traits such as antibiotic resistance or the ability to degrade certain types of pollutants.

Plasmids are capable of replicating independently of the chromosomal DNA and can be transferred between bacteria through a process called conjugation. They often contain genes that provide resistance to antibiotics, heavy metals, and other environmental stressors. Plasmids have also been engineered for use in molecular biology as cloning vectors, allowing scientists to replicate and manipulate specific DNA sequences.

Plasmids are important tools in genetic engineering and biotechnology because they can be easily manipulated and transferred between organisms. They have been used to produce vaccines, diagnostic tests, and genetically modified organisms (GMOs) for various applications, including agriculture, medicine, and industry.

The liver is a large, solid organ located in the upper right portion of the abdomen, beneath the diaphragm and above the stomach. It plays a vital role in several bodily functions, including:

1. Metabolism: The liver helps to metabolize carbohydrates, fats, and proteins from the food we eat into energy and nutrients that our bodies can use.
2. Detoxification: The liver detoxifies harmful substances in the body by breaking them down into less toxic forms or excreting them through bile.
3. Synthesis: The liver synthesizes important proteins, such as albumin and clotting factors, that are necessary for proper bodily function.
4. Storage: The liver stores glucose, vitamins, and minerals that can be released when the body needs them.
5. Bile production: The liver produces bile, a digestive juice that helps to break down fats in the small intestine.
6. Immune function: The liver plays a role in the immune system by filtering out bacteria and other harmful substances from the blood.

Overall, the liver is an essential organ that plays a critical role in maintaining overall health and well-being.

Immunohistochemistry (IHC) is a technique used in pathology and laboratory medicine to identify specific proteins or antigens in tissue sections. It combines the principles of immunology and histology to detect the presence and location of these target molecules within cells and tissues. This technique utilizes antibodies that are specific to the protein or antigen of interest, which are then tagged with a detection system such as a chromogen or fluorophore. The stained tissue sections can be examined under a microscope, allowing for the visualization and analysis of the distribution and expression patterns of the target molecule in the context of the tissue architecture. Immunohistochemistry is widely used in diagnostic pathology to help identify various diseases, including cancer, infectious diseases, and immune-mediated disorders.

Radiation-sensitizing agents are drugs that make cancer cells more sensitive to radiation therapy. These agents work by increasing the ability of radiation to damage the DNA of cancer cells, which can lead to more effective tumor cell death. This means that lower doses of radiation may be required to achieve the same therapeutic effect, reducing the potential for damage to normal tissues surrounding the tumor.

Radiation-sensitizing agents are often used in conjunction with radiation therapy to improve treatment outcomes for patients with various types of cancer. They can be given either systemically (through the bloodstream) or locally (directly to the tumor site). The choice of agent and the timing of administration depend on several factors, including the type and stage of cancer, the patient's overall health, and the specific radiation therapy protocol being used.

It is important to note that while radiation-sensitizing agents can enhance the effectiveness of radiation therapy, they may also increase the risk of side effects. Therefore, careful monitoring and management of potential toxicities are essential during treatment.

Sprague-Dawley rats are a strain of albino laboratory rats that are widely used in scientific research. They were first developed by researchers H.H. Sprague and R.C. Dawley in the early 20th century, and have since become one of the most commonly used rat strains in biomedical research due to their relatively large size, ease of handling, and consistent genetic background.

Sprague-Dawley rats are outbred, which means that they are genetically diverse and do not suffer from the same limitations as inbred strains, which can have reduced fertility and increased susceptibility to certain diseases. They are also characterized by their docile nature and low levels of aggression, making them easier to handle and study than some other rat strains.

These rats are used in a wide variety of research areas, including toxicology, pharmacology, nutrition, cancer, and behavioral studies. Because they are genetically diverse, Sprague-Dawley rats can be used to model a range of human diseases and conditions, making them an important tool in the development of new drugs and therapies.

Aging is a complex, progressive and inevitable process of bodily changes over time, characterized by the accumulation of cellular damage and degenerative changes that eventually lead to increased vulnerability to disease and death. It involves various biological, genetic, environmental, and lifestyle factors that contribute to the decline in physical and mental functions. The medical field studies aging through the discipline of gerontology, which aims to understand the underlying mechanisms of aging and develop interventions to promote healthy aging and extend the human healthspan.

A biological marker, often referred to as a biomarker, is a measurable indicator that reflects the presence or severity of a disease state, or a response to a therapeutic intervention. Biomarkers can be found in various materials such as blood, tissues, or bodily fluids, and they can take many forms, including molecular, histologic, radiographic, or physiological measurements.

In the context of medical research and clinical practice, biomarkers are used for a variety of purposes, such as:

1. Diagnosis: Biomarkers can help diagnose a disease by indicating the presence or absence of a particular condition. For example, prostate-specific antigen (PSA) is a biomarker used to detect prostate cancer.
2. Monitoring: Biomarkers can be used to monitor the progression or regression of a disease over time. For instance, hemoglobin A1c (HbA1c) levels are monitored in diabetes patients to assess long-term blood glucose control.
3. Predicting: Biomarkers can help predict the likelihood of developing a particular disease or the risk of a negative outcome. For example, the presence of certain genetic mutations can indicate an increased risk for breast cancer.
4. Response to treatment: Biomarkers can be used to evaluate the effectiveness of a specific treatment by measuring changes in the biomarker levels before and after the intervention. This is particularly useful in personalized medicine, where treatments are tailored to individual patients based on their unique biomarker profiles.

It's important to note that for a biomarker to be considered clinically valid and useful, it must undergo rigorous validation through well-designed studies, including demonstrating sensitivity, specificity, reproducibility, and clinical relevance.

Recombination is a natural process that occurs in cells to exchange genetic information between two similar or identical strands of DNA. This process helps to maintain the stability and diversity of the genome. RecA (RecA protein) is a type of recombinase enzyme found in bacteria, including Escherichia coli, that plays a crucial role in this process.

RecA recombinases are proteins that facilitate the exchange of genetic information between two DNA molecules by promoting homologous pairing and strand exchange. Homologous pairing is the alignment of similar or identical sequences of nucleotides on two different DNA molecules, while strand exchange refers to the physical transfer of one strand of DNA from one molecule to another.

RecA recombinases work by forming a nucleoprotein filament on single-stranded DNA (ssDNA) and then searching for complementary sequences on double-stranded DNA (dsDNA). Once a complementary sequence is found, the RecA protein facilitates the invasion of the ssDNA into the dsDNA, leading to strand exchange and the formation of a joint molecule. This joint molecule can then be used as a template for DNA replication or repair.

RecA recombinases have been extensively studied due to their importance in genetic recombination and DNA repair. They also have potential applications in biotechnology, such as in the development of genome engineering tools and methods for detecting and quantifying specific DNA sequences.

In medical terms, the skin is the largest organ of the human body. It consists of two main layers: the epidermis (outer layer) and dermis (inner layer), as well as accessory structures like hair follicles, sweat glands, and oil glands. The skin plays a crucial role in protecting us from external factors such as bacteria, viruses, and environmental hazards, while also regulating body temperature and enabling the sense of touch.

Alkylation, in the context of medical chemistry and toxicology, refers to the process of introducing an alkyl group (a chemical moiety made up of a carbon atom bonded to one or more hydrogen atoms) into a molecule, typically a biomolecule such as a protein or DNA. This process can occur through various mechanisms, including chemical reactions with alkylating agents.

In the context of cancer therapy, alkylation is used to describe a class of chemotherapeutic drugs known as alkylating agents, which work by introducing alkyl groups onto DNA molecules in rapidly dividing cells. This can lead to cross-linking of DNA strands and other forms of DNA damage, ultimately inhibiting cell division and leading to the death of cancer cells. However, these agents can also affect normal cells, leading to side effects such as nausea, hair loss, and increased risk of infection.

It's worth noting that alkylation can also occur through non-chemical means, such as in certain types of radiation therapy where high-energy particles can transfer energy to electrons in biological molecules, leading to the formation of reactive radicals that can react with and alkylate DNA.

Cricetinae is a subfamily of rodents that includes hamsters, gerbils, and relatives. These small mammals are characterized by having short limbs, compact bodies, and cheek pouches for storing food. They are native to various parts of the world, particularly in Europe, Asia, and Africa. Some species are popular pets due to their small size, easy care, and friendly nature. In a medical context, understanding the biology and behavior of Cricetinae species can be important for individuals who keep them as pets or for researchers studying their physiology.

E2F1 is a member of the E2F family of transcription factors, which are involved in the regulation of cell cycle progression and apoptosis (programmed cell death). Specifically, E2F1 plays a role as a transcriptional activator, binding to specific DNA sequences and promoting the expression of genes required for the G1/S transition of the cell cycle.

In more detail, E2F1 forms a complex with a retinoblastoma protein (pRb) in the G0 and early G1 phases of the cell cycle. When pRb is phosphorylated by cyclin-dependent kinases during the late G1 phase, E2F1 is released and can then bind to its target DNA sequences and activate transcription of genes involved in DNA replication and cell cycle progression.

However, if E2F1 is overexpressed or activated inappropriately, it can also promote apoptosis, making it a key player in both cell proliferation and cell death pathways. Dysregulation of E2F1 has been implicated in the development of various human cancers, including breast, lung, and prostate cancer.

Medical Definition of "Multiprotein Complexes" :

Multiprotein complexes are large molecular assemblies composed of two or more proteins that interact with each other to carry out specific cellular functions. These complexes can range from relatively simple dimers or trimers to massive structures containing hundreds of individual protein subunits. They are formed through a process known as protein-protein interaction, which is mediated by specialized regions on the protein surface called domains or motifs.

Multiprotein complexes play critical roles in many cellular processes, including signal transduction, gene regulation, DNA replication and repair, protein folding and degradation, and intracellular transport. The formation of these complexes is often dynamic and regulated in response to various stimuli, allowing for precise control of their function.

Disruption of multiprotein complexes can lead to a variety of diseases, including cancer, neurodegenerative disorders, and infectious diseases. Therefore, understanding the structure, composition, and regulation of these complexes is an important area of research in molecular biology and medicine.

Ubiquitin-conjugating enzymes (UBCs or E2 enzymes) are a family of enzymes that play a crucial role in the ubiquitination process, which is a post-translational modification of proteins. This process involves the covalent attachment of the protein ubiquitin to specific lysine residues on target proteins, ultimately leading to their degradation by the 26S proteasome.

Ubiquitination is a multi-step process that requires the coordinated action of three types of enzymes: E1 (ubiquitin-activating), E2 (ubiquitin-conjugating), and E3 (ubiquitin ligases). Ubiquitin-conjugating enzymes are responsible for transferring ubiquitin from the E1 enzyme to the target protein, which is facilitated by an E3 ubiquitin ligase. The human genome encodes around 40 different UBCs, each with unique substrate specificities and functions in various cellular processes, such as protein degradation, DNA repair, and signal transduction.

Ubiquitination is a highly regulated process that can be reversed by the action of deubiquitinating enzymes (DUBs), which remove ubiquitin molecules from target proteins. Dysregulation of the ubiquitination pathway has been implicated in various diseases, including cancer, neurodegenerative disorders, and inflammatory conditions.

A phenotype is the physical or biochemical expression of an organism's genes, or the observable traits and characteristics resulting from the interaction of its genetic constitution (genotype) with environmental factors. These characteristics can include appearance, development, behavior, and resistance to disease, among others. Phenotypes can vary widely, even among individuals with identical genotypes, due to differences in environmental influences, gene expression, and genetic interactions.

Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) is a laboratory technique used in molecular biology to amplify and detect specific DNA sequences. This technique is particularly useful for the detection and quantification of RNA viruses, as well as for the analysis of gene expression.

The process involves two main steps: reverse transcription and polymerase chain reaction (PCR). In the first step, reverse transcriptase enzyme is used to convert RNA into complementary DNA (cDNA) by reading the template provided by the RNA molecule. This cDNA then serves as a template for the PCR amplification step.

In the second step, the PCR reaction uses two primers that flank the target DNA sequence and a thermostable polymerase enzyme to repeatedly copy the targeted cDNA sequence. The reaction mixture is heated and cooled in cycles, allowing the primers to anneal to the template, and the polymerase to extend the new strand. This results in exponential amplification of the target DNA sequence, making it possible to detect even small amounts of RNA or cDNA.

RT-PCR is a sensitive and specific technique that has many applications in medical research and diagnostics, including the detection of viruses such as HIV, hepatitis C virus, and SARS-CoV-2 (the virus that causes COVID-19). It can also be used to study gene expression, identify genetic mutations, and diagnose genetic disorders.

'Escherichia coli' (E. coli) is a type of gram-negative, facultatively anaerobic, rod-shaped bacterium that commonly inhabits the intestinal tract of humans and warm-blooded animals. It is a member of the family Enterobacteriaceae and one of the most well-studied prokaryotic model organisms in molecular biology.

While most E. coli strains are harmless and even beneficial to their hosts, some serotypes can cause various forms of gastrointestinal and extraintestinal illnesses in humans and animals. These pathogenic strains possess virulence factors that enable them to colonize and damage host tissues, leading to diseases such as diarrhea, urinary tract infections, pneumonia, and sepsis.

E. coli is a versatile organism with remarkable genetic diversity, which allows it to adapt to various environmental niches. It can be found in water, soil, food, and various man-made environments, making it an essential indicator of fecal contamination and a common cause of foodborne illnesses. The study of E. coli has contributed significantly to our understanding of fundamental biological processes, including DNA replication, gene regulation, and protein synthesis.

Physiological stress is a response of the body to a demand or threat that disrupts homeostasis and activates the autonomic nervous system and hypothalamic-pituitary-adrenal (HPA) axis. This results in the release of stress hormones such as adrenaline, cortisol, and noradrenaline, which prepare the body for a "fight or flight" response. Increased heart rate, rapid breathing, heightened sensory perception, and increased alertness are some of the physiological changes that occur during this response. Chronic stress can have negative effects on various bodily functions, including the immune, cardiovascular, and nervous systems.

Up-regulation is a term used in molecular biology and medicine to describe an increase in the expression or activity of a gene, protein, or receptor in response to a stimulus. This can occur through various mechanisms such as increased transcription, translation, or reduced degradation of the molecule. Up-regulation can have important functional consequences, for example, enhancing the sensitivity or response of a cell to a hormone, neurotransmitter, or drug. It is a normal physiological process that can also be induced by disease or pharmacological interventions.

Neoplastic cell transformation is a process in which a normal cell undergoes genetic alterations that cause it to become cancerous or malignant. This process involves changes in the cell's DNA that result in uncontrolled cell growth and division, loss of contact inhibition, and the ability to invade surrounding tissues and metastasize (spread) to other parts of the body.

Neoplastic transformation can occur as a result of various factors, including genetic mutations, exposure to carcinogens, viral infections, chronic inflammation, and aging. These changes can lead to the activation of oncogenes or the inactivation of tumor suppressor genes, which regulate cell growth and division.

The transformation of normal cells into cancerous cells is a complex and multi-step process that involves multiple genetic and epigenetic alterations. It is characterized by several hallmarks, including sustained proliferative signaling, evasion of growth suppressors, resistance to cell death, enabling replicative immortality, induction of angiogenesis, activation of invasion and metastasis, reprogramming of energy metabolism, and evading immune destruction.

Neoplastic cell transformation is a fundamental concept in cancer biology and is critical for understanding the molecular mechanisms underlying cancer development and progression. It also has important implications for cancer diagnosis, prognosis, and treatment, as identifying the specific genetic alterations that underlie neoplastic transformation can help guide targeted therapies and personalized medicine approaches.

Immunoprecipitation (IP) is a research technique used in molecular biology and immunology to isolate specific antigens or antibodies from a mixture. It involves the use of an antibody that recognizes and binds to a specific antigen, which is then precipitated out of solution using various methods, such as centrifugation or chemical cross-linking.

In this technique, an antibody is first incubated with a sample containing the antigen of interest. The antibody specifically binds to the antigen, forming an immune complex. This complex can then be captured by adding protein A or G agarose beads, which bind to the constant region of the antibody. The beads are then washed to remove any unbound proteins, leaving behind the precipitated antigen-antibody complex.

Immunoprecipitation is a powerful tool for studying protein-protein interactions, post-translational modifications, and signal transduction pathways. It can also be used to detect and quantify specific proteins in biological samples, such as cells or tissues, and to identify potential biomarkers of disease.

Caspase-3 is a type of protease enzyme that plays a central role in the execution-phase of cell apoptosis, or programmed cell death. It's also known as CPP32 (CPP for ced-3 protease precursor) or apopain. Caspase-3 is produced as an inactive protein that is activated when cleaved by other caspases during the early stages of apoptosis. Once activated, it cleaves a variety of cellular proteins, including structural proteins, enzymes, and signal transduction proteins, leading to the characteristic morphological and biochemical changes associated with apoptotic cell death. Caspase-3 is often referred to as the "death protease" because of its crucial role in executing the cell death program.

Malondialdehyde (MDA) is a naturally occurring organic compound that is formed as a byproduct of lipid peroxidation, a process in which free radicals or reactive oxygen species react with polyunsaturated fatty acids. MDA is a highly reactive aldehyde that can modify proteins, DNA, and other biomolecules, leading to cellular damage and dysfunction. It is often used as a marker of oxidative stress in biological systems and has been implicated in the development of various diseases, including cancer, cardiovascular disease, and neurodegenerative disorders.

Infrared rays are not typically considered in the context of medical definitions. They are a type of electromagnetic radiation with longer wavelengths than those of visible light, ranging from 700 nanometers to 1 millimeter. In the field of medicine, infrared radiation is sometimes used in therapeutic settings for its heat properties, such as in infrared saunas or infrared therapy devices. However, infrared rays themselves are not a medical condition or diagnosis.

Ascorbic acid is the chemical name for Vitamin C. It is a water-soluble vitamin that is essential for human health. Ascorbic acid is required for the synthesis of collagen, a protein that plays a role in the structure of bones, tendons, ligaments, and blood vessels. It also functions as an antioxidant, helping to protect cells from damage caused by free radicals.

Ascorbic acid cannot be produced by the human body and must be obtained through diet or supplementation. Good food sources of vitamin C include citrus fruits, strawberries, bell peppers, broccoli, and spinach.

In the medical field, ascorbic acid is used to treat or prevent vitamin C deficiency and related conditions, such as scurvy. It may also be used in the treatment of various other health conditions, including common cold, cancer, and cardiovascular disease, although its effectiveness for these uses is still a matter of scientific debate.

Proto-oncogene proteins are normal cellular proteins that play crucial roles in various cellular processes, such as signal transduction, cell cycle regulation, and apoptosis (programmed cell death). They are involved in the regulation of cell growth, differentiation, and survival under physiological conditions.

When proto-oncogene proteins undergo mutations or aberrations in their expression levels, they can transform into oncogenic forms, leading to uncontrolled cell growth and division. These altered proteins are then referred to as oncogene products or oncoproteins. Oncogenic mutations can occur due to various factors, including genetic predisposition, environmental exposures, and aging.

Examples of proto-oncogene proteins include:

1. Ras proteins: Involved in signal transduction pathways that regulate cell growth and differentiation. Activating mutations in Ras genes are found in various human cancers.
2. Myc proteins: Regulate gene expression related to cell cycle progression, apoptosis, and metabolism. Overexpression of Myc proteins is associated with several types of cancer.
3. EGFR (Epidermal Growth Factor Receptor): A transmembrane receptor tyrosine kinase that regulates cell proliferation, survival, and differentiation. Mutations or overexpression of EGFR are linked to various malignancies, such as lung cancer and glioblastoma.
4. Src family kinases: Intracellular tyrosine kinases that regulate signal transduction pathways involved in cell proliferation, survival, and migration. Dysregulation of Src family kinases is implicated in several types of cancer.
5. Abl kinases: Cytoplasmic tyrosine kinases that regulate various cellular processes, including cell growth, differentiation, and stress responses. Aberrant activation of Abl kinases, as seen in chronic myelogenous leukemia (CML), leads to uncontrolled cell proliferation.

Understanding the roles of proto-oncogene proteins and their dysregulation in cancer development is essential for developing targeted cancer therapies that aim to inhibit or modulate these aberrant signaling pathways.

Gene expression regulation in fungi refers to the complex cellular processes that control the production of proteins and other functional gene products in response to various internal and external stimuli. This regulation is crucial for normal growth, development, and adaptation of fungal cells to changing environmental conditions.

In fungi, gene expression is regulated at multiple levels, including transcriptional, post-transcriptional, translational, and post-translational modifications. Key regulatory mechanisms include:

1. Transcription factors (TFs): These proteins bind to specific DNA sequences in the promoter regions of target genes and either activate or repress their transcription. Fungi have a diverse array of TFs that respond to various signals, such as nutrient availability, stress, developmental cues, and quorum sensing.
2. Chromatin remodeling: The organization and compaction of DNA into chromatin can influence gene expression. Fungi utilize ATP-dependent chromatin remodeling complexes and histone modifying enzymes to alter chromatin structure, thereby facilitating or inhibiting the access of transcriptional machinery to genes.
3. Non-coding RNAs: Small non-coding RNAs (sncRNAs) play a role in post-transcriptional regulation of gene expression in fungi. These sncRNAs can guide RNA-induced transcriptional silencing (RITS) complexes to specific target loci, leading to the repression of gene expression through histone modifications and DNA methylation.
4. Alternative splicing: Fungi employ alternative splicing mechanisms to generate multiple mRNA isoforms from a single gene, thereby increasing proteome diversity. This process can be regulated by RNA-binding proteins that recognize specific sequence motifs in pre-mRNAs and promote or inhibit splicing events.
5. Protein stability and activity: Post-translational modifications (PTMs) of proteins, such as phosphorylation, ubiquitination, and sumoylation, can influence their stability, localization, and activity. These PTMs play a crucial role in regulating various cellular processes, including signal transduction, stress response, and cell cycle progression.

Understanding the complex interplay between these regulatory mechanisms is essential for elucidating the molecular basis of fungal development, pathogenesis, and drug resistance. This knowledge can be harnessed to develop novel strategies for combating fungal infections and improving agricultural productivity.

Neoplastic gene expression regulation refers to the processes that control the production of proteins and other molecules from genes in neoplastic cells, or cells that are part of a tumor or cancer. In a normal cell, gene expression is tightly regulated to ensure that the right genes are turned on or off at the right time. However, in cancer cells, this regulation can be disrupted, leading to the overexpression or underexpression of certain genes.

Neoplastic gene expression regulation can be affected by a variety of factors, including genetic mutations, epigenetic changes, and signals from the tumor microenvironment. These changes can lead to the activation of oncogenes (genes that promote cancer growth and development) or the inactivation of tumor suppressor genes (genes that prevent cancer).

Understanding neoplastic gene expression regulation is important for developing new therapies for cancer, as targeting specific genes or pathways involved in this process can help to inhibit cancer growth and progression.

In the context of medicine and pharmacology, "kinetics" refers to the study of how a drug moves throughout the body, including its absorption, distribution, metabolism, and excretion (often abbreviated as ADME). This field is called "pharmacokinetics."

1. Absorption: This is the process of a drug moving from its site of administration into the bloodstream. Factors such as the route of administration (e.g., oral, intravenous, etc.), formulation, and individual physiological differences can affect absorption.

2. Distribution: Once a drug is in the bloodstream, it gets distributed throughout the body to various tissues and organs. This process is influenced by factors like blood flow, protein binding, and lipid solubility of the drug.

3. Metabolism: Drugs are often chemically modified in the body, typically in the liver, through processes known as metabolism. These changes can lead to the formation of active or inactive metabolites, which may then be further distributed, excreted, or undergo additional metabolic transformations.

4. Excretion: This is the process by which drugs and their metabolites are eliminated from the body, primarily through the kidneys (urine) and the liver (bile).

Understanding the kinetics of a drug is crucial for determining its optimal dosing regimen, potential interactions with other medications or foods, and any necessary adjustments for special populations like pediatric or geriatric patients, or those with impaired renal or hepatic function.

'Escherichia coli (E. coli) proteins' refer to the various types of proteins that are produced and expressed by the bacterium Escherichia coli. These proteins play a critical role in the growth, development, and survival of the organism. They are involved in various cellular processes such as metabolism, DNA replication, transcription, translation, repair, and regulation.

E. coli is a gram-negative, facultative anaerobe that is commonly found in the intestines of warm-blooded organisms. It is widely used as a model organism in scientific research due to its well-studied genetics, rapid growth, and ability to be easily manipulated in the laboratory. As a result, many E. coli proteins have been identified, characterized, and studied in great detail.

Some examples of E. coli proteins include enzymes involved in carbohydrate metabolism such as lactase, sucrase, and maltose; proteins involved in DNA replication such as the polymerases, single-stranded binding proteins, and helicases; proteins involved in transcription such as RNA polymerase and sigma factors; proteins involved in translation such as ribosomal proteins, tRNAs, and aminoacyl-tRNA synthetases; and regulatory proteins such as global regulators, two-component systems, and transcription factors.

Understanding the structure, function, and regulation of E. coli proteins is essential for understanding the basic biology of this important organism, as well as for developing new strategies for combating bacterial infections and improving industrial processes involving bacteria.

DNA primers are short single-stranded DNA molecules that serve as a starting point for DNA synthesis. They are typically used in laboratory techniques such as the polymerase chain reaction (PCR) and DNA sequencing. The primer binds to a complementary sequence on the DNA template through base pairing, providing a free 3'-hydroxyl group for the DNA polymerase enzyme to add nucleotides and synthesize a new strand of DNA. This allows for specific and targeted amplification or analysis of a particular region of interest within a larger DNA molecule.

DNA\ {\xrightarrow {}}\ {}^{3}O_{2}+damaged\ DNA} } Unlike direct DNA damage, which occurs in areas directly exposed to UV-B ... unlike direct DNA damage which causes sunburn, indirect DNA damage does not result in any warning signal or pain in the human ... These reactive chemical species can reach DNA by diffusion and the bimolecular reaction damages the DNA (oxidative stress). It ... Indirect DNA damage occurs when a UV-photon is absorbed in the human skin by a chromophore that does not have the ability to ...
... brain AP site Direct DNA damage DNA DNA adduct DNA damage theory of aging DNA repair DNA replication Free radical damage to DNA ... This reflects the accumulation of DNA damage with age. DNA damage accumulation with age is further described in DNA damage ... DNA damage is an abnormal chemical structure in DNA, while a mutation is a change in the sequence of base pairs. DNA damages ... One indication that DNA damages are a major problem for life is that DNA repair processes, to cope with DNA damages, have been ...
October 2012). "Damaged DNA induced UV-damaged DNA-binding protein (UV-DDB) dimerization and its roles in chromatinized DNA ... DNA damage-binding protein or UV-DDB is a protein complex that is responsible for repair of UV-damaged DNA. This complex is ... UV-DDB is not only responsible for the repair of damaged DNA, it can also function by acting as a damage sensor. In base ... During DNA damage, proteins OGG1 and APE 1 encounter difficulty in repairing the lesions in a DNA wrapped nucleosome. ...
DNA damages and mutations are related because DNA damages often cause errors of DNA synthesis during replication or repair and ... If a DNA repair protein is deficient, unrepaired DNA damages tend to accumulate. Such accumulated DNA damages appear to cause ... a common type of oxidative DNA damage. DNA strand breaks also increased in atherosclerotic plaques, thus linking DNA damage to ... Some DNA damage may remain in any cell despite the action of repair processes. The accumulation of unrepaired DNA damage is ...
Calonge, T. M.; O'Connell, M. J. (2007). "Turning off the G2 DNA damage checkpoint". DNA Repair (Amst). 7 (2): 136-140. doi: ... causes recruitment of these proteins to sites of DNA damage where they mediate the activity of DNA polymerases involved in DNA ... The G2-M DNA damage checkpoint is an important cell cycle checkpoint in eukaryotic organisms that ensures that cells don't ... The main rad3 effector is the kinase Chk1, which is required for the G2-M arrest in response to DNA-damaging agents. Chk1 is an ...
... has been shown to interact with [proteins]: ATF3, C-Fos, C-jun and CEBPB, CSNK2A1, JunD, and ... "Entrez Gene: DDIT3 DNA-damage-inducible transcript 3". Ubeda M, Wang XZ, Zinszner H, Wu I, Habener JF, Ron D (April 1996). " ... DNA damage-inducible transcript 3, also known as C/EBP homologous protein (CHOP), is a pro-apoptotic transcription factor that ... March 2002). "Activation of peroxisome proliferator-activated receptor-gamma stimulates the growth arrest and DNA-damage ...
... can occur as a result of exposure to ionizing radiation or to radiomimetic compounds. Damage to DNA ... Radical damage to DNA can also occur through the interaction of DNA with certain natural products known as radiomimetic ... Malignant melanoma can be caused by indirect DNA damage because it is found in parts of the body not exposed to sunlight. DNA ... Povirk LF (1996). "DNA damage and mutagenesis by radiomimetic DNA-cleaving agents: Bleomycin, neocarzinostatin and other ...
Combined with the damage pattern, this short fragment length can also help differentiate between modern and ancient DNA. ... As DNA degrades over time, the nucleotides that make up the DNA may change, especially at the ends of the DNA molecules. The ... Another problem with ancient DNA samples is contamination by modern human DNA and by microbial DNA (most of which is also ... Frequency measurement of the C-T level, ancient DNA damage, can be made using various software such as mapDamage2.0 or PMDtools ...
Friedberg, Errol C. (2003-01-23). "DNA damage and repair". Nature. 421 (6921): 436-440. doi:10.1038/nature01408. ISSN 0028-0836 ... Replication machineries include primosotors are replication enzymes; DNA polymerase, DNA helicases, DNA clamps and DNA ... DNA replication is the biological process of producing two identical replicas of DNA from one original DNA molecule. DNA ... In contrast, DNA Pol I is the enzyme responsible for replacing RNA primers with DNA. DNA Pol I has a 5′ to 3′ exonuclease ...
Exposure to them can directly or indirectly cause DNA damage. In the direct case, a carcinogen can bind to DNA and cause it to ... This allows ELISA to quantify DNA adducts as well as map an inverse relationship between DNA damage and the intensity of the ... "Airborne particulate matter induces oxidative damage, DNA adduct formation and alterations in DNA repair pathways". ... Kastan MB (April 2008). "DNA damage responses: mechanisms and roles in human disease: 2007 G.H.A. Clowes Memorial Award Lecture ...
The type of DNA damage produced depends on the type of mutagen. For example, UV light can damage DNA by producing thymine ... DNA exists in many possible conformations that include A-DNA, B-DNA, and Z-DNA forms, although only B-DNA and Z-DNA have been ... In DNA replication, DNA-dependent DNA polymerases make copies of DNA polynucleotide chains. To preserve biological information ... "Cancer and aging as consequences of un-repaired DNA damage". In Kimura H, Suzuki A (eds.). New Research on DNA Damage. New York ...
This damage is in the form of DNA lesions that arise spontaneously or due to DNA damaging agents. DNA replication machinery is ... There are several different definitions for DNA synthesis: it can refer to DNA replication - DNA biosynthesis (in vivo DNA ... However, DNA synthesis in vitro can be a very error-prone process. Damaged DNA is subject to repair by several different ... DNA Damage and Associated DNA Repair Defects in Disease and Premature Aging. Am J Hum Genet. 2019 Aug 1;105(2):237-257. doi: ...
"DNA damage theory of aging" for a discussion of the evidence that DNA damage is the primary underlying cause of aging.) Some ... Such DNA damages can cause errors during DNA synthesis leading to mutations, some of which may give rise to cancer. Germ-line ... A DNA repair-deficiency disorder is a medical condition due to reduced functionality of DNA repair. DNA repair defects can ... "The human Werner syndrome protein stimulates repair of oxidative DNA base damage by the DNA glycosylase NEIL1". J. Biol. Chem. ...
In E. coli, DNA AP lyase (endonuclease III) helps repair oxidative damage to DNA bases by catalyzing the excision of the ... Clancy, Suzanne (2008). "DNA damage & repair: mechanisms for maintaining DNA integrity". Nature Education. 1 (1): 103. ... DNA damage is ubiquitous amongst all forms of life. There is an estimated 1 x 10−4 to 1 x 10−6 mutations per human gamete, ... damaged pyrimidines and purines from ring saturation or opening from the DNA backbone. This damage can be caused by non- ...
Biology portal Accelerated aging disease Aging DNA Cell cycle DNA damage (naturally occurring) DNA damage theory of aging DNA ... Reduced expression of DNA repair genes causes deficient DNA repair. When DNA repair is deficient DNA damages remain in cells at ... It is important to distinguish between DNA damage and mutation, the two major types of error in DNA. DNA damage and mutation ... UV damage, alkylation/methylation, X-ray damage and oxidative damage are examples of induced damage. Spontaneous damage can ...
Lindahl, T. (1986). "DNA Glycosylases in DNA Repair". Mechanisms of DNA Damage and Repair. 38: 335-340. doi:10.1007/978-1-4615- ... Ide H, Kotera M (April 2004). "Human DNA glycosylases involved in the repair of oxidatively damaged DNA". Biol. Pharm. Bull. 27 ... Uracil-DNA glycosylases are DNA repair enzymes that excise uracil residues from DNA by cleaving the N-glycosydic bond, ... This was the most frequent DNA repair abnormality found among the 8 DNA repair genes tested. NEIL1 was also one of six DNA ...
These compounds mutate DNA strands and produce genetic damage, inducing newly lysis and subsequent cell death. Its action on ... DNA damage. Cancer, particularly leukemia. Leukopenia. Growth reduction. Reproductive deficiencies: sterility, reduction in ... D. radiodurans is capable to resist oxidative stress and DNA damage from radiation, and reduces technetium, uranium and ... This effect is accentuated if the absorbed dose is between 0.5 and 2 Sv, in whose first damage, nausea and hair loss are ...
DNA damage; Nucleophosmin; Cancer; Ribosomal protein; p53; MDM2; p14-ARF Abdol-Hossein Rezaeian, Hiroyuki Inuzuka, Wenyi Wei, ... The role of the nucleolus in regulating the cell cycle and the DNA damage response, Editor(s): Rossen Donev, Advances in ... https://www.sciencedirect.com/science/article/pii/S1876162323000330) Keywords: DNA replication; DNA replication stress; Genome ... Chromatin regulators in DNA replication and genome stability maintenance during S-phase, Editor(s): Rossen Donev, Advances in ...
If DNA repair is deficient, DNA damage tends to accumulate. Such excess DNA damage may increase mutations due to error-prone ... DNA-PK also cooperates with ATR and ATM to phosphorylate proteins involved in the DNA damage checkpoint. DNA-PKcs knockout mice ... Cancer and aging as consequences of un-repaired DNA damage. In: New Research on DNA Damages (Editors: Honoka Kimura and Aoi ... DNA damage appears to be the primary underlying cause of cancer, and deficiencies in DNA repair genes likely underlie many ...
... resulting in deficient DNA repair, DNA damages will accumulate. Increased DNA damage tends to cause increased errors during DNA ... DNA damages can also give rise to epigenetic alterations during faulty DNA repair processes. The DNA damages that accumulate ... July 2007). "DNA damage, homology-directed repair, and DNA methylation". PLOS Genetics. 3 (7): e110. doi:10.1371/journal.pgen. ... Jin B, Robertson KD (2013). "DNA Methyltransferases, DNA Damage Repair, and Cancer". Epigenetic Alterations in Oncogenesis. ...
This enzyme is involved in DNA damage repair and targets hypoxanthine bases. DNA is constantly exposed to chemical reactions ... DNA damage is caused by environmental and endogenous agents, that can create highly mutagenic lesions or compromise genomic ... In order to preserve genomic sequence information, cells must counteract DNA damage through one of its five major pathways: ... DNA polymerase and DNA ligase then completes the process by incorporating the correct base pair and closing the nick. The UDG ...
Exogenous factors in DNA, particularly DNA damage, are more known as environmental factors that cause progression in the ... Friedberg, Errol C.; McDaniel, Lisa D.; Schultz, Roger A. (February 2004). "The role of endogenous and exogenous DNA damage and ... Hakem, Razqallah (January 16, 2008). "DNA-damage repair; the good, the bad, and the ugly". The EMBO Journal. 27 (4): 589-605. ... DNA introduced to cells via transfection or viral transduction is an exogenous factor. ...
S and G2 phase to check if DNA is normal, and withdraws the cell from the cycle if the DNA is damaged or has undergone ... Another type of negative regulator is p53, which halts the cell cycle process upon detection of DNA damage so as to provide to ... This regulator can also induce apoptosis when the DNA damage is too large and cannot be repaired. Checkpoints in cell cycles ... Kaina, Bernd (2011). "DNA Damage-Induced Apoptosis". Encyclopedia of Cancer. pp. 1136-1139. doi:10.1007/978-3-642-16483-5_1673 ...
... (TS) plays a crucial role in the early stages of DNA biosynthesis. DNA damage or deletion occur on a daily ... Both cause DNA damage. The following reaction is catalyzed by thymidylate synthase: 5,10-methylenetetrahydrofolate + dUMP ⇌ {\ ... The enzyme is essential for regulating the balanced supply of the 4 DNA precursors in normal DNA replication: defects in the ... Therefore, synthesis and insertion of healthy DNA is vital for normal body functions and avoidance of cancerous activity. In ...
DNA Damage and Repair. Contemporary Cancer Research. pp. 107-124. doi:10.1007/978-1-59259-095-7_5 (inactive 1 August 2023). ... The coding sequence is on the Crick strand of the DNA. YDL102W (aka POL3 encoding a subunit of DNA polymerase delta) is located ... and are significant tools in the study of DNA damage and repair mechanisms. S. cerevisiae has developed as a model organism ... reactive oxygen species increase leading to the accumulation of DNA damages such as apurinic/apyrimidinic sites and double- ...
"Benzophenone Photosensitized DNA Damage". Acc. Chem. Res. 45 (9): 1558-1570. doi:10.1021/ar300054e. PMID 22698517. Doug Brunk ( ... The interaction with DNA and the successive photo-induced energy transfer is at the base of the benzophenone activity as a DNA ... Benzophenone prevents ultraviolet (UV) light from damaging scents and colors in products such as perfumes and soaps. ... From a molecular chemistry point of view interaction of benzophenone with B-DNA has been demonstrated experimentally. ...
DNA damage was proposed in a 2021 review to be the underlying cause of aging because of the mechanistic link of DNA damage to ... Free radicals can damage proteins, lipids or DNA. Glycation mainly damages proteins. Damaged proteins and lipids accumulate in ... damage to DNA, and damage to tissues and cells by oxygen radicals (widely known as free radicals), and some of this damage is ... DNA) damage (DNA damage theory of aging) and dysdifferentiation. The system theories include the immunologic approach to ageing ...
Most long-term effects are caused by damage to DNA. DNA damage induced by ionizing radiation is similar to oxidative stress, ... Further information on the association of oxidative DNA damage with aging is presented in the article DNA damage theory of ... Oxidative stress can cause DNA damage in neurons. In neuronal progenitor cells, DNA damage is associated with increased ... and DNA. Oxidative stress from oxidative metabolism causes base damage, as well as strand breaks in DNA. Base damage is mostly ...
Acetaldehyde induces DNA interstrand crosslinks, a form of DNA damage. These can be repaired by an inaccurate replication- ... doi:10.1093/toxsci/kfp133 Li L, Jiang L, Geng C, Cao J, Zhong L. The role of oxidative stress in acrolein-induced DNA damage in ... The basic cause of sporadic (non-familial) cancers is DNA damage and genomic instability. A minority of cancers are due to ... Infectious organisms that increase the risk of cancer are frequently a source of DNA damage or genomic instability. Viral ...
"Cancer and Aging as Consequences of Unrepaired DNA Damage". In Kimura H, Suzuki A (eds.). New Research on DNA Damages. New York ... Oxidative DNA damage 8-hydroxydeoxyguanosine in houseflies was found in one study to increase with age and reduce life ... Holmes GE, Bernstein C, Bernstein H (September 1992). "Oxidative and other DNA damages as the basis of aging: a review". ... ISBN 978-1-4020-6242-1. Agarwal S, Sohal RS (December 1994). "DNA oxidative damage and life expectancy in houseflies". ...
DNA\ {\xrightarrow {}}\ {}^{3}O_{2}+damaged\ DNA} } Unlike direct DNA damage, which occurs in areas directly exposed to UV-B ... unlike direct DNA damage which causes sunburn, indirect DNA damage does not result in any warning signal or pain in the human ... These reactive chemical species can reach DNA by diffusion and the bimolecular reaction damages the DNA (oxidative stress). It ... Indirect DNA damage occurs when a UV-photon is absorbed in the human skin by a chromophore that does not have the ability to ...
diets high in selenium had less DNA damage in their prostates compared with dogs on a normal diet, enhancing the view that the ... DNA damage: Pharma meets nutrition in healthy ageing product 07-Oct-2013. By Annie-Rose Harrison-Dunn ... Increased intakes of vitamins C and E and other antioxidants from the diet may protect against DNA damage in people exposed to ... Supplements of the omega-3 fatty acid DHA (docosahexaenoic acid) may reduce the amount of DNA damage in sperm, says a new ...
... researchers witness how molecules find and fix damaged DNA. ... It identifies DNA damage so that another crew can come in and ... Molecular sensor scouts DNA damage and supervises repair. Date:. July 23, 2019. Source:. University of Pittsburgh. Summary:. ... Using single-molecule imaging, researchers witness how molecules find and fix damaged DNA. Share:. Facebook Twitter Pinterest ... "Molecular sensor scouts DNA damage and supervises repair." ScienceDaily. www.sciencedaily.com. /. releases. /. 2019. /. 07. /. ...
A poorly understood feature of these responses is the delayed onset (days), in contrast to the acute DNA-damage responses that ... Roger Greenberg and colleagues report a link between mitosis, the formation of micronuclei and DNA-damage-induced inflammatory ... Such dichotomous kinetics implicate additional rate-limiting steps that are essential for DNA-damage-induced inflammation. Here ... the formation of micronuclei and DNA-damage-induced cGAS-dependent inflammation. Ionizing radiation and genotoxic cancer ...
... the DNA damage checkpoint remains active. The treatment of female cancer patients with DNA damaging irradiation or ... Thymine DNA Glycosylase (TDG) is an enzyme of the base excision repair mechanism and removes damaged or mispaired bases from ... Thymine DNA Glycosylase (TDG) is an enzyme of the base excision repair mechanism and removes damaged or mispaired bases from ... Based on this new adaptation of the MSA model, we have predicted cell-specific levels of initial complex DNA damage and cell ...
UV light can trigger a slow-acting chemisensitization mechanism that involves melanin and keeps damaging DNA for hours, long ... UV light, it turns out, may act via a "light" or a "dark" biochemical pathway to cause a type of DNA damage known as a ... Home Topics Cancer Even in the Dark, Sunlight Can Damage Your Skins DNA ... "As suggested by the authors, UV DNA damage arising in the dark might also be reduced by agents that scavenge or interfere with ...
Infographic: DNA Damage Viewed with Unprecedented Clarity. A new genetic sensor called PRISM makes use of a host cells DNA ... Infographic: DNA Damage Viewed with Unprecedented Clarity. A new genetic sensor called PRISM makes use of a host cells DNA ... From DNA damage to cellular miscommunication, aging is a mysterious and multifarious process. ... sensor called PRISM makes use of a host cells DNA replication machinery to trigger fluorescence in neurons with damaged DNA. ...
A new study found that a compound in sucralose may damage DNA, impact gene expression, and contribute to leaky gut. ... Chemical in Artificial Sweetener May Damage DNA. Published on June 05, 2023 . ... The chemicals damage the tight junctions, or interfaces, where cells in the gut wall connect to each other." ... Now, new research by the same scientific team found that sucralose-6-acetate may be genotoxic and negatively impact DNA. They ...
A new animal study has revealed food DNA as a novel potential source of genetic damage. ... the lab-grown cells showed significant DNA damage resulting from taking up heat-damaged DNA components. As for the mice, DNA ... suggesting a plausible and potentially significant pathway for damaged food DNA to inflict damage on other DNA downstream in ... showed that all three foods exhibited DNA damage when boiled and roasted, and higher temperatures increased DNA damage in ...
Molecular Sensor Scouts DNA Damage and Supervises RepairVeronica Hinman Appointed Head of Department of Biological Sciences ... UV-DDB Finds Damaged DNA in Living Cells. When telomeres (green) undergo damage (bottom row), UV-DDB (red) rushes to the scene ... It identifies DNA damage so that another crew can come in and patch and seal it," said study senior author Bennett Van Houten, ... Molecular Sensor Scouts DNA Damage and Supervises Repair. By Jocelyn Duffy jhduffy(through)andrew.cmu.edu ...
However, it remains unexplored if TIGAR plays an important role in DNA damage and repair. This study investigated the role of ... Knockdown of TIGAR exacerbated DNA damage and the effects were partly reversed by the supplementation of PPP products NADPH, ... TIGAR in DNA damage response (DDR) induced by genotoxic drugs and hypoxia in tumor cells. Results showed that TIGAR was ... TIGAR knockdown increased DNA damage. To define a role of TIGAR in DNA damage, TIGAR expression was knocked down with siRNA in ...
Long-term exposure to low-dose radiation can cause DNA damage to lymphocyte cells in interventional radiologists. ... Long-term exposure to low-dose radiation can cause DNA damage to lymphocyte cells in interventional radiologists - yet ... A group at the Slovak Academy of Sciences in Bratislava investigated DNA damage in peripheral blood lymphocytes of ... Ultimately, whether the increased presence of micronuclei due to DNA damage translates to higher health risks in interventional ...
Upon DNA damage, Mec1 and Tel1 can transduce the DNA damage signal to Rad53, which ultimately results in arrest of the cell ... in response to DNA damage and identified factors that bind these DNA elements to mediate the response of RNR2 to DNA damage. ... "Many biologists used to think that DNA damage was just about arresting the cell cycle, but the fact is that the DNA-damage ... upon exposure of cells to DNA damage and that mutations in RNR2 resulted in hypersensitivity to DNA damage. ...
This study links melatonin to 8-OH-dG which repairs DNA damage. Night shift workers show lower levels than day shift workers ... abstract 32ZCLEW0BYJLRG4631BW6YXBBW8PJ0 39PAAFCODMZVXNV1R9312G5RHKFTVD A2EZY9OFIH2XQH !!Could Shift Work Damage Your Dna?!!)( ... Working at night resulted in lower melatonin levels and a less of the chemical which repairs DNA in the urine. The melatonin ... Do you work at night? Recent research shows that working the night shift has negative effects on your DNA! See how melatonin ...
In conclusion, CD44v6 and ALDH1A1 are candidate stem cell markers for NPC, and the increased formation of DNA lesions by ... We also measured 8-nitroguanine formation as an indicator of inflammation-related DNA lesions. The staining intensity of 8- ... Nitrative and oxidative DNA damage plays an important role in inflammation-related carcinogenesis. To investigate the ... Inflammation-Related DNA Damage and Cancer Stem Cell Markers in Nasopharyngeal Carcinoma. Shumin Wang. ,1,2Ning Ma. ,3Weilin ...
Learn about the Center for DNA Damage and Repair at Dana-Farber, including its research and expert team of scientists. ... The Center for DNA Damage and Repair (CDDR) explores the various DNA repair defects in human cancers which underlie genomic ... CDDR will collaborate with both basic researchers and clinicians to develop innovative research involving DNA damage and DNA ... The Center for DNA Damage and Repair will function as a central resource for Dana-Farber Cancer Institute in supporting ...
Gene context of DNA Damage. *EP300 acetylation of TP53 in response to DNA damage regulates its DNA-binding and transcription ... Biological context of DNA Damage. *Hence, p53 is not the only mediator of apoptosis provoked by DNA damage [20]. ... Disease relevance of DNA Damage. *Fanconi anemia proteins function in a DNA damage response pathway involving breast cancer ... diagnostic and therapeutic context of DNA Damage. *These results indicate that RAD53 is a signal transducer in the DNA damage ...
Environmental exposures may damage DNA in as few as three days. May 14, 2021. May 17, 2009. American Thoracic Society ... Blood DNA samples were collected on the morning of the first day of the work week, and again after three days of work. ... "The changes in DNA methylation we observed are reversible and some of them are currently being used as targets of cancer drugs ... "We aimed at investigating whether exposure to particulate matter induced changes in DNA methylation in blood from healthy ...
Folates, a group of water-soluble B vitamins present in high concentrations in green, leafy vegetables, maintain DNA stability ... Folate and cancer: how DNA damage, repair and methylation impact on colon carcinogenesis J Inherit Metab Dis. 2011 Feb;34(1): ... Folates, a group of water-soluble B vitamins present in high concentrations in green, leafy vegetables, maintain DNA stability ... Folate maintains genomic stability by regulating DNA biosynthesis, repair and methylation. Folate deficiency induces and ...
Avhandlingar om DNA DAMAGE. Sök bland 109715 avhandlingar från svenska högskolor och universitet på Avhandlingar.se. ... DNA damage. . *. 1. Modeling DNA Damage. Författare :Patric Schyman; Aatto Laaksonen; Sason Shaik; Stockholms universitet; []. ... DNA damage; DNA repair; DSB; NHEJ; DNA-PK; ionizing radiation; heat-labile sites; Biomedical Radiation Science; Biomedicinsk ... DNA damage; DNA replication; homologous recombination; DNA double strand-breaks; ionizing radiation; alkylating agents; NATURAL ...
Scientists revealed that damaged DNA in overheated food can be absorbed during digestion and incorporated into the DNA of those ... How DNA becomes damaged. For those reactive species to cause DNA damage, they must physically encounter DNA in a cell to ... the lab-grown cells showed significant DNA damage resulting from taking up heat-damaged DNA components. As for the mice, DNA ... Eating very hot food can cause damage to your DNA, new study finds. Scientists revealed that damaged DNA in overheated food can ...
University of Utah chemists devised a new way to detect chemical damage to DNA that sometimes leads to genetic mutations ... Method detects DNA damage that can lead to disease. DNA is a molecule, shaped like a double-stranded helix that contains the ... "We have a way of marking and copying DNA damage sites so that we can preserve the information of where and what the damage was ... This graphic shows a new method, developed at the University of Utah, for identifying DNA lesions, or sites of damage on DNA ...
Even modest drinking may be enough to induce DNA damage in the form of oxidative stress. ... Even modest drinking may be enough to induce DNA damage in the form of oxidative stress. Photo Courtesy of Shutterstock *Share ... Modest Alcohol Consumption Tied To DNA Damage In College Kids. Dec 30, 2013 04:45 PM. By ... Researchers have found that smoking causes chromosomal damage and speeds up aging. The risk of damage can be reduced by ...
... a complex summary of GVHD pathogenesis with focus on references considering basic biological processes such as DNA damage ... Graft versus Host Disease: From Basic Pathogenic Principles to DNA Damage Response and Cellular Senescence. Adam Kuba. 1and ... ATM kinase has a crucial role in the initiation of SASP in the DNA damage response and is required for the secretion of the two ... S. Gasser, S. Orsulic, E. J. Brown, and D. H. Raulet, "The DNA damage pathway regulates innate immune system ligands of the ...
Diabetes Damages Sperm DNA. By Will Parker on May 3, 2007 in News ... Diabetic men have greater levels of DNA damage to their sperm, which may affect their fertility and their offsprings future ... Other studies have already shown that, while the female egg has a limited ability to repair damaged sperm DNA, fragmentation ... "This [study] is important, particularly given the overwhelming evidence that sperm DNA damage impairs male fertility and ...
UVA radiation is believed to tremendously damage the skin but this harm may be reduced or completely avoided by using a sun ... They then evaluated DNA damage and the ability of these cells to repair damaged DNA. The authors claimed to be successful in ... Transcription and DNA replication in melanocytes may be adversely damaged due to this oxidative DNA. UVA-induced oxidative DNA ... DNA damage is assumed to be led by sunlight in the form of UVA radiation, as it is responsible for the oxygen caused in ...
DNA distortion and damage. Cells are regularly exposed to mutagens-factors in the environment which can damage DNA and generate ... DNA polymerase fills the gap with new nucleotides, and then the DNA ligase enzyme seals the edges between the new and old DNA. ... Damaged DNA can be detected and repaired in a process called nucleotide excision repair (NER). NER employs a set of specialized ... Exposure to mutagens can damage DNA and result in bulky lesions that distort the double-helix structure or impede proper ...
Research has shown that when DNA damage occurs, a key enzyme - called ataxia telangiectasia mutated protein, or ATM - becomes ... Mouse Study Finds That Mutant Enzyme is Able to Help Protect DNA From Damage. Research has shown that when DNA damage occurs, a ... to sites of DNA damage and phosphorylates other proteins that are necessary for halting the cell cycle and repairing DNA damage ... Bringing the cell cycle to a standstill after DNA damage occurs allows time for the cells machinery to fix the errors before ...
"The proposed project aims to identify the dynamics of RNA-protein interactions during DNA damage response (DDR) using high- ... These activate the ""late"" transcriptional reprogramming influencing DNA repair, cell cycle regulation and apoptosis. However ...
  • The published original articles or reviews will open new paths toward the understanding of the generation and repair of DNA lesions, thus making it possible to advance knowledge of the processes that are at the origin of tumorigenesis. (mdpi.com)
  • We also measured 8-nitroguanine formation as an indicator of inflammation-related DNA lesions. (hindawi.com)
  • In conclusion, CD44v6 and ALDH1A1 are candidate stem cell markers for NPC, and the increased formation of DNA lesions by inflammation may result in the mutation of stem cells, leading to tumor development in NPC. (hindawi.com)
  • DNA lesions such as 8-nitroguanine and 8-oxodG with mutagenic properties occur in several types of inflammation-related cancer tissues [ 10 ]. (hindawi.com)
  • Ionizing radiation induces a range of different DNA lesions. (avhandlingar.se)
  • In terms of mutation frequency and mammalian cell survival, the most critical of these lesions is the DNA double-strand break (DSB). (avhandlingar.se)
  • This is demonstrated by the formation of DNA double strand breaks (DSBs), considered to be the most toxic DNA lesions, at stalled replication forks. (avhandlingar.se)
  • Jan Riedl, a University of Utah postdoctoral fellow and the study's first author, says 99 percent of DNA lesions - damage to the chemical bases known as A, C, G and T that help form the DNA double helix - are repaired naturally. (utah.edu)
  • The chemists say their new method will let researchers study chemical details of DNA lesions or damage. (utah.edu)
  • The new method for finding DNA lesions combines other, existing techniques. (utah.edu)
  • This graphic shows a new method, developed at the University of Utah, for identifying DNA lesions, or sites of damage on DNA strands that can lead to disease-causing mutations. (utah.edu)
  • Exposure to mutagens can damage DNA and result in bulky lesions that distort the double-helix structure or impede proper transcription. (jove.com)
  • Taking advantage of this finding, we proposed a paper-assisted TdT (PAT) assay for absolute quantification of alkylated DNA lesions (N7-methylguanine), DNA deamination (cytosine-to-uracil) and DNA oxidation (8-oxo-7,8-dihydroguanine) by combining various DNA glycosylases. (rsc.org)
  • This PAT assay provides a low-cost, high throughput and easy to use method for quantifying the absolute levels of various types of DNA lesions, thus making it well-suited for drug development, genotoxicity testing, and environmental toxicology. (rsc.org)
  • A byproduct of alcohol consumption causes mutations in the DNA of mouse blood stem cells, and some of the breaks are not repaired. (the-scientist.com)
  • That uptake directly places damage in the consumer's DNA, potentially triggering genetic mutations that may eventually lead to cancer and other diseases. (stanford.edu)
  • He found that RNR2 RNA levels increased dramatically, even more than the protein levels, upon exposure of cells to DNA damage and that mutations in RNR2 resulted in hypersensitivity to DNA damage. (the-scientist.com)
  • A prerequisite for a multicellular organism to survive is the ability to correctly replicate and repair DNA while minimizing the number of heritable mutations. (avhandlingar.se)
  • University of Utah chemists devised a new way to detect chemical damage to DNA that sometimes leads to genetic mutations responsible for many diseases, including various cancers and neurological disorders. (utah.edu)
  • Experts from the NYU School of Medicine claim that UVA radiation damages the DNA in human melanocyte cells further leading mutations to cause melanoma. (healthjockey.com)
  • Cells are regularly exposed to mutagens-factors in the environment which can damage DNA and generate mutations. (jove.com)
  • If these errors are not fixed, the damage can cause mutations which in turn can result in cancer or disease depending on which sequences are disrupted. (jove.com)
  • Antioxidant enzymes are deployed to mop up reactive oxygen species at their source before they reach DNA, a defensive strategy that protects the roughly 3 billion nucleotides from suffering potentially catastrophic mutations. (technologynetworks.com)
  • The study appeared February 19 in Science, in an article entitled, "Chemiexcitation of melanin derivatives induces DNA photoproducts long after UV exposure. (genengnews.com)
  • This creates a quantum triplet state that has the energy of a UV photon but induces CPDs by energy transfer to DNA in a radiation-independent manner. (genengnews.com)
  • MMC induces DNA damage to quiescent corneal keratocytes, which remains unrepaired, resulting in abnormal cell replication and gene transcription that leads to long-term effects on corneal repair. (molvis.org)
  • Mitomycin C (MMC) belongs to a family of anti-tumor quinolone antibiotics derived from Streptomyces caespitosus , and functions as a powerful bifunctional alkylating agent that induces DNA interstrand crosslinks (ICL). (molvis.org)
  • On the other hand, etoposide treatment reliably induces DNA damage-related senescence in human articular chondrocytes evidenced by loss of proliferative capacity, DNA damage accumulation, and expression of some SASP components. (aging-us.com)
  • Increased intakes of niacin (vitamin B3) from the diet may protect against DNA damage in people exposed to ionizing radiation such as pilots, says a new study. (nutraingredients.com)
  • However, they did find a comparatively increased frequency of micronuclei in the radiologists, which are fragments of DNA produced when ionizing radiation breaks DNA strands. (auntminnie.com)
  • In this thesis methods of computational chemistry have been used to examine DNA damaging processes initiated by ionizing radiation, free radicals, or Low-Energy Electrons (LEE). (avhandlingar.se)
  • Fourth, another chemical label, named 18-crown-6 ether, is affixed to the unnatural base pair on all the DNA strands, which are then read or sequenced using a kind of nanopore sequencing developed a few years ago by Burrows and Utah chemist Henry White. (utah.edu)
  • At that location the strands are separated and endonuclease enzymes such as UvrC cleave the strand on both sides of the damage. (jove.com)
  • Next, NER proteins separate the strands and excise the damaged area. (jove.com)
  • The first two proteins work together as a complex, traveling along the DNA strands to detect any physical aberrations. (jove.com)
  • Once identified, the strands at the damaged location are separated, and endonuclease enzymes such as UvrC cut and excise the affected region. (jove.com)
  • Both are responsible for producing breaks in DNA strands. (laserfocusworld.com)
  • Importantly, we observed that TdT efficiently incorporates fluorescently labeled dUTP on to 3′-OH ends of DNA strands in a strictly controllable manner on cellulose paper, in comparison to its distributive mode of DNA synthesis in solution. (rsc.org)
  • Etoposide works by breaking DNA strands and blocking an enzyme which helps repair the damage. (technologynetworks.com)
  • A new genetic sensor called PRISM makes use of a host cell's DNA replication machinery to trigger fluorescence in neurons with damaged DNA. (the-scientist.com)
  • Epirubicin was reported to block DNA replication either through direct DNA damage or indirectly through inhibition of replication proteins such as topoisomerase. (nature.com)
  • Elledge's idea that eukaryotic cells sense the progress of DNA replication and transform that information into a DNA-damage response was new. (the-scientist.com)
  • Those results led him to study how cells monitor roadblocks to replication and DNA damage, such as nicks and double-stranded breaks, and how the cell handles that information. (the-scientist.com)
  • Faithful DNA replication is essential and the induction of replication stress may have profound effects on genomic integrity. (avhandlingar.se)
  • Transcription and DNA replication in melanocytes may be adversely damaged due to this oxidative DNA. (healthjockey.com)
  • These include bends or kinks in the structure which can block DNA replication or transcription. (jove.com)
  • These data indicate that the effect of MMC on corneal scarring and haze is related to the generation of DNA ICLs leading to defective cell replication and gene expression. (molvis.org)
  • Rabbit polyclonal to Parp.Poly(ADP-ribose) polymerase-1 (PARP-1), also designated PARP, is a nuclear DNA-bindingzinc finger protein that influences DNA repair, DNA replication, modulation of chromatin structure,and apoptosis. (2011globalhealth.org)
  • Without vigilant repair, cancer would run rampant, and now scientists at the University of Pittsburgh have gotten a glimpse of how one protein in particular keeps DNA damage in check. (sciencedaily.com)
  • According to a study published today in Nature Structural and Molecular Biology , a protein called UV-DDB -- which stands for ultraviolet-damaged DNA-binding -- is useful beyond safeguarding against the sun. (sciencedaily.com)
  • Buildup of a DNA-repair protein in brain cells spurs shut-eye in the fish, a study finds, and similar results in mice suggest the mechanism is widespread in animals. (the-scientist.com)
  • Researchers find that temporary double-stranded DNA breaks commonly result from normal neuron activation-but expression of an Alzheimer's-linked protein increases the damage. (the-scientist.com)
  • The oversight is understandable, since DNA does not appear on nutrition labels in the same manner as protein, carbohydrates, fat, vitamins, and minerals. (stanford.edu)
  • Elledge had found that Rnr2 protein levels increased when yeast cells were grown in the presence of agents that damaged DNA. (the-scientist.com)
  • Studying RNR2' s regulatory elements, he found those that were necessary to induce the production of higher protein levels in response to DNA damage and identified factors that bind these DNA elements to mediate the response of RNR2 to DNA damage. (the-scientist.com)
  • Nucleotide excision repair relies on specific protein complexes to recognize damaged regions of DNA and flag them for removal and repair. (jove.com)
  • Research has shown that when DNA damage occurs, a key enzyme - called ataxia telangiectasia mutated protein, or ATM - becomes activated. (nih.gov)
  • ATM is a protein that functions to maintain the stability of DNA. (nih.gov)
  • This mutant ATM protein also migrated to the site of DNA breaks and phosphorylated other proteins appropriately. (nih.gov)
  • The proposed project aims to identify the dynamics of RNA-protein interactions during DNA damage response (DDR) using high-throughput "omics" approaches. (europa.eu)
  • Jeffrey, I. W. , Bushell, M. , Tilleray, V. J. , Morley, S. and Clemens, M. J. (2002) Inhibition of protein synthesis in apoptosis: differential requirements by the tumor necrosis factor alpha family and a DNA-damaging agent for caspases and the double-stranded RNA-dependent protein kinase. (gla.ac.uk)
  • In contrast, treatment of cells with the DNA-damaging agent etoposide inhibits protein synthesis equally well in the presence of a pan-specific caspase inhibitor and in the presence or absence of PKR. (gla.ac.uk)
  • In contrast, key components of DNA known as nucleotides that are made available through normal breakdown of biomolecules - for instance, during digestion - are readily incorporated into the DNA of cells, suggesting a plausible and potentially significant pathway for damaged food DNA to inflict damage on other DNA downstream in consumers. (stanford.edu)
  • The phosphopentose pathway (PPP), which converts glucose-6-phosphate to ribose-5-phosphate for synthesis of nucleotides and NADPH to reduce DNA damage caused by ROS was reported to be activated in DDR 2 . (nature.com)
  • Another enzyme, DNA polymerase fills the gap with new nucleotides. (jove.com)
  • DNA polymerase fills the gap with new nucleotides, and then the DNA ligase enzyme seals the edges between the new and old DNA. (jove.com)
  • PRDX1 was also found to repair the damage by regulating the cellular availability of aspartate, a raw material that is critical for synthesising nucleotides, the building blocks of DNA. (technologynetworks.com)
  • The growth defect and the hypersensitivity to DNA-damaging agents of mdo1-1 were enhanced significantly when combined with a lesion of the ATAXIA-TELANGIECTASIA MUTATED (ATM) gene, but not of the ATM/RAD3-RELATED (ATR) gene, suggesting that the function of the MDO1 gene is closely related to that of ATM kinase. (nih.gov)
  • The excited state lifetime of compounds used in sunscreens such as menthyl anthranilate, avobenzone or padimate O is 1,000 to 1,000,000 times longer than that of melanin, and therefore they may cause damage to living cells that come in contact with them. (wikipedia.org)
  • The potential heart health benefits of omega-3s may be linked (in part at least) to their ability to protect the DNA in the cells lining blood vessels, according to a study from Japanese researchers. (nutraingredients.com)
  • As in the DNA tightrope experiment, UV-DDB rushed to the scene, and when it wasn't available, cells were more sensitive to oxidative stress. (sciencedaily.com)
  • The DNA forms known as G-quadruplexes are finally discovered in human cells. (the-scientist.com)
  • The team observed that the DNA of cells exposed to sucralose-6-acetate showed breaks, which indicates the compound is genotoxic. (healthnews.com)
  • The chemicals damage the 'tight junctions,' or interfaces, where cells in the gut wall connect to each other. (healthnews.com)
  • While it's too soon to say this occurs in humans - the study only observed heat-damaged DNA component uptake and increased DNA injury in lab-grown cells and mice - the findings could have important implications for dietary choices and public health. (stanford.edu)
  • This study investigated the role of TIGAR in DNA damage response (DDR) induced by genotoxic drugs and hypoxia in tumor cells. (nature.com)
  • To define a role of TIGAR in DNA damage, TIGAR expression was knocked down with siRNA in HepG2 cells. (nature.com)
  • Epirubicin, a DNA damaging anticancer agent and CoCl 2 , which was used to imitate hypoxia condition, was applied to induce DNA damage in TIGAR knockdown HepG2 cells. (nature.com)
  • DNA damage in HepG2 cells after TIGAR knockdown with or without treatment of epirubicin or CoCl 2 was detected by Comet assay. (nature.com)
  • Long-term exposure to low-dose radiation can cause DNA damage to lymphocyte cells in interventional radiologists - yet importantly, the doctors are likely not vulnerable to an increased risk of leukemia, according to researchers in Slovakia. (auntminnie.com)
  • His first experimental results contained a serendipitous artifact that laid the foundation for a scientific career studying how eukaryotic cells deal with damage to their DNA. (the-scientist.com)
  • To achieve this, cells need a balanced supply of deoxyribonucleoside triphosphates (dNTPs), the precursors for DNA synthesis. (avhandlingar.se)
  • Researchers identified that an abundance of fungi in the gut, particularly strains of Candida albicans yeast, could trigger an increase in immune cells, which could worsen lung damage. (medicaldaily.com)
  • Besides the secretion of pro-inflammatory cytokines (TNF-alpha, IL-1beta, and IL-6), the increased expression of receptor repertoire (pattern recognition receptors, PRR) on APCs, mostly macrophages and dendritic cells, occurs as a result of the release of endogenous and exogenous antigens (damage-associated molecular patterns, DAMPs, and pathogen-associated molecular patterns, PAMPs). (hindawi.com)
  • Moon-shong Tang, PhD, professor of environmental medicine, pathology and medicine at NYU School of Medicine and a member of the NYU Cancer Institute enlightened, "For the first time, UVA rays have been shown to cause significant damage to the DNA of human melanocyte skin cells. (healthjockey.com)
  • They then evaluated DNA damage and the ability of these cells to repair damaged DNA. (healthjockey.com)
  • The authors claimed to be successful in identifying DNA damage in all melanocyte cells. (healthjockey.com)
  • They further revealed that these cells were unable to repair the damage. (healthjockey.com)
  • When normal skin cells were exposed to UVA light no harm was seemingly discovered in their DNA. (healthjockey.com)
  • UVA-induced oxidative DNA damage in melanocytes and the inherently reduced repair capacity in these cells are assumed to be the two key factors responsible for melanoma on the skin. (healthjockey.com)
  • Although enzyme mechanisms may differ between humans and mice, gaining a better understanding of DNA damage repair might someday allow us to specifically alter ATM in cancer cells. (nih.gov)
  • Bringing the cell cycle to a standstill after DNA damage occurs allows time for the cell's machinery to fix the errors before abnormal cells are generated. (nih.gov)
  • Even when immune cells from Atm-S1987A-rescued mice are treated with radiation, several signs of normal cell cycle inhibition were observed, including decreased rate of DNA synthesis and decreased cell proliferation. (nih.gov)
  • The mutant plants also showed several phenotypes related to DNA damage, suggesting that the mutant cells are exposed constitutively to DNA damage even without external genotoxic stress. (nih.gov)
  • The results thus suggested that the MDO1 gene product is required for the maintenance of stem cells through a reduction in DNA damage. (nih.gov)
  • The image illustrates the location of DNA damage (in the nucleus of these four cells - green) and the colocalization of PRDX1 (in red, same place). (technologynetworks.com)
  • Cells are thought to delicately balance their energy needs and avoid damaging DNA by containing metabolic activity outside the nucleus and within the cytoplasm and mitochondria. (technologynetworks.com)
  • If DNA damage occurs anyway, cells pause momentarily and carry out repairs, synthesising new building blocks and filling in the gaps. (technologynetworks.com)
  • These experiments revealed that cells order the enzyme PRDX1, an antioxidant enzyme also normally found in mitochondria, to travel to the nucleus and scavenge reactive oxygen species present to prevent further damage. (technologynetworks.com)
  • Using fast DNA tracking microscopy in S. cerevisiae cells and improved analysis of mean square displacements, we quantified DNA motion at time scales ranging from 10 milliseconds to minute and found that following DNA damage, DNA exhibits distinct sub-diffusive regimes. (escholarship.org)
  • Radiation can damage the DNA in our cells. (cdc.gov)
  • UV light, it turns out, may act via a "light" or a "dark" biochemical pathway to cause a type of DNA damage known as a cyclobutane pyrimidine dimer (CPD). (genengnews.com)
  • A new tool called PRISM draws on virus-host interactions and a DNA repair pathway to help researchers visualize how cellular stress may contribute to neurodegenerative disease. (the-scientist.com)
  • Then I thought, there must be a sensory pathway that recognizes the DNA damage that's going on in the cell," says Elledge. (the-scientist.com)
  • It was published in ACS Central Science under the title "Possible Genetic Risks from Heat-Damaged DNA in Food. (jpost.com)
  • Researchers have warned manufacturers of functional foods and supplements to keep polyphenol doses low, following conclusions that low concentrations protect but high concentrations may damage DNA. (nutraingredients.com)
  • Using single-molecule imaging, researchers witness how molecules find and fix damaged DNA. (sciencedaily.com)
  • The researchers described how they tested the extent of damage that occurred after sun exposure by preventing normal DNA repair in mouse samples. (genengnews.com)
  • The researchers proceeded to cook foods - namely, ground beef, ground pork, and potatoes - through either 15-minute boils at 100 degrees Celsius (212 degrees Fahrenheit) or 20-minute mild roastings at 220 C (about 430 F). The Stanford researchers then extracted DNA from these foods and sent the samples to collaborators at NIST. (stanford.edu)
  • CDDR will collaborate with both basic researchers and clinicians to develop innovative research involving DNA damage and DNA repair experimentation. (dana-farber.org)
  • These methodologies will be especially helpful to Dana-Farber researchers working in related fields of cancer biology, as well as to the Institute's clinical investigators developing new drugs for cancers with specific underlying DNA repair defects. (dana-farber.org)
  • First, the researchers find the damage and cut it out of the DNA the same way a cell does naturally, using what is called "base excision repair," the discovery of which won a Nobel Prize in Chemistry this year for Tomas Lindahl, a scientist in England. (utah.edu)
  • Researchers have found that smoking causes chromosomal damage and speeds up aging. (medicaldaily.com)
  • Diabetic men have greater levels of DNA damage to their sperm, which may affect their fertility and their offspring's future health, say researchers at Queen's University, Belfast. (scienceagogo.com)
  • Earlier work carried out by the researchers showed that OH radicals were four times more likely than electrons to produce double-strand breaks in DNA. (laserfocusworld.com)
  • Memorial Sloan Kettering Cancer Center (MSK) researchers have discovered that innate immune signaling drives late cardiac toxicity after DNA-damaging cancer therapies. (mskcc.org)
  • The researchers experimentally induced DNA damage in human cell lines using a common chemotherapy medication known as etoposide. (technologynetworks.com)
  • The researchers observed that cellular respiratory enzymes, a major source of reactive oxygen species, relocated from the mitochondria to the nucleus in response to DNA damage. (technologynetworks.com)
  • This study used etoposide, to induce DNA damage-related senescence or chronic exposure to IL-1β to entail inflammation-related senescence in human OA chondrocytes. (aging-us.com)
  • A research team led by Sara Sdelci at the Centre for Genomic Regulation (CRG) in Barcelona and Joanna Loizou at the CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences in Vienna and the Medical University of Vienna addressed this challenge by carrying out various experiments to identify which metabolic enzymes and processes are essential for a cell's DNA damage response. (technologynetworks.com)
  • DNA is the cell's "instruction manual" that controls a cell's normal growth and function. (cdc.gov)
  • From DNA damage to cellular miscommunication, aging is a mysterious and multifarious process. (the-scientist.com)
  • Folates, a group of water-soluble B vitamins present in high concentrations in green, leafy vegetables, maintain DNA stability through their ability to donate one-carbon units for cellular metabolism. (nih.gov)
  • This review presents a complex summary of GVHD pathogenesis with focus on references considering basic biological processes such as DNA damage response and cellular senescence. (hindawi.com)
  • This review summarizes the updated GVHD pathogenesis linking GVHD with biological processes such as DNA damage response (DDR) and cellular senescence (Figure 1 ). (hindawi.com)
  • The current understanding of aGVHD pathogenesis can be summarized as (1) initial tissue damage induced by the conditioning regimen followed by the denudation of auto- and alloantigens accompanied by massive inflammatory cytokine secretion ("cytokine storm") activating APCs, (2) auto- and alloantigen presentation mediated by APCs together with the costimulatory signaling prime donor's cytotoxic T lymphocytes and their proliferation, and (3) the migration of activated cellular effectors toward GVHD target tissues. (hindawi.com)
  • If the damage cannot be accurately fixed, the cell may commit cellular suicide rather than lose control of growth. (nih.gov)
  • Several hallmarks of cellular senescence, such as cell cycle arrest, expression of cyclin-dependent kinase inhibitors, DNA damages, and senescence-associated secretory profile were evaluated. (aging-us.com)
  • Despite the central role of cellular metabolism in maintaining genome integrity, there has been no systematic, unbiased study on how metabolic perturbations affect the DNA damage and repair process. (technologynetworks.com)
  • Human mediator of DNA damage checkpoint 1 (hMDC1) is an essential component of the cellular response to DNA double strand breaks. (ox.ac.uk)
  • Instead, this caspase activity leads to DNA damage that, in turn, promotes genomic instability , cellular transformation, and tumorigenesis . (bvsalud.org)
  • Further study of reactive oxygen species was conducted using analysis of cellular H2O2 generation, lipid peroxidation of cellular membranes and DNA damage. (cdc.gov)
  • Bimolecular reactions can occur either between the excited chromophore and DNA or between the excited chromophore and another species, to produce free radicals and reactive oxygen species. (wikipedia.org)
  • These reactions also create reactive oxygen species, dangerous by-products like hydrogen peroxide which damage the building blocks of DNA in the same way oxygen and water corrode metal and form rust. (technologynetworks.com)
  • Surprisingly, inducing DNA damage resulted in reactive oxygen species being generated and accumulating inside the nucleus. (technologynetworks.com)
  • Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. (nature.com)
  • SnCs exhibit irreversible growth arrest accompanied by increased expression of cyclin-dependent kinase inhibitors (CDKi) such as p16 INK4a , and p21 Cip1 , accumulation of DNA damages, and secretion of diverse bioactive molecules known as the senescence-associated secretory phenotype (SASP). (aging-us.com)
  • The process begins when an enzyme complex, shown here in E. coli, containing the proteins UvrA and UvrB, travels along the DNA and detects the physical aberration of the lesion. (jove.com)
  • The enzyme DNA helicase then removes the cut pieces. (jove.com)
  • Finally, the enzyme DNA ligase seals the edges between the new and old DNA completing the repair. (jove.com)
  • OxiSelectâ„¢ Oxidative UV-induced DNA Damage ELISA Kit (CPD Quantitation) is an enzyme immunoassay developed for rapid detection and quantitation of CPD in any DNA samples. (antibodies-online.com)
  • Discoveries span from disrupted gene patterns and epigenomic changes to the significance of microglia and DNA damage in neurons. (scitechdaily.com)
  • These reactive chemical species can reach DNA by diffusion and the bimolecular reaction damages the DNA (oxidative stress). (wikipedia.org)
  • Eight weeks of supplementation with watercress may reduce markers of oxidative stress and damage after exhaustive exercise, suggests a new study. (nutraingredients.com)
  • There's damage from oxidative stress due to inflammation and infection, too much metabolism, or environmental chemicals. (neurosciencenews.com)
  • This [study] is important, particularly given the overwhelming evidence that sperm DNA damage impairs male fertility and reproductive health. (scienceagogo.com)
  • Since sperm DNA damage can cause miscarriage and infertility, it can be one of the causes of hatching failure in birds. (lu.se)
  • However, it has been shown in humans that sperm DNA damage can cause infertility, we do not know whether pollution can affect sperm DNA integrity in bird populations. (lu.se)
  • It also has the ability to find damage buried in chromosomes and help DNA repair molecules go places they otherwise couldn't, the way a helicopter can navigate really hilly areas. (sciencedaily.com)
  • Many studies link the consumption of charred and fried foods to DNA damage, and attribute the harm to certain small molecules that form so-called reactive species in the body. (stanford.edu)
  • Of note, however, those small molecules produced in typical cooking number many thousands of times less than the amount of DNA occurring naturally in foods, Kool says. (stanford.edu)
  • Investigating the nitty-gritty of how complex DNA molecules are repaired - both after unavoidable natural errors, as well as damage induced by environmental exposures - is a chief aim of Kool's lab at Stanford. (stanford.edu)
  • For example, a roughly 500 gram (16 ounce) beef steak contains over a gram (0.04 ounce) of cow DNA, suggesting that human exposure to potentially heat-damaged DNA is likewise not negligible. (stanford.edu)
  • We aimed at investigating whether exposure to particulate matter induced changes in DNA methylation in blood from healthy subjects who were exposed to high levels of particulate matter in a foundry facility. (scienceblog.com)
  • Exposure of DNA to ultraviolet light and certain chemical carcinogens can cause a bulky lesion. (jove.com)
  • Here we show that cell cycle progression through mitosis following double-stranded DNA breaks leads to the formation of micronuclei, which precede activation of inflammatory signalling and are a repository for the pattern-recognition receptor cyclic GMP-AMP synthase (cGAS). (nature.com)
  • It identifies DNA damage so that another crew can come in and patch and seal it," said study senior author Bennett Van Houten, Ph.D., professor of pharmacology and chemical biology at the Pitt School of Medicine and UPMC Hillman Cancer Center. (sciencedaily.com)
  • Chemical Found in Common Sweetener Damages DNA. (healthnews.com)
  • For those reactive species to cause DNA damage, they must physically encounter DNA in a cell to trigger a deleterious chemical reaction - a rare event, in all likelihood. (stanford.edu)
  • Working at night resulted in lower melatonin levels and a less of the chemical which repairs DNA in the urine. (healthstatus.com)
  • These results indicate that the radical generation and the damage caused by them is not only a function of surface area but is also influenced by changing chemical and other characteristics due to particle size. (cdc.gov)
  • Note: Samples with high concentrations of CPD may be further diluted 2-4 fold in 4 μg/mL Reduced DNA. (antibodies-online.com)
  • These outcomes correlated with nFe2O3 dissolution, increased intracellular iron, and genotoxicity, as well as significant changes in pathways related to DNA damage repair and autophagic processes. (cdc.gov)
  • We report on a paper-based sensor capable of performing template-independent DNA synthesis by terminal deoxynucleotidyl transferase (TdT). (rsc.org)
  • Indirect DNA damage occurs when a UV-photon is absorbed in the human skin by a chromophore that does not have the ability to convert the energy into harmless heat very quickly. (wikipedia.org)
  • A collaborative project between TIFR Mumbai, the Centre for Excellence in Basic Sciences, Mumbai, and Manipal University, the experiments used different incident laser energies and various external focusing conditions to establish that DNA damage occurs in two distinct regimes. (laserfocusworld.com)
  • As a start to designing those gene-targeting tools, Elledge, now a professor of genetics at Harvard Medical School, began by trying to clone the mammalian homolog of recA , a bacterial gene required for DNA repair via recombination. (the-scientist.com)
  • The gene p53 suppresses cancer and inflammation in the body, and NIEHS investigators speculate that changes in p53 lead to changes in inflammation and the ability to repair DNA damage. (nih.gov)
  • Cis ligation of CD137 relative to the TCR-CD3 complex results in more intense canonical and non-canonical NF-κB signaling and provides a more robust induction of cell cycle and DNA damage repair gene expression programs. (lu.se)
  • Daily supplements of astaxanthin - the pigment that gives salmon its pink colour - may protect DNA from damage and boost the immune response in healthy young women, says a new study. (nutraingredients.com)
  • CDDR employs a wide range of technologies relevant to the measurement of DNA damage response in primary human cancers and cell lines, including cytotoxicity measurements, radiation sensitivity testing, and proteomic biomarker measurements. (dana-farber.org)
  • Radiation and other factors can create double-stranded breaks in DNA, and evidence suggests that ATM becomes activated in response to this DNA damage. (nih.gov)
  • Such a pattern of dynamics is consistent with a global increase in chromatin persistence length in response to DNA damage. (escholarship.org)
  • Mediator of DNA damage checkpoint 1 (MDC1) regulates mitotic progression. (ox.ac.uk)
  • Third, the DNA with the damage site labeled by an unnatural third base pair is then amplified or copied millions of times using a well-known existing method called PCR, or polymerase chain reaction. (utah.edu)
  • The reactions were conducted in initiation of the study, had received no antiretroviral a 50-L PCR mixture containing Taq DNA polymerase, treatment. (cdc.gov)
  • CDDR Director Alan D'Andrea, MD, is an expert in the area of DNA repair and genomic stability. (dana-farber.org)
  • Folate maintains genomic stability by regulating DNA biosynthesis, repair and methylation. (nih.gov)
  • Studies conducted in the Center will also facilitate the development of new classes of anti-cancer drugs, such as DNA Repair inhibitors and Cell Cycle inhibitors. (dana-farber.org)
  • The new promising marker Schlafen 11 seems to correlate with sensitivity or resistance to DNA-damaging agents, including platinum compounds or PARP inhibitors in various types of cancer. (muni.cz)
  • NER employs a set of specialized proteins that first scan DNA to detect a damaged region. (jove.com)
  • In prokaryotes, the NER complex consists of the three Uvr proteins, but in eukaryotes, more than a dozen proteins operate to regulate DNA repair. (jove.com)
  • Subsequently, activated ATM then migrates to sites of DNA damage and phosphorylates other proteins that are necessary for halting the cell cycle and repairing DNA damage. (nih.gov)
  • Once ATM is recruited to the double-stranded DNA break, lots of other proteins also relocate to the same site for phosphorylation. (nih.gov)
  • We investigated the presence of HGyV DNA in 1, VP2, and VP3) that encode proteins of 465, 231, and blood samples of 301 persons in Italy using specifi c PCRs. (cdc.gov)
  • The conditioning-induced damage of recipients' tissues leads to danger signal secretion [ 13 ]. (hindawi.com)
  • However, our data suggest that neither increased phosphorylation of eIF2α nor increased [eIF4E.4E-BP1] complex formation is essential for the inhibition of overall translation by the DNA-damaging agent. (gla.ac.uk)
  • dubious - discuss] The traveling nature of the indirect DNA damage can be seen in the fact that the malignant melanoma can occur in places that are not directly illuminated by the sun-in contrast to basal-cell carcinoma and squamous cell carcinoma, which appear only on directly illuminated locations on the body. (wikipedia.org)
  • Now, new research by the same scientific team found that sucralose-6-acetate may be genotoxic and negatively impact DNA . (healthnews.com)
  • Such dichotomous kinetics implicate additional rate-limiting steps that are essential for DNA-damage-induced inflammation. (nature.com)
  • Nitrative and oxidative DNA damage plays an important role in inflammation-related carcinogenesis. (hindawi.com)
  • Moreover, 8-nitroguanine is a potentially mutagenic DNA lesion and has been reported to play a significant role in and to be a biomarker for inflammation-related carcinogenesis [ 5 ]. (hindawi.com)
  • In the time it takes you to read this sentence, every cell in your body suffers some form of DNA damage. (sciencedaily.com)
  • But there was also evidence from studies suggesting that melanin was associated with skin cell damage. (genengnews.com)
  • Comparing the quality of DNA in sperm from diabetic and non-diabetic men, the study found that DNA fragmentation was greater in diabetic men (52 percent, versus 32 percent in non-diabetic men), and that there were more deletions of DNA in the cell mitochondria. (scienceagogo.com)
  • These alterations could make tumors more sensitive to DNA damage and cell death radiation therapy for cancer," said senior author André Nussenzweig, Ph.D., senior investigator in NCI's Experimental Immunology Branch. (nih.gov)
  • These activate the ""late"" transcriptional reprogramming influencing DNA repair, cell cycle regulation and apoptosis. (europa.eu)
  • Extract DNA from cell or tissue samples using a commercial DNA Extraction kit or other desired method. (antibodies-online.com)
  • The aim of the present investigation was to study the effect of the intake of a single portion of blood orange juice (BOJ, 300 ml, providing 150 mg vitamin C) on mononuclear blood cell (MNBC) DNA damage, compared with a drink supplemented with the same amount of vitamin C (C-drink) or sugars (S-drink). (cambridge.org)
  • The consumption of alcohol is always a controversial topic and a Reuter's article explored how drinking can damage stem cell DNA. (mercola.com)
  • The purpose of this study was to determine the acute and long-term effects of mitomycin C (MMC) on quiescent rabbit corneal keratocytes regarding cell proliferation, myofibroblast differentiation and DNA repair. (molvis.org)
  • MMC also induced phosphorylation of the nuclear histone marker of DNA damage, γH2AX (a member of the H2A histone family), without induction of cell cycle entry or immediate DNA repair measured by Comet assay. (molvis.org)
  • Limited mitochondrial permeabilization causes DNA damage and genomic instability in the absence of cell death. (bvsalud.org)
  • When DNA is damaged, a cell can begin growing out of control and create a cancer tumor. (cdc.gov)
  • For the first time, Van Houten's team witnessed this molecular tango along a "tightrope" of DNA slung between two silica beads, using real-time, single-molecule imaging. (sciencedaily.com)
  • DNA is a molecule of paramount importance due to its fundamental biological role in carrying and storing genetic information. (mdpi.com)
  • DNA is a molecule, shaped like a double-stranded helix that contains the genetic instructions that living organisms use to develop, function and reproduce. (utah.edu)
  • which makes it easier to detect the site of DNA damage by passing the strand of DNA through molecule-size nanopore. (utah.edu)
  • Such sequencing involves determining the order and location of bases on a DNA strand - including damage sites labeled by unnatural bases - by passing the strand through a molecule-size pore or nanopore. (utah.edu)
  • The characteristics of its genome--a aplastic anemia, hemorrhage, and lymphoid depletion) and single, closed molecule of circular, negative-sense DNA increased death in young chicken ( 11 ). (cdc.gov)
  • The changes in DNA methylation we observed are reversible and some of them are currently being used as targets of cancer drugs. (scienceblog.com)
  • Other studies have already shown that, while the female egg has a limited ability to repair damaged sperm DNA, fragmentation beyond this threshold may result in increased rates of embryonic failure and pregnancy loss," said co-researcher Sheena Lewis. (scienceagogo.com)
  • It was shown in medical studies that in a highly polluted area the percentage of sperm DNA fragmentation (measurement for DNA damage) can be above 30%, which can lead to miscarriage in women or lead to infertility in men. (lu.se)
  • Daily folic acid supplements could improve DNA stability and reduce the risk of certain cancers linked to faults in the genetic code, suggests new research. (nutraingredients.com)
  • As such, its stability is key for safeguarding DNA integrity from external stress. (mdpi.com)
  • A better understanding of these DNA repair defects will lead to improved diagnostic methods for predicting the best course of cytotoxic chemotherapy and radiation for a given cancer patient ( "precision" or "personalized" medicine ). (dana-farber.org)
  • This is achieved, inter alia, by means of specific repair mechanisms that ensure an efficient and prompt correction of the DNA defects. (mdpi.com)
  • The Center for DNA Damage and Repair (CDDR) explores the various DNA repair defects in human cancers which underlie genomic instability. (dana-farber.org)
  • These PFGs can form due to DNA double-strand breaks caused by radiation. (auntminnie.com)
  • One of the powerful things about our method is we can read more than a single damaged site [and up to dozens] on the same strand of DNA. (utah.edu)
  • Ultimately, whether the increased presence of micronuclei due to DNA damage translates to higher health risks in interventional radiologists will require further research, the authors noted. (auntminnie.com)
  • Together they inflicted oxidative damage to the chromosomes' protective endcaps -- called telomeres. (sciencedaily.com)
  • But what has never been documented before our study is the potentially large quantities of heat-damaged DNA available for uptake into a consumer's own DNA. (stanford.edu)
  • And because melanocytes have a reduced capacity to repair DNA damage from UVA radiation, they mutate more frequently, potentially leading to the development of melanoma. (healthjockey.com)
  • When telomeres (green) undergo damage (bottom row), UV-DDB (red) rushes to the scene (yellow). (cmu.edu)
  • This new evidence points to UV-DDB being a scout for general DNA damage and an overseer of the molecular repair crew that fixes it. (sciencedaily.com)
  • This type of damage can be fixed by a process called nucleotide excision repair. (jove.com)
  • Damaged DNA can be detected and repaired in a process called nucleotide excision repair (NER). (jove.com)