Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Diabetes ; 72(12): 1809-1819, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37725835

ABSTRACT

The importance of glucokinase (GK) in the regulation of insulin secretion has been highlighted by the phenotypes of individuals with activating and inactivating mutations in the glucokinase gene (GCK). Here we report 10 individuals with congenital hyperinsulinism (HI) caused by eight unique activating mutations of GCK. Six are novel and located near previously identified activating mutations sites. The first recognized episode of hypoglycemia in these patients occurred between birth and 24 years, and the severity of the phenotype was also variable. Mutant enzymes were expressed and purified for enzyme kinetics in vitro. Mutant enzymes had low glucose half-saturation concentration values and an increased enzyme activity index compared with wild-type GK. We performed functional evaluation of islets from the pancreata of three children with GCK-HI who required pancreatectomy. Basal insulin secretion in perifused GCK-HI islets was normal, and the response to glyburide was preserved. However, the threshold for glucose-stimulated insulin secretion in perifused glucokinase hyperinsulinism (GCK-HI) islets was decreased, and glucagon secretion was greatly suppressed. Our evaluation of novel GCK disease-associated mutations revealed that the detrimental effects of these mutations on glucose homeostasis can be attributed not only to a lowering of the glucose threshold of insulin secretion but also to a decreased counterregulatory glucagon secretory response. ARTICLE HIGHLIGHTS: Our evaluation of six novel and two previously published activating GCK mutations revealed that the detrimental effects of these mutations on glucose homeostasis can be attributed not only to a lowering of the glucose threshold of insulin secretion but also to a decreased counterregulatory glucagon secretory response. These studies provide insights into the pathophysiology of GCK-hyperinsulinism and the dual role of glucokinase in ß-cells and α-cells to regulate glucose homeostasis.


Subject(s)
Congenital Hyperinsulinism , Hyperinsulinism , Child , Humans , Glucokinase/genetics , Glucagon , Congenital Hyperinsulinism/genetics , Hyperinsulinism/genetics , Glucose , Mutation , Phenotype
2.
Horm Res Paediatr ; 95(5): 492-498, 2022.
Article in English | MEDLINE | ID: mdl-35952631

ABSTRACT

INTRODUCTION: The hyperinsulinemia-hyperammonemia syndrome (HIHA) is the second most common cause of congenital hyperinsulinism and is caused by activating heterozygous missense mutations in GLUD1. In the majority of HIHA cases, the GLUD1 mutation is found to be de novo. We have identified 3 patients in whom clinical evaluation was suggestive of HIHA but with negative mutation analysis in peripheral blood DNA for GLUD1 as well as other known HI genes. METHODS: We performed next-generation sequencing (NGS) on peripheral blood DNA from two children with clinical features of HIHA in order to look for mosaic mutations in GLUD1. Pancreas tissue was also available in one of these cases for NGS. In addition, NGS was performed on peripheral blood DNA from a woman with a history of HI in infancy whose child had HIHA due to a presumed de novo GLUD1 mutation. RESULTS: Mosaic GLUD1 mutations were identified in these 3 cases at percent mosaicism ranging from 2.7% to 10.4% in peripheral blood. In one case with pancreas tissue available, the mosaic GLUD1 mutation was present at 17.9% and 28.9% in different sections of the pancreas. Two unique GLUD1 mutations were identified in these cases, both of which have been previously reported (c.1493c>t/p.Ser445Leu and c.820c>t/p.Arg221Cys). CONCLUSION: The results suggest that low-level mosaic mutations in known HI genes may be the underlying molecular mechanism in some children with HI who have negative genetic testing in peripheral blood DNA.


Subject(s)
Congenital Hyperinsulinism , Hyperammonemia , Hyperinsulinism , Child , Female , Humans , Hyperammonemia/genetics , Glutamate Dehydrogenase/genetics , Hyperinsulinism/genetics , Mutation , DNA , Congenital Hyperinsulinism/genetics
3.
Eur J Endocrinol ; 187(2): 301-313, 2022 Aug 01.
Article in English | MEDLINE | ID: mdl-35674212

ABSTRACT

Objective: Congenital hyperinsulinism (HI) is the most common cause of persistent hypoglycemia in children. In addition to typical focal or diffuse HI, some cases with diazoxide-unresponsive congenital HI have atypical pancreatic histology termed Localized Islet Nuclear Enlargement (LINE) or mosaic HI, characterized by histologic features similar to diffuse HI, but confined to only a region of pancreas. Our objective was to characterize the phenotype and genotype of children with LINE-HI. Design: The phenotype and genotype features of 12 children with pancreatic histology consistent with LINE-HI were examined. Methods: We compiled clinical features of 12 children with LINE-HI and performed next-generation sequencing on specimens of pancreas from eight of these children to look for mosaic mutations in genes known to be associated with diazoxide-unresponsive HI (ABCC8, KCNJ11, and GCK). Results: Children with LINE-HI had lower birth weights and later ages of presentation compared to children with typical focal or diffuse HI. Partial pancreatectomy in LINE-HI cases resulted in euglycemia in 75% of cases; no cases have developed diabetes. Low-level mosaic mutations were identified in the pancreas of six cases with LINE-HI (three in ABCC8, three in GCK). Expression studies confirmed that all novel mutations were pathogenic. Conclusion: These results indicate that post-zygotic low-level mosaic mutations of known HI genes are responsible for some cases of LINE-HI that lack an identifiable germ-line mutation and that partial pancreatectomy may be curative for these cases.


Subject(s)
Congenital Hyperinsulinism , Germinal Center Kinases , Sulfonylurea Receptors , Child , Congenital Hyperinsulinism/genetics , Diazoxide , Genotype , Germinal Center Kinases/genetics , Humans , Mutation , Phenotype , Sulfonylurea Receptors/genetics
4.
Am J Med Genet A ; 179(11): 2214-2227, 2019 11.
Article in English | MEDLINE | ID: mdl-31464105

ABSTRACT

Inactivating mutations in the genes encoding the two subunits of the pancreatic beta-cell KATP channel, ABCC8 and KCNJ11, are the most common finding in children with congenital hyperinsulinism (HI). Interpreting novel missense variants in these genes is problematic, because they can be either dominant or recessive mutations, benign polymorphisms, or diabetes mutations. This report describes six novel missense variants in ABCC8 and KCNJ11 that were identified in 11 probands with congenital HI. One of the three ABCC8 mutations (p.Ala1458Thr) and all three KCNJ11 mutations were associated with responsiveness to diazoxide. Sixteen family members carried the ABCC8 or KCNJ11 mutations; only two had hypoglycemia detected at birth and four others reported symptoms of hypoglycemia. Phenotype testing of seven adult mutation carriers revealed abnormal protein-induced hypoglycemia in all; fasting hypoketotic hypoglycemia was demonstrated in four of the seven. All of six mutations were confirmed to cause dominant pathogenic defects based on in vitro expression studies in COSm6 cells demonstrating normal trafficking, but reduced responses to MgADP and diazoxide. These results indicate a combination of in vitro and in vivo phenotype tests can be used to differentiate dominant from recessive KATP channel HI mutations and personalize management of children with congenital HI.


Subject(s)
Congenital Hyperinsulinism/diagnosis , Congenital Hyperinsulinism/genetics , Genetic Association Studies , Genetic Predisposition to Disease , KATP Channels/genetics , Mutation , Alleles , DNA Mutational Analysis , Female , Gene Frequency , Genotype , Humans , Infant , KATP Channels/chemistry , Male , Pedigree , Phenotype , Structure-Activity Relationship
5.
Horm Res Paediatr ; 89(6): 413-422, 2018.
Article in English | MEDLINE | ID: mdl-29902804

ABSTRACT

BACKGROUND: Previous case reports have suggested a possible association of congenital hyperinsulinism with Turner syndrome. OBJECTIVE: We examined the clinical and molecular features in girls with both congenital hyperinsulinism and Turner syndrome seen at The Children's Hospital of Philadelphia (CHOP) between 1974 and 2017. METHODS: Records of girls with hyperinsulinism and Turner syndrome were reviewed. Insulin secretion was studied in pancreatic islets and in mouse islets treated with an inhibitor of KDM6A, an X chromosome gene associated with hyperinsulinism in Kabuki syndrome. RESULTS: Hyperinsulinism was diagnosed in 12 girls with Turner syndrome. Six were diazoxide-unresponsive; 3 had pancreatectomies. The incidence of Turner syndrome among CHOP patients with hyperinsulinism (10 of 1,050 from 1997 to 2017) was 48 times more frequent than expected. The only consistent chromosomal anomaly in these girls was the presence of a 45,X cell line. Studies of isolated islets from 1 case showed abnormal elevated cytosolic calcium and heightened sensitivity to amino acid-stimulated insulin release; similar alterations were demonstrated in mouse islets treated with a KDM6A inhibitor. CONCLUSION: These results demonstrate a higher than expected frequency of Turner syndrome among children with hyperinsulinism. Our data suggest that haploinsufficiency for KDM6A due to mosaic X chromosome monosomy may be responsible for hyperinsulinism in Turner syndrome.


Subject(s)
Congenital Hyperinsulinism/genetics , Haploinsufficiency , Histone Demethylases/genetics , Nuclear Proteins/genetics , Turner Syndrome/genetics , Animals , Congenital Hyperinsulinism/diagnosis , Congenital Hyperinsulinism/metabolism , Female , Humans , Infant , Infant, Newborn , Mice , Retrospective Studies , Turner Syndrome/diagnosis , Turner Syndrome/metabolism
6.
Diabetes ; 66(7): 1901-1913, 2017 07.
Article in English | MEDLINE | ID: mdl-28442472

ABSTRACT

Loss-of-function mutations of ß-cell KATP channels cause the most severe form of congenital hyperinsulinism (KATPHI). KATPHI is characterized by fasting and protein-induced hypoglycemia that is unresponsive to medical therapy. For a better understanding of the pathophysiology of KATPHI, we examined cytosolic calcium ([Ca2+] i ), insulin secretion, oxygen consumption, and [U-13C]glucose metabolism in islets isolated from the pancreases of children with KATPHI who required pancreatectomy. Basal [Ca2+] i and insulin secretion were higher in KATPHI islets compared with controls. Unlike controls, insulin secretion in KATPHI islets increased in response to amino acids but not to glucose. KATPHI islets have an increased basal rate of oxygen consumption and mitochondrial mass. [U-13C]glucose metabolism showed a twofold increase in alanine levels and sixfold increase in 13C enrichment of alanine in KATPHI islets, suggesting increased rates of glycolysis. KATPHI islets also exhibited increased serine/glycine and glutamine biosynthesis. In contrast, KATPHI islets had low γ-aminobutyric acid (GABA) levels and lacked 13C incorporation into GABA in response to glucose stimulation. The expression of key genes involved in these metabolic pathways was significantly different in KATPHI ß-cells compared with control, providing a mechanism for the observed changes. These findings demonstrate that the pathophysiology of KATPHI is complex, and they provide a framework for the identification of new potential therapeutic targets for this devastating condition.


Subject(s)
Calcium/metabolism , Congenital Hyperinsulinism/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Oxygen Consumption , Potassium Channels, Inwardly Rectifying/metabolism , Sulfonylurea Receptors/metabolism , Alanine/metabolism , Carbon Isotopes , Case-Control Studies , Congenital Hyperinsulinism/genetics , Congenital Hyperinsulinism/surgery , Female , Flow Cytometry , Gene Expression , Glutamine/biosynthesis , Glycine/biosynthesis , Glycolysis/genetics , Humans , Immunohistochemistry , Infant , Infant, Newborn , Insulin Secretion , Insulin-Secreting Cells/ultrastructure , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , KATP Channels/genetics , KATP Channels/metabolism , Male , Metabolomics , Microscopy, Electron, Transmission , Mutation , Pancreatectomy , Potassium Channels, Inwardly Rectifying/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Serine/biosynthesis , Sulfonylurea Receptors/genetics , gamma-Aminobutyric Acid/metabolism
7.
Pediatr Diabetes ; 18(8): 911-916, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28165182

ABSTRACT

Activating mutations in the GLUD1 gene, which encodes glutamate dehydrogenase (GDH), result in the hyperinsulinism-hyperammonemia syndrome. GDH is an allosterically regulated enzyme responsible for amino acid-mediated insulin secretion via the oxidative deamination of glutamate to 2-oxoglutarate, leading to ATP production and insulin release. This study characterizes a novel combination of mutations in GLUD1 found in a neonate who presented on the first day of life with severe hypoglycemia, hyperammonemia, and seizures. Mutation analysis revealed a novel frameshift mutation (c.37delC) inherited from the asymptomatic mother that results in a truncated protein and a de novo activating mutation (p.S445L) close to the GTP binding site that has previously been reported. GTP inhibition of GDH enzyme activity in 293T cells expressing the p.S445L or wild-type GDH showed that the half-maximal inhibitory concentration (IC50 ) for GTP was approximately 800 times higher for p.S445L compared to wild type. GTP inhibition of GDH activity in lymphoblasts from the patient, from a heterozygote for the p.S445L mutation, and in wild-type lymphoblasts showed that the IC50 for GTP of the patient was approximately 200 times that of wild type and 7 times that of heterozygote. However, while the patient had a loss of GTP inhibition of GDH that was more severe than that of heterozygotes, the patient's clinical phenotype is similar to typical heterozygous mutations of GDH. This is the first time we have observed a functionally homozygous activating mutation of GDH in a human.


Subject(s)
Congenital Hyperinsulinism/genetics , Glutamate Dehydrogenase/genetics , Female , HEK293 Cells , Humans , Infant, Newborn , Inheritance Patterns , Mutation
8.
J Clin Endocrinol Metab ; 102(3): 942-949, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27967291

ABSTRACT

Context: The rarest genetic form of congenital hyperinsulinism (HI) has been associated with dominant inactivating mutations in uncoupling protein 2 (UCP2), a mitochondrial inner membrane carrier that modulates oxidation of glucose vs amino acids. Objective: To evaluate the frequency of UCP2 mutations in children with HI and phenotypic features of this form of HI. Design: We examined 211 children with diazoxide-responsive HI seen at The Children's Hospital of Philadelphia (CHOP) between 1997 and October 2016. Setting: CHOP Clinical and Translational Research Center. Results: Of 211 cases of diazoxide-responsive HI, we identified 5 unrelated children with UCP2 mutations (5 of 211; 2.4%). All 5 were diagnosed with HI before 6 months of age; diazoxide treatment was only partly effective in 3 of the 5. Among the 5 cases, 4 unique mutations (3 missense and 1 splicing) were identified. Three mutations were novel; 1 was previously reported. In vitro functional assays showed 30% to 75% decrease in UCP2 activity. Two of the children, when not taking diazoxide, developed hypoketotic-hypoglycemia after fasting 15 to 20 hours; a similar trend toward hypoglycemia after fasting 24 hours occurred in 4 adult carriers. In contrast, both children and 2 of the 4 carriers developed symptomatic hypoglycemia 4 hours following oral glucose. Unusual oscillating glucose and insulin responses to oral glucose were seen in both cases and carriers. Conclusions: These data indicate that dominant UCP2 mutations are a more important cause of HI than has been recognized and that affected individuals are markedly hypersensitive to glucose-induced hypoglycemia.


Subject(s)
Blood Glucose/metabolism , Congenital Hyperinsulinism/genetics , Fasting/metabolism , Insulin/metabolism , Uncoupling Protein 2/genetics , Child , Child, Preschool , Congenital Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/metabolism , DNA Mutational Analysis , Diazoxide/therapeutic use , Female , Glucose Tolerance Test , Heterozygote , Humans , Infant , Infant, Newborn , Male , Mutation , Mutation, Missense , Phenotype
9.
J Biol Chem ; 291(42): 21971-21983, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27573238

ABSTRACT

ATP-sensitive potassium (KATP) channels play a key role in mediating glucose-stimulated insulin secretion by coupling metabolic signals to ß-cell membrane potential. Loss of KATP channel function due to mutations in ABCC8 or KCNJ11, genes encoding the sulfonylurea receptor 1 (SUR1) or the inwardly rectifying potassium channel Kir6.2, respectively, results in congenital hyperinsulinism. Many SUR1 mutations prevent trafficking of channel proteins from the endoplasmic reticulum to the cell surface. Channel inhibitors, including sulfonylureas and carbamazepine, have been shown to correct channel trafficking defects. In the present study, we identified 13 novel SUR1 mutations that cause channel trafficking defects, the majority of which are amenable to pharmacological rescue by glibenclamide and carbamazepine. By contrast, none of the mutant channels were rescued by KATP channel openers. Cross-linking experiments showed that KATP channel inhibitors promoted interactions between the N terminus of Kir6.2 and SUR1, whereas channel openers did not, suggesting the inhibitors enhance intersubunit interactions to overcome channel biogenesis and trafficking defects. Functional studies of rescued mutant channels indicate that most mutants rescued to the cell surface exhibited WT-like sensitivity to ATP, MgADP, and diazoxide. In intact cells, recovery of channel function upon trafficking rescue by reversible sulfonylureas or carbamazepine was facilitated by the KATP channel opener diazoxide. Our study expands the list of KATP channel trafficking mutations whose function can be recovered by pharmacological ligands and provides further insight into the structural mechanism by which channel inhibitors correct channel biogenesis and trafficking defects.


Subject(s)
Carbamazepine/pharmacology , Glyburide/pharmacology , Mutation , Potassium Channels, Inwardly Rectifying/metabolism , Sulfonylurea Compounds/pharmacology , Sulfonylurea Receptors/metabolism , Female , Humans , Male , Potassium Channels, Inwardly Rectifying/genetics , Protein Transport/drug effects , Protein Transport/genetics , Sulfonylurea Receptors/genetics
10.
J Clin Endocrinol Metab ; 101(3): 914-22, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26756113

ABSTRACT

CONTEXT: Acquired insulinomas are rare causes of hyperinsulinemic hypoglycemia in children and are much less common than focal lesions of congenital hyperinsulinism. The latter are known to be associated with isodisomy for paternally transmitted ATP-sensitive potassium channel mutations on 11p15; however, the molecular basis for pediatric insulinomas is not well characterized. OBJECTIVE: The purpose of this study was to characterize the histopathological and molecular defects in a large group of 12 pediatric insulinomas seen at The Children's Hospital of Philadelphia. RESULTS: Twelve children with insulinomas were seen between 1971 and 2013, compared to 201 cases with focal congenital hyperinsulinism seen between 1997 and 2014. The age of insulinoma patients ranged from 4-16 years at the time of surgery. Features of MEN1 syndrome were present in five of the 12, including four cases with heterozygous mutations of MEN1 on 11q. Immunohistochemical analysis revealed nuclear loss of p57 staining consistent with loss of the maternal 11p15 allele in 11 of the 12 insulinomas, including all five MEN1-associated tumors. Imbalance of the paternal 11p allele was confirmed by single nucleotide polymorphism genotyping and methylation assays of the 11p imprinting control loci in four of five MEN1-associated tumors and six of seven sporadic insulinomas. In addition, single nucleotide polymorphism genotyping revealed extensive tumor aneuploidy beyond chromosome 11. CONCLUSIONS: These data indicate that MEN1 mutations are more common in insulinomas in children than in adults. Aneuploidy of chromosome 11 and other chromosomes is common in both MEN1 and non-MEN1 insulinomas. The novel observation of a paternal parent-of-origin effect in all MEN1 and most non-MEN1 tumors suggests a critical role for imprinted growth-regulatory genes in the 11p region in the genesis of ß-cell endocrine tumors in children.


Subject(s)
Insulinoma/genetics , Pancreatic Neoplasms/genetics , Adolescent , Aneuploidy , Child , Child, Preschool , Chromosomes, Human, Pair 11 , DNA Methylation , Female , Humans , Insulinoma/pathology , Male , Mutation , Pancreatic Neoplasms/pathology , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins/genetics
11.
J Med Genet ; 53(1): 53-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26545876

ABSTRACT

BACKGROUND: Congenital hyperinsulinism (HI) can have monogenic or syndromic causes. Although HI has long been recognised to be common in children with Beckwith-Wiedemann syndrome (BWS), the underlying mechanism is not known. METHODS: We characterised the clinical features of children with both HI and BWS/11p overgrowth spectrum, evaluated the contribution of KATP channel mutations to the molecular pathogenesis of their HI and assessed molecular pathogenesis associated with features of BWS. RESULTS: We identified 28 children with HI and BWS/11p overgrowth from 1997 to 2014. Mosaic paternal uniparental isodisomy for chromosome 11p (pUPD11p) was noted in 26/28 cases. Most were refractory to diazoxide treatment and half required subtotal pancreatectomies. Patients displayed a wide range of clinical features from classical BWS to only mild hemihypertrophy (11p overgrowth spectrum). Four of the cases had a paternally transmitted KATP mutation and had a much more severe HI course than patients with pUPD11p alone. CONCLUSIONS: We found that patients with pUPD11p-associated HI have a persistent and severe HI phenotype compared with transient hypoglycaemia of BWS/11p overgrowth patients caused by other aetiologies. Testing for pUPD11p should be considered in all patients with persistent congenital HI, especially for those without an identified HI gene mutation.


Subject(s)
Beckwith-Wiedemann Syndrome/genetics , Chromosomes, Human, Pair 11 , Congenital Hyperinsulinism/genetics , Uniparental Disomy , Beckwith-Wiedemann Syndrome/diagnosis , Beckwith-Wiedemann Syndrome/therapy , Child , Child, Preschool , Congenital Hyperinsulinism/diagnosis , Congenital Hyperinsulinism/drug therapy , DNA Methylation , DNA Mutational Analysis , Epigenesis, Genetic , Female , Humans , Infant , KATP Channels/genetics , Male , Mutation , Pancreas/metabolism , Pancreas/pathology , Phenotype , Polymorphism, Single Nucleotide
12.
Turk J Pediatr ; 58(5): 541-544, 2016.
Article in English | MEDLINE | ID: mdl-28621098

ABSTRACT

Hyperinsulinism/hyperammonemia (HI/HA) syndrome is a rare disorder presented with recurrent hypoglycemia and elevated serum ammonia, which may lead to development delays, permanent neurologic damages, if it remains underdiagnosed. It is caused by activating mutations in the GLUD1 gene which encodes the intra-mitochondrial enzyme glutamate dehydrogenase (GDH). HI/HA syndrome is considered the second most common form of hyperinsulinism (HI), and usually associated with epileptic seizures, mental retardation and generalized dystonia. We reported a patient who was diagnosed as HI/HA with multiple episodes of seizures; and previously had been diagnosed and treated for epilepsy. She has heterozygous mutation in GLUD1 gene. Treatment with diazoxide enabled complete resolution of the seizures. One year later, when her brother was six months old, he was also diagnosed with HI/HA. Later, the same mutation of GLUD1 was detected in both her father and brother too.


Subject(s)
Glutamate Dehydrogenase/genetics , Hyperinsulinism/diagnosis , Hypoglycemia/diagnosis , Seizures/etiology , Child, Preschool , Developmental Disabilities/complications , Female , Heterozygote , Humans , Male , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...