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.
Surgical removal of the pancreas. (Dorland, 28th ed)
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.
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.
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.
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 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).
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.
A type of pancreatic cell representing about 50-80% of the islet cells. Beta cells secrete INSULIN.
An infant during the first month after birth.
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 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)
An individual having different alleles at one or more loci regarding a specific character.
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.

Hyperinsulinism in infancy: from basic science to clinical disease. (1/62)

Ion channelopathies have now been described in many well-characterized cell types including neurons, myocytes, epithelial cells, and endocrine cells. However, in only a few cases has the relationship between altered ion channel function, cell biology, and clinical disease been defined. Hyperinsulinism in infancy (HI) is a rare, potentially lethal condition of the newborn and early childhood. The causes of HI are varied and numerous, but in almost all cases they share a common target protein, the ATP-sensitive K+ channel. From gene defects in ion channel subunits to defects in beta-cell metabolism and anaplerosis, this review describes the relationship between pathogenesis and clinical medicine. Until recently, HI was generally considered an orphan disease, but as parallel defects in ion channels, enzymes, and metabolic pathways also give rise to diabetes and impaired insulin release, the HI paradigm has wider implications for more common disorders of the endocrine pancreas and the molecular physiology of ion transport.  (+info)

Characterization of hyperinsulinism in infancy assessed with PET and 18F-fluoro-L-DOPA. (2/62)

Hyperinsulinism (HI) of infancy is a neuroendocrine disease secondary to either focal adenomatous hyperplasia or a diffuse abnormality of insulin secretion of the pancreas. HI with focal lesions can revert by selective surgical resection in contrast to the diffuse form, which requires subtotal pancreatectomy when resistant to medical treatment. Neuroendocrine diseases are a heterogeneous group of entities with the ability to take up amine precursors and to convert them into biogenic amines. Therefore, the aim of this study was (a) to evaluate the use of PET with 18F-fluoro-L-dihydroxyphenylalanine (18F-fluoro-L-DOPA) and (b) to distinguish between focal and diffuse HI. METHODS: Fifteen patients (11 boys, 4 girls) with neonatal HI were enrolled in this study. All patients fasted for at least 6 h before the PET examination and their medication was discontinued for at least 72 h. The examination was performed under light sedation (pentobarbital associated with or without chloral). The dynamic acquisition started 45-65 min after the injection of 18F-fluoro-L-DOPA (4.0 MBq/kg weight). Four or 6 scans of 5 min each (2 or 3 steps according to the height of the patient) were acquired from the neck to the upper legs. RESULTS: An abnormal focal pancreatic uptake of 18F-fluoro-L-DOPA was observed in 5 patients, whereas a diffuse uptake of the radiotracer was observed in the pancreatic area of the other patients. All patients with focal radiotracer uptake and also 4 of 10 patients with pancreatic diffuse radiotracer accumulation, unresponsive to medical treatment, underwent surgery. The histopathologic results confirmed the PET findings--that is, focal versus diffuse HI. CONCLUSION: The results of this study suggest that 18F-fluoro-L-DOPA could be an accurate noninvasive technique to distinguish between focal and diffuse forms of HI.  (+info)

Low temperature completely rescues the function of two misfolded K ATP channel disease-mutants. (3/62)

The pancreatic ATP-sensitive potassium channels comprise two subunits: SUR1 and Kir6.2. Two SUR1 mutations, A116P and V187D, reduce channel activity causing persistent hyperinsulinemic hypoglycemia of infancy. We investigated whether these mutations cause temperature sensitive misfolding. We show that the processing defect of these mutants is temperature sensitive and these two mutations disrupt the association between SUR1 and Kir6.2 by causing misfolding in SUR1 at 37 degrees C but can be rescued at 18 degrees C. Extensive electrophysiological characterization of these mutants indicated that low temperature largely, if not completely, corrects the folding defect of these two SUR1 mutants observed at 37 degrees C.  (+info)

A novel KCNJ11 mutation associated with congenital hyperinsulinism reduces the intrinsic open probability of beta-cell ATP-sensitive potassium channels. (4/62)

The beta-cell ATP-sensitive potassium (KATP) channel controls insulin secretion by linking glucose metabolism to membrane excitability. Loss of KATP 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. Here, we report identification of a novel KCNJ11 mutation associated with the disease that renders a missense mutation, F55L, in the Kir6.2 protein. Mutant channels reconstituted in COS cells exhibited a wild-type-like surface expression level and normal sensitivity to ATP, MgADP, and diazoxide. However, the intrinsic open probability of the mutant channel was greatly reduced, by approximately 10-fold. This low open probability defect could be reversed by application of phosphatidylinositol 4,5-bisphosphates or oleoyl-CoA to the cytoplasmic face of the channel, indicating that reduced channel response to membrane phospholipids and/or long chain acyl-CoAs underlies the low intrinsic open probability in the mutant. Our findings reveal a novel molecular mechanism for loss of KATP channel function and congenital hyperinsulinism and support the importance of phospholipids and/or long chain acyl-CoAs in setting the physiological activity of beta-cell KATP channels. The F55L mutation is located in the slide helix of Kir6.2. Several permanent neonatal diabetes-associated mutations found in the same structure have the opposite effect of increasing intrinsic channel open probability. Our results also highlight the critical role of the Kir6.2 slide helix in determining the intrinsic open probability of KATP channels.  (+info)

Molecular and immunohistochemical analyses of the focal form of congenital hyperinsulinism. (5/62)

Congenital hyperinsulinism is a rare pancreatic endocrine cell disorder that has been categorized histologically into diffuse and focal forms. In focal hyperinsulinism, the pancreas contains a focus of endocrine cell adenomatous hyperplasia, and the patients have been reported to possess paternally inherited mutations of the ABCC8 and KCNJ11 genes, which encode subunits of an ATP-sensitive potassium channel (K(ATP)). In addition, the hyperplastic endocrine cells show loss of maternal 11p15, where imprinted genes such as p57(kip2) reside. In order to evaluate whether all cases of focal hyperinsulinism are caused by this mechanism, 56 pancreatectomy specimens with focal hyperinsulinism were tested for the loss of maternal allele by two methods: immunohistochemistry for p57(kip2) (n=56) and microsatellite marker analysis (n=27). Additionally, 49 patients were analyzed for K(ATP) mutations. Out of 56 focal lesions, 48 demonstrated clear loss of p57(kip2) expression by immunohistochemistry. The other eight lesions similarly showed no nuclear labeling, but the available tissue was not ideal for definitive interpretation. Five of these eight patients had paternal K(ATP) mutations, of which four demonstrated loss of maternal 11p15 within the lesion by microsatellite marker analysis. All of the other three without a paternal K(ATP) mutation showed loss of maternal 11p15. K(ATP) mutation analysis identified 32/49 cases with paternal mutations. There were seven patients with nonmaternal mutations whose paternal DNA material was not available, and one patient with a mutation that was not present in either parent's DNA. These eight patients showed either loss of p57(kip2) expression or loss of maternal 11p15 region by microsatellite marker analysis, as did the remaining nine patients with no identifiable K(ATP) coding region mutations. The combined results from the immunohistochemical and molecular methods indicate that maternal 11p15 loss together with paternal K(ATP) mutation is the predominant causative mechanism of focal hyperinsulinism.  (+info)

Noninvasive diagnosis of focal hyperinsulinism of infancy with [18F]-DOPA positron emission tomography. (6/62)

Congenital hyperinsulinism of infancy (CHI) is characterized by severe hypoglycemia due to dysregulated insulin secretion, associated with either focal or diffuse pathology of the endocrine pancreas. The focal condition is caused by a paternally inherited mutation in one of the genes encoding the subunits of the beta-cell ATP-sensitive potassium channel (SUR1/ABCC8 or Kir6.2/KCNJ11) and somatic loss of maternal 11p15 alleles within the affected area. Until now, preoperative diagnostics have relied on technically demanding and invasive catheterization techniques. We evaluated the utility of fluorine-18 l-3,4-dihydroxyphenylalanine ([(18)F]-DOPA) positron emission tomography (PET) to identify focal pancreatic lesions in 14 CHI patients, 11 of which carried mutations in the ABCC8 gene (age 1-42 months). To reduce bias in PET image interpretation, quantitative means for evaluation of pancreatic [(18)F]-DOPA uptake were established. Five patients had a visually apparent focal accumulation of [(18)F]-DOPA and standardized uptake value (SUV) >50% higher (mean 1.8-fold) than the maximum SUV of the unaffected part of the pancreas. When these patients were operated on, a focus of 4-5 x 5-8 mm matching with the PET scan was found, and all were normoglycemic after resection of the focus. The remaining nine patients had diffuse accumulation of [(18)F]-DOPA in the pancreas (SUV ratio <1.5). Diffuse histology was verified in four of these, and pancreatic catheterization was consistent with diffuse pathology in four cases. In conclusion, [(18)F]-DOPA PET is a promising noninvasive method for the identification and localization of the focal form of CHI.  (+info)

Molecular mechanisms of neonatal hyperinsulinism. (7/62)

Congenital hyperinsulinism (CHI), characterized by profound hypoglycaemia related to inappropriate insulin secretion, may be associated histologically with either diffuse insulin hypersecretion or focal adenomatous hyperplasia, which share a similar clinical presentation, but result from different molecular mechanisms. Whereas diffuse CHI is of autosomal recessive, or less frequently of autosomal dominant, inheritance, focal CHI is sporadic. The most common mechanism underlying CHI is dysfunction of the pancreatic ATP-sensitive potassium channel (K(+)(ATP)). The two subunits of the K(+)(ATP) channel are encoded by the sulfonylurea receptor gene (SUR1 or ABCC8) and the inward-rectifying potassium channel gene (KIR6.2 or KCNJ11), both located in the 11p15.1 region. Germ-line, paternally inherited, mutations of the SUR1 or KIR6.2 genes, together with somatic maternal haplo-insufficiency for 11p15.5, were shown to result in focal CHI. Diffuse CHI results from germ-line mutations in the SUR1 or KIR6.2 genes, but also from mutations in several other genes, namely glutamate dehydrogenase (with associated hyperammonaemia), glucokinase, short-chain L-3-hydroxyacyl-CoA dehydrogenase, and insulin receptor gene. Hyperinsulinaemic hypoglycaemia may be observed in several overlapping syndromes, such as Beckwith-Wiedemann syndrome (BWS), Perlman syndrome, and, more rarely, Sotos syndrome. Mosaic genome-wide paternal isodisomy has recently been reported in patients with clinical signs of BWS and CHI. The primary causes of CHI are genetically heterogeneous and have not yet been completely unveiled. However, secondary causes of hyperinsulinism have to be considered such as fatty acid oxidation deficiency, congenital disorders of glycosylation and factitious hypoglycaemia secondary to Munchausen by proxy syndrome.  (+info)

Macrosomia and hyperinsulinaemic hypoglycaemia in patients with heterozygous mutations in the HNF4A gene. (8/62)

BACKGROUND: Macrosomia is associated with considerable neonatal and maternal morbidity. Factors that predict macrosomia are poorly understood. The increased rate of macrosomia in the offspring of pregnant women with diabetes and in congenital hyperinsulinaemia is mediated by increased foetal insulin secretion. We assessed the in utero and neonatal role of two key regulators of pancreatic insulin secretion by studying birthweight and the incidence of neonatal hypoglycaemia in patients with heterozygous mutations in the maturity-onset diabetes of the young (MODY) genes HNF4A (encoding HNF-4alpha) and HNF1A/TCF1 (encoding HNF-1alpha), and the effect of pancreatic deletion of Hnf4a on foetal and neonatal insulin secretion in mice. METHODS AND FINDINGS: We examined birthweight and hypoglycaemia in 108 patients from families with diabetes due to HNF4A mutations, and 134 patients from families with HNF1A mutations. Birthweight was increased by a median of 790 g in HNF4A-mutation carriers compared to non-mutation family members (p < 0.001); 56% (30/54) of HNF4A-mutation carriers were macrosomic compared with 13% (7/54) of non-mutation family members (p < 0.001). Transient hypoglycaemia was reported in 8/54 infants with heterozygous HNF4A mutations, but was reported in none of 54 non-mutation carriers (p = 0.003). There was documented hyperinsulinaemia in three cases. Birthweight and prevalence of neonatal hypoglycaemia were not increased in HNF1A-mutation carriers. Mice with pancreatic beta-cell deletion of Hnf4a had hyperinsulinaemia in utero and hyperinsulinaemic hypoglycaemia at birth. CONCLUSIONS: HNF4A mutations are associated with a considerable increase in birthweight and macrosomia, and are a novel cause of neonatal hypoglycaemia. This study establishes a key role for HNF4A in determining foetal birthweight, and uncovers an unanticipated feature of the natural history of HNF4A-deficient diabetes, with hyperinsulinaemia at birth evolving to decreased insulin secretion and diabetes later in life.  (+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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

"Syndromic Causes of Congenital Hyperinsulinism", Congenital Hyperinsulinism, Cham: Springer International Publishing, pp. 49-59 ... Congenital Hyperinsulinism International. Retrieved 2023-08-22. "Congenital Hyperinsulinism Center". The Children's Hospital of ... Hyperinsulinism or congenital hyperinsulinism can be a genetic or acquired condition. Acquired HI may be secondary to factors ... Hyperinsulinism Genes. Information on the genetic mechanisms of CHI. Congenital Hyperinsulinism International. Nonprofit ...
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 ...
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/ ... "Congenital hyperinsulinism: current trends in diagnosis and therapy". Orphanet Journal of Rare Diseases. 6 (1): 63. doi:10.1186 ...
Online Mendelian Inheritance in Man (OMIM): 605285 Hewat TI, Johnson MB, Flanagan SE (7 July 2022). "Congenital Hyperinsulinism ... 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 ...
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- ... 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- ...
Meissner T, Beinbrech B, Mayatepek E (1999). "Congenital hyperinsulinism: molecular basis of a heterogeneous disease". Human ... and congenital hyperinsulinism. Alternative splicing of this gene has been observed; however, the transcript variants have not ... "Clinical and molecular characterisation of 300 patients with congenital hyperinsulinism". European Journal of Endocrinology. ... June 1994). "Familial hyperinsulinism maps to chromosome 11p14-15.1, 30 cM centromeric to the insulin gene". Nature Genetics. 7 ...
Mazor-Aronovitch K, Landau H, Gillis D (Mar 2009). "Surgical versus non-surgical treatment of congenital hyperinsulinism". ... "Preoperative evaluation of infants with focal or diffuse congenital hyperinsulinism by intravenous acute insulin response tests ... "Clinical characteristics and biochemical mechanisms of congenital hyperinsulinism associated with dominant KATP channel ... "Long-term neurodevelopmental outcome in conservatively treated congenital hyperinsulinism". European Journal of Endocrinology. ...
The gene encoding the channel is called KCNJ11 and mutations in this gene are associated with congenital hyperinsulinism. It is ... 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 ... "Clinical and molecular characterisation of 300 patients with congenital hyperinsulinism". European Journal of Endocrinology. ...
Congenital hyperinsulinism Hyperinsulinemic hypoglycemia Wong's nursing care of infants and children. Hockenberry, Marilyn J ...
"Clinical characteristics and biochemical mechanisms of congenital hyperinsulinism associated with dominant KATP channel ... Hyperinsulinism-hyperammonemia syndrome (HI/HA) is an autosomal dominant disorder that results in the excess production of ...
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. ...
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 ... In more severe cases of persistent congenital hyperinsulinism unresponsive to drugs, a near-total pancreatectomy may be needed ...
Three CDG subtypes PMM2-CDG, PMI-CDG, ALG6-CDG can cause congenital hyperinsulinism with hyperinsulinemic hypoglycemia in ... Congenital Disorder of Glycosylation Type 1a; Jaeken Syndrome GeneReviews/NIH/NCBI/UW entry on Congenital Disorders of ... "Congenital disorder of glycosylation due to DPM1 mutations presenting with dystroglycanopathy-type congenital muscular ... A congenital disorder of glycosylation (previously called carbohydrate-deficient glycoprotein syndrome) is one of several rare ...
... or congenital hyperinsulinism. Diazoxide acts as a positive allosteric modulator of the AMPA and kainate receptors, suggesting ...
... is also used in the treatment of refractory hypoglycemia or congenital hyperinsulinism in neonates and sulphonylurea ... McMahon AW, Wharton GT, Thornton P, De Leon DD (January 2017). "Octreotide use and safety in infants with hyperinsulinism". ...
This creates hypoglycemia of varying patterns, including transient or persistent congenital hyperinsulinism, or fasting or ... Homozygosity for GCK alleles with reduced function can cause severe congenital insulin deficiency, resulting in persistent ... Glaser B (2013-01-24). "Familial Hyperinsulinism". GeneReviews. Seattle WA: University of Washington, Seattle. PMID 20301549. ...
Most congenital hyperinsulinism is now known to be caused by different mechanisms than excessive proliferation of beta cells in ... Congenital hyperinsulinism Neonatal hypoglycemia Raffel A, Krausch MM, Anlauf M, Wieben D, Braunstein S, Klöppel G, Röher H, ... in most cases from the 1970s until the 1980s it was used as a synonym for what is now referred to as congenital hyperinsulinism ... Clancy T, Moore F, Zinner M (2006). "Post-gastric bypass hyperinsulinism with nesidioblastosis: subtotal or total ...
A meter can occasionally be useful in the monitoring of severe types of hypoglycemia (e.g., congenital hyperinsulinism) to ...
... determined the physiological role of cardiac KATP channels and identified one type of congenital hyperinsulinism. Animal models ...
Congenital Disorder of Glycosylation (CDG) Congenital hyperinsulinism Congenital insensitivity to pain with anhidrosis (CIPA) ... congenital anomalies Rare disease (Articles with short description, Short description is different from Wikidata, Congenital ... Congenital heart defects) Hemifacial microsomia Holoprosencephaly Huntington's disease Hirschsprung's disease, or congenital ... Congenital central hypoventilation syndrome Congenital diaphragmatic hernia (CDH) ...
Congenital hyperinsulinism Minor findings associated with BWS Pregnancy-related findings including polyhydramnios and ... Gosden R, Trasler J, Lucifero D, Faddy M (2003). "Rare congenital disorders, imprinted genes, and assisted reproductive ... "Congenital mesoblastic nephroma 50 years after its recognition: A narrative review" (PDF). Pediatric Blood & Cancer. 64 (7): ... characterized by an increased risk of childhood cancer and certain congenital features. A minority (. 97th centile) ...
... or congenital hyperinsulinism, increases blood glucose and decreases insulin secretion and glucagon accelerates breakdown of ...
... from which he sought to donate proceeds for research towards diseases such congenital hyperinsulinism. In December 2020, Jesé ...
All of the congenital metabolic defects, congenital forms of hyperinsulinism, and congenital hypopituitarism are likely to have ... congenital hypopituitarism, or congenital hyperinsulinism.[citation needed] A list of common causes: Prolonged fasting ... Congenital hypopituitarism Congenital hyperinsulinism, several types, both transient and persistent Inborn errors of ... Hyperinsulinism due to several congenital disorders of insulin secretion Insulin injected for type 1 diabetes Hyperinsulinism- ...
... congenital Hyperinsulinism, diffuse Hyperinsulinism, focal Hyperkalemia Hyperkalemic periodic paralysis Hyperkeratosis ... E Hyperinsulinism due to focal adenomatous hyperplasia Hyperinsulinism due to glucokinase deficiency Hyperinsulinism due to ... congenital Hillig syndrome Hing-Torack-Dowston syndrome Hinson-Pepys disease Hip dislocation Hip dysplasia Beukes type Hip ... congenital essential Hemeralopia, familial Hemi 3 syndrome Hemifacial atrophy agenesis of the caudate nucleus Hemifacial ...
... but the crucial role of the fetal hyperinsulinism and monitoring of motherly glucose was nevertheless stressed. Recent studies ... among other things the incidence of congenital malformations, supporting the Hypothesis, that even moderately increased blood ...
Congenital hyperammonemia is usually due to genetic defects in one of the enzymes of the urea cycle, such as ornithine ... hyperinsulinism-hyperammonemia syndrome (glutamate dehydrogenase 1) Online Mendelian Inheritance in Man (OMIM): 238970 - ...
May 2020). "Fetal antisense oligonucleotide therapy for congenital deafness and vestibular dysfunction". Nucleic Acids Research ... 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 MeSH C18.452.394.968.500 - insulin resistance MeSH C18.452.394.968.500.570 - metabolic syndrome x MeSH C18.452. ... congenital MeSH C18.452.648.925.500 - mineralocorticoid excess syndrome, apparent MeSH C18.452.648.925.750 - ichthyosis, x- ...
Hyperinsulinism Too high a level of insulin in the blood. This often involves a condition in which the body produces too much ... Congenital defect problems or conditions that are present at birth. Congestive heart failure heart failure caused by loss of ...
"Syndromic Causes of Congenital Hyperinsulinism", Congenital Hyperinsulinism, Cham: Springer International Publishing, pp. 49-59 ... Congenital Hyperinsulinism International. Retrieved 2023-08-22. "Congenital Hyperinsulinism Center". The Childrens Hospital of ... Hyperinsulinism or congenital hyperinsulinism can be a genetic or acquired condition. Acquired HI may be secondary to factors ... Hyperinsulinism Genes. Information on the genetic mechanisms of CHI. Congenital Hyperinsulinism International. Nonprofit ...
... many authors prefer the term congenital hyperinsulinism (CHI). It was first identified in 1938, when Laidlaw coined the term ... represents the most common cause of hyperinsulinism in neonates; currently, ... encoded search term (Congenital Hyperinsulinism) and Congenital Hyperinsulinism What to Read Next on Medscape ... Congenital Hyperinsulinism. Pancreatic specimen showing congenital hyperinsulinism (CHI) viewed at low power. Paler-staining ...
"A Very High Unmet Need" for the Most Severe Hyperinsulinism Types The most common and severe form of congenital hyperinsulinism ... A rare disorder, occurring in 1 in 20,000 to 1 in 50,000 US live births, congenital hyperinsulinism is caused by several ... Cite this: Early Promise for New Agent in Congenital Hyperinsulinism in Kids - Medscape - Apr 21, 2022. ... which includes congenital hyperinsulinism). It has also been granted a rare pediatric disease designation by FDA. ...
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 ...
Congenital hyperinsulinism is a condition of dysregulated insulin secretion often caused by inactivating mutations of the ATP- ... Clinical characteristics and biochemical mechanisms of congenital hyperinsulinism associated with dominant KATP channel ... Clinical characteristics and biochemical mechanisms of congenital hyperinsulinism associated with dominant KATP channel ... In contrast to a previous report of increased diabetes risk in dominant KATP hyperinsulinism, only 4 of 29 adults had diabetes ...
Clinical practice guidelines for congenital hyperinsulinism.. Yorifuji T, Horikawa R, Hasegawa T, Adachi M, Soneda S, Minagawa ...
HM15136 Treatment for 8 Weeks in Subjects Aged ≥2 Years With Congenital Hyperinsulinism (CHI) ...
Angelman syndrome, Beckwith-Wiedemann syndrome, Congenital hyperinsulinism, Genome-wide uniparental disomy, Mosaicism. in ... We report of an apparently non-syndromic infant with severe congenital hyperinsulinism (CHI) and diffuse pancreatic labelling ... We report of an apparently non-syndromic infant with severe congenital hyperinsulinism (CHI) and diffuse pancreatic labelling ... We report of an apparently non-syndromic infant with severe congenital hyperinsulinism (CHI) and diffuse pancreatic labelling ...
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 ...
Nonprofit Overview: Congenital Hyperinsulinism International (CHI) is a health non-profit dedicated to improving the lives of ... Familiarity with congenital hyperinsulinism or the rare disease patient experience.. *A strong interest in rare diseases, ... Nonprofit Overview: Congenital Hyperinsulinism International (CHI) is the leading organization dedicated to improving the lives ... The Development Manager will work to support the mission, vision and priority goals of Congenital Hyperinsulinism International ...
Welcome to hyperinsulinism.dk. Welcome to hyperinsulinism.dk, a site dedicated to the condition called CHI - Congenital ... hyperinsulinism in infants.. CHI is a condition that causes newborns to have abnormally high levels of insulin, which is a ...
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.. ...
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. ... HM15136 for the Treatment of Congenital Hyperinsulinism Research Study. The purpose of this study is to look at the safety and ... The Congenital Hyperinsulinism Center at the Childrens Hospital of Philadelphia is working on a research study to better ... The Congenital Hyperinsulinism Center at the Childrens Hospital of Philadelphia is working on a research study to better ...
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, also known as persistent hyperinsulinemic hypoglycemia of infancy, is the most frequent cause of ... Real-Time Continuous Glucose Monitoring in a Newborn with Congenital Hyperinsulinism Date of submission: 19-03-2020 , Date of ... particularly in children with congenital hyperinsulinism.. In this case report, the authors present their experience with real- ... time continuous glucose monitoring system in a neonate with congenital hyperinsulinism, associated with a novel missense ...
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 ... in patients with congenital hyperinsulinism (CHI). The Company has screened the first patient and expects to make significant ...
Congenital hyperinsulinism: MedlinePlus Genetics (National Library of Medicine) * Dihydropyrimidinase deficiency: MedlinePlus ... ALG1-congenital disorder of glycosylation: MedlinePlus Genetics (National Library of Medicine) * ALG12-congenital disorder of ... NGLY1-congenital disorder of deglycosylation: MedlinePlus Genetics (National Library of Medicine) * Ornithine transcarbamylase ... ALG6-congenital disorder of glycosylation: MedlinePlus Genetics (National Library of Medicine) * Aspartylglucosaminuria: ...
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). ...
Home , Archives , Vol 13, No 2 (2023) , Successful laparoscopic resection of a rare focal form of congenital hyperinsulinism: ... Successful laparoscopic resection of a rare focal form of congenital hyperinsulinism: Case report and literature review - PDF ( ...
Background: Congenital Hyperinsulinism (CHI) is the most frequent cause of severe, persistent hypoglycemia in children. ... Results from a Global, Multi-Center, Phase 2b Study (RIZE) in Congenital Hyperinsulinism: Characterization of a High Unmet ... Results from a Global, Multi-Center, Phase 2b Study (RIZE) in Congenital Hyperinsulinism: Characterization of a High Unmet ... Results from a Global, Multi-Center, Phase 2b Study (RIZE) in Congenital Hyperinsulinism: Characterization of a High Unmet ...
Our findings reveal a novel molecular mechanism for loss of K ATP channel function and congenital hyperinsulinism and support ... Our findings reveal a novel molecular mechanism for loss of K ATP channel function and congenital hyperinsulinism and support ... Our findings reveal a novel molecular mechanism for loss of K ATP channel function and congenital hyperinsulinism and support ... Our findings reveal a novel molecular mechanism for loss of K ATP channel function and congenital hyperinsulinism and support ...
Heterogeneity in Phenotype of Usher-Congenital Hyperinsulinism Syndrome: Hearing Loss, Retinitis Pigmentosa, and ... Heterogeneity in Phenotype of Usher-Congenital Hyperinsulinism Syndrome: Hearing Loss, Retinitis Pigmentosa, and ...
Watch the video to learn more about the expert care provided by the Congenital Hyperinsulinism Center at The Childrens ... What is Hyperinsulinism (HI)?. Diva D. De León-Crutchlow, MD: Hyperinsulinism is a congenital disorder of regulation of insulin ... Congenital hyperinsulinism (HI) is a disorder that causes low blood sugar (hypoglycemia) in infants and children. Congenital ... to what is hyperinsulinism? What can be causing the hyperinsulinism? And how can we treat the hyperinsulinism? ...
C-Path and Congenital Hyperinsulinism International Announce Data Sharing Agreement. TUCSON, Ariz., October 26, 2023 - Critical ... 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 ...
Study Title: Insulin Receptor Antagonists for Treating Congenital Hyperinsulinism. Maya Kasowski, MD, PhD. Assistant Professor ... Study Title: Evaluation of Congenital Unilateral Hydronephrosis using Artificial Intelligence. Katrin Svensson, PhD. Assistant ... Study Title: Identification and Functional Validation of Pathogenic Noncoding Loci in Congenital Heart Disease. ... Project: Bilayered Nanofibrillar Vascular Graft for Treatment of Congenital Heart Defect. Prasanna Jagannathan, M.D.. Assistant ...

No FAQ available that match "congenital hyperinsulinism"

No images available that match "congenital hyperinsulinism"