A familial, nontransient HYPOGLYCEMIA with defects in negative feedback of GLUCOSE-regulated INSULIN release. Clinical phenotypes include HYPOGLYCEMIA; HYPERINSULINEMIA; SEIZURES; COMA; and often large BIRTH WEIGHT. Several sub-types exist with the most common, type 1, associated with mutations on an ATP-BINDING CASSETTE TRANSPORTERS (subfamily C, member 8).
A syndrome with excessively high INSULIN levels in the BLOOD. It may cause HYPOGLYCEMIA. Etiology of hyperinsulinism varies, including hypersecretion of a beta cell tumor (INSULINOMA); autoantibodies against insulin (INSULIN ANTIBODIES); defective insulin receptor (INSULIN RESISTANCE); or overuse of exogenous insulin or HYPOGLYCEMIC AGENTS.
ATP-BINDING CASSETTE PROTEINS that are highly conserved and widely expressed in nature. They form an integral part of the ATP-sensitive potassium channel complex which has two intracellular nucleotide folds that bind to sulfonylureas and their analogs.
Proteins that bind specific drugs with high affinity and trigger intracellular changes influencing the behavior of cells. Drug receptors are generally thought to be receptors for some endogenous substance not otherwise specified.
Potassium channels where the flow of K+ ions into the cell is greater than the outward flow.
A benzothiadiazine derivative that is a peripheral vasodilator used for hypertensive emergencies. It lacks diuretic effect, apparently because it lacks a sulfonamide group.
A syndrome of abnormally low BLOOD GLUCOSE level. Clinical hypoglycemia has diverse etiologies. Severe hypoglycemia eventually lead to glucose deprivation of the CENTRAL NERVOUS SYSTEM resulting in HUNGER; SWEATING; PARESTHESIA; impaired mental function; SEIZURES; COMA; and even DEATH.
Methods and procedures for the diagnosis of diseases or dysfunction of the endocrine glands or demonstration of their physiological processes.
Elevated level of AMMONIA in the blood. It is a sign of defective CATABOLISM of AMINO ACIDS or ammonia to UREA.
An enzyme that catalyzes the conversion of L-glutamate and water to 2-oxoglutarate and NH3 in the presence of NAD+. (From Enzyme Nomenclature, 1992) EC 1.4.1.2.
A family of MEMBRANE TRANSPORT PROTEINS that require ATP hydrolysis for the transport of substrates across membranes. The protein family derives its name from the ATP-binding domain found on the protein.
Surgical removal of the pancreas. (Dorland, 28th ed)
A 51-amino acid pancreatic hormone that plays a major role in the regulation of glucose metabolism, directly by suppressing endogenous glucose production (GLYCOGENOLYSIS; GLUCONEOGENESIS) and indirectly by suppressing GLUCAGON secretion and LIPOLYSIS. Native insulin is a globular protein comprised of a zinc-coordinated hexamer. Each insulin monomer containing two chains, A (21 residues) and B (30 residues), linked by two disulfide bonds. Insulin is used as a drug to control insulin-dependent diabetes mellitus (DIABETES MELLITUS, TYPE 1).
A benign tumor of the PANCREATIC BETA CELLS. Insulinoma secretes excess INSULIN resulting in HYPOGLYCEMIA.
Heteromultimers of Kir6 channels (the pore portion) and sulfonylurea receptor (the regulatory portion) which affect function of the HEART; PANCREATIC BETA CELLS; and KIDNEY COLLECTING DUCTS. KATP channel blockers include GLIBENCLAMIDE and mitiglinide whereas openers include CROMAKALIM and minoxidil sulfate.
Antibodies specific to INSULIN.
Sulfonylurea compounds are a class of medications used in the treatment of diabetes mellitus type 2 that promote insulin secretion from pancreatic beta-cells by closing ATP-sensitive potassium channels in their membranes.
A syndrome of multiple defects characterized primarily by umbilical hernia (HERNIA, UMBILICAL); MACROGLOSSIA; and GIGANTISM; and secondarily by visceromegaly; HYPOGLYCEMIA; and ear abnormalities.
Enzymes that reversibly catalyze the oxidation of a 3-hydroxyacyl CoA to 3-ketoacyl CoA in the presence of NAD. They are key enzymes in the oxidation of fatty acids and in mitochondrial fatty acid synthesis.
Cell membrane glycoproteins that are selectively permeable to potassium ions. At least eight major groups of K channels exist and they are made up of dozens of different subunits.
An infant during the first month after birth.
A genetically heterogeneous group of heritable disorders resulting from defects in protein N-glycosylation.
A potent inhibitor of CYCLIN-DEPENDENT KINASES in G1 PHASE and S PHASE. In humans, aberrant expression of p57 is associated with various NEOPLASMS as well as with BECKWITH-WIEDEMANN SYNDROME.
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.
A specific pair of GROUP C CHROMOSOMES of the human chromosome classification.
Irregular microscopic structures consisting of cords of endocrine cells that are scattered throughout the PANCREAS among the exocrine acini. Each islet is surrounded by connective tissue fibers and penetrated by a network of capillaries. There are four major cell types. The most abundant beta cells (50-80%) secrete INSULIN. Alpha cells (5-20%) secrete GLUCAGON. PP cells (10-35%) secrete PANCREATIC POLYPEPTIDE. Delta cells (~5%) secrete SOMATOSTATIN.
A sulphonylurea hypoglycemic agent with actions and uses similar to those of CHLORPROPAMIDE. (From Martindale, The Extra Pharmacopoeia, 30th ed, p290)
A type of pancreatic cell representing about 50-80% of the islet cells. Beta cells secrete INSULIN.
A condition caused by the excessive secretion of ANDROGENS from the ADRENAL CORTEX; the OVARIES; or the TESTES. The clinical significance in males is negligible. In women, the common manifestations are HIRSUTISM and VIRILISM as seen in patients with POLYCYSTIC OVARY SYNDROME and ADRENOCORTICAL HYPERFUNCTION.
Glucose in blood.
A mutation in which a codon is mutated to one directing the incorporation of a different amino acid. This substitution may result in an inactive or unstable product. (From A Dictionary of Genetics, King & Stansfield, 5th ed)
A benzenesulfonamide-phthalimidine that tautomerizes to a BENZOPHENONES form. It is considered a thiazide-like diuretic.
A potent, long-acting synthetic SOMATOSTATIN octapeptide analog that inhibits secretion of GROWTH HORMONE and is used to treat hormone-secreting tumors; DIABETES MELLITUS; HYPOTENSION, ORTHOSTATIC; HYPERINSULINISM; hypergastrinemia; and small bowel fistula.
A pancreatic polypeptide of about 110 amino acids, depending on the species, that is the precursor of insulin. Proinsulin, produced by the PANCREATIC BETA CELLS, is comprised sequentially of the N-terminal B-chain, the proteolytically removable connecting C-peptide, and the C-terminal A-chain. It also contains three disulfide bonds, two between A-chain and B-chain. After cleavage at two locations, insulin and C-peptide are the secreted products. Intact proinsulin with low bioactivity also is secreted in small amounts.
A nodular organ in the ABDOMEN that contains a mixture of ENDOCRINE GLANDS and EXOCRINE GLANDS. The small endocrine portion consists of the ISLETS OF LANGERHANS secreting a number of hormones into the blood stream. The large exocrine portion (EXOCRINE PANCREAS) is a compound acinar gland that secretes several digestive enzymes into the pancreatic ductal system that empties into the DUODENUM.
A focal malformation resembling a neoplasm, composed of an overgrowth of mature cells and tissues that normally occur in the affected area.
The middle segment of proinsulin that is between the N-terminal B-chain and the C-terminal A-chain. It is a pancreatic peptide of about 31 residues, depending on the species. Upon proteolytic cleavage of proinsulin, equimolar INSULIN and C-peptide are released. C-peptide immunoassay has been used to assess pancreatic beta cell function in diabetic patients with circulating insulin antibodies or exogenous insulin. Half-life of C-peptide is 30 min, almost 8 times that of insulin.
An individual having different alleles at one or more loci regarding a specific character.
The occurrence in an individual of two or more cell populations of different chromosomal constitutions, derived from a single ZYGOTE, as opposed to CHIMERISM in which the different cell populations are derived from more than one zygote.
Tumors or cancer of the PANCREAS. Depending on the types of ISLET CELLS present in the tumors, various hormones can be secreted: GLUCAGON from PANCREATIC ALPHA CELLS; INSULIN from PANCREATIC BETA CELLS; and SOMATOSTATIN from the SOMATOSTATIN-SECRETING CELLS. Most are malignant except the insulin-producing tumors (INSULINOMA).
Diminution or cessation of secretion of one or more hormones from the anterior pituitary gland (including LH; FOLLICLE STIMULATING HORMONE; SOMATOTROPIN; and CORTICOTROPIN). This may result from surgical or radiation ablation, non-secretory PITUITARY NEOPLASMS, metastatic tumors, infarction, PITUITARY APOPLEXY, infiltrative or granulomatous processes, and other conditions.
The record of descent or ancestry, particularly of a particular condition or trait, indicating individual family members, their relationships, and their status with respect to the trait or condition.
A colorless alkaline gas. It is formed in the body during decomposition of organic materials during a large number of metabolically important reactions. Note that the aqueous form of ammonia is referred to as AMMONIUM HYDROXIDE.
Clinical or subclinical disturbances of cortical function due to a sudden, abnormal, excessive, and disorganized discharge of brain cells. Clinical manifestations include abnormal motor, sensory and psychic phenomena. Recurrent seizures are usually referred to as EPILEPSY or "seizure disorder."
Genes that influence the PHENOTYPE both in the homozygous and the heterozygous state.
A characteristic symptom complex.
An adenine nucleotide containing three phosphate groups esterified to the sugar moiety. In addition to its crucial roles in metabolism adenosine triphosphate is a neurotransmitter.
A primary source of energy for living organisms. It is naturally occurring and is found in fruits and other parts of plants in its free state. It is used therapeutically in fluid and nutrient replacement.
An individual in which both alleles at a given locus are identical.
A test to determine the ability of an individual to maintain HOMEOSTASIS of BLOOD GLUCOSE. It includes measuring blood glucose levels in a fasting state, and at prescribed intervals before and after oral glucose intake (75 or 100 g) or intravenous infusion (0.5 g/kg).
A mutation caused by the substitution of one nucleotide for another. This results in the DNA molecule having a change in a single base pair.

Effect of hyperglycemia-hyperinsulinemia on whole body and regional fatty acid metabolism. (1/1504)

The effects of combined hyperglycemia-hyperinsulinemia on whole body, splanchnic, and leg fatty acid metabolism were determined in five volunteers. Catheters were placed in a femoral artery and vein and a hepatic vein. U-13C-labeled fatty acids were infused, once in the basal state and, on a different occasion, during infusion of dextrose (clamp; arterial glucose 8.8 +/- 0.5 mmol/l). Lipids and heparin were infused together with the dextrose to maintain plasma fatty acid concentrations at basal levels. Fatty acid availability in plasma and fatty acid uptake across the splanchnic region and the leg were similar during the basal and clamp experiments. Dextrose infusion decreased fatty acid oxidation by 51.8% (whole body), 47.4% (splanchnic), and 64.3% (leg). Similarly, the percent fatty acid uptake oxidized decreased at the whole body level (53 to 29%), across the splanchnic region (30 to 13%), and in the leg (48 to 22%) during the clamp. We conclude that, in healthy men, combined hyperglycemia-hyperinsulinemia inhibits fatty acid oxidation to a similar extent at the whole body level, across the leg, and across the splanchnic region, even when fatty acid availability is constant.  (+info)

Relative contribution of insulin and its precursors to fibrinogen and PAI-1 in a large population with different states of glucose tolerance. The Insulin Resistance Atherosclerosis Study (IRAS). (2/1504)

Hyperinsulinemia is associated with the development of coronary heart disease. However, the underlying mechanisms are still poorly understood. Hypercoagulability and impaired fibrinolysis are possible candidates linking hyperinsulinism with atherosclerotic disease, and it has been suggested that proinsulin rather than insulin is the crucial pathophysiological agent. The aim of this study was to investigate the relationship of insulin and its precursors to markers of coagulation and fibrinolysis in a large triethnic population. A strong and independent relationship between plasminogen activator inhibitor-1 (PAI-1) antigen and insulin and its precursors (proinsulin, 32-33 split proinsulin) was found consistently across varying states of glucose tolerance (PAI-1 versus fasting insulin [proinsulin], r=0.38 [r=0.34] in normal glucose tolerance; r=0.42 [r=0.43] in impaired glucose tolerance; and r=0.38 [r=0.26] in type 2 diabetes; all P<0.001). The relationship remained highly significant even after accounting for insulin sensitivity as measured by a frequently sampled intravenous glucose tolerance test. In a stepwise multiple regression model after adjusting for age, sex, ethnicity, and clinic, both insulin and its precursors were significantly associated with PAI-1 levels. The relationship between fibrinogen and insulin and its precursors was significant in the overall population (r=0.20 for insulin and proinsulin; each P<0.001) but showed a more inconsistent pattern in subgroup analysis and after adjustments for demographic and metabolic variables. Stepwise multiple regression analysis showed that proinsulin (split products) but not fasting insulin significantly contributed to fibrinogen levels after adjustment for age, sex, clinic, and ethnicity. Decreased insulin sensitivity was independently associated with higher PAI-1 and fibrinogen levels. In summary, we were able to demonstrate an independent relationship of 2 crucial factors of hemostasis, fibrinogen and PAI-1, to insulin and its precursors. These findings may have important clinical implications in the risk assessment and prevention of macrovascular disease, not only in patients with overt diabetes but also in nondiabetic subjects who are hyperinsulinemic.  (+info)

The contributions of oestrogen and growth factors to increased adrenal androgen secretion in polycystic ovary syndrome. (3/1504)

Adrenal hyperandrogenism is prevalent in many women with polycystic ovary syndrome (PCOS), although the expression of this enhanced secretion may be heterogeneous. Since no single factor acts in isolation, this study was performed to assess the influence of oestradiol (total and unbound), insulin, insulin-like growth factor (IGF)-I, IGF-II and the binding proteins IGFBP-I, and IGFBP-3, on basal and adrenocorticotrophic hormone (ACTH) stimulated adrenal androgen secretion in 25 women with PCOS and 10 matched ovulatory controls. Women with PCOS exhibited elevations of all androgens as well as unbound oestradiol, insulin and non-IGFBP-1 bound IGF-I. Positive correlations were noted between oestrogen and basal and ACTH stimulated delta 5 adrenal androgens. Serum IGF-I was only correlated with basal dehydroepiandrosterone sulphate (DHEA-S), while insulin exhibited a strong correlation with the delta 4 pathway and androstenedione formation in particular. This correlation was also confirmed by dividing the PCOS group into those women with and without hyperinsulinaemia. The activity of 17,20 lyase favouring androstenedione was increased in the hyperinsulinaemic women. By multivariate analyses, body mass index did not influence these findings. Although there are inherent difficulties in making major conclusions based on correlative analyses, it is suggested that oestrogen may have a greater influence on enhancing delta 5 adrenal androgen secretion, and insulin a greater effect on the delta 4 pathway. In turn, the relative importance of these influences may contribute to the heterogeneous nature of adrenal hyperandrogenism in PCOS.  (+info)

Pancreatic exocrine and endocrine function after pancreatectomy for persistent hyperinsulinaemic hypoglycaemia of infancy. (4/1504)

AIM: To evaluate long term detailed pancreatic endocrine and exocrine function in children with persistent hyperinsulinaemic hypoglycaemia of infancy (PHHI) after 85-95% pancreatectomy. METHODS: Six children with PHHI between 0.9 and 12.7 years after pancreatic resection underwent clinical and investigative follow up at 1.0 to 14.9 years of age. One child with PHHI who had not had pancreatectomy was also assessed. Standard endocrine assessment, pancreatic magnetic resonance imaging (MRI), and detailed direct and indirect tests of exocrine pancreatic function were performed. RESULTS: Pancreozymin-secretin stimulation test results were normal in only one child, borderline in two, and deficient in four, one of whom requires daily pancreatic enzyme supplements. Pancreolauryl tests performed in three children were borderline in two and abnormal in the other. Only one child had low faecal chymotrypsin values. One child developed insulin dependent diabetes at 9 years and two children at 1.0 and 13.3 years require diazoxide to maintain normoglycaemia. MRI showed no major regrowth of the pancreatic remnant after resection (n = 5). CONCLUSIONS: Clinical evidence of endocrine or exocrine dysfunction has developed in only two patients to date, but detailed pancreatic function testing suggests subclinical deficiency in all but one of our patients with PHHI. Although 95% pancreatectomy results in postoperative control of blood glucose, subclinical pancreatic insufficiency is present on long term follow up and development of diabetes mellitus and exocrine failure remain ongoing risks.  (+info)

Long-term follow up of persistent hyperinsulinaemic hypoglycaemia of infancy. (5/1504)

Twenty six children with hypoglycaemia were diagnosed and followed between 1975 and 1995. Diagnosis was confirmed by a high insulin:glucose ratio, and low free fatty acid and 3-hydroxybutyrate on fasting. All patients were treated with diazoxide at a maximum dose of 20 mg/kg/day. Requirement of a higher dose was considered as a failure of medical treatment and an indication for surgery. Sixteen children Responded to diazoxide; 10 failed to respond and underwent pancreatic resection. Six of the latter group started with symptoms in the neonatal period. Eleven of the 26 children have neurological sequelae. Head growth and neurological outcome correlated well. Additionally, non-specific electroencephalogram abnormalities (slow waves) appear to be indicative of subclinical hypoglycaemia during follow up.  (+info)

Hyperinsulinism: molecular aetiology of focal disease. (6/1504)

Persistent hypoglycaemia in infancy is most commonly caused by hyperinsulinism. A case is reported of the somatic loss of the maternal 11p in an insulin secreting focal adenoma in association with a germline SUR-1 mutation on the paternal allele in a baby boy with hyperinsulinism diagnosed at 49 days old. A reduction to homozygosity of an SUR-1 mutation is proposed as a critical part of the cause of focal hyperinsulinism.  (+info)

The structure and function of the ATP-sensitive K+ channel in insulin-secreting pancreatic beta-cells. (7/1504)

ATP-sensitive K+ channels (KATP channels) play important roles in many cellular functions by coupling cell metabolism to electrical activity. The KATP channels in pancreatic beta-cells are thought to be critical in the regulation of glucose-induced and sulfonylurea-induced insulin secretion. Until recently, however, the molecular structure of the KATP channel was not known. Cloning members of the novel inwardly rectifying K+ channel subfamily Kir6.0 (Kir6.1 and Kir6.2) and the sulfonylurea receptors (SUR1 and SUR2) has clarified the molecular structure of KATP channels. The pancreatic beta-cell KATP channel comprises two subunits: a Kir6.2 subunit and an SUR1 subunit. Molecular biological and molecular genetic studies have provided insights into the physiological and pathophysiological roles of the pancreatic beta-cell KATP channel in insulin secretion.  (+info)

Surgery-induced insulin resistance in human patients: relation to glucose transport and utilization. (8/1504)

To investigate the underlying molecular mechanisms for surgery-induced insulin resistance in skeletal muscle, six otherwise healthy patients undergoing total hip replacement were studied before, during, and after surgery. Patients were studied under basal conditions and during physiological hyperinsulinemia (60 microU/ml). Biopsies of vastus lateralis muscle were used to measure GLUT-4 translocation, glucose transport, and glycogen synthase activities. Surgery reduced insulin-stimulated glucose disposal (P < 0.05) without altering the insulin-stimulated increase in glucose oxidation or suppression of endogenous glucose production. Preoperatively, insulin infusion increased plasma membrane GLUT-4 in all six subjects (P < 0.05), whereas insulin-stimulated GLUT-4 translocation only occurred in three patients postoperatively (not significant). Moreover, nonoxidative glucose disposal rates and basal levels of glycogen synthase activities in muscle were reduced postoperatively (P < 0.05). These findings demonstrate that peripheral insulin resistance develops immediately postoperatively and that this condition might be associated with perturbations in insulin-stimulated GLUT-4 translocation as well as nonoxidative glucose disposal, presumably at the level of glycogen synthesis.  (+info)

Congenital hyperinsulinism is a medical condition that is present at birth and characterized by the excessive production and release of insulin from the beta cells of the pancreas. Insulin is a hormone that regulates blood sugar levels, and an overproduction of it can lead to low blood sugar (hypoglycemia).

There are two main types of congenital hyperinsulinism: diffuse and focal. Diffuse hyperinsulinism affects the entire pancreas, while focal hyperinsulinism affects only a small part of it. The condition can be caused by genetic mutations that affect the way insulin is produced or released from the beta cells.

Symptoms of congenital hyperinsulinism may include hypoglycemia, which can cause symptoms such as seizures, lethargy, irritability, and poor feeding. If left untreated, severe hypoglycemia can lead to brain damage or even death. Treatment for congenital hyperinsulinism typically involves medication to control blood sugar levels, as well as dietary modifications and, in some cases, surgery to remove the affected part of the pancreas.

Hyperinsulinism is a medical condition characterized by an excess production and release of insulin from the pancreas. Insulin is a hormone that helps regulate blood sugar levels by allowing cells in the body to take in sugar (glucose) for energy or storage. In hyperinsulinism, the increased insulin levels can cause low blood sugar (hypoglycemia), which can lead to symptoms such as sweating, shaking, confusion, and in severe cases, seizures or loss of consciousness.

There are several types of hyperinsulinism, including congenital forms that are present at birth and acquired forms that develop later in life. Congenital hyperinsulinism is often caused by genetic mutations that affect the way insulin is produced or released from the pancreas. Acquired hyperinsulinism can be caused by factors such as certain medications, hormonal disorders, or tumors of the pancreas.

Treatment for hyperinsulinism depends on the underlying cause and severity of the condition. Treatment options may include dietary changes, medication to reduce insulin secretion, or surgery to remove part or all of the pancreas.

Sulfonylurea receptors (SURs) are a type of transmembrane protein found in the beta cells of the pancreas. They are part of the ATP-sensitive potassium (KATP) channel complex, which plays a crucial role in regulating insulin secretion.

SURs have two subtypes, SUR1 and SUR2, which are associated with different KATP channel subunits. SUR1 is primarily found in the pancreas and brain, while SUR2 is expressed in various tissues, including skeletal muscle and heart.

Sulfonylurea drugs, used to treat type 2 diabetes, bind to SURs and stimulate insulin secretion by closing the KATP channel, which leads to membrane depolarization and subsequent calcium influx, triggering insulin release from beta cells.

Drug receptors are specific protein molecules found on the surface of cells, to which drugs can bind. These receptors are part of the cell's communication system and are responsible for responding to neurotransmitters, hormones, and other signaling molecules in the body. When a drug binds to its corresponding receptor, it can alter the receptor's function and trigger a cascade of intracellular events that ultimately lead to a biological response.

Drug receptors can be classified into several types based on their function, including:

1. G protein-coupled receptors (GPCRs): These are the largest family of drug receptors and are involved in various physiological processes such as vision, olfaction, neurotransmission, and hormone signaling. They activate intracellular signaling pathways through heterotrimeric G proteins.
2. Ion channel receptors: These receptors form ion channels that allow the flow of ions across the cell membrane when activated. They are involved in rapid signal transduction and can be directly gated by ligands or indirectly through G protein-coupled receptors.
3. Enzyme-linked receptors: These receptors have an intracellular domain that functions as an enzyme, activating intracellular signaling pathways when bound to a ligand. Examples include receptor tyrosine kinases and receptor serine/threonine kinases.
4. Nuclear receptors: These receptors are located in the nucleus and function as transcription factors, regulating gene expression upon binding to their ligands.

Understanding drug receptors is crucial for developing new drugs and predicting their potential therapeutic and adverse effects. By targeting specific receptors, drugs can modulate cellular responses and produce desired pharmacological actions.

Inwardly rectifying potassium channels (Kir) are a type of potassium channel that allow for the selective passage of potassium ions (K+) across cell membranes. The term "inwardly rectifying" refers to their unique property of allowing potassium ions to flow more easily into the cell (inward current) than out of the cell (outward current). This characteristic is due to the voltage-dependent blockage of these channels by intracellular magnesium and polyamines at depolarized potentials.

These channels play crucial roles in various physiological processes, including:

1. Resting membrane potential maintenance: Kir channels help establish and maintain the negative resting membrane potential in cells by facilitating potassium efflux when the membrane potential is near the potassium equilibrium potential (Ek).
2. Action potential repolarization: In excitable cells like neurons and muscle fibers, Kir channels contribute to the rapid repolarization phase of action potentials, allowing for proper electrical signaling.
3. Cell volume regulation: Kir channels are involved in regulating cell volume by mediating potassium influx during osmotic stress or changes in intracellular ion concentrations.
4. Insulin secretion: In pancreatic β-cells, Kir channels control the membrane potential and calcium signaling necessary for insulin release.
5. Renal function: Kir channels are essential for maintaining electrolyte balance and controlling renal tubular transport in the kidneys.

There are several subfamilies of inwardly rectifying potassium channels (Kir1-7), each with distinct biophysical properties, tissue distributions, and functions. Mutations in genes encoding these channels can lead to various human diseases, including cardiac arrhythmias, epilepsy, and Bartter syndrome.

Diazoxide is a medication that is primarily used to treat hypoglycemia (low blood sugar) in newborns and infants. It works by inhibiting the release of insulin from the pancreas, which helps to prevent the blood sugar levels from dropping too low. Diazoxide may also be used in adults with certain rare conditions that cause hypoglycemia.

In addition to its use as a hypoglycemic agent, diazoxide has been used off-label for other indications, such as the treatment of hypertension (high blood pressure) that is resistant to other medications. It works as a vasodilator, relaxing the smooth muscle in the walls of blood vessels and causing them to widen, which reduces the resistance to blood flow and lowers blood pressure.

Diazoxide is available as an injection and is typically administered in a hospital setting under the close supervision of a healthcare professional. Common side effects of diazoxide include fluid retention, headache, nausea, and vomiting. It may also cause rare but serious side effects such as heart rhythm disturbances and allergic reactions.

Hypoglycemia is a medical condition characterized by an abnormally low level of glucose (sugar) in the blood. Generally, hypoglycemia is defined as a blood glucose level below 70 mg/dL (3.9 mmol/L), although symptoms may not occur until the blood sugar level falls below 55 mg/dL (3.0 mmol/L).

Hypoglycemia can occur in people with diabetes who are taking insulin or medications that increase insulin production, as well as those with certain medical conditions such as hormone deficiencies, severe liver illnesses, or disorders of the adrenal glands. Symptoms of hypoglycemia include sweating, shaking, confusion, rapid heartbeat, and in severe cases, loss of consciousness or seizures.

Hypoglycemia is typically treated by consuming fast-acting carbohydrates such as fruit juice, candy, or glucose tablets to rapidly raise blood sugar levels. If left untreated, hypoglycemia can lead to serious complications, including brain damage and even death.

Diagnostic techniques in endocrinology are methods used to identify and diagnose various endocrine disorders. These techniques include:

1. Hormone measurements: Measuring the levels of hormones in blood, urine, or saliva can help identify excess or deficiency of specific hormones. This is often done through immunoassays, which use antibodies to detect and quantify hormones.

2. Provocative and suppression tests: These tests involve administering a medication that stimulates or suppresses the release of a particular hormone. Blood samples are taken before and after the medication is given to assess changes in hormone levels. Examples include the glucose tolerance test for diabetes, the ACTH stimulation test for adrenal insufficiency, and the thyroid suppression test for hyperthyroidism.

3. Imaging studies: Various imaging techniques can be used to visualize endocrine glands and identify structural abnormalities such as tumors or nodules. These include X-rays, ultrasound, computed tomography (CT), magnetic resonance imaging (MRI), and nuclear medicine scans using radioactive tracers.

4. Genetic testing: Molecular genetic tests can be used to identify genetic mutations associated with certain endocrine disorders, such as multiple endocrine neoplasia type 1 or 2, or congenital adrenal hyperplasia.

5. Biopsy: In some cases, a small sample of tissue may be removed from an endocrine gland for microscopic examination (biopsy). This can help confirm the presence of cancer or other abnormalities.

6. Functional tests: These tests assess the ability of an endocrine gland to produce and secrete hormones in response to various stimuli. Examples include the glucagon stimulation test for gastrinoma and the calcium infusion test for hyperparathyroidism.

7. Wearable monitoring devices: Continuous glucose monitoring systems (CGMS) are wearable devices that measure interstitial glucose levels continuously over several days, providing valuable information about glycemic control in patients with diabetes.

Hyperammonemia is a medical condition characterized by an excessively high level of ammonia (a toxic byproduct of protein metabolism) in the blood. This can lead to serious neurological symptoms and complications, as ammonia is highly toxic to the brain. Hyperammonemia can be caused by various underlying conditions, including liver disease, genetic disorders that affect ammonia metabolism, certain medications, and infections. It is important to diagnose and treat hyperammonemia promptly to prevent long-term neurological damage or even death. Treatment typically involves addressing the underlying cause of the condition, as well as providing supportive care such as administering medications that help remove ammonia from the blood.

Glutamate Dehydrogenase (GLDH or GDH) is a mitochondrial enzyme that plays a crucial role in the metabolism of amino acids, particularly within liver and kidney tissues. It catalyzes the reversible oxidative deamination of glutamate to alpha-ketoglutarate, which links amino acid metabolism with the citric acid cycle and energy production. This enzyme is significant in clinical settings as its levels in blood serum can be used as a diagnostic marker for diseases that damage liver or kidney cells, since these cells release GLDH into the bloodstream upon damage.

ATP-binding cassette (ABC) transporters are a family of membrane proteins that utilize the energy from ATP hydrolysis to transport various substrates across extra- and intracellular membranes. These transporters play crucial roles in several biological processes, including detoxification, drug resistance, nutrient uptake, and regulation of cellular cholesterol homeostasis.

The structure of ABC transporters consists of two nucleotide-binding domains (NBDs) that bind and hydrolyze ATP, and two transmembrane domains (TMDs) that form the substrate-translocation pathway. The NBDs are typically located adjacent to each other in the cytoplasm, while the TMDs can be either integral membrane domains or separate structures associated with the membrane.

The human genome encodes 48 distinct ABC transporters, which are classified into seven subfamilies (ABCA-ABCG) based on their sequence similarity and domain organization. Some well-known examples of ABC transporters include P-glycoprotein (ABCB1), multidrug resistance protein 1 (ABCC1), and breast cancer resistance protein (ABCG2).

Dysregulation or mutations in ABC transporters have been implicated in various diseases, such as cystic fibrosis, neurological disorders, and cancer. In cancer, overexpression of certain ABC transporters can contribute to drug resistance by actively effluxing chemotherapeutic agents from cancer cells, making them less susceptible to treatment.

A pancreatectomy is a surgical procedure in which all or part of the pancreas is removed. There are several types of pancreatectomies, including:

* **Total pancreatectomy:** Removal of the entire pancreas, as well as the spleen and nearby lymph nodes. This type of pancreatectomy is usually done for patients with cancer that has spread throughout the pancreas or for those who have had multiple surgeries to remove pancreatic tumors.
* **Distal pancreatectomy:** Removal of the body and tail of the pancreas, as well as nearby lymph nodes. This type of pancreatectomy is often done for patients with tumors in the body or tail of the pancreas.
* **Partial (or segmental) pancreatectomy:** Removal of a portion of the head or body of the pancreas, as well as nearby lymph nodes. This type of pancreatectomy is often done for patients with tumors in the head or body of the pancreas that can be removed without removing the entire organ.
* **Pylorus-preserving pancreaticoduodenectomy (PPPD):** A type of surgery used to treat tumors in the head of the pancreas, as well as other conditions such as chronic pancreatitis. In this procedure, the head of the pancreas, duodenum, gallbladder, and bile duct are removed, but the stomach and lower portion of the esophagus (pylorus) are left in place.

After a pancreatectomy, patients may experience problems with digestion and blood sugar regulation, as the pancreas plays an important role in these functions. Patients may need to take enzyme supplements to help with digestion and may require insulin therapy to manage their blood sugar levels.

Insulin is a hormone produced by the beta cells of the pancreatic islets, primarily in response to elevated levels of glucose in the circulating blood. It plays a crucial role in regulating blood glucose levels and facilitating the uptake and utilization of glucose by peripheral tissues, such as muscle and adipose tissue, for energy production and storage. Insulin also inhibits glucose production in the liver and promotes the storage of excess glucose as glycogen or triglycerides.

Deficiency in insulin secretion or action leads to impaired glucose regulation and can result in conditions such as diabetes mellitus, characterized by chronic hyperglycemia and associated complications. Exogenous insulin is used as a replacement therapy in individuals with diabetes to help manage their blood glucose levels and prevent long-term complications.

Insulinoma is a rare type of neuroendocrine tumor that originates from the beta cells of the pancreatic islets (islets of Langerhans). These tumors produce and secrete excessive amounts of insulin, leading to hypoglycemia (low blood sugar levels) even when the person hasn't eaten for a while. Insulinomas are typically slow-growing and benign (noncancerous), but about 10% of them can be malignant (cancerous) and may spread to other parts of the body. Common symptoms include sweating, confusion, dizziness, and weakness due to low blood sugar levels. The diagnosis is often confirmed through imaging tests like CT scans or MRI, and measuring insulin and C-peptide levels in the blood during a fasting test. Treatment usually involves surgical removal of the tumor.

ATP-sensitive potassium (KATP) channels are a type of ion channel found in the membranes of cells, including those in the heart, muscle, and pancreas. These channels are unique because their opening and closing are regulated by the levels of adenosine triphosphate (ATP) and adenosine diphosphate (ADP) in the cell.

Under normal conditions, when ATP levels are high and ADP levels are low, the KATP channels are closed, which allows the cells to maintain their normal electrical activity. However, during times of metabolic stress or ischemia (a lack of blood flow), the levels of ATP in the cell decrease while the levels of ADP increase. This change in the ATP-to-ADP ratio causes the KATP channels to open, which allows potassium ions to flow out of the cell. The efflux of potassium ions then leads to hyperpolarization of the cell membrane, which helps to protect the cells from damage.

In the pancreas, KATP channels play a crucial role in regulating insulin secretion. In the beta cells of the pancreas, an increase in blood glucose levels leads to an increase in ATP production and a decrease in ADP levels, which causes the KATP channels to close. This closure of the KATP channels leads to depolarization of the cell membrane, which triggers the release of insulin.

Overall, KATP channels are important regulators of cellular electrical activity and play a critical role in protecting cells from damage during times of metabolic stress or ischemia.

Insulin antibodies are proteins produced by the immune system that recognize and bind to insulin. They are typically formed in response to an exposure to exogenous insulin, such as in people with diabetes who use insulin therapy. In some cases, the presence of insulin antibodies can affect insulin absorption, distribution, metabolism, and elimination, leading to variable insulin requirements, reduced glycemic control, and potentially an increased risk of hypoglycemia or hyperglycemia. However, not all individuals with insulin antibodies experience clinical consequences, and the significance of their presence can vary between individuals.

Sulfonylurea compounds are a group of medications used in the management of type 2 diabetes. They work by stimulating the release of insulin from the pancreas, thereby lowering blood glucose levels. These compounds bind to specific receptors on the beta cells of the pancreas, which triggers the release of insulin.

Examples of sulfonylurea compounds include glipizide, glyburide, and glimepiride. It's important to note that these medications can cause hypoglycemia (low blood sugar) if not properly monitored and dosed. They are often used in combination with other medications, such as metformin, to achieve optimal blood glucose control.

As with any medication, sulfonylurea compounds should be taken under the supervision of a healthcare provider, who can monitor their effectiveness and potential side effects.

Beckwith-Wiedemann syndrome (BWS) is a genetic overgrowth disorder that affects several parts of the body. It is characterized by an increased risk of developing certain tumors, especially during the first few years of life. The symptoms and features of BWS can vary widely among affected individuals.

The medical definition of Beckwith-Wiedemann syndrome includes the following major criteria:

1. Excessive growth before birth (macrosomia) or in infancy (infantile gigantism)
2. Enlargement of the tongue (macroglossia)
3. Abdominal wall defects, such as an omphalocele (protrusion of abdominal organs through the belly button) or a diastasis recti (separation of the abdominal muscles)
4. Enlargement of specific internal organs, like the kidneys, liver, or pancreas
5. A distinctive facial appearance, which may include ear creases or pits, wide-set eyes, and a prominent jaw

Additional findings in BWS can include:

1. Increased risk of developing embryonal tumors, such as Wilms tumor (a type of kidney cancer), hepatoblastoma (a liver cancer), and neuroblastoma (a nerve tissue cancer)
2. Hypoglycemia (low blood sugar) in infancy due to hyperinsulinism (overproduction of insulin)
3. Asymmetric growth, where one side of the body or a specific region is significantly larger than the other
4. Ear abnormalities, such as cupped ears or low-set ears
5. Developmental delays and learning disabilities in some cases

Beckwith-Wiedemann syndrome is caused by changes in the chromosome 11p15 region, which contains several genes that regulate growth and development. The most common cause of BWS is an epigenetic abnormality called paternal uniparental disomy (UPD), where both copies of this region come from the father instead of one copy from each parent. Other genetic mechanisms, such as mutations in specific genes or imprinting center defects, can also lead to BWS.

The diagnosis of Beckwith-Wiedemann syndrome is typically based on clinical findings and confirmed by molecular testing. Management includes regular monitoring for tumor development, controlling hypoglycemia, and addressing any other complications as needed. Surgical intervention may be required in cases of organ enlargement or structural abnormalities. Genetic counseling is recommended for affected individuals and their families to discuss the risks of recurrence and available reproductive options.

3-Hydroxyacyl CoA Dehydrogenases (3-HADs) are a group of enzymes that play a crucial role in the beta-oxidation of fatty acids. These enzymes catalyze the third step of the beta-oxidation process, which involves the oxidation of 3-hydroxyacyl CoA to 3-ketoacyl CoA. This reaction is an essential part of the energy-generating process that occurs in the mitochondria of cells and allows for the breakdown of fatty acids into smaller molecules, which can then be used to produce ATP, the primary source of cellular energy.

There are several different isoforms of 3-HADs, each with specific substrate preferences and tissue distributions. The most well-known isoform is the mitochondrial 3-hydroxyacyl CoA dehydrogenase (M3HD), which is involved in the oxidation of medium and long-chain fatty acids. Other isoforms include the short-chain 3-hydroxyacyl CoA dehydrogenase (SCHAD) and the long-chain 3-hydroxyacyl CoA dehydrogenase (LCHAD), which are involved in the oxidation of shorter and longer chain fatty acids, respectively.

Deficiencies in 3-HADs can lead to serious metabolic disorders, such as 3-hydroxyacyl-CoA dehydrogenase deficiency (3-HAD deficiency), which is characterized by the accumulation of toxic levels of 3-hydroxyacyl CoAs in the body. Symptoms of this disorder can include hypoglycemia, muscle weakness, cardiomyopathy, and developmental delays. Early diagnosis and treatment of 3-HAD deficiency are essential to prevent serious complications and improve outcomes for affected individuals.

Potassium channels are membrane proteins that play a crucial role in regulating the electrical excitability of cells, including cardiac, neuronal, and muscle cells. These channels facilitate the selective passage of potassium ions (K+) across the cell membrane, maintaining the resting membrane potential and shaping action potentials. They are composed of four or six subunits that assemble to form a central pore through which potassium ions move down their electrochemical gradient. Potassium channels can be modulated by various factors such as voltage, ligands, mechanical stimuli, or temperature, allowing cells to fine-tune their electrical properties and respond to different physiological demands. Dysfunction of potassium channels has been implicated in several diseases, including cardiac arrhythmias, epilepsy, and neurodegenerative disorders.

A newborn infant is a baby who is within the first 28 days of life. This period is also referred to as the neonatal period. Newborns require specialized care and attention due to their immature bodily systems and increased vulnerability to various health issues. They are closely monitored for signs of well-being, growth, and development during this critical time.

Congenital Disorders of Glycosylation (CDG) are a group of genetic disorders that affect the body's ability to add sugar molecules (glycans) to proteins and lipids. This process, known as glycosylation, is essential for the proper functioning of many cellular processes, including protein folding, trafficking, and signaling.

CDG can be caused by mutations in genes that are involved in the synthesis or transport of glycans. These genetic defects can lead to abnormal glycosylation patterns, which can result in a wide range of clinical manifestations, including developmental delay, intellectual disability, seizures, movement disorders, hypotonia, coagulation abnormalities, and multi-organ involvement.

CDG are typically classified into two main types: type I CDG, which involves defects in the synthesis of the lipid-linked oligosaccharide precursor used for N-glycosylation, and type II CDG, which involves defects in the processing and transfer of glycans to proteins.

The diagnosis of CDG is often based on clinical features, laboratory tests, and genetic analysis. Treatment is typically supportive and multidisciplinary, focusing on addressing specific symptoms and improving quality of life. In some cases, dietary modifications or supplementation with mannose or other sugars may be beneficial.

Cyclin-dependent kinase inhibitor p57, also known as CDKN1C or p57KIP2, is a protein that regulates the cell cycle and acts as a tumor suppressor. It inhibits the activity of cyclin-dependent kinases (CDKs), which are enzymes that play crucial roles in regulating the cell cycle and transitioning from one phase to another.

The p57 protein is encoded by the CDKN1C gene, which is located on chromosome 11p15.5. This region is known as an imprinted gene cluster, meaning that only one copy of the gene is active, depending on whether it is inherited from the mother or father. In the case of p57, the paternal allele is usually silenced, and only the maternal allele is expressed.

Mutations in the CDKN1C gene can lead to several developmental disorders, including Beckwith-Wiedemann syndrome (BWS), a condition characterized by overgrowth, abdominal wall defects, and an increased risk of childhood tumors. Loss of function mutations in CDKN1C have also been associated with an increased risk of cancer, particularly Wilms' tumor, a type of kidney cancer that typically affects children.

In summary, cyclin-dependent kinase inhibitor p57 is a protein that regulates the cell cycle and acts as a tumor suppressor by inhibiting the activity of CDKs. Mutations in the CDKN1C gene can lead to developmental disorders and an increased risk of cancer.

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.

Human chromosome pair 11 consists of two rod-shaped structures present in the nucleus of each cell in the human body. Each member of the pair is a single chromosome, and together they contain the genetic material that is inherited from both parents. They are located on the eleventh position in the standard karyotype, which is a visual representation of the 23 pairs of human chromosomes.

Chromosome 11 is one of the largest human chromosomes and contains an estimated 135 million base pairs. It contains approximately 1,400 genes that provide instructions for making proteins, as well as many non-coding RNA molecules that play a role in regulating gene expression.

Chromosome 11 is known to contain several important genes and genetic regions associated with various human diseases and conditions. For example, it contains the Wilms' tumor 1 (WT1) gene, which is associated with kidney cancer in children, and the neurofibromatosis type 1 (NF1) gene, which is associated with a genetic disorder that causes benign tumors to grow on nerves throughout the body. Additionally, chromosome 11 contains the region where the ABO blood group genes are located, which determine a person's blood type.

It's worth noting that human chromosomes come in pairs because they contain two copies of each gene, one inherited from the mother and one from the father. This redundancy allows for genetic diversity and provides a backup copy of essential genes, ensuring their proper function and maintaining the stability of the genome.

The Islets of Langerhans are clusters of specialized cells within the pancreas, an organ located behind the stomach. These islets are named after Paul Langerhans, who first identified them in 1869. They constitute around 1-2% of the total mass of the pancreas and are distributed throughout its substance.

The Islets of Langerhans contain several types of cells, including:

1. Alpha (α) cells: These produce and release glucagon, a hormone that helps to regulate blood sugar levels by promoting the conversion of glycogen to glucose in the liver when blood sugar levels are low.
2. Beta (β) cells: These produce and release insulin, a hormone that promotes the uptake and utilization of glucose by cells throughout the body, thereby lowering blood sugar levels.
3. Delta (δ) cells: These produce and release somatostatin, a hormone that inhibits the release of both insulin and glucagon and helps regulate their secretion in response to changing blood sugar levels.
4. PP cells (gamma or γ cells): These produce and release pancreatic polypeptide, which plays a role in regulating digestive enzyme secretion and gastrointestinal motility.

Dysfunction of the Islets of Langerhans can lead to various endocrine disorders, such as diabetes mellitus, where insulin-producing beta cells are damaged or destroyed, leading to impaired blood sugar regulation.

Tolbutamide is defined as a first-generation sulfonylurea oral hypoglycemic agent used in the management of type 2 diabetes mellitus. It acts by stimulating the release of insulin from the pancreas, thereby reducing blood glucose levels. Tolbutamide is metabolized and excreted rapidly, with a half-life of about 6 hours, making it useful in patients with renal impairment.

Common side effects of tolbutamide include gastrointestinal symptoms such as nausea, vomiting, and diarrhea, as well as skin reactions such as rash and itching. Hypoglycemia is a potential adverse effect, particularly if the medication is dosed improperly or if the patient skips meals. Tolbutamide should be used with caution in patients with hepatic impairment, kidney disease, and the elderly due to an increased risk of hypoglycemia.

It's important to note that tolbutamide is not commonly used as a first-line treatment for type 2 diabetes mellitus due to the availability of newer medications with more favorable side effect profiles and efficacy.

Insulin-secreting cells, also known as beta cells, are a type of cell found in the pancreas. They are responsible for producing and releasing insulin, a hormone that regulates blood glucose levels by allowing cells in the body to take in glucose from the bloodstream. Insulin-secreting cells are clustered together in the pancreatic islets, along with other types of cells that produce other hormones such as glucagon and somatostatin. In people with diabetes, these cells may not function properly, leading to an impaired ability to regulate blood sugar levels.

Hyperandrogenism is a medical condition characterized by excessive levels of androgens (male sex hormones) in the body. This can lead to various symptoms such as hirsutism (excessive hair growth), acne, irregular menstrual periods, and infertility in women. It can be caused by conditions like polycystic ovary syndrome (PCOS), congenital adrenal hyperplasia, and tumors in the ovaries or adrenal glands. Proper diagnosis and management of hyperandrogenism is important to prevent complications and improve quality of life.

Blood glucose, also known as blood sugar, is the concentration of glucose in the blood. Glucose is a simple sugar that serves as the main source of energy for the body's cells. It is carried to each cell through the bloodstream and is absorbed into the cells with the help of insulin, a hormone produced by the pancreas.

The normal range for blood glucose levels in humans is typically between 70 and 130 milligrams per deciliter (mg/dL) when fasting, and less than 180 mg/dL after meals. Levels that are consistently higher than this may indicate diabetes or other metabolic disorders.

Blood glucose levels can be measured through a variety of methods, including fingerstick blood tests, continuous glucose monitoring systems, and laboratory tests. Regular monitoring of blood glucose levels is important for people with diabetes to help manage their condition and prevent complications.

A missense mutation is a type of point mutation in which a single nucleotide change results in the substitution of a different amino acid in the protein that is encoded by the affected gene. This occurs when the altered codon (a sequence of three nucleotides that corresponds to a specific amino acid) specifies a different amino acid than the original one. The function and/or stability of the resulting protein may be affected, depending on the type and location of the missense mutation. Missense mutations can have various effects, ranging from benign to severe, depending on the importance of the changed amino acid for the protein's structure or function.

Chlorthalidone is a diuretic medication, which is a type of drug that helps the body get rid of excess salt and water by increasing urine production. It is a type of sulfonamide, and it works by blocking the reabsorption of sodium and chloride in the distal convoluted tubules of the kidneys, which leads to increased excretion of these ions and water in the urine.

Chlorthalidone is used to treat hypertension (high blood pressure) and edema (fluid retention) associated with various medical conditions, such as heart failure, cirrhosis, and kidney disease. It may be used alone or in combination with other medications to achieve better blood pressure control.

Like all medications, chlorthalidone can cause side effects, including electrolyte imbalances, dehydration, dizziness, headache, muscle cramps, and gastrointestinal disturbances. It is important to take this medication as directed by a healthcare provider and to report any bothersome or persistent symptoms promptly.

Octreotide is a synthetic analogue of the natural hormone somatostatin, which is used in medical treatment. It is a octapeptide with similar effects to somatostatin, but with a longer duration of action. Octreotide is primarily used in the management of acromegaly, gastroenteropancreatic neuroendocrine tumors (GEP-NETs), and diarrhea and flushing associated with carcinoid syndrome.

It works by inhibiting the release of several hormones, including growth hormone, insulin, glucagon, and gastrin. This results in a decrease in symptoms caused by excessive hormone secretion, such as reduced growth hormone levels in acromegaly, decreased tumor size in some GEP-NETs, and improved diarrhea and flushing in carcinoid syndrome.

Octreotide is available in several forms, including short-acting subcutaneous injections (Sandostatin®), long-acting depot intramuscular injections (Sandostatin LAR®), and a slow-release formulation for the treatment of diarrhea associated with AIDS (Mycapssa™).

The medical definition of Octreotide is:

A synthetic octapeptide analogue of somatostatin, used in the management of acromegaly, gastroenteropancreatic neuroendocrine tumors (GEP-NETs), and diarrhea and flushing associated with carcinoid syndrome. Octreotide inhibits the release of several hormones, including growth hormone, insulin, glucagon, and gastrin, leading to symptomatic improvement in these conditions. It is available as short-acting subcutaneous injections, long-acting depot intramuscular injections, and a slow-release formulation for diarrhea associated with AIDS.

Proinsulin is the precursor protein to insulin, produced in the beta cells of the pancreas. It has a molecular weight of around 9,000 daltons and is composed of three distinct regions: the A-chain, the B-chain, and the C-peptide. The A-chain and B-chain are linked together by disulfide bonds and will eventually become the insulin molecule after a series of enzymatic cleavages. The C-peptide is removed during this process and is released into the bloodstream in equimolar amounts to insulin. Proinsulin levels can be measured in the blood and are sometimes used as a marker for beta cell function in certain clinical settings, such as diagnosing or monitoring insulinoma (a tumor of the pancreas that produces insulin) or assessing the risk of diabetes-related complications.

The pancreas is a glandular organ located in the abdomen, posterior to the stomach. It has both exocrine and endocrine functions. The exocrine portion of the pancreas consists of acinar cells that produce and secrete digestive enzymes into the duodenum via the pancreatic duct. These enzymes help in the breakdown of proteins, carbohydrates, and fats in food.

The endocrine portion of the pancreas consists of clusters of cells called islets of Langerhans, which include alpha, beta, delta, and F cells. These cells produce and secrete hormones directly into the bloodstream, including insulin, glucagon, somatostatin, and pancreatic polypeptide. Insulin and glucagon are critical regulators of blood sugar levels, with insulin promoting glucose uptake and storage in tissues and glucagon stimulating glycogenolysis and gluconeogenesis to raise blood glucose when it is low.

A hamartoma is a benign tumor-like growth that is composed of an unusual mixture of cells and tissues that are normally found in the affected area. These growths can occur anywhere in the body, but they are most commonly found in the skin, lungs, and brain. Hamartomas are typically slow growing and do not spread to other parts of the body (metastasize). They are usually harmless, but in some cases, they may cause symptoms or complications depending on their size and location. In general, hamartomas do not require treatment unless they are causing problems.

C-peptide is a byproduct that is produced when the hormone insulin is generated in the body. Insulin is a hormone that helps regulate blood sugar levels, and it is produced in the pancreas by specialized cells called beta cells. When these cells produce insulin, they also generate C-peptide as a part of the same process.

C-peptide is often used as a marker to measure the body's insulin production. By measuring C-peptide levels in the blood, healthcare providers can get an idea of how much insulin the body is producing on its own. This can be helpful in diagnosing and monitoring conditions such as diabetes, which is characterized by impaired insulin production or function.

It's worth noting that C-peptide is not typically used as a treatment for any medical conditions. Instead, it is primarily used as a diagnostic tool to help healthcare providers better understand their patients' health status and make informed treatment decisions.

A heterozygote is an individual who has inherited two different alleles (versions) of a particular gene, one from each parent. This means that the individual's genotype for that gene contains both a dominant and a recessive allele. The dominant allele will be expressed phenotypically (outwardly visible), while the recessive allele may or may not have any effect on the individual's observable traits, depending on the specific gene and its function. Heterozygotes are often represented as 'Aa', where 'A' is the dominant allele and 'a' is the recessive allele.

Mosaicism, in the context of genetics and medicine, refers to the presence of two or more cell lines with different genetic compositions in an individual who has developed from a single fertilized egg. This means that some cells have one genetic makeup, while others have a different genetic makeup. This condition can occur due to various reasons such as errors during cell division after fertilization.

Mosaicism can involve chromosomes (where whole or parts of chromosomes are present in some cells but not in others) or it can involve single genes (where a particular gene is present in one form in some cells and a different form in others). The symptoms and severity of mosaicism can vary widely, depending on the type and location of the genetic difference and the proportion of cells that are affected. Some individuals with mosaicism may not experience any noticeable effects, while others may have significant health problems.

Pancreatic neoplasms refer to abnormal growths in the pancreas that can be benign or malignant. The pancreas is a gland located behind the stomach that produces hormones and digestive enzymes. Pancreatic neoplasms can interfere with the normal functioning of the pancreas, leading to various health complications.

Benign pancreatic neoplasms are non-cancerous growths that do not spread to other parts of the body. They are usually removed through surgery to prevent any potential complications, such as blocking the bile duct or causing pain.

Malignant pancreatic neoplasms, also known as pancreatic cancer, are cancerous growths that can invade and destroy surrounding tissues and organs. They can also spread (metastasize) to other parts of the body, such as the liver, lungs, or bones. Pancreatic cancer is often aggressive and difficult to treat, with a poor prognosis.

There are several types of pancreatic neoplasms, including adenocarcinomas, neuroendocrine tumors, solid pseudopapillary neoplasms, and cystic neoplasms. The specific type of neoplasm is determined through various diagnostic tests, such as imaging studies, biopsies, and blood tests. Treatment options depend on the type, stage, and location of the neoplasm, as well as the patient's overall health and preferences.

Hypopituitarism is a medical condition characterized by deficient secretion of one or more hormones produced by the pituitary gland, a small endocrine gland located at the base of the brain. The pituitary gland controls several other endocrine glands in the body, including the thyroid, adrenals, and sex glands (ovaries and testes).

Hypopituitarism can result from damage to the pituitary gland due to various causes such as tumors, surgery, radiation therapy, trauma, or inflammation. In some cases, hypopituitarism may also be caused by a dysfunction of the hypothalamus, a region in the brain that regulates the pituitary gland's function.

The symptoms and signs of hypopituitarism depend on which hormones are deficient and can include fatigue, weakness, decreased appetite, weight loss, low blood pressure, decreased sex drive, infertility, irregular menstrual periods, intolerance to cold, constipation, thinning hair, dry skin, and depression.

Treatment of hypopituitarism typically involves hormone replacement therapy to restore the deficient hormones' normal levels. The type and dosage of hormones used will depend on which hormones are deficient and may require regular monitoring and adjustments over time.

I must clarify that the term "pedigree" is not typically used in medical definitions. Instead, it is often employed in genetics and breeding, where it refers to the recorded ancestry of an individual or a family, tracing the inheritance of specific traits or diseases. In human genetics, a pedigree can help illustrate the pattern of genetic inheritance in families over multiple generations. However, it is not a medical term with a specific clinical definition.

Ammonia is a colorless, pungent-smelling gas with the chemical formula NH3. It is a compound of nitrogen and hydrogen and is a basic compound, meaning it has a pH greater than 7. Ammonia is naturally found in the environment and is produced by the breakdown of organic matter, such as animal waste and decomposing plants. In the medical field, ammonia is most commonly discussed in relation to its role in human metabolism and its potential toxicity.

In the body, ammonia is produced as a byproduct of protein metabolism and is typically converted to urea in the liver and excreted in the urine. However, if the liver is not functioning properly or if there is an excess of protein in the diet, ammonia can accumulate in the blood and cause a condition called hyperammonemia. Hyperammonemia can lead to serious neurological symptoms, such as confusion, seizures, and coma, and is treated by lowering the level of ammonia in the blood through medications, dietary changes, and dialysis.

A seizure is an uncontrolled, abnormal firing of neurons (brain cells) that can cause various symptoms such as convulsions, loss of consciousness, altered awareness, or changes in behavior. Seizures can be caused by a variety of factors including epilepsy, brain injury, infection, toxic substances, or genetic disorders. They can also occur without any identifiable cause, known as idiopathic seizures. Seizures are a medical emergency and require immediate attention.

Dominant genes refer to the alleles (versions of a gene) that are fully expressed in an individual's phenotype, even if only one copy of the gene is present. In dominant inheritance patterns, an individual needs only to receive one dominant allele from either parent to express the associated trait. This is in contrast to recessive genes, where both copies of the gene must be the recessive allele for the trait to be expressed. Dominant genes are represented by uppercase letters (e.g., 'A') and recessive genes by lowercase letters (e.g., 'a'). If an individual inherits one dominant allele (A) from either parent, they will express the dominant trait (A).

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.

Adenosine Triphosphate (ATP) is a high-energy molecule that stores and transports energy within cells. It is the main source of energy for most cellular processes, including muscle contraction, nerve impulse transmission, and protein synthesis. ATP is composed of a base (adenine), a sugar (ribose), and three phosphate groups. The bonds between these phosphate groups contain a significant amount of energy, which can be released when the bond between the second and third phosphate group is broken, resulting in the formation of adenosine diphosphate (ADP) and inorganic phosphate. This process is known as hydrolysis and can be catalyzed by various enzymes to drive a wide range of cellular functions. ATP can also be regenerated from ADP through various metabolic pathways, such as oxidative phosphorylation or substrate-level phosphorylation, allowing for the continuous supply of energy to cells.

Glucose is a simple monosaccharide (or single sugar) that serves as the primary source of energy for living organisms. It's a fundamental molecule in biology, often referred to as "dextrose" or "grape sugar." Glucose has the molecular formula C6H12O6 and is vital to the functioning of cells, especially those in the brain and nervous system.

In the body, glucose is derived from the digestion of carbohydrates in food, and it's transported around the body via the bloodstream to cells where it can be used for energy. Cells convert glucose into a usable form through a process called cellular respiration, which involves a series of metabolic reactions that generate adenosine triphosphate (ATP)—the main currency of energy in cells.

Glucose is also stored in the liver and muscles as glycogen, a polysaccharide (multiple sugar) that can be broken down back into glucose when needed for energy between meals or during physical activity. Maintaining appropriate blood glucose levels is crucial for overall health, and imbalances can lead to conditions such as diabetes mellitus.

A homozygote is an individual who has inherited the same allele (version of a gene) from both parents and therefore possesses two identical copies of that allele at a specific genetic locus. This can result in either having two dominant alleles (homozygous dominant) or two recessive alleles (homozygous recessive). In contrast, a heterozygote has inherited different alleles from each parent for a particular gene.

The term "homozygote" is used in genetics to describe the genetic makeup of an individual at a specific locus on their chromosomes. Homozygosity can play a significant role in determining an individual's phenotype (observable traits), as having two identical alleles can strengthen the expression of certain characteristics compared to having just one dominant and one recessive allele.

A Glucose Tolerance Test (GTT) is a medical test used to diagnose prediabetes, type 2 diabetes, and gestational diabetes. It measures how well your body is able to process glucose, which is a type of sugar.

During the test, you will be asked to fast (not eat or drink anything except water) for at least eight hours before the test. Then, a healthcare professional will take a blood sample to measure your fasting blood sugar level. After that, you will be given a sugary drink containing a specific amount of glucose. Your blood sugar levels will be measured again after two hours and sometimes also after one hour.

The results of the test will indicate how well your body is able to process the glucose and whether you have normal, impaired, or diabetic glucose tolerance. If your blood sugar levels are higher than normal but not high enough to be diagnosed with diabetes, you may have prediabetes, which means that you are at increased risk of developing type 2 diabetes in the future.

It is important to note that a Glucose Tolerance Test should be performed under the supervision of a healthcare professional, as high blood sugar levels can be dangerous if not properly managed.

A point mutation is a type of genetic mutation where a single nucleotide base (A, T, C, or G) in DNA is altered, deleted, or substituted with another nucleotide. Point mutations can have various effects on the organism, depending on the location of the mutation and whether it affects the function of any genes. Some point mutations may not have any noticeable effect, while others might lead to changes in the amino acids that make up proteins, potentially causing diseases or altering traits. Point mutations can occur spontaneously due to errors during DNA replication or be inherited from parents.

... refers to an above normal level of insulin in the blood of a person or animal. Normal insulin secretion and ... Hyperinsulinism can be associated with several types of medical problems, which can be roughly divided into two broad and ... Hyperinsulinism due to reduced insulin sensitivity is usually asymptomatic. In contrast, hyperinsulinemic hypoglycemia can ...
Hyperinsulinism or congenital hyperinsulinism can be a genetic or acquired condition. Acquired HI may be secondary to factors ... "Children's Hyperinsulinism Charity". www.hyperinsulinism.co.uk. Retrieved 2023-08-22. "HI Global Registry". www. ... Hyperinsulinism Genes. Information on the genetic mechanisms of CHI. Congenital Hyperinsulinism International. Nonprofit ... "Congenital Hyperinsulinism Center". The Children's Hospital of Philadelphia. 2014-05-05. Retrieved 2023-08-22. "Hyperinsulinism ...
... (HI/HA) is an autosomal dominant disorder that results in the excess production of ... "Clinical characteristics and biochemical mechanisms of congenital hyperinsulinism associated with dominant KATP channel ...
Ferry, R.J. (2010). Hyperinsulinism "Medscape". "Hyperinsulinemia means that the amount of insulin in the blood is higher than ... It can also occur in congenital hyperinsulinism, including nesidioblastosis. Hyperinsulinemia is associated with hypertension, ... Demirbilek, Hüseyin; Hussain, Khalid (2017-12-30). "Congenital Hyperinsulinism: Diagnosis and Treatment Update". Journal of ... "Congenital hyperinsulinism: current trends in diagnosis and therapy". Orphanet Journal of Rare Diseases. 6: 63. doi:10.1186/ ...
This creates hypoglycemia of varying patterns, including transient or persistent congenital hyperinsulinism, or fasting or ... Glaser B (2013-01-24). "Familial Hyperinsulinism". GeneReviews. Seattle WA: University of Washington, Seattle. PMID 20301549. ...
Conclusions 1. Hyperinsulinism is a condition, perhaps a disease entity, with definite symptoms; i.e. those described as being ... His research on hyperinsulinism and its control brought international recognition to Harris, including the Distinguished ... 1924: "Hyperinsulinism and Dysinsulinism", Journal of the American Medical Association 83(10): 729-33. 1928: The Sugar Fed ... Harris, Seale (1924). "Hyperinsulinism and disinsulinism". Journal of the American Medical Association. 83 (10): 729-33. doi: ...
SLC16A1 Hyperinsulinism-hyperammonemia syndrome; 606762; GLUD1 Hyperkalemic periodic paralysis, type 2; 613345; SCN4A ...
Hyperinsulinism Perry, Julian C.; Bourne, Blanche; Lester Henry, W. (January 1957). "Idiopathic Hypoglycemia in Childhood: ...
Fournet JC, Junien C (2004). "Genetics of congenital hyperinsulinism". Endocrine Pathology. 15 (3): 233-40. doi:10.1385/EP:15:3 ... SUR2A and SUR2B The SUR1 protein is coded by the ABCC8 gene and is associated with congenital hyperinsulinism and ...
The same signs may be caused by hyperinsulinism not caused by insulinoma. The criteria date back to the 1930s, when a few ... Hyperinsulinism Hypoglycemia Causes of hypoglycemia Melmed, Shlomo (2016). Williams textbook of endocrinology (13 ed.). ... Whipple, Allen O (1938). "The surgical therapy of hyperinsulinism". International Journal of Surgery. 3: 237-276 - via Scopus ...
June 1994). "Familial hyperinsulinism maps to chromosome 11p14-15.1, 30 cM centromeric to the insulin gene". Nature Genetics. 7 ... Meissner T, Beinbrech B, Mayatepek E (1999). "Congenital hyperinsulinism: molecular basis of a heterogeneous disease". Human ... November 1996). "Mutations in the sulonylurea receptor gene are associated with familial hyperinsulinism in Ashkenazi Jews". ... July 1998). "Genetic heterogeneity in familial hyperinsulinism". Human Molecular Genetics. 7 (7): 1119-1128. doi:10.1093/hmg/ ...
Rosenfeld E, Ganguly A, De Leon DD (December 2019). "Congenital hyperinsulinism disorders: Genetic and clinical characteristics ... Changes in hexokinase 1 have also been identified to cause both mild and severe forms of congenital hyperinsulinism. Due to the ... "Congenital Hyperinsulinism: Current Laboratory-Based Approaches to the Genetic Diagnosis of a Heterogeneous Disease". Frontiers ...
McMahon AW, Wharton GT, Thornton P, De Leon DD (January 2017). "Octreotide use and safety in infants with hyperinsulinism". ... Octreotide is also used in the treatment of refractory hypoglycemia or congenital hyperinsulinism in neonates and sulphonylurea ...
1998). "Hyperinsulinism and hyperammonemia in infants with regulatory mutations of the glutamate dehydrogenase gene". N. Engl. ... 2001). "Hyperinsulinism/hyperammonemia syndrome in children with regulatory mutations in the inhibitory guanosine triphosphate- ... 2000). "Novel missense mutations in the glutamate dehydrogenase gene in the congenital hyperinsulinism-hyperammonemia syndrome ... "Congenital hyperinsulinism: molecular basis of a heterogeneous disease". Hum. Mutat. 13 (5): 351-61. doi:10.1002/(SICI)1098- ...
Hyperammonemia/hyperinsulinism (HA/HI) is associated with mild-to-moderate hyperammonemia and with relatively mild, late-onset ... Familial hyperinsulinism, linked to mutations in GLUD1, is characterized by hypoglycemia that ranges from severe neonatal-onset ... GeneReviews/NCBI/NIH/UW entry on Familial Hyperinsulinism Glutamate+dehydrogenase at the U.S. National Library of Medicine ... Individuals with autosomal recessive familial hyperinsulinism, caused by mutations in either ABCC8 or KCNJ11 (FHI-KATP), tend ...
... due to causes other than hyperinsulinism; in various inborn errors of metabolism, and intentionally induced via a ketogenic ...
It causes hyperinsulinemic hypoglycemia, where hyperinsulinism is exercise-induced. Monocarboxylate transporter 1 deficiency ( ... in mouse pancreatic β-cells leads to relative hyperinsulinism during exercise". Diabetes. 61 (7): 1719-25. doi:10.2337/db11- ... Overexpression of MCT1 in pancreatic beta cells leads to hyperinsulinism during exercise. Hyperinsulinemic hypoglycemia, ...
September 2000). "A recessive contiguous gene deletion causing infantile hyperinsulinism, enteropathy and deafness identifies ... "Hyperinsulinism in short-chain L-3-hydroxyacyl-CoA dehydrogenase deficiency reveals the importance of β-oxidation in insulin ... "Hyperinsulinism in Infancy: From Basic Science to Clinical Disease". Physiological Reviews. 84 (1): 239-275. doi:10.1152/ ... "A recessive contiguous gene deletion causing infantile hyperinsulinism, enteropathy and deafness identifies the Usher type 1C ...
GDM results in maternal hyperglycemia, fetal hyperinsulinism, and fetal overnutrition. DNA methylation can act as the mechanism ...
Hyperinsulinism Too high a level of insulin in the blood. This often involves a condition in which the body produces too much ...
The gene encoding the channel is called KCNJ11 and mutations in this gene are associated with congenital hyperinsulinism. It is ... GeneReviews/NCBI/NIH/UW entry on Familial Hyperinsulinism GeneReviews/NCBI/NIH/UW entry on Permanent Neonatal Diabetes Mellitus ... Mutations in this gene are a cause of congenital hyperinsulinism (CHI), an autosomal recessive disorder characterized by ... Meissner T, Beinbrech B, Mayatepek E (1999). "Congenital hyperinsulinism: molecular basis of a heterogeneous disease". Human ...
Congenital hyperinsulinism Hyperinsulinemic hypoglycemia Wong's nursing care of infants and children. Hockenberry, Marilyn J ...
Ricquier has demonstrated that mutations in the UCP2 protein induce congenital hyperinsulinism in children at birth. He also ... "Mutations in UCP2 in congenital hyperinsulinism reveal a role for regulation of insulin secretion". PLOS ONE. 3 (12): e3850. ...
"Clinical features and insulin regulation in infants with a syndrome of prolonged neonatal hyperinsulinism". The Journal of ...
Due to Diabetogenic Hyperinsulinism: Observations on 131 Patients". Journal of the American Geriatrics Society. 13 (7): 602-38 ...
Hyperinsulinism due to diffuse overactivity of beta cells, such as in many of the forms of congenital hyperinsulinism, and more ... Hypoglycemia due to endogenous insulin Congenital hyperinsulinism Transient neonatal hyperinsulinism (mechanism not known) ... When congenital hyperinsulinism is due to focal defects of the insulin-secretion mechanism, surgical removal of that part of ... Detectable amounts of insulin are abnormal and indicate that hyperinsulinism is likely to be the cause. Other aspects of the ...
The first case of hyperinsulinism due to a tumor of this type was reported in 1927. Recognition of a non-insulin-secreting type ...
In the 1920s, after the discovery of insulin and its use in the treatment of diabetics, hyperinsulinism was suspected to be a ... A pioneering description of hyperinsulinism as a cause of hypoglycemia was published by Seale Harris in 1924. The first report ...
Most congenital hyperinsulinism is now known to be caused by different mechanisms than excessive proliferation of beta cells in ... Clancy T, Moore F, Zinner M (2006). "Post-gastric bypass hyperinsulinism with nesidioblastosis: subtotal or total ... Congenital hyperinsulinism Neonatal hypoglycemia Raffel A, Krausch MM, Anlauf M, Wieben D, Braunstein S, Klöppel G, Röher H, ... the term has been revived to describe a form of acquired hyperinsulinism with beta cell hyperplasia found in adults, especially ...
September 2000). "A recessive contiguous gene deletion causing infantile hyperinsulinism, enteropathy and deafness identifies ... September 2000). "A recessive contiguous gene deletion causing infantile hyperinsulinism, enteropathy and deafness identifies ...
Hyperinsulinism refers to an above normal level of insulin in the blood of a person or animal. Normal insulin secretion and ... Hyperinsulinism can be associated with several types of medical problems, which can be roughly divided into two broad and ... Hyperinsulinism due to reduced insulin sensitivity is usually asymptomatic. In contrast, hyperinsulinemic hypoglycemia can ...
Primary hyperinsulinism is a rare but important cause of hypoglycemia in infants and children. It is the most common cause of ... encoded search term (Hyperinsulinism) and Hyperinsulinism What to Read Next on Medscape ... Hyperinsulinism-hyperammonemia (HH) syndrome due to GLUD1 mutation, as follows:. * GLUD1 mutations reported to date have been ... Hyperinsulinism (HI) is the most common cause of severe, persistent hypoglycemia in infants and children. [1, 2] Infants with ...
"A Very High Unmet Need" for the Most Severe Hyperinsulinism Types The most common and severe form of congenital hyperinsulinism ... One drug, diazoxide, is a KATP channel agonist that suppresses insulin release in some types of hyperinsulinism but not in the ... A rare disorder, occurring in 1 in 20,000 to 1 in 50,000 US live births, congenital hyperinsulinism is caused by several ... De León holds patents in the United States and Europe for the use of exendin-(9-39) for treating hyperinsulinism and ...
What causes Hyperinsulinism?. There is an organ in the tummy called a Pancreas. Its job is to help digest food and control ... Hyperinsulinism is where the body produces too much insulin, causing blood sugar levels to drop too low. When this happens it ... Hyperinsulinism can mean that their brother or sister has time away from school for appointments or has to spend time in ... Children with Hyperinsulinism need to check their blood sugars by using a handheld blood sugar monitor. They will put a very ...
In congenital hyperinsulinism, the insulin cells of the pancreas (beta cells) secrete too much insulin and at the wrong time. ... What is congenital hyperinsulinism (HI)?. Congenital hyperinsulinism (HI) is a genetic disorder in which the insulin cells of ... Causes of congenital hyperinsulinism (HI). Congenital hyperinsulinism is caused by genetic mutations that result in ... Symptoms of congenital hyperinsulinism (HI). Congenital hyperinsulinism causes low plasma sugar (hypoglycemia).The symptoms of ...
Watch the video to learn more about the expert care provided by the Congenital Hyperinsulinism Center at The Childrens ... to what is hyperinsulinism? What can be causing the hyperinsulinism? And how can we treat the hyperinsulinism? ... What is Hyperinsulinism (HI)?. Diva D. De León-Crutchlow, MD: Hyperinsulinism is a congenital disorder of regulation of insulin ... After Hyperinsulinism Surgery. Andrew Palladino, MD: As it stands now, about 50 percent of our babies that have the near total ...
Join Cook Childrens team of experts on blazing new trails in treating and curing hyperinsulinism (HI) for children across the ... 2021 Congenital Hyperinsulinism Virtual Family Conference. Congenital Hyperinsulinism Collaborative Research Network (CRN) ... But also the more common acquired forms of hyperinsulinism like perinatal stress hyperinsulinism, which is far more common. And ... Host: So weve talked about focal hyperinsulinism, which is curable. There are other types of hyperinsulinism we cant ...
Congenital hyperinsulinism is a condition of dysregulated insulin secretion often caused by inactivating mutations of the ATP- ... In contrast to a previous report of increased diabetes risk in dominant KATP hyperinsulinism, only 4 of 29 adults had diabetes ... These data highlight distinctive features of dominant KATP hyperinsulinism relative to the more common and more severe ... Clinical characteristics and biochemical mechanisms of congenital hyperinsulinism associated with dominant KATP channel ...
"Hyperinsulinism" is a descriptor in the National Library of Medicines controlled vocabulary thesaurus, MeSH (Medical Subject ... Etiology of hyperinsulinism varies, including hypersecretion of a beta cell tumor (INSULINOMA); autoantibodies against insulin ... This graph shows the total number of publications written about "Hyperinsulinism" by people in this website by year, and ... Below are the most recent publications written about "Hyperinsulinism" by people in Profiles. ...
Although her overgrowth may be a result of hyperinsulinism, her serum contains a factor (neither insulin nor IGF-1) which is ... Our patient has postnatal overgrowth associated with severe hyperinsulinism progressing to type 2 diabetes mellitus. She also ...
Clinical practice guidelines for congenital hyperinsulinism.. Yorifuji T, Horikawa R, Hasegawa T, Adachi M, Soneda S, Minagawa ...
Congenital hyperinsulinism (CHI), previously termed persistent hyperinsulinemic hyperglycemia of infancy (PHHI), is a spectrum ... Congenital hyperinsulinism (CHI), previously termed persistent hyperinsulinemic hyperglycemia of infancy (PHHI), is a spectrum ...
... due to inappropriate secretion, associated with hypoglycemia. Hyperinsulinism may also refer to forms of ... The treatment of this form of hyperinsulinism depends on the cause and the severity of the hyperinsulinism, and may include ... Hyperinsulinism refers to an above normal level of insulin in the blood of a person or animal. Normal insulin secretion and ... That spontaneous hyperinsulinism might be a cause of symptomatic hypoglycemia was first proposed by Seale Harris, MD, 1924, in ...
Familiarity with congenital hyperinsulinism or the rare disease patient experience.. *A strong interest in rare diseases, ... Nonprofit Overview: Congenital Hyperinsulinism International (CHI) is a health non-profit dedicated to improving the lives of ... The Development Manager will work to support the mission, vision and priority goals of Congenital Hyperinsulinism International ... Nonprofit Overview: Congenital Hyperinsulinism International (CHI) is the leading organization dedicated to improving the lives ...
What is ABCC8-related Hyperinsulinism?. ABCC8-related hyperinsulinism, also called congenital hyperinsulinism, is an inherited ... How common is ABCC8-related Hyperinsulinism?. ABCC8-related hyperinsulinism affects roughly 1 in 50,000 Europeans. It is ... Other names for ABCC8-related Hyperinsulinism. *Persistent Hyperinsulinemic Hypoglycemia of Infancy. *Familial Hyperinsulinism ... How is ABCC8-related Hyperinsulinism treated?. Treatments for ABCC8-related hyperinsulinism include dietary modification, ...
For further information on the HI Global Registry please view the registry study communication material, and feel free to contact the CHI team with any questions or concerns you may have. Thank you for your interest and support with this incredibly important research initiative.. ...
Primary hyperinsulinism is a rare but important cause of hypoglycemia in infants and children. It is the most common cause of ... encoded search term (Hyperinsulinism) and Hyperinsulinism What to Read Next on Medscape ... Hyperinsulinism Treatment & Management. Updated: Dec 16, 2015 * Author: Sunil Sinha, MD; Chief Editor: Stephen Kemp, MD, PhD ... Hyperinsulinism in infancy and childhood: when an insulin level is not always enough. Clin Chem. 2008. 54:256-63. [QxMD MEDLINE ...
Share info and advice with people concerned by Isolated congenital hyperinsulinism ✓ The leading social network for patients, ...
Studies related to the Congenital Hyperinsulinism Research Laboratory. ... 18F-DOPA PET/CT scan for Hyperinsulinism. The purpose of this research is to study 18F-L-Fluoro-DOPA (F-DOPA), a tracer, which ... AAV Vectors, Hyperinsulinism, Adeno-associated Viral Vectors, Viral Vectors, Hypoglycemia, Clinical Trials, Rare Disorder ... HM15136 for the Treatment of Congenital Hyperinsulinism Research Study. The purpose of this study is to look at the safety and ...
Fish Oil Helps Treat Congenital Hyperinsulinism. Purified fish oils could help treat congenital hyperinsulinism - a rare and ...
Hepatitis in an infant treated with octreotide for congenital hyperinsulinism. Ilana Koren*, Arieh Riskin, Winfried Barthlen, ... Dive into the research topics of Hepatitis in an infant treated with octreotide for congenital hyperinsulinism. Together they ...
Congenital hyperinsulinism. *Congenital hypoaldosteronism, see Corticosterone methyloxidase deficiency. *Congenital hypoplasia ...
Familial Hyperinsulinism (ABCC8-Related) and our test ABCC8-related familial hyperinsulinism is a rare genetic disorder. It is ... Familial Hyperinsulinism (ABCC8-Related) Familial Mediterranean Fever Fanconi Anemia Group C GRACILE Syndrome Gaucher Disease ... Symptoms of familial hyperinsulinism may vary between people with the condition even if they have the same genetic variants. ... However, the American College of Obstetricians and Gynecologists (ACOG) notes that testing for familial hyperinsulinism may be ...
About Congenital Hyperinsulinism. Congenital hyperinsulinism is a rare, genetic, pediatric endocrine disorder that leads to the ... "Congenital hyperinsulinism is the most common cause of persistent low blood sugars in infants and children and often leads to ... Rezolute Initiates Phase 2b Clinical Trial of Lead Candidate RZ358 in Congenital Hyperinsulinism. February 10, 2020 8:30am EST ... Therefore, RZ358 is ideally suited as a potential therapy for hyperinsulinism, and it is being developed to treat the ...
Evidence for adaptive hyperinsulinism. Lancet i, 690-692.Google Scholar. Randle, PJ, Garland, PB, Hales, CN & Newsholme, EA ( ...
Hyperinsulinism was hypothesized to link PCOS to PP, triggering androgen hypersecretion by the ovaries and adrenal glands [24, ... Hyperinsulinism may be the consequence of IR and may derive from the production of metabolically active molecules by visceral ... Ibáñez, L.; Potau, N.; Francois, I.; de Zegher, F. Precocious Pubarche, Hyperinsulinism, and Ovarian Hyperandrogenism in Girls ... IR and the resulting compensatory hyperinsulinism are linked to hyperandrogenemia in various ways. Firstly, insulin promotes ...
Among 13 children with congenital hyperinsulinism, elevated incretin hormone concentrations were detected in 2 with atypical, ... Congenital hyperinsulinism causes profound hypoglycemia, which may persist or resolve spontaneously. ... Congenital hyperinsulinism causes profound hypoglycemia, which may persist or resolve spontaneously. Among 13 children with ... Increased plasma incretin concentrations identifies a subset of patients with persistent congenital hyperinsulinism without ...
About congenital hyperinsulinism. Congenital hyperinsulinism is a severe, ultra-rare genetic disease, primarily affecting ... Zealand Pharma announces designation of priority review by the US FDA for dasiglucagon in congenital hyperinsulinism ... 2011) Congenital hyperinsulinism: current trends in diagnosis and therapy, Orphanet J Rare Dis. 2011; 6:63.. 4) Yau et al. ( ... Zealand Pharma announces designation of priority review by the US FDA for dasiglucagon in congenital hyperinsulinism. Zealand ...
Congenital Hyperinsulinism and Cochlear Hypoplasia in a Rare Case of Pallister-Hall Syndrome ... Congenital Hyperinsulinism and Cochlear Hypoplasia in a Rare Case of Pallister-Hall Syndrome. ... level during hypoglycemia with low free fatty acids and beta-hydroxy butyrate suggestive of congenital hyperinsulinism (CHI). ...
  • Even today with all we know, that's the frequency," said De León, chief of the Division of Endocrinology and Diabetes and director of the Congenital Hyperinsulinism Center at Children's Hospital of Philadelphia (CHOP). (medscape.com)
  • Since most children's hospitals encounter only one or two cases a year, it is important to receive medical care from an experienced treatment center, such as the Congenital Hyperinsulinism Center at CHOP. (chop.edu)
  • Since October 1998 the Congenital Hyperinsulinism Center at Children's Hospital of Philadelphia has evaluated and treated more than 400 patients - making it the largest program of its kind in North America. (chop.edu)
  • The Congenital Hyperinsulinism Center at the Children's Hospital of Philadelphia is working on a research study to better understand how people with hyperinsulinism may have different blood sugar responses to certain tests (like fasting or drinking a high-protein shake) when compared to people without hyperinsulinism. (chop.edu)
  • At The Children's Hospital of Philadelphia Hyperinsulinism Center, we see approximately two-thirds of all the cases that are born in this country with hyperinsulinism. (chop.edu)
  • A center within the Children's Hospital of Philadelphia that specializes in the treatment of hyperinsulinism in children. (jscreen.org)
  • The remainder of patients, with diffuse hyperinsulinism, "are the most severe and most difficult group to treat. (medscape.com)
  • By doing genetic screening and finding out what gene is defective in these children we can very-with high accuracy-determine who has focal hyperinsulinism and who would have diffuse hyperinsulinism. (chop.edu)
  • [ 15 ] One recent study showed almost 94% of focal hyperinsulinism cases required no further treatment, versus 41% with diffuse hyperinsulinism that showed continued hypoglycemia postoperatively. (medscape.com)
  • A non-profit dedicated to improving the lives of children, adults, and families living with congenital hyperinsulinism. (jscreen.org)
  • TUCSON, Ariz., October 26, 2023 - Critical Path Institute (C-Path) and Congenital Hyperinsulinism International (CHI), a leading nonprofit dedicated to improving the lives of children and adults living with Congenital Hyperinsulinism (HI), today announced a data sharing agreement to incorporate rare disease patient-level data from CHI's HI Global Registry into C-Path's Rare Disease Cures Accelerator-Data. (c-path.org)
  • Hyperinsulinism can be associated with several types of medical problems, which can be roughly divided into two broad and largely non-overlapping categories: those tending toward reduced sensitivity to insulin and high blood glucose levels (hyperglycemia), and those tending toward excessive insulin secretion and low glucose levels (hypoglycemia). (wikipedia.org)
  • The most common and severe form of congenital hyperinsulinism is due to inactivating mutations of two genes that encode subunits of the beta-cell K ATP channel, leading to dysregulated insulin secretion. (medscape.com)
  • Congenital hyperinsulinism is caused by genetic mutations that result in inappropriate and excess insulin secretion from the beta cells of the pancreas. (chop.edu)
  • Hyperinsulinism is a congenital disorder of regulation of insulin secretion. (chop.edu)
  • The first line of therapy is Diazoxide, a medication that is used in an oral preparation is very effective in suppressing insulin secretion in milder forms of hyperinsulinism. (chop.edu)
  • Congenital hyperinsulinism is a condition of dysregulated insulin secretion often caused by inactivating mutations of the ATP-sensitive K+ (KATP) channel in the pancreatic β cell. (jci.org)
  • Hyperinsulinism may also refer to forms of hypoglycemia caused by excessive insulin secretion. (ipfs.io)
  • The treatment of this form of hyperinsulinism depends on the cause and the severity of the hyperinsulinism, and may include surgical removal of the source of insulin, or a drug such as diazoxide or octreotide that reduces insulin secretion. (ipfs.io)
  • Dysregulation of insulin secretion in children with congenital hyperinsulinism due to sulfonylurea receptor mutations. (medscape.com)
  • Congenital hyperinsulinism is a rare, genetic, pediatric endocrine disorder that leads to the inappropriate secretion of the hormone insulin by the pancreas. (rezolutebio.com)
  • One drug, diazoxide, is a K ATP channel agonist that suppresses insulin release in some types of hyperinsulinism but not in the majority of patients who have those two particular K ATP channel mutations. (medscape.com)
  • There are two types of hyperinsulinism. (chop.edu)
  • Children that have hyperinsulinism produce too much insulin and that causes severe low blood glucose. (chop.edu)
  • Congenital hyperinsulinism is a condition in which the beta cells of the pancreas make too much insulin, and more importantly, have lost the ability to regulate insulin based on the plasma glucose levels. (cookchildrens.org)
  • Insulin levels above 3 μU/mL are inappropriate when the glucose level is below 50 mg/dL (2.8 mM), and may indicate hyperinsulinism as the cause of the hypoglycemia. (ipfs.io)
  • If a child shows symptoms of ABCC8-related hyperinsulinism at birth, intravenous glucose is often given to raise and stabilize the blood sugar level. (jscreen.org)
  • Patients with hyperinsulinism may require very high glucose infusion rates (20-30 mg/kg/min) to maintain normoglycemia. (medscape.com)
  • Hyperinsulinism (HI) is the most common cause of severe, persistent hypoglycemia in infants and children. (medscape.com)
  • Congenital hyperinsulinism causes low plasma sugar (hypoglycemia).The symptoms of hypoglycemia in infants are often difficult to identify, as they can be similar to normal infant activities. (chop.edu)
  • Congenital hyperinsulinism (HI) is a disorder that causes low blood sugar (hypoglycemia) in infants and children. (chop.edu)
  • Some of those that occur in infants and young children are termed congenital hyperinsulinism . (ipfs.io)
  • Infants with ABCC8-related hyperinsulinism tend to have significantly low blood sugar within the first few days of life. (jscreen.org)
  • Infants with hyperinsulinism are at higher risk of neurologic sequelae than infants with hypoglycemia from other causes. (medscape.com)
  • Congenital hyperinsulinism is the most common cause of persistent low blood sugars in infants and children and often leads to serious neurologic complications," said Brian Roberts, M.D., head of clinical development at Rezolute. (rezolutebio.com)
  • Delayed-Onset Transient Hyperinsulinism in Infants with Very Low and Extremely Low Birth Weights: A Cohort Study. (bvsalud.org)
  • Congenital hyperinsulinism (HI) is the most common cause of persistent hypoglycaemia in infants. (biomedcentral.com)
  • This study is a randomized, double-blind, placebo-controlled study that will evaluate the safety and efficacy of dasiglucagon in children between the ages of 7 days and 12 months who have congenital hyperinsulinism. (chop.edu)
  • US FDA has granted priority review designation to dasiglucagon for the prevention and treatment of hypoglycemia in pediatric patients 7 days of age and older with congenital hyperinsulinism (CHI) for up to 3 weeks of dosing with a Prescription Drug User Fee Act (PDUFA) date on December 30, 2023. (biotech-365.com)
  • Press release - No. 8 / 2023 Zealand Pharma submits New Drug Application to US FDA for dasiglucagon in congenital hyperinsulinism Copenhagen, Denmark, June 30, 2023 - Zealand Pharma A/S. (globenewswire.com)
  • These data highlight distinctive features of dominant KATP hyperinsulinism relative to the more common and more severe recessive form, including retention of normal subunit trafficking, impaired channel activity, and a milder hypoglycemia phenotype that may escape detection in infancy and is often responsive to diazoxide medical therapy, without the need for surgical pancreatectomy. (jci.org)
  • 18F-fluoro-L-DOPA PET-CT imaging combined with genetic analysis for optimal classification and treatment in a child with severe congenital hyperinsulinism. (medscape.com)
  • Congenital Hyperinsulinism (CHI) is the most frequent cause of severe, persistent hypoglycemia in children. (eurospe.org)
  • The current study involved 16 children (6 girls, 10 boys), aged 10 months through 15 years with persistent hypoglycemia due to congenital hyperinsulinism. (medscape.com)
  • Evaluation, Medical Therapy, and Course of Adult Persistent Hyperinsulinemic Hypoglycemia After Roux-en-Y Gastric Bypass Surgery: A Case Series. (umassmed.edu)
  • Increased plasma incretin concentrations identifies a subset of patients with persistent congenital hyperinsulinism without KATP channel gene defects. (dspace-express.com)
  • Among 13 children with congenital hyperinsulinism, elevated incretin hormone concentrations were detected in 2 with atypical, persistent disease. (dspace-express.com)
  • In contrast to a previous report of increased diabetes risk in dominant KATP hyperinsulinism, only 4 of 29 adults had diabetes. (jci.org)
  • We are doing this study to see if the patterns of abnormalities in controlling insulin in children and adults with hyperinsulinism are related to any underlying genetic cause and to identify possible new genetic causes. (chop.edu)
  • Exendin-(9-39) has been granted an orphan drug designation by the European Medicines Agency for the treatment of congenital hyperinsulinism and by the US Food and Drug Administration for the treatment of hyperinsulinemic hypoglycemia (which includes congenital hyperinsulinism). (medscape.com)
  • Novel de novo mutation in sulfonylurea receptor 1 presenting as hyperinsulinism in infancy followed by overt diabetes in early adolescence. (medscape.com)
  • Familial hyperinsulinism caused by an activating glucokinase mutation. (medscape.com)
  • Conversely, hypoglycemia associated with ketonuria makes hyperinsulinism less likely. (medscape.com)
  • Therefore, RZ358 is ideally suited as a potential therapy for hyperinsulinism, and it is being developed to treat the hypoglycemia associated with diseases such as CHI. (rezolutebio.com)
  • REDWOOD CITY, Calif., Feb. 10, 2020 (GLOBE NEWSWIRE) -- Rezolute, Inc. ("Rezolute" or "the Company") (OTCQB:RZLT), today announced the initiation of the RIZE study (RZ358-606), a Phase 2b clinical trial of its lead candidate, RZ358, in patients with congenital hyperinsulinism (CHI). (rezolutebio.com)
  • A rare disorder, occurring in 1 in 20,000 to 1 in 50,000 US live births, congenital hyperinsulinism is caused by several different genetic mutations. (medscape.com)
  • Loss of K ATP channel function due to mutations in ABCC8 or KCNJ11, genes that encode the sulfonylurea receptor 1 or the inward rectifier Kir6.2 subunit of the channel, is a major cause of congenital hyperinsulinism. (elsevierpure.com)
  • We hypothesize that postnatal stress caused by prematurity and associated problems may contribute to development of delayed-onset transient hyperinsulinism . (bvsalud.org)
  • In both cases, the hyperinsulinism is transient. (merckmanuals.com)
  • In addition to being a risk factor for type 2 diabetes , hyperinsulinism due to insulin resistance may increase blood pressure and contribute to hypertension by direct action on vascular endothelial cells (the cells lining blood vessels). (ipfs.io)
  • However, in Hyperinsulinism the pancreas makes too much insulin, removing too much sugar from the blood causing low blood sugar (hypoglycaemia). (sibs.org.uk)
  • Congenital hyperinsulinism (HI) is a genetic disorder in which the insulin cells of the pancreas, called beta cells, secrete too much insulin. (chop.edu)
  • ABCC8-related hyperinsulinism, also called congenital hyperinsulinism, is an inherited condition in which the pancreas releases inappropriately large quantities of the hormone insulin, leading to low blood sugar (hypoglycemia). (jscreen.org)
  • In people with ABCC8-related hyperinsulinism, the pancreas secretes insulin even without sugar consumption, thereby removing too much sugar from the blood. (jscreen.org)
  • Our team is working on a study to better understand neurological problems, including seizures and developmental delays, that occur in children with hyperinsulinism, including HI/HA syndrome, and type 1 diabetes mellitus. (chop.edu)
  • Today we're talking about hyperinsulinism with pediatric endocrinologist Dr. Paul Thornton, pediatric surgeon Dr. John Huffman and neonatologist Dr. Jonathan Nedrelow. (cookchildrens.org)
  • A distinct clinical syndrome of hyperinsulinism and autosomal recessive polycystic kidney disease (HIPKD) arises in the context of a specific variant in the PMM2 promotor, either in homozygosity, or compound heterozygous with a deleterious PMM2 variant. (ox.ac.uk)
  • An investigational glucagon-like peptide-1 (GLP-1) antagonist, exendin-(9-39), prevents fasting and protein-induced hypoglycemia among children with congenital hyperinsulinism, new data suggest. (medscape.com)
  • Some children with Hyperinsulinism do not show any symptoms even when their sugars are dangerously low. (sibs.org.uk)
  • Children with Hyperinsulinism need to check their blood sugars by using a handheld blood sugar monitor. (sibs.org.uk)
  • Some children with Hyperinsulinism may need to take their medicine and feeds regularly on time to help them control their blood sugars. (sibs.org.uk)
  • We have different treatment options for children with hyperinsulinism. (chop.edu)
  • Join Cook Children's team of experts on blazing new trails in treating and curing hyperinsulinism (HI) for children across the nation and around the globe. (cookchildrens.org)
  • Congenital Hyperinsulinism International (CHI) is a health non-profit dedicated to improving the lives of children with a rare disease, congenital hyperinsulinism (HI). (congenitalhi.org)
  • The diagnosis of congenital hyperinsulinism is based on history, laboratory findings, and genetic testing. (chop.edu)
  • Once a diagnosis of hyperinsulinism is made, we need to educate families, to what is hyperinsulinism? (chop.edu)
  • An established clinical diagnosis of congenital HI (hyperinsulinism), with or without identification of a known monogenic variant by genetic testing. (who.int)
  • A large-for-gestational-age infant without any other somatic features of BWS presented with medically refractory hyperinsulinism (HI) requiring 80% pancreatectomy. (biomedcentral.com)
  • After an extended period of successful treatment, many people with ABCC8-related hyperinsulinism find their symptoms lessen in severity or even go into remission. (jscreen.org)
  • However with careful treatment, people with ABCC8-related hyperinsulinism can live normal lifespans. (jscreen.org)
  • The purpose of this study is to look at the safety and tolerability (how well you will react) of the study drug (HM15136) and to determine if it is effective for the treatment of Congenital Hyperinsulinism (HI). (chop.edu)
  • and Avexitide for the treatment of congenital hyperinsulinism, as well as has completed Phase II clinical trials to treat post-bariatric hypoglycemia. (yahoo.com)
  • citation needed] Hyperinsulinism due to reduced insulin sensitivity is usually asymptomatic. (wikipedia.org)
  • The principal treatments of hyperinsulinism due to insulin resistance are measures that improve insulin sensitivity, such as weight loss, physical exercise, and drugs such as thiazolidinediones or metformin . (ipfs.io)
  • The long-term outlook for someone with ABCC8-related hyperinsulinism depends upon the severity of the symptoms and the vigilance of the efforts to treat it. (jscreen.org)
  • Hyperinsulinism refers to an above normal level of insulin in the blood of a person or animal. (wikipedia.org)
  • Hyperinsulinism is where the body produces too much insulin, causing blood sugar levels to drop too low. (sibs.org.uk)
  • In some people with ABCC8-related hyperinsulinism, symptoms do not appear until later in childhood. (jscreen.org)
  • People with ABCC8-related hyperinsulinism may find their symptoms aggravated by viral infections and should take particular precautions when they become ill, even if their symptoms have gone into remission. (jscreen.org)
  • Treatments for ABCC8-related hyperinsulinism include dietary modification, medications, and surgical intervention. (jscreen.org)
  • There are several types of medication to treat ABCC8-related hyperinsulinism. (jscreen.org)
  • He is recognized the world over for his work in hyperinsulinism, serves on many national and international boards and committees and is a much sought after speaker and teacher. (cookchildrens.org)
  • He was recognized as a Rare Disease Hero for his work in hyperinsulinism. (cookchildrens.org)
  • Familiarity with congenital hyperinsulinism or the rare disease patient experience. (congenitalhi.org)
  • Purified fish oils could help treat congenital hyperinsulinism - a rare and potentially lethal disease of newborn babies, says research. (medindia.net)
  • And in congenital hyperinsulinism, none of those fuels are available, and hence a very high risk of 20 to 40% of brain damage that can occur in this condition. (cookchildrens.org)
  • Hyperinsulinism has also been implicated as a contributing factor in the excessive production of androgens in polycystic ovary syndrome . (ipfs.io)