A chromosome instability syndrome resulting from a defective response to DNA double-strand breaks. In addition to characteristic FACIES and MICROCEPHALY, patients have a range of findings including RADIOSENSITIVITY, immunodeficiency, increased cancer risk, and growth retardation. Causative mutations occur in the NBS1 gene, located on human chromosome 8q21. NBS1 codes for nibrin, the key regulator protein of the R/M/N (RAD50/MRE11/NBS1) protein complex which senses and mediates cellular response to DNA DAMAGE caused by IONIZING RADIATION.
A congenital abnormality in which the CEREBRUM is underdeveloped, the fontanels close prematurely, and, as a result, the head is small. (Desk Reference for Neuroscience, 2nd ed.)
A type of chromosomal aberration involving DNA BREAKS. Chromosome breakage can result in CHROMOSOMAL TRANSLOCATION; CHROMOSOME INVERSION; or SEQUENCE DELETION.
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).
A characteristic symptom complex.
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.
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.
Enzymes that are involved in the reconstruction of a continuous two-stranded DNA molecule without mismatch from a molecule, which contained damaged regions.
Clinical conditions caused by an abnormal chromosome constitution in which there is extra or missing chromosome material (either a whole chromosome or a chromosome segment). (from Thompson et al., Genetics in Medicine, 5th ed, p429)
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.
'Abnormalities, Multiple' is a broad term referring to the presence of two or more structural or functional anomalies in an individual, which may be genetic or environmental in origin, and can affect various systems and organs of the body.
Syndromes in which there is a deficiency or defect in the mechanisms of immunity, either cellular or humoral.
ELECTROMAGNETIC RADIATION or particle radiation (high energy ELEMENTARY PARTICLES) capable of directly or indirectly producing IONS in its passage through matter. The wavelengths of ionizing electromagnetic radiation are equal to or smaller than those of short (far) ultraviolet radiation and include gamma and X-rays.
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.
A specific pair of GROUP C CHROMOSOMES of the human chromosome classification.
An increased tendency to acquire CHROMOSOME ABERRATIONS when various processes involved in chromosome replication, repair, or segregation are dysfunctional.
Genes that influence the PHENOTYPE only in the homozygous state.
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.
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.
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.
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.
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.
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 group of enzymes that catalyzes the phosphorylation of serine or threonine residues in proteins, with ATP or other nucleotides as phosphate donors.
Interruptions in the sugar-phosphate backbone of DNA, across both strands adjacently.
Connective tissue cells which secrete an extracellular matrix rich in collagen and other macromolecules.
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.
Abnormal number or structure of chromosomes. Chromosome aberrations may result in CHROMOSOME DISORDERS.
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.
Disorders resulting from defective DNA REPAIR processes or the associated cellular responses to DNA DAMAGE.
The introduction of a phosphoryl group into a compound through the formation of an ester bond between the compound and a phosphorus moiety.
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.
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.
Established cell cultures that have the potential to propagate indefinitely.

T-cell lymphoblastic leukemia/lymphoma in Nijmegen breakage syndrome. (1/32)

Nijmegen breakage syndrome (NBS) is a rare autosomal-recessive disorder characterized by microcephaly, immunodeficiency and predisposition to cancer, mainly B-cell lymphomas. Our 10-years-old female patient with NBS developed T-cell lymphoblastic leukemia/lymphoma (TLBL/ALL). The use of standard chemotherapy in our patient, except for cranial irradiation, led to complete and sustained remission of TLBL/ALL. In patients with chromosomal instability syndromes chemotherapy must be modified and radiotherapy must be omitted because of potentially serious toxic complications. Careful prevention of infections, including the use of intravenous immunoglobulin is also essential for successful treatment of lymphoid malignancies in NBS. Immunodeficiency in NBS is profound affecting both humoral and cellular immune system. During long-term follow-up after treatment of malignancy our patient remained free of major infections However, the rise of oligoclonal serum IgM was detected recently. Monitoring of serum IgM concentration may be a useful indicator for early detection of lymphomas in NBS.  (+info)

Werner syndrome protein and the MRE11 complex are involved in a common pathway of replication fork recovery. (2/32)

Werner syndrome (WS) is an autosomal recessive disease that predisposes individuals to a wide range of cancers. The gene mutated in WS, WRN, encodes a member of the RecQ family of DNA helicases. The precise DNA metabolic processes in which WRN participates remain to be elucidated. However, it has been proposed that WRN might play an important role in the maintenance of genetic stability during DNA replication, possibly cooperating with other proteins. Here, we show that, following DNA replication arrest, WRN associates and colocalizes with the MRE11 complex at PCNA sites. We also provide evidence that both WRN/MRE11 complex association and proper WRN relocalization after HU treatment require a functional MRE11 complex. We demonstrate that mutations altering the functionality of WRN or that of the MRE11 complex result in chromosomal breakage during DNA replication and enhanced cell death following replication arrest. Finally, we show that the DNA breakage in replicating cells and apoptosis observed in WS are not enhanced by concomitant knock down of MRE11 by RNAi, indicating that WRN and MRE11 complex act in a common pathway. These results suggest a functional relationship between WRN and the MRE11 complex in response to replication fork arrest, disclosing a common action of WRN and the MRE11 complex in the pathway(s) preserving genome stability during DNA replication.  (+info)

Nijmegen breakage syndrome (NBS) with neurological abnormalities and without chromosomal instability. (3/32)

BACKGROUND: Nijmegen breakage syndrome (NBS) is an autosomal recessive chromosomal instability disorder with hypersensitivity to ionising radiation. The clinical phenotype is characterised by congenital microcephaly, mild dysmorphic facial appearance, growth retardation, immunodeficiency, and greatly increased risk for lymphoreticular malignancy. Most NBS patients are of Slavic origin and homozygous for the founder mutation 657del5. The frequency of 657del5 heterozygotes in the Czech population is 1:150. Recently, another NBS1 mutation, 643C>T(R215W), with uncertain pathogenicity was found to have higher frequency among tumour patients of Slavic origin than in controls. This alteration results in the substitution of the basic amino acid arginine with the non-polar tryptophan and thus could potentially interfere with the function of the NBS1 protein, nibrin. METHODS AND RESULTS: Children with congenital microcephaly are routinely tested for the 657del5 mutation in the Czech and Slovak Republics. Here, we describe for the first time a severe form of NBS without chromosomal instability in monozygotic twin brothers with profound congenital microcephaly and developmental delay who are compound heterozygotes for the 657del5 and 643C>T(R215W) NBS1 mutations. Both children showed reduced expression of full length nibrin when compared with a control and a heterozygote for the 657del5 mutation. Radiation response processes such as phosphorylation of ATM and phosphorylation/stabilisation of p53, which are promoted by NBS1, are strongly reduced in cells from these patients. CONCLUSIONS: Interestingly, the patients are more severely affected than classical NBS patients. Consequently, we postulate that homozygosity for the 643C>T(R215W) mutation will also lead to a, possibly very, severe disease phenotype.  (+info)

Molecular basis of ataxia telangiectasia and related diseases. (4/32)

Ataxia telangiectasia (AT) is a rare human disease characterized by extreme cellular sensitivity to radiation and a predisposition to cancer, with a hallmark of onset in early childhood. Several human diseases also share similar symptoms with AT albeit with different degrees of severity and different associated disorders. While all AT patients contain mutations in the AT-mutated gene (ATM), most other AT-like disorders are defective in genes encoding an MRN protein complex consisting of Mre11, Rad50 and Nbs1. Both ATM and MRN function as cellular sensors to DNA double-strand breaks, which lead to the recruitment and phosphorylation of an array of substrate proteins involved in DNA repair, apoptosis and cell-cycle checkpoints, as well as gene regulation, translation initiation and telomere maintenance. ATM is a member of the family of phosphatidylinositol 3-kinase-like protein kinases (PIKK), and the discovery of many ATM substrates provides the underlying mechanisms of heterologous symptoms among AT patients. This review article focuses on recent findings related to the initial recognition of double-strand breaks by ATM and MRN, as well as a DNA-dependent protein kinase complex consisting of the heterodimer Ku70/Ku80 and its catalytic subunit DNA-PKcs, another member of PIKK. This possible interaction suggests that a much greater complex is involved in sensing, transducing and co-ordinating cellular events in response to genome instability.  (+info)

Importin KPNA2 is required for proper nuclear localization and multiple functions of NBS1. (5/32)

Nijmegen breakage syndrome (NBS) is a chromosomal-instability syndrome associated with cancer predisposition, radiosensitivity, microcephaly, and growth retardation. The NBS gene product, NBS1, is a component of the MRE11-RAD50-NBS1 (MRN) complex, a central player associated with double strand break (DSB) repair. In response to radiation, NBS1 is phosphorylated by ATM, and the MRN complex relocalizes to form punctate nuclear foci for DNA repair. NBS1 controls both the nuclear localization of the MRN complexes and radiation-induced focus formation. We report here that the KPNA2 (importin alpha1) is important for the normal nuclear localization of the MRN complex and its proper formation of the nuclear foci. KPNA2 is the only member of the importin alpha family that physically interacts with NBS1, and the KPNA2-mediated nucleus localization sequence (NLS) is mapped to amino acid residues 461-467 of NBS1 that is sufficient for both the interaction with KPNA2 and the proper nuclear localization. Inhibition of KPNA2 or blockage of the KPNA2 interaction with NBS1 results in a reduction of radiation-induced nuclear focus accumulation, DSB repair, and cell cycle checkpoint signaling of NBS1. Collectively, our results strongly suggest that an interaction with KPNA2 contributes to nuclear localization and multiple tumor suppression functions of the NBS1 complex.  (+info)

Role of the Nijmegen breakage syndrome 1 gene in familial and sporadic prostate cancer. (6/32)

The Nijmegen breakage syndrome 1 (NBS1) gene, which participates in DNA double strand break repair, has been postulated to be a susceptibility factor for a number of cancers, including prostate cancer. Numerous mutations have been identified in NBS1, including the founder mutation 657del5. In this study, a number of analyses were done to determine whether mutations in NBS1 are associated with an increased risk for prostate cancer. The frequency of the 657del5 mutation in both familial prostate cancer cases (1,819 affected men among 909 families) and sporadic prostate cancer cases (1,218 affected men) collected from five centers participating in the International Consortium for Prostate Cancer Genetics were compared with that found in 697 normal controls. Seven individuals were identified to carry the mutation among the 3,037 cases screened: four in the familial group (three from one family and one from another) and three in the sporadic cases. The carrier frequency was 0.22% (2 of 909) for the probands and 0.25% (3 of 1,218) for the sporadic cases of prostate cancer. The 657del5 mutation was not detected in either the 293 unaffected members of the prostate cancer families or in the 697 control samples tested. The entire NBS1 gene was also sequenced in 20 of the youngest affected individuals from the Finnish group of familial cases to identify the presence of possible mutations in this high-risk group. One rare (D95N) and one common (E185Q) missense alteration was identified. More detailed analyses of the E185Q polymorphism, along with a third rare variant (R215W), failed to show an association with prostate cancer. Because the 657del5 mutation was absent from the control population, we are unable to determine if this alteration predisposes to prostate cancer. However, our data does suggest that mutations within NBS1, and in particular, 657del5, do not significantly contribute to the overall prostate cancer burden within our patient samples.  (+info)

Heterozygous carriers of Nijmegen Breakage Syndrome have a distinct gene expression phenotype. (7/32)

Autosomal recessive diseases are those that require mutations in both alleles to exhibit the disorder. Although most recessive conditions are rare, heterozygous carriers of recessive mutations are quite common. In this study, we show that carriers of Nijmegen Breakage Syndrome (NBS) have a distinct gene expression phenotype that differs from that of noncarriers and also from that of carriers of a similar syndrome, Ataxia Telangiectasia (AT). We found 520 genes whose expression levels differ significantly (P < or = 0.001) between NBS carriers and controls. By linear discriminant analysis, we found a combination of 16 genes that allows 100% correct classification of individuals as either NBS carriers or noncarriers in a training set with 25 individuals, and in a test set with 52 individuals. When applied to AT carriers, the discriminant function misclassified only one out of 18 AT carriers as an NBS carrier. Our result shows that NBS carriers have a specific gene expression phenotype. It suggests that heterozygous mutations can contribute significantly to natural variation in gene expression. This has implications for the role that heterozygosity for recessive diseases plays in the overall genetic architecture of complex human traits and diseases.  (+info)

Cancer incidence in Nijmegen breakage syndrome is modulated by the amount of a variant NBS protein. (8/32)

The human genetic disorder, Nijmegen breakage syndrome (NBS), is characterized by radiosensitivity, immunodeficiency and an increased risk for cancer, particularly B-cell non-Hodgkin lymphoma. The NBS1 gene codes for a protein, nibrin, involved in the processing/repair of DNA double-strand breaks and in cell cycle checkpoints. The majority of patients are homozygous for a founder mutation, a 5 bp deletion. This mutation is actually hypomorphic, since a functionally relevant truncated protein, of approximately 70 kDa, is produced by alternative translation. Null mutation of the homologous gene in mice is lethal; however, null-mutant murine cells can be rescued by a human NBS1 cDNA carrying the founder mutation. Clearly, the truncated p70-nibrin is able to sustain vital cellular functions of the full-length protein. We have used semi-quantitative immunoprecipitation to examine a panel of 26 lymphoblastoid B-cell lines from NBS patients for their level of p70-nibrin expression and correlate this with details of clinical phenotype provided by the two contributing centres. We find considerable variation in the amount of p70-nibrin in cell lines from different patients. Examination of clinical history indicated a clear and statistically significant correlation between p70-nibrin expression levels and lymphoma incidence. The variation in p70-nibrin levels between patients probably reflects the susceptibility of the alternative translation process to other genetic and non-genetic factors. Patients whose cells are able to maintain particularly high levels of the truncated p70-nibrin protein are at a lower risk for lymphoma than those patients with low levels of p70-nibrin in their cells.  (+info)

Nijmegen Breakage Syndrome (NBS) is a rare autosomal recessive disorder characterized by extreme sensitivity to ionizing radiation, progressive microcephaly, short stature, immunodeficiency, and an increased risk of developing malignancies, particularly lymphoid tumors. The syndrome is caused by mutations in the NBN gene, which encodes a protein called nibrin that plays a critical role in DNA repair and maintenance of genomic stability.

Individuals with NBS typically have microcephaly at birth or develop it in early childhood, accompanied by developmental delay, intellectual disability, and characteristic facial features such as a prominent forehead, recessed jaw, and widely spaced eyes. They may also have skin abnormalities, skeletal anomalies, and hearing loss.

Immunodeficiency is a common feature of NBS, with patients often experiencing recurrent infections due to impaired immune function. They may have low levels of immunoglobulins and T-cell lymphopenia, which can increase their susceptibility to infections.

NBS is associated with an increased risk of malignancies, particularly lymphoid tumors such as B-cell non-Hodgkin lymphoma and leukemia. The risk of cancer increases with age, and most patients develop a malignancy by their mid-20s.

The diagnosis of NBS is typically made based on clinical features, genetic testing, and confirmation of biallelic mutations in the NBN gene. Treatment may involve management of infections, immunoglobulin replacement therapy, and chemotherapy or radiation therapy for malignancies. However, these treatments can be challenging due to the increased sensitivity to ionizing radiation and potential toxicity of chemotherapeutic agents.

Overall, NBS is a rare but serious disorder that requires multidisciplinary care from specialists in genetics, immunology, oncology, and other fields.

Microcephaly is a medical condition where an individual has a smaller than average head size. The circumference of the head is significantly below the normal range for age and sex. This condition is typically caused by abnormal brain development, which can be due to genetic factors or environmental influences such as infections or exposure to harmful substances during pregnancy.

Microcephaly can be present at birth (congenital) or develop in the first few years of life. People with microcephaly often have intellectual disabilities, delayed development, and other neurological problems. However, the severity of these issues can vary widely, ranging from mild to severe. It is important to note that not all individuals with microcephaly will experience significant impairments or challenges.

Chromosome breakage is a medical term that refers to the breaking or fragmentation of chromosomes, which are thread-like structures located in the nucleus of cells that carry genetic information. Normally, chromosomes are tightly coiled and consist of two strands called chromatids, joined together at a central point called the centromere.

Chromosome breakage can occur spontaneously or be caused by environmental factors such as radiation or chemicals, or inherited genetic disorders. When a chromosome breaks, it can result in various genetic abnormalities, depending on the location and severity of the break.

For instance, if the break occurs in a region containing important genes, it can lead to the loss or alteration of those genes, causing genetic diseases or birth defects. In some cases, the broken ends of the chromosome may rejoin incorrectly, leading to chromosomal rearrangements such as translocations, deletions, or inversions. These rearrangements can also result in genetic disorders or cancer.

Chromosome breakage is commonly observed in individuals with certain inherited genetic conditions, such as Bloom syndrome, Fanconi anemia, and ataxia-telangiectasia, which are characterized by an increased susceptibility to chromosome breakage due to defects in DNA repair mechanisms.

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.

A syndrome, in medical terms, is a set of symptoms that collectively indicate or characterize a disease, disorder, or underlying pathological process. It's essentially a collection of signs and/or symptoms that frequently occur together and can suggest a particular cause or condition, even though the exact physiological mechanisms might not be fully understood.

For example, Down syndrome is characterized by specific physical features, cognitive delays, and other developmental issues resulting from an extra copy of chromosome 21. Similarly, metabolic syndromes like diabetes mellitus type 2 involve a group of risk factors such as obesity, high blood pressure, high blood sugar, and abnormal cholesterol or triglyceride levels that collectively increase the risk of heart disease, stroke, and diabetes.

It's important to note that a syndrome is not a specific diagnosis; rather, it's a pattern of symptoms that can help guide further diagnostic evaluation and management.

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.

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.

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.

Chromosome disorders are a group of genetic conditions caused by abnormalities in the number or structure of chromosomes. Chromosomes are thread-like structures located in the nucleus of cells that contain most of the body's genetic material, which is composed of DNA and proteins. Normally, humans have 23 pairs of chromosomes, for a total of 46 chromosomes.

Chromosome disorders can result from changes in the number of chromosomes (aneuploidy) or structural abnormalities in one or more chromosomes. Some common examples of chromosome disorders include:

1. Down syndrome: a condition caused by an extra copy of chromosome 21, resulting in intellectual disability, developmental delays, and distinctive physical features.
2. Turner syndrome: a condition that affects only females and is caused by the absence of all or part of one X chromosome, resulting in short stature, lack of sexual development, and other symptoms.
3. Klinefelter syndrome: a condition that affects only males and is caused by an extra copy of the X chromosome, resulting in tall stature, infertility, and other symptoms.
4. Cri-du-chat syndrome: a condition caused by a deletion of part of the short arm of chromosome 5, resulting in intellectual disability, developmental delays, and a distinctive cat-like cry.
5. Fragile X syndrome: a condition caused by a mutation in the FMR1 gene on the X chromosome, resulting in intellectual disability, behavioral problems, and physical symptoms.

Chromosome disorders can be diagnosed through various genetic tests, such as karyotyping, chromosomal microarray analysis (CMA), or fluorescence in situ hybridization (FISH). Treatment for these conditions depends on the specific disorder and its associated symptoms and may include medical interventions, therapies, and educational support.

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.

'Abnormalities, Multiple' is a broad term that refers to the presence of two or more structural or functional anomalies in an individual. These abnormalities can be present at birth (congenital) or can develop later in life (acquired). They can affect various organs and systems of the body and can vary greatly in severity and impact on a person's health and well-being.

Multiple abnormalities can occur due to genetic factors, environmental influences, or a combination of both. Chromosomal abnormalities, gene mutations, exposure to teratogens (substances that cause birth defects), and maternal infections during pregnancy are some of the common causes of multiple congenital abnormalities.

Examples of multiple congenital abnormalities include Down syndrome, Turner syndrome, and VATER/VACTERL association. Acquired multiple abnormalities can result from conditions such as trauma, infection, degenerative diseases, or cancer.

The medical evaluation and management of individuals with multiple abnormalities depend on the specific abnormalities present and their impact on the individual's health and functioning. A multidisciplinary team of healthcare professionals is often involved in the care of these individuals to address their complex needs.

Immunologic deficiency syndromes refer to a group of disorders characterized by defective functioning of the immune system, leading to increased susceptibility to infections and malignancies. These deficiencies can be primary (genetic or congenital) or secondary (acquired due to environmental factors, medications, or diseases).

Primary immunodeficiency syndromes (PIDS) are caused by inherited genetic mutations that affect the development and function of immune cells, such as T cells, B cells, and phagocytes. Examples include severe combined immunodeficiency (SCID), common variable immunodeficiency (CVID), Wiskott-Aldrich syndrome, and X-linked agammaglobulinemia.

Secondary immunodeficiency syndromes can result from various factors, including:

1. HIV/AIDS: Human Immunodeficiency Virus infection leads to the depletion of CD4+ T cells, causing profound immune dysfunction and increased vulnerability to opportunistic infections and malignancies.
2. Medications: Certain medications, such as chemotherapy, immunosuppressive drugs, and long-term corticosteroid use, can impair immune function and increase infection risk.
3. Malnutrition: Deficiencies in essential nutrients like protein, vitamins, and minerals can weaken the immune system and make individuals more susceptible to infections.
4. Aging: The immune system naturally declines with age, leading to an increased incidence of infections and poorer vaccine responses in older adults.
5. Other medical conditions: Chronic diseases such as diabetes, cancer, and chronic kidney or liver disease can also compromise the immune system and contribute to immunodeficiency syndromes.

Immunologic deficiency syndromes require appropriate diagnosis and management strategies, which may include antimicrobial therapy, immunoglobulin replacement, hematopoietic stem cell transplantation, or targeted treatments for the underlying cause.

Ionizing radiation is a type of radiation that carries enough energy to ionize atoms or molecules, which means it can knock electrons out of their orbits and create ions. These charged particles can cause damage to living tissue and DNA, making ionizing radiation dangerous to human health. Examples of ionizing radiation include X-rays, gamma rays, and some forms of subatomic particles such as alpha and beta particles. The amount and duration of exposure to ionizing radiation are important factors in determining the potential health effects, which can range from mild skin irritation to an increased risk of cancer and other diseases.

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.

Human chromosome pair 8 consists of two rod-shaped structures present in the nucleus of each cell of the human body. Each chromosome is made up of DNA tightly coiled around histone proteins, forming a complex structure known as a chromatin.

Human cells have 23 pairs of chromosomes, for a total of 46 chromosomes. Pair 8 is one of the autosomal pairs, meaning that it is not a sex chromosome (X or Y). Each member of chromosome pair 8 has a similar size, shape, and banding pattern, and they are identical in males and females.

Chromosome pair 8 contains several genes that are essential for various cellular functions and human development. Some of the genes located on chromosome pair 8 include those involved in the regulation of metabolism, nerve function, immune response, and cell growth and division.

Abnormalities in chromosome pair 8 can lead to genetic disorders such as Wolf-Hirschhorn syndrome, which is caused by a partial deletion of the short arm of chromosome 4, or partial trisomy 8, which results from an extra copy of all or part of chromosome 8. Both of these conditions are associated with developmental delays, intellectual disability, and various physical abnormalities.

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.

Recessive genes refer to the alleles (versions of a gene) that will only be expressed when an individual has two copies of that particular allele, one inherited from each parent. If an individual inherits one recessive allele and one dominant allele for a particular gene, the dominant allele will be expressed and the recessive allele will have no effect on the individual's phenotype (observable traits).

Recessive genes can still play a role in determining an individual's genetic makeup and can be passed down through generations even if they are not expressed. If two carriers of a recessive gene have children, there is a 25% chance that their offspring will inherit two copies of the recessive allele and exhibit the associated recessive trait.

Examples of genetic disorders caused by recessive genes include cystic fibrosis, sickle cell anemia, and albinism.

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.

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.

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.

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.

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.

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.

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.

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.

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).

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.

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.

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.

DNA repair-deficiency disorders are a group of genetic conditions that result from mutations in genes responsible for the repair and maintenance of DNA. These disorders are characterized by increased sensitivity to environmental factors that can damage DNA, such as ultraviolet (UV) radiation, chemicals, and free radicals. As a result, individuals with these disorders have an increased risk of developing various types of cancer, neurological disorders, premature aging, and other health problems.

Examples of DNA repair-deficiency disorders include xeroderma pigmentosum, Cockayne syndrome, trichothiodystrophy, and Bloom syndrome. These conditions are 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 disorder.

Treatment for DNA repair-deficiency disorders is focused on managing symptoms and preventing complications. This may include avoiding exposure to UV radiation, using sunscreens and protective clothing, and undergoing regular cancer screenings. In some cases, medications or other therapies may be used to treat specific symptoms or complications of the disorder.

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.

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.

"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.

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.

The I (2000). "Nijmegen breakage syndrome. The International Nijmegen Breakage Syndrome Study Group". Arch Dis Child. 82 (5): ... Group, The International Nijmegen Breakage Syndrome Study (2000-05-01). "Nijmegen breakage syndrome". Archives of Disease in ... nijmegen at NIH/UW GeneTests Nijmegen Breakage Syndrome at NIHs Office of Rare Diseases (CS1 maint: multiple names: authors ... Other syndromes with clinical features similar to Nijmegen Breakage Syndrome include RAD50 deficiency and Cernunnos/NHEJ ...
Nijmegen breakage syndrome is a condition characterized by short stature, an unusually small head size ( microcephaly ), ... medlineplus.gov/genetics/condition/nijmegen-breakage-syndrome/ Nijmegen breakage syndrome. ... Nijmegen breakage syndrome gets its name from numerous breaks in DNA that occur in affected peoples cells. A buildup of these ... Mutations in the NBN gene cause Nijmegen breakage syndrome. The NBN gene provides instructions for making a protein called ...
Nijmegen breakage syndrome (NBS) is a rare autosomal recessive condition of chromosomal instability that is clinically ... Nijmegen breakage syndrome. The International Nijmegen Breakage Syndrome Study Group. Arch Dis Child. 2000 May. 82(5):400-6. [ ... encoded search term (Nijmegen Breakage Syndrome) and Nijmegen Breakage Syndrome What to Read Next on Medscape ... gene located in band 8q21 are responsible for Nijmegen breakage syndrome. Nijmegen breakage syndrome is identified as entries ...
Nijmegen breakage syndrome. Disease definition A rare, genetic chromosomal instability syndrome presenting at birth with ... Nijmegen breakage syndrome (NBS) is caused by mutations in the NBN gene (8q21-q24), specifically within exons 6-10, which lead ... Differential diagnosis includes Fanconi anemia, LIG4 syndrome, Cernunnos-XLF deficiency, NBS-like disorder, ataxia- ... telangiectasia-like disorder, Bloom syndrome.. Antenatal diagnosis Affected families may be offered prenatal diagnosis by ...
Nijmegen breakage syndrome (NBS) is a rare autosomal recessive chromosomal instability syndrome characterized by severe ... Nijmegen breakage syndrome complicated with primary cutaneous tuberculosis.. Melinda Erdös, Beáta Tóth, Imre Veres, Mária Kiss ...
2014 Protein Structure and Bioinformatics, last updated 11.11.2010 10:51 ...
Nijmegen Breakage Syndrome (NBS) is a rare autosomal recessive DNA repair disorder that increases risk of hematological ... such as Nijmegen breakage syndrome (NBS), Ataxia telangiectasia, Fanconi anemia, and Bloom syndrome, can predispose pediatric ... A rare case of primary gastric Hodgkin lymphoma in an adolescent with Nijmegen breakage syndrome. *Elizabeth Batiuk. ORCID: ... Nijmegen Breakage Syndrome (NBS) is a rare autosomal recessive DNA repair disorder that increases risk of hematological ...
Nijmegen Breakage Syndrome. Varon R, Demuth I, Chrzanowska KH. Varon R, et al. Among authors: chrzanowska kh. 1999 May 17 [ ... Nijmegen breakage syndrome. van der Burgt I, Chrzanowska KH, Smeets D, Weemaes C. van der Burgt I, et al. Among authors: ... Nijmegen breakage syndrome (NBS). Chrzanowska KH, Gregorek H, Dembowska-Bagińska B, Kalina MA, Digweed M. Chrzanowska KH, et al ... The Dubowitz syndrome--one more case. Chrzanowska KH, Krajewska-Walasek M. Chrzanowska KH, et al. Klin Padiatr. 1987 Sep-Oct; ...
Nijmegen breakage syndrome; SCID, severe combined immunodeficiency; XLA, X-linked agammaglobulinemia.. †Reference range for ...
Comparison of Selected Parameters of Redox Homeostasis in Patients with Ataxia-Telangiectasia and Nijmegen Breakage Syndrome. ...
Nijmegen Breakage Syndrome and our test Nijmegen breakage syndrome is a rare genetic disorder. It is characterized by ... Nijmegen Breakage Syndrome Nonsyndromic Hearing Loss and Deafness, DFNB1 (GJB2-Related) Pendred Syndrome and DFNB4 Hearing Loss ... Sjögren-Larsson Syndrome Tay-Sachs Disease Tyrosinemia Type I Usher Syndrome Type 1F Usher Syndrome Type 3A Zellweger Spectrum ... Bloom Syndrome and our test Bloom syndrome is a rare genetic disorder characterized by impaired growth and increased risk of ...
Nijmegen breakage syndrome. Breast cancer. Aplastic anemia. Defects in NBN might play a role in the pathogenesis of childhood ... Nijmegen breakage syndrome 1 (nibrin) antibody. *Nijmegen breakage syndrome antibody. *Nijmegen breakage syndrome protein 1 ...
Immortalization and characterization of Nijmegen Breakage Syndrome fibroblasts. Mutat. Res. ... Nijmegen Breakage Syndrome. ...
Nijmegen Breakage Syndrome - neglected primary immunodeficiency. Eva Kafková, Pavel Seeman, Alexandra Martinčeková, Zdeňka. ... Nijmegen Breakage Syndrom - opomíjená primární imunodeficience. Eva Kafková, Pavel Seeman, Alexandra Martinčeková, Zdeňka. ... X-linked hyper-IgM syndrome (CD40 ligand deficiency).. Patients in the Czech Republic and literature review. Aleš Janda, ...
Impaired DNA double strand break repair in cells from Nijmegen Breakage Syndrome patients. DNA Repair, 5, 251-257. ... induces chromosomal instability and common fragile site breakage. Oncogene, 25, 4381-4388. ...
The Nijmegen Breakage Syndrome is a chromosomal instability disorder characterized by microcephaly, growth retardation, ... Varon R, Reis A, Henze G, von Einsiedel HG, Sperling K, Seeger K: Mutations in the Nijmegen Breakage Syndrome gene (NBS1) in ... Seemanova E, Sperling K, Neitzel H, Varon R, Hadac J, Butova O, Schrock E, Seeman P, Digweed M: Nijmegen breakage syndrome (NBS ... The Nijmegen Breakage Syndrome is a chromosomal instability disorder characterized by microcephaly, growth retardation, ...
Guidelines focus on syndromes associated with an increased risk of breast and/or ovarian cancer. The NCCN Genetic/Familial High ... Breast and Ovarian provide recommendations for genetic testing and counseling for hereditary cancer syndromes and risk ... management recommendations for patients who are diagnosed with a syndrome. ... Nijmegen Breakage Syndrome mutations and risk of breast cancer. Int J Cancer 2008;122:802-806. ...
Nijmegen breakage syndrome 1 (NBS1) antibody Product code : pab50002 Raised in : Rabbit Clonality : Polyclonal antibody ...
Mutations in this gene are associated with Nijmegen breakage syndrome, an autosomal recessive chromosomal instability syndrome ...
... and Nijmegen breakage syndrome. These syndromes are rarely associated with NK cell deficiency, and their association with ... Bloom syndrome, RIDDLE syndrome, immunodeficiency centromeric instability facial anomalies syndrome, Fanconi anemia, LIG4 ... Complementation of the breakage phenotype was analyzed in a blinded manner.. For lymphocyte DNA breakage experiments, blood ... Breakage analysis. Primary or SV40 fibroblasts were treated with 0.3 μM aphidicolin for 24 hours, arrested by incubation with ...
Nijmegen Breakage Syndrome. Hemolytic anemia, Abnormality of chromosome stability, Autoimmune hemolytic anemia, Acute leukemi ... Anemia, Chromosome breakage, Chromosomal breakage induced by crosslinking agents. OMIM:614083. Fructose-1,6-Bisphosphatase ... Anemia, Pancytopenia, Chromosomal breakage induced by crosslinking agents. OMIM:613951. Fanconi Anemia, Complementation Group N ... Chromosome breakage, Abnormality of chromosome stability. OMIM:181750. Hemolytic Anemia, Nonspherocytic, Due To Hexokinase ...
3. Nijmegen Breakage Syndrome. Nijmegen breakage syndrome (NBS) is another rare AR defect in DSBR. It is most commonly seen in ... The International Nijmegen Breakage Syndrome Study Group. Nijmegen breakage syndrome. Arch. Dis. Child. 2000, 82, 400-406. [ ... Nijmegen breakage syndrome, Bloom syndrome, and even more rare disorders reported in only one or less than 10 patients. In ... Nijmegen breakage syndrome. NBS1. AR. Bacterial respiratory tract infections. Mycobacterial infections (only a few patients). ...
Nijmegen breakage syndrome (chromosome breakage analysis) This test is available for the following conditions: ... Bloom syndrome (chromosome breakage analysis) This test is available for the following conditions: ... Fanconi anemia (chromosome breakage analysis, MMC test) This test is available for the following conditions: ... ataxia telangiectasia (chromosome breakage analysis) This test is available for the following conditions: ...
Cancer Proneness in Nijmegen Breakage Syndrome Carriers. - Pp. 101-119 (19). Alessandra di Masi, Francesco Berardinelli, ... Nijmegen Breakage Syndrome, Tumors and Malaria among other diseases. The volume also features some chapters on plant genetics. ...
Clinical variability and novel mutations in the NHEJ1 gene in patients with a Nijmegen breakage syndrome-like phenotype. Hum ... Jyonouchi S, Orange J, Sullivan KE, Krantz I, Deardorff M. Immunologic features of Cornelia de Lange syndrome. Pediatrics. 2013 ...
Nijmegen Breakage Syndrome and Ataxia-Telangiectasia). The latter also raises the question what actually constitutes a curable/ ...
Association of Nijmegen Breakage Syndrome 1 Genotypes With Bladder Cancer Risk. Chen Meng, et al. Anticancer research 2020 4 (4 ... Contribution of Double-strand Break Repair Gene Nijmegen Breakage Syndrome 1 Genotypes, Gender Difference and Smoking Status to ... Chromosome instability syndromes. Taylor A Malcolm R et al. Nature reviews. Disease primers 2019 Sep 5(1) 64 ... Clonal Evolution and Outcomes in 144 Mayo Clinic Patients with Germline Predisposition Syndromes. St Martin Emma et al. ...
In culture, cells from affected individuals exhibit elevated rates of chromosomal breakage or instability, leading to ... Chromosomal breakage syndromes are a group of genetic disorders that are typically transmitted in an autosomal recessive mode ... However, radiosensitivity is seen in patients with Nijmegen breakage syndrome, X-linked agammaglobulinemia, Fanconi anemia, ... encoded search term (Chromosomal Breakage Syndromes) and Chromosomal Breakage Syndromes What to Read Next on Medscape ...
Other variable symptoms of this probably under-diagnosed syndrome include mild facial dysmorphism, growth retardation, failure ... Facial anomalies syndrome (ICF) is a rare autosomal recessive disease described in about 50 patients worldwide and ... Like Bloom syndrome (BS), ataxia-telangiectasia (AT), and Nijmegen breakage syndrome (NBS), ICF is usually diagnosed in ... van der Burgt I, Chrzanowska KH, Smeets D, Weemaes C: Nijmegen breakage syndrome. J Med Genet. 1996, 33: 153-156. ...
  • This explains why mutations in the NBS1 gene lead to higher levels of cancer (see Fanconi anemia, Cockayne syndrome. (wikipedia.org)
  • Mutations in the NBN gene cause Nijmegen breakage syndrome. (medlineplus.gov)
  • The NBN gene mutations that cause Nijmegen breakage syndrome typically lead to the production of an abnormally short version of the nibrin protein, which prevents it from responding to DNA damage effectively. (medlineplus.gov)
  • [ 2 ] Mutations in the NBN ( NBS1) gene located in band 8q21 are responsible for Nijmegen breakage syndrome. (medscape.com)
  • Nijmegen breakage syndrome (NBS) is caused by mutations in the NBN gene (8q21-q24), specifically within exons 6-10, which lead to partially functional truncated fragments of nibrin, the gene product involved in repairing DNA double strand breaks. (orpha.net)
  • Omenn syndrome is the result of mutations in the genes coding for recombinases (recombination activating genes). (medscape.com)
  • These 2 syndromes, AT and NBS, are part of a family of mutations involving proteins involved in DNA repair. (medscape.com)
  • Mutations in this gene are associated with Nijmegen breakage syndrome, an autosomal recessive chromosomal instability syndrome characterized by microcephaly, growth retardation, immunodeficiency, and cancer predisposition. (origene.com)
  • Griscelli syndrome type II - GS type II is caused by mutations in the gene RAB27A. (immunodeficiencysearch.com)
  • Unsurprisingly, many of the features are similar to ataxia telangiectasia (AT) and this syndrome was sometimes termed AT-variant 1, as the protein mutated in AT, ATM, interacts with the MRE11/RAD50/NBS1 (MRN) complex. (wikipedia.org)
  • Further investigations revealed that in vitro cells derived from patients with Nijmegen breakage syndrome display characteristic abnormalities similar to those observed in ataxia-telangiectasia (A-T) , including spontaneous chromosomal instability, sensitivity to ionizing radiation (IR), and radioresistant DNA synthesis (RDS). (medscape.com)
  • Underlying genetic chromosome instability disorders, such as Nijmegen breakage syndrome (NBS), Ataxia telangiectasia, Fanconi anemia, and Bloom syndrome, can predispose pediatric patients to malignancy and clinicians should consider diagnostic testing in these patients Footnote 3 . (biomedcentral.com)
  • Patients with ataxia telangiectasia, also known as Louis-Bar syndrome, are hypersensitive to ionizing radiation, while patients with Bloom syndrome, Fanconi anemia, and xeroderma pigmentosum are sensitive to UV radiation. (medscape.com)
  • Constitutional syndromes such as Down syndrome and ataxia-telangiectasia are associated with increased risk of B-cell-ALL (with CRLF2 rearrangement) and T-cell-ALL, respectively. (haematologica.org)
  • However, whereas MRE11 and NBN gene variants are known to induce various illnesses, such as ataxia telangiectasia-like condition and Nijmegen breakage syndrome, the pathogenic implications of RAD50 gene variants have remained fairly unknown - until now. (thejapannews.net)
  • While some of those genes encode proteins with a direct involvement in a DNA repair machinery, such as nucleotide excision repair (NER), others destabilize the genome by compromising the stability of the nuclear envelope, when lamin A is dysfunctional in Hutchinson-Gilford progeria syndrome (HGPS) or regulate the DNA damage response (DDR) such as the ataxia telangiectasia-mutated (ATM) gene. (bvsalud.org)
  • Neuromascular abnormality presenting with ataxia(ataxia-telangiectasia) , flaccid paralysis after live poliovirus immunization (combined or antibody deficiencies) ,pernicious anaemia (CVID), cognitive impairment, nystagmus and cerebellar, spinal and peripheral neuropathies(Chediac-Higashi syndrome), seizures, ataxia and occulomotor and reflex abnormalities(Griscelli syndrome) are examples of neurologic features seen in different immunodeficiency syndromes. (ac.ir)
  • Abstract The 657del5 mutation of the NBS1 gene has been demonstrated in most patients with Nijmegen breakage syndrome (NBS). (the-medical-dictionary.com)
  • Expression of a large genomic locus (NBS1 [Nijmegen breakage syndrome], encoding nibrin) was assessed by immunofluorescence. (ox.ac.uk)
  • Nijmegen breakage syndrome (NBS) is a rare autosomal recessive congenital disorder causing chromosomal instability, probably as a result of a defect in the double Holliday junction DNA repair mechanism and/or the synthesis dependent strand annealing mechanism for repairing double strand breaks in DNA (see Homologous recombination). (wikipedia.org)
  • Nijmegen breakage syndrome (NBS) is a rare autosomal recessive condition of chromosomal instability that is clinically characterized by microcephaly, a distinct facial appearance, short stature, immunodeficiency, radiation sensitivity, and a strong predisposition to lymphoid malignancy. (medscape.com)
  • A rare, genetic chromosomal instability syndrome presenting at birth with microcephaly, dysmorphic facial features which become more noticeable with age, growth delay, recurring sinopulmonary infections and extremely high frequency of malignancies. (orpha.net)
  • Nijmegen breakage syndrome (NBS) is a rare autosomal recessive chromosomal instability syndrome characterized by severe immunodeficiency, growth retardation, microcephaly, a distinct facial appearance, and a high predisposition to lymphoid malignancy. (qxmd.com)
  • Nijmegen breakage syndrome (NBS) is also an autosomal recessive chromosomal instability syndrome. (medscape.com)
  • Depletion of CHK1, but not CHK2, induces chromosomal instability and common fragile site breakage. (uri.edu)
  • The Nijmegen Breakage Syndrome is a chromosomal instability disorder characterized by microcephaly, growth retardation, immunodeficiency, and increased frequency of cancers. (biomedcentral.com)
  • In culture, cells from affected individuals exhibit elevated rates of chromosomal breakage or instability, leading to chromosomal rearrangements. (medscape.com)
  • The Immunodeficiency, Centromeric region instability, Facial anomalies syndrome (ICF) is a rare autosomal recessive disease described in about 50 patients worldwide and characterized by immunodeficiency, although B cells are present, and by characteristic rearrangements in the vicinity of the centromeres (the juxtacentromeric heterochromatin) of chromosomes 1 and 16 and sometimes 9. (biomedcentral.com)
  • [ 7 ] first delineated the syndrome in two siblings with microcephaly, short stature, skin pigmentation abnormalities, mental retardation, immunologic defects, and a high prevalence of chromosome 7 and/or chromosome 14 rearrangements in cultured lymphocytes. (medscape.com)
  • Together, the findings from our case and the three previously reported cases suggest that RAD50 deficiency/Nijmegen breakage syndrome-like disorder is characterized by growth retardation and microcephaly, which may coexist with bone marrow failure and immunodeficiency in some patients," said senior author of the study Hirokazu Kanegane. (thejapannews.net)
  • Nibrin's role in regulating cell division and cell growth (proliferation) is thought to lead to the immunodeficiency seen in individuals with Nijmegen breakage syndrome. (medlineplus.gov)
  • When the research team came across a patient with progressive bone marrow failure and immunodeficiency combined with Nijmegen breakage syndrome-like manifestations, they decided to perform whole-exome sequencing to see if they could identify any gene variants that might lead to the observed symptoms. (thejapannews.net)
  • The aim of this study is to review the neurological manifestations of different primary immunodeficiency syndromes. (ac.ir)
  • Table 1 provides a summary outline of the gene symbols, chromosomal locations, radiation sensitivity characteristics, immunodeficiencies, chromosome breakage characteristics, and major cancer risk for each of these disorders. (medscape.com)
  • or other CHROMOSOME BREAKAGE related aberrations. (lookformedical.com)
  • Spontaneous CHROMOSOME BREAKAGE is a feature of this disease along with predisposition to LEUKEMIA. (lookformedical.com)
  • Nijmegen Breakage Syndrome (NBS) is a rare autosomal recessive DNA repair disorder that increases risk of hematological malignancy. (biomedcentral.com)
  • Ataxialike disorder (ATLD) syndrome involves a mutation in meiotic recombination 11 homolog (MRE11). (medscape.com)
  • Chediak-Higashi syndrome - Patients are characterized by partial oculocutaneous albinism, bleeding disorder from platelet dysfunction, chronic neutropenia, and defective cell cytotoxicity. (immunodeficiencysearch.com)
  • Hermansky-Pudlak syndrome II - HPS type II is characterized by oculocutaneous albinism, bleeding disorder due to platelet dysfunction, and chronic neutropenia. (immunodeficiencysearch.com)
  • The second volume of this ebook series contains a selection of articles on intracellular gene regulation and expressoion in human disease such as arthritis, Alzheimer's disease, Nijmegen Breakage Syndrome, Tumors and Malaria among other diseases. (benthambooks.com)
  • Two autosomal recessive syndromes involving DNA repair indicate some interaction between the immune system and neurologic function. (medscape.com)
  • Chromosomal breakage syndromes are a group of genetic disorders that are typically transmitted in an autosomal recessive mode of inheritance. (medscape.com)
  • Familial cancer syndromes such as Li-Fraumeni syndrome, constitutional mismatch repair deficiency syndrome, or DNA repair syndromes (e.g. (haematologica.org)
  • Cockayne syndrome (CS) and UV-sensitive syndrome (UVSS) are rare genetic disorders caused by mutation of the DNA repair and multifunctional CSA or CSB protein, but only CS patients display a progeroid and neurodegenerative phenotype, providing a unique conceptual and experimental paradigm. (bvsalud.org)
  • Progeroid syndromes (PSs) are characterized by the premature onset of age-related pathologies. (bvsalud.org)
  • Bloom syndrome - Patients can develop patchy areas of hypopigmentation or hyperpigmentation, especially on the trunk. (immunodeficiencysearch.com)
  • Other variable symptoms of this probably under-diagnosed syndrome include mild facial dysmorphism, growth retardation, failure to thrive, and psychomotor retardation. (biomedcentral.com)
  • Nijmegen breakage syndrome is identified as entries 251260 in and 602667 in Online Mendelian Inheritance in Man (OMIM). (medscape.com)
  • See Omenn Syndrome and Purine Nucleoside Phosphorylase Deficiency for a discussion of other forms of SCID. (medscape.com)
  • Other cancers seen in people with Nijmegen breakage syndrome include brain tumors such as medulloblastoma and glioma, and a cancer of muscle tissue called rhabdomyosarcoma. (medlineplus.gov)
  • Nijmegen breakage syndrome complicated with primary cutaneous tuberculosis. (qxmd.com)
  • The ARID1B spectrum in 143 patients: from nonsyndromic intellectual disability to Coffin-Siris syndrome. (nih.gov)
  • Male patients with thrombocytopenia and eczema may have Wiskott-Aldrich syndrome with defective T-cell function and resultant recurrent infections. (medscape.com)
  • Impaired DNA double strand break repair in cells from Nijmegen Breakage Syndrome patients. (uri.edu)
  • When your patients are looking to understand if they are a carrier for specific genetic conditions like cystic fibrosis, spinal muscular atrophy, fragile X syndrome, or Tay-Sachs disease, appropriate genetic screening and actionable results are essential . (questwomenshealth.com)
  • Nijmegen breakage syndrome - Patients can develop café au lait spots as well as vitiligo. (immunodeficiencysearch.com)
  • Other syndromes with clinical features similar to Nijmegen Breakage Syndrome include RAD50 deficiency and Cernunnos/NHEJ deficiency. (wikipedia.org)
  • When we looked at the literature, we realized that only three cases of RAD50 deficiency, which leads to symptoms similar to those of Nijmegen breakage syndrome, had been reported," explained Masatoshi Takagi, lead author of the study. (thejapannews.net)
  • Hb Bart syndrome is a severe form of anemia secondary to alpha thalassemia. (ghcgenetics.com)
  • The exact prevalence of Nijmegen breakage syndrome is unknown. (medlineplus.gov)
  • Nijmegen breakage) have an increased incidence of malignancy in general. (haematologica.org)
  • Myelodysplastic syndrome (MDS), a heterogeneous group of hematopoietic malignancy, has been shown to present different cytogenetic abnormalities, risk factors, and clinico-hematological features in different p. (biomedcentral.com)
  • Additionally, medical radiation or chemotherapy, which is often used to treat cancers, can cause further DNA damage in people with Nijmegen breakage syndrome. (medlineplus.gov)
  • Sometimes referred to as "Poland syndrome," it is an uncommon condition present at birth (congenital). (the-medical-dictionary.com)
  • Consequently, Nijmegen breakage syndrome has long been considered a variant of A-T. (medscape.com)
  • Nijmegen breakage syndrome gets its name from numerous breaks in DNA that occur in affected people's cells. (medlineplus.gov)
  • A buildup of these breaks lead to errors in DNA that can trigger cells to grow and divide abnormally, increasing the risk of cancer in people with Nijmegen breakage syndrome. (medlineplus.gov)
  • The name derives from the Dutch city Nijmegen, where the condition was first described. (wikipedia.org)
  • People with Nijmegen breakage syndrome are 50 times more likely to develop cancer than people without this condition. (medlineplus.gov)

No images available that match "nijmegen breakage syndrome"