Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 753
Filter
1.
Int J Mol Sci ; 25(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38791571

ABSTRACT

Congenital hyperinsulinism (CHI) is a rare disorder of glucose metabolism and is the most common cause of severe and persistent hypoglycemia (hyperinsulinemic hypoglycemia, HH) in the neonatal period and childhood. Most cases are caused by mutations in the ABCC8 and KCNJ11 genes that encode the ATP-sensitive potassium channel (KATP). We present the correlation between genetic heterogeneity and the variable phenotype in patients with early-onset HH caused by ABCC8 gene mutations. In the first patient, who presented persistent severe hypoglycemia since the first day of life, molecular genetic testing revealed the presence of a homozygous mutation in the ABCC8 gene [deletion in the ABCC8 gene c.(2390+1_2391-1)_(3329+1_3330-1)del] that correlated with a diffuse form of hyperinsulinism (the parents being healthy heterozygous carriers). In the second patient, the onset was on the third day of life with severe hypoglycemia, and genetic testing identified a heterozygous mutation in the ABCC8 gene c.1792C>T (p.Arg598*) inherited on the paternal line, which led to the diagnosis of the focal form of hyperinsulinism. To locate the focal lesions, (18)F-DOPA (3,4-dihydroxy-6-[18F]fluoro-L-phenylalanine) positron emission tomography/computed tomography (PET/CT) was recommended (an investigation that cannot be carried out in the country), but the parents refused to carry out the investigation abroad. In this case, early surgical treatment could have been curative. In addition, the second child also presented secondary adrenal insufficiency requiring replacement therapy. At the same time, she developed early recurrent seizures that required antiepileptic treatment. We emphasize the importance of molecular genetic testing for diagnosis, management and genetic counseling in patients with HH.


Subject(s)
Congenital Hyperinsulinism , Genetic Heterogeneity , Hypoglycemia , Mutation , Phenotype , Sulfonylurea Receptors , Humans , Congenital Hyperinsulinism/genetics , Sulfonylurea Receptors/genetics , Female , Infant, Newborn , Male , Hypoglycemia/genetics , Infant , Potassium Channels, Inwardly Rectifying/genetics
2.
BMJ Case Rep ; 17(5)2024 May 22.
Article in English | MEDLINE | ID: mdl-38782444

ABSTRACT

Glycogen storage disease type 1A (GSD1A), also known as Von Gierke's disease, is a rare autosomal recessive disorder affecting glycogen metabolism in the liver. It most commonly presents in infancy with hypoglycaemia and failure to thrive, but cases have been reported as undiagnosed until adulthood. A woman in her early 20s with diabetes mellitus presented with right upper quadrant pain and was found to have several haemorrhagic hepatic adenomas. This patient had insulin-dependent diabetes since a pancreatectomy at age 9 months due to continued episodes of hypoglycaemia and suspected insulinoma. During the hospital stay, the hepatic adenomas were embolised, but significant lactic acidosis and hypoglycaemia continued. Further workup revealed a chronic lactic acid level, during several hospital stays, of above 5 mmol/L. After cytology of hepatic tissue ruled out hepatocellular carcinoma, the patient was discharged and recommended to follow-up for genetic testing, which confirmed the diagnosis of GSD1A.


Subject(s)
Congenital Hyperinsulinism , Glycogen Storage Disease Type I , Liver Neoplasms , Humans , Female , Liver Neoplasms/genetics , Liver Neoplasms/diagnosis , Congenital Hyperinsulinism/genetics , Congenital Hyperinsulinism/diagnosis , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/diagnosis , Adult , Adenoma, Liver Cell/genetics , Adenoma, Liver Cell/diagnosis , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/genetics , Young Adult , Adenoma/genetics , Adenoma/diagnosis , Adenoma/complications , Adenoma/surgery , Diagnosis, Differential
3.
BMJ Open Diabetes Res Care ; 12(2)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38575153

ABSTRACT

INTRODUCTION: Congenital hyperinsulinism (HI) is the leading cause of persistent hypoglycemia in infants. Current models to study the most common and severe form of HI resulting from inactivating mutations in the ATP-sensitive potassium channel (KATP) are limited to primary islets from patients and the Sur1 -/- mouse model. Zebrafish exhibit potential as a novel KATPHI model since they express canonical insulin secretion pathway genes and those with identified causative HI mutations. Moreover, zebrafish larvae transparency provides a unique opportunity for in vivo visualization of pancreatic islets. RESEARCH DESIGN AND METHODS: We evaluated zebrafish as a model for KATPHI using a genetically encoded Ca2+ sensor (ins:gCaMP6s) expressed under control of the insulin promoter in beta cells of an abcc8 -/- zebrafish line. RESULTS: We observed significantly higher islet cytosolic Ca2+ in vivo in abcc8 -/- compared with abcc8 +/+ zebrafish larvae. Additionally, abcc8 -/- larval zebrafish had significantly lower whole body glucose and higher whole body insulin levels compared with abcc8 +/+ controls. However, adult abcc8 -/- zebrafish do not show differences in plasma glucose, plasma insulin, or glucose tolerance when compared with abcc8 +/+ zebrafish. CONCLUSIONS: Our results identify that zebrafish larvae, but not adult fish, are a demonstrable novel model for advancement of HI research.


Subject(s)
Congenital Hyperinsulinism , Potassium Channels, Inwardly Rectifying , Infant , Adult , Animals , Mice , Humans , KATP Channels/genetics , Zebrafish/genetics , Zebrafish/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Congenital Hyperinsulinism/genetics , Insulin/metabolism , Glucose , Adenosine Triphosphate
4.
Rev Med Liege ; 79(3): 168-174, 2024 Mar.
Article in French | MEDLINE | ID: mdl-38487911

ABSTRACT

Congenital hyperinsulinism is the most common cause of recurrent hypoglycemia in newborns and children. Early diagnosis and rapid management are essential to avoid hypoglycaemic brain injury and later neurological complications. Management of those patients involves biological evaluation, molecular genetics, imaging techniques and surgical advances. We report the case of a newborn with recurrent hypoglycemia due to congenital hyperinsulinism (CHI) caused by a new variant in the ABCC8 gene. Fluorine 18-L-3,4 Dihydroxyphenylalanine Positron Emission Tomography (18F-DOPA PET/CT scan) reported a focal lesion at the isthmus of the pancreas which has been removed by laparoscopic surgery with a complete recovery for the patient.


L'hyperinsulinisme congénital est la cause la plus fréquente d'hypoglycémies récidivantes chez le nouveau-né et l'enfant. Un diagnostic et une prise en charge précoces sont primordiaux pour éviter les conséquences potentielles sur le développement neurologique. Ces derniers reposent sur la conjonction d'éléments biologiques, génétiques et d'imagerie. Nous rapportons le cas d'un nouveau-né présentant des hypoglycémies récidivantes. La mise au point mettra en évidence un hyperinsulinisme congénital (CHI) lié à un variant non encore décrit au sein du gène ABCC8. L'imagerie par Fluorine 18-L-3,4 Dihydroxyphenylalanine Positron Emission Tomography/Computed Tomography-scanner (18F-DOPA PET/CT scan) a mis en évidence une forme focale de l'hyperinsulinisme justifiant une prise en charge chirurgicale amenant à une guérison complète et à l'arrêt de tout traitement médicamenteux.


Subject(s)
Congenital Hyperinsulinism , Laparoscopy , Child , Humans , Infant, Newborn , Infant , Positron Emission Tomography Computed Tomography , Congenital Hyperinsulinism/diagnostic imaging , Congenital Hyperinsulinism/genetics , Congenital Hyperinsulinism/pathology , Pancreas/pathology , Pancreas/surgery , Positron-Emission Tomography/methods
5.
Diabetologia ; 67(5): 940-951, 2024 May.
Article in English | MEDLINE | ID: mdl-38366195

ABSTRACT

AIMS/HYPOTHESIS: The ATP-sensitive potassium (KATP) channel couples beta cell electrical activity to glucose-stimulated insulin secretion. Loss-of-function mutations in either the pore-forming (inwardly rectifying potassium channel 6.2 [Kir6.2], encoded by KCNJ11) or regulatory (sulfonylurea receptor 1, encoded by ABCC8) subunits result in congenital hyperinsulinism, whereas gain-of-function mutations cause neonatal diabetes. Here, we report a novel loss-of-function mutation (Ser118Leu) in the pore helix of Kir6.2 paradoxically associated with sulfonylurea-sensitive diabetes that presents in early adult life. METHODS: A 31-year-old woman was diagnosed with mild hyperglycaemia during an employee screen. After three pregnancies, during which she was diagnosed with gestational diabetes, the patient continued to show elevated blood glucose and was treated with glibenclamide (known as glyburide in the USA and Canada) and metformin. Genetic testing identified a heterozygous mutation (S118L) in the KCNJ11 gene. Neither parent was known to have diabetes. We investigated the functional properties and membrane trafficking of mutant and wild-type KATP channels in Xenopus oocytes and in HEK-293T cells, using patch-clamp, two-electrode voltage-clamp and surface expression assays. RESULTS: Functional analysis showed no changes in the ATP sensitivity or metabolic regulation of the mutant channel. However, the Kir6.2-S118L mutation impaired surface expression of the KATP channel by 40%, categorising this as a loss-of-function mutation. CONCLUSIONS/INTERPRETATION: Our data support the increasing evidence that individuals with mild loss-of-function KATP channel mutations may develop insulin deficiency in early adulthood and even frank diabetes in middle age. In this case, the patient may have had hyperinsulinism that escaped detection in early life. Our results support the importance of functional analysis of KATP channel mutations in cases of atypical diabetes.


Subject(s)
Congenital Hyperinsulinism , Diabetes, Gestational , Potassium Channels, Inwardly Rectifying , Infant, Newborn , Adult , Middle Aged , Female , Pregnancy , Humans , Potassium Channels, Inwardly Rectifying/genetics , Sulfonylurea Receptors/genetics , Sulfonylurea Receptors/metabolism , Congenital Hyperinsulinism/genetics , Sulfonylurea Compounds/therapeutic use , Mutation/genetics , Glyburide , Adenosine Triphosphate/metabolism
6.
Int J Mol Sci ; 25(2)2024 01 20.
Article in English | MEDLINE | ID: mdl-38279270

ABSTRACT

The BiP co-chaperone DNAJC3 protects cells during ER stress. In mice, the deficiency of DNAJC3 leads to beta-cell apoptosis and the gradual onset of hyperglycemia. In humans, biallelic DNAJC3 variants cause a multisystem disease, including early-onset diabetes mellitus. Recently, hyperinsulinemic hypoglycemia (HH) has been recognized as part of this syndrome. This report presents a case study of an individual with HH caused by DNAJC3 variants and provides an overview of the metabolic phenotype of individuals with HH and DNAJC3 variants. The study demonstrates that HH may be a primary symptom of DNAJC3 deficiency and can persist until adolescence. Additionally, glycemia and insulin release were analyzed in young DNACJ3 knockout (K.O.) mice, which are equivalent to human infants. In the youngest experimentally accessible age group of 4-week-old mice, the in vivo glycemic phenotype was already dominated by a reduced total insulin secretion capacity. However, on a cellular level, the degree of insulin release of DNAJC3 K.O. islets was higher during periods of increased synthetic activity (high-glucose stimulation). We propose that calcium leakage from the ER into the cytosol, due to disrupted DNAJC3-controlled gating of the Sec61 channel, is the most likely mechanism for HH. This is the first genetic mechanism explaining HH solely by the disruption of intracellular calcium homeostasis. Clinicians should screen for HH in DNAJC3 deficiency and consider DNAJC3 variants in the differential diagnosis of congenital hyperinsulinism.


Subject(s)
Congenital Hyperinsulinism , HSP40 Heat-Shock Proteins , Adolescent , Animals , Humans , Mice , Calcium/metabolism , Congenital Hyperinsulinism/genetics , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Insulin/metabolism , Insulin Secretion , Molecular Chaperones/genetics , Molecular Chaperones/metabolism
7.
Clin Genet ; 105(5): 549-554, 2024 05.
Article in English | MEDLINE | ID: mdl-38225536

ABSTRACT

Congenital hyperinsulinism (CHI; OMIM: 256450) is characterized by persistent insulin secretion despite severe hypoglycemia. The most common causes are variants in the ATP-binding cassette subfamily C member 8(ABCC8) and potassium inwardly-rectifying channel subfamily J member 11(KCNJ11) genes. These encode ATP-sensitive potassium (KATP) channel subunit sulfonylurea receptor 1 (SUR1) and inwardly rectifying potassium channel (Kir6.2) proteins. A 7-day-old male infant presented with frequent hypoglycemic episodes and was clinically diagnosed with CHI, underwent trio-whole-exome sequencing, revealing compound heterozygous ABCC8 variants (c.307C>T, p.His103Tyr; and c.3313_3315del, p.Ile1105del) were identified. In human embryonic kidney 293 (HEK293) and rat insulinoma cells (INS-1) transfected with wild-type and variant plasmids, KATP channels formed by p.His103Tyr were delivered to the plasma membrane, whereas p.Ile1105del or double variants (p.His103Tyr coupled with p.Ile1105del) failed to be transported to the plasma membrane. Compared to wild-type channels, the channels formed by the variants (p.His103Tyr; p.Ile1105del) had elevated basal [Ca2+]i, but did not respond to stimulation by glucose. Our results provide evidence that the two ABCC8 variants may be related to CHI owing to defective trafficking and dysfunction of KATP channels.


Subject(s)
Congenital Hyperinsulinism , Potassium Channels, Inwardly Rectifying , Infant , Animals , Rats , Male , Humans , Sulfonylurea Receptors/genetics , Sulfonylurea Receptors/metabolism , Potassium Channels, Inwardly Rectifying/genetics , HEK293 Cells , Receptors, Drug/genetics , Receptors, Drug/metabolism , Mutation/genetics , Congenital Hyperinsulinism/genetics , Adenosine Triphosphate , Potassium/metabolism
9.
BMC Endocr Disord ; 24(1): 8, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38212772

ABSTRACT

BACKGROUND: ABCC8 variants can cause hyperinsulinemia by activating or deactivating gene expression. This study used targeted exon sequencing to investigate genetic variants of ABCC8 and the associated phenotypic features in Chinese patients with hyperinsulinemic hypoglycemia (HH). METHODS: We enrolled eight Chinese children with HH and analyzed their clinical characteristics, laboratory results, and genetic variations. RESULTS: The age at presentation among the patients ranged from neonates to 0.6 years old, and the age at diagnosis ranged from 1 month to 5 years, with an average of 1.3 ± 0.7 years. Among these patients, three presented with seizures, and five with hypoglycemia. One patient (Patient 7) also had microcephaly. All eight patients exhibited ABCC8 abnormalities, including six missense mutations (c. 2521 C > G, c. 3784G > A, c. 4478G > A, c. 4532T > C, c. 2669T > C, and c. 331G > A), two deletion-insertion mutations (c. 3126_3129delinsTC and c. 3124_3126delins13), and one splicing mutation (c. 1332 + 2T > C). Two of these mutations (c. 3126_3129delinsTC and c. 4532T > C) are novel. Six variations were paternal, two were maternal, and one was de novo. Three patients responded to diazoxide and one patient responded to octreotide treatment. All there patients had diazoxide withdrawal with age. Two patients (patients 3 and 7) were unresponsive to both diazoxide and octreotide and had mental retardation. CONCLUSIONS: Gene analysis can aid in the classification, treatment, and prognosis of children with HH. In this study, the identification of seven known and two novel variants in the ABCC8 gene further enriched the variation spectrum of the gene.


Subject(s)
Congenital Hyperinsulinism , Infant, Newborn , Child , Humans , Congenital Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/genetics , Congenital Hyperinsulinism/diagnosis , Diazoxide/therapeutic use , Octreotide/therapeutic use , Mutation , China/epidemiology , Sulfonylurea Receptors/genetics
11.
Clin Endocrinol (Oxf) ; 100(2): 132-137, 2024 02.
Article in English | MEDLINE | ID: mdl-38059644

ABSTRACT

OBJECTIVE: Transient hyperinsulinism (THI) is the most common form of recurrent hypoglycaemia in neonates beyond the first week of life. Although self-resolving, treatment can be required. Consensus guidelines recommend the lower end of the diazoxide 5-15 mg/kg/day range in THI to reduce the risk of adverse events. We sought to determine if doses <5 mg/kg/day of diazoxide can be effective in THI. DESIGN, PATIENTS, MEASURMENTS: Infants with THI (duration <6 months) were treated with low-dose diazoxide from October 2015 to February 2021. Dosing was based on weight at diazoxide start: 2 mg/kg/day in infants 1000-2000 g (cohort 1), 3 mg/kg/day in those 2000-3500 g (cohort 2) and 5 mg/kg/day in those >3500 g. RESULTS: A total of 73 infants with THI (77% male, 33% preterm, 52% small-for-gestational age) were commenced on diazoxide at a median age of 11 days (range 3-43) for a median duration of 4 months (0.3-6.8), with no difference between cohorts. The mean effective diazoxide dose was 3 mg/kg/day (range 1.5-10); 35% (26/73) required an increase from their starting dose, including 60% (9/15) of cohort 1. There was no association between perinatal stress risk factors or treatment-related characteristics and dose increase. Adverse events occurred in 13 patients (18%); oedema (12%) and hyponatraemia (5%) were the most common. Two infants developed suspected necrotising enterocolitis (NEC); none had pulmonary hypertension. CONCLUSION: Diazoxide doses <5 mg/kg/day are effective in THI. While the nature of the association between diazoxide and NEC was unclear, other adverse events were mild. We suggest considering starting doses as low as 2-3 mg/kg/day in THI to balance the side effect risk while maintaining euglycaemia.


Subject(s)
Congenital Hyperinsulinism , Hyperinsulinism , Hypoglycemia , Infant , Female , Infant, Newborn , Humans , Male , Diazoxide/adverse effects , Hypoglycemia/drug therapy , Infant, Small for Gestational Age , Risk Factors , Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/drug therapy
12.
J Pediatr (Rio J) ; 100(2): 163-168, 2024.
Article in English | MEDLINE | ID: mdl-37866397

ABSTRACT

OBJECTIVE: Congenital hyperinsulinism (CHI) is a heterogeneous genetic disease characterized by increased insulin secretion and causes persistent hypoglycemia in neonates and infants due to dysregulation of insulin secretion by pancreatic ß cells. Babies with severe hypoglycemia and for whom medical treatment has been ineffective usually require surgical treatment with near-total pancreatectomy. To evaluate the clinical and surgical aspects affecting survival outcomes in babies diagnosed with CHI in a single tertiary care center. METHODS: Retrospective Cohort study involving a single university tertiary center for the treatment of CHI. The authors study the demographics, clinical, laboratory, and surgical outcomes of this casuistic. RESULTS: 61 % were female, 39 % male, Birth weight: 3576 g (±313); Age of onset of symptoms: from the 2nd hour of life to 28 days; Time between diagnosis and surgery ranged between 10 and 60 days; Medical clinical treatment, all patients received glucose solution with a continuous glucose infusion and diazoxide. 81 % of the patients used corticosteroids, 77 %. thiazide, 72 % octreotide, 27 % nifedipine; Neurological sequelae during development and growth: 54 % had some degree of delay in neuropsychomotor development, 27 % obesity. Surgery was performed open in 6 and 12 minimally invasive surgery (MIS). HISTOPATHOLOGY: 2 focal and 16 diffuse, Length of stay (days) was lower in MIS (p < 0.05). Survival was 100 %. CONCLUSIONS: CHI is a rare and difficult-to-manage tumor that must be performed in a multidisciplinary and tertiary center. Most surgical results are good and the laparoscopic approach to disease has been the best choice for patients.


Subject(s)
Congenital Hyperinsulinism , Infant , Infant, Newborn , Humans , Male , Female , Retrospective Studies , Brazil , Congenital Hyperinsulinism/surgery , Congenital Hyperinsulinism/diagnosis , Congenital Hyperinsulinism/drug therapy , Glucose/therapeutic use , Treatment Outcome
13.
J Clin Endocrinol Metab ; 109(4): 1071-1079, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-37930757

ABSTRACT

CONTEXT: Congenital hyperinsulinism (CHI) is characterized by dysregulated insulin secretion causing hypoglycemia and consequent brain damage. Dasiglucagon is a glucagon analogue under investigation to treat CHI. OBJECTIVE: To evaluate the efficacy and safety of dasiglucagon delivered via continuous subcutaneous infusion to children with CHI and persistent hypoglycemia as add-on to standard of care (SoC). METHODS: In this open-label trial, patients were randomized 1:1 to SoC or SoC + dasiglucagon (10-70 µg/h) for 4 weeks. In the following 4 weeks, all patients received dasiglucagon + SoC. Hypoglycemia was assessed by self-monitored plasma glucose (SMPG) and blinded continuous glucose monitoring (CGM). Primary endpoint was average number of SMPG-detected hypoglycemia episodes/week (SMPG <3.9 mmol/L) during Weeks 2 to 4. RESULTS: Thirty-two patients (0.6-10.9 years) were randomly assigned to dasiglucagon + SoC (n = 16) or SoC (n = 16). The rate of SMPG-detected hypoglycemia decreased from baseline in both groups, but with no statistically significant difference during Weeks 2 to 4 (event rate ratio: 0.85 [0.54; 1.36], P = .5028). However, dasiglucagon administration resulted in a 43% reduction in CGM-detected hypoglycemia (<3.9 mmol/L) vs SoC alone during Weeks 2 to 4 (post hoc analysis; event rate ratio: 0.57 [0.39; 0.83], P = .0029). Dasiglucagon enabled reductions (of 37% to 61%) in all other measures of hypoglycemia assessed by CGM vs SoC alone including extent and percent time in hypoglycemia (post hoc analyses). Dasiglucagon appeared safe and well tolerated. Skin and gastrointestinal events were more frequent with dasiglucagon + SoC than SoC only. CONCLUSION: Clinically meaningful reductions in all CGM-recorded measures of hypoglycemia support using dasiglucagon as a potential treatment for CHI.


Subject(s)
Congenital Hyperinsulinism , Diabetes Mellitus, Type 1 , Glucagon/analogs & derivatives , Infant , Child , Humans , Glucagon/therapeutic use , Blood Glucose , Blood Glucose Self-Monitoring , Congenital Hyperinsulinism/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin/adverse effects
15.
Tidsskr Nor Laegeforen ; 143(18)2023 12 12.
Article in English, Norwegian | MEDLINE | ID: mdl-38088279

ABSTRACT

This clinical review will give doctors who work with children and neonates an introduction to the diagnosis and treatment of congenital hyperinsulinism, the most common cause of persistent neonatal hypoglycaemia. The condition is a rare monogenic disorder characterised by elevated insulin secretion and is a result of mutations in genes that regulate insulin secretion from pancreatic beta cells. The anabolic effect of insulin induces systemic glucose uptake and inhibits gluconeogenesis, glycogenolysis, ketogenesis and lipolysis. Low levels of glucose and ketone bodies in the blood are harmful to the central nervous system and can lead to brain damage or death. Early diagnosis and treatment of congenital hyperinsulinism are therefore crucial for a good prognosis.


Subject(s)
Congenital Hyperinsulinism , Child , Infant, Newborn , Humans , Congenital Hyperinsulinism/diagnosis , Congenital Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/genetics , Ketone Bodies , Insulin
16.
Femina ; 51(12): 666-673, 20231230. ilus, tab
Article in Portuguese | LILACS | ID: biblio-1532469

ABSTRACT

Objetivo: Atualizar a estatística do serviço, reconhecendo a prevalência de amnior- rexe prematura no pré-termo e seus principais desfechos materno-fetais. Méto- dos: Estudo transversal realizado pela análise de prontuários médicos de pacien- tes internadas devido a amniorrexe prematura no pré-termo e de seus respectivos conceptos no Hospital Universitário da Faculdade de Medicina de Jundiaí durante o período de janeiro de 2020 a dezembro de 2021. Resultados: Participaram da pesquisa 161 pacientes e 166 conceptos, resultando em uma prevalência de 2,12% no período estudado, com intervalo de confiança de 95% (1,80-2,47). Entre os des- fechos maternos, 2,5% das gestantes compunham critérios para near miss mater- no; enquanto entre os desfechos fetais, o resultado foi de 54,8% dos conceptos apresentando complicações, sendo as mais prevalentes a síndrome do desconfor- to respiratório (36,3%), icterícia (39,5%), baixo peso (27,5%) e hipoglicemia (24,2%). Além disso, 40,4% necessitaram de internação na unidade de terapia intensiva, 22,9% foram classificados como near miss neonatal e 4,4% foram a óbito. Conclu- são: Os resultados seguiram os padrões nacionais e internacionais esperados para prevalência de amniorrexe prematura no pré-termo e seus desfechos materno-fe- tais, com alta porcentagem de internações e complicações neonatais e baixa taxa de complicações maternas.


Objective: To update service statistics, recognizing the preva- lence of the pathology and its main outcomes. Methods: Cros- s-sectional study carried out through the analysis of medical records of patients hospitalized due to preterm premature rup- ture of membranes and their respective fetuses at the Univer- sity Hospital of Jundiaí's Medical School during the period from January 2020 to December 2021. Results: A total of 161 patients and 166 fetuses participated in the research, resulting in a pre- valence of 2.12% in the period studied with 95% confidence in- terval (1.80-2.47). About the outcomes, 2.5% of the pregnant wo- men composed the criteria for maternal near miss; as for the fetus, complications evolved in 54.8% of the fetuses, the most prevalent being respiratory distress syndrome (36.3%), jaundice (39.5%), low birth weight (27.5%) and hypoglycemia (24.2%). In addition, 40.4% required admission to the intensive care unit, 22.9% were neonatal near miss and 4.4% died. Conclusion: The results followed the expected national and international standards for the prevalence of preterm premature rupture of membranes and its maternal and fetal outcomes, with a high percentage of hospitalizations and neonatal complications, and a low rate of maternal complications.


Subject(s)
Humans , Female , Pregnancy , Fetal Membranes, Premature Rupture , Obstetric Labor, Premature , Respiratory Distress Syndrome, Newborn/diagnosis , Infant, Low Birth Weight , Maternal Mortality/trends , Medical Records/statistics & numerical data , Statistics , Congenital Hyperinsulinism/diagnosis , Near Miss, Healthcare/statistics & numerical data , Jaundice/complications
17.
Orv Hetil ; 164(47): 1877-1884, 2023 Nov 26.
Article in Hungarian | MEDLINE | ID: mdl-38007812

ABSTRACT

In congenital hyperinsulinemic hypoglycemia - the most common cause of persistent hypoglycemia in infancy - a focal lesion can be identified in 50% of the cases. With appropriate medical care based upon early diagnosis, these patients can be cured by the resection of the lesion rendering unnecessary long time medical care, and avoiding serious brain damage from recurrent hypoglycemic episodes. Genetic testing and 18F-fluoro-dihydroxyphenylalanine PET/CT imaging are essential for determining the best possible treatment. We report 2 cases of focal congenital hyperinsulinism - both male infants: 22 and 2 months of age - treated successfully with enucleation of the pancreas lesion (Semmelweis University, Budapest). Both patients had the pathognomonic mutation of the ABCC8 gene of the ATP-sensitive potassium channel. Radiologic imaging and histology confirmed the diagnosis, and after the operation, pharmacological treatment was terminated in both cases. During the follow-up period (5 and 1.5 years, respectively) they are euglycemic, with no morbidities attributed to the operation. We believe that these two operations for focal hyperinsulinism - diagnosed and localised by the above detailed genetic and specific radiological testing - were the first of their kind in Hungary. Based on the acquired experience, every necessary examination can be achieved in our country to improve patient care, reduce morbidity and medical costs. Orv Hetil. 2023; 164(47): 1877-1884.


Subject(s)
Congenital Hyperinsulinism , Hyperinsulinism , Infant , Humans , Male , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography/methods , Congenital Hyperinsulinism/diagnosis , Congenital Hyperinsulinism/genetics , Congenital Hyperinsulinism/surgery , Pancreas/pathology , Mutation , Hyperinsulinism/pathology
18.
J Pediatr Endocrinol Metab ; 36(12): 1133-1139, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37988593

ABSTRACT

OBJECTIVES: Unrecognized hypoglycemia, especially in the neonatal population, is a significant cause of morbidity and poor neurologic outcomes. Children with congenital hyperinsulinism (HI) are at risk of hypoglycemia and point of care testing (POCT) is the standard of care. Studies have shown that continuous glucose monitoring (CGM) improves glycemic control and reduces the frequency of hypoglycemia among children with type 1 diabetes. There is limited experience with the use of CGM in children with HI. To assess the glycemic pattern of children with HI on stable therapy and evaluate the frequency of undetected hypoglycemia using Dexcom G6® CGM. METHODS: A cross-sectional, observational pilot study was done in 10 children, ages 3 months to 17 years. Each child had a clinical or genetic diagnosis of HI on stable medical therapy. Participants were asked to continue their usual POCT blood glucose monitoring, as well as wear a blinded Dexcom G6® CGM during a 20-day study period with the potential of unblinding if there was severe hypoglycemia detected during the study trial. RESULTS: During the study period, 26 hypoglycemic events were noted by CGM in 60 % of the participants with 45 % occurring between 0600 and 0800. CONCLUSIONS: CGM can help detect hypoglycemia and blood glucose trends during a time when there is usually no POCT, which can guide medical management. 30 % of our population had a dose adjustment in their medications. This study was limited by population size.


Subject(s)
Congenital Hyperinsulinism , Diabetes Mellitus, Type 1 , Child , Infant, Newborn , Humans , Blood Glucose , Blood Glucose Self-Monitoring , Cross-Sectional Studies , Hypoglycemic Agents/adverse effects , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Congenital Hyperinsulinism/diagnosis , Congenital Hyperinsulinism/drug therapy
19.
Orphanet J Rare Dis ; 18(1): 360, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37974153

ABSTRACT

BACKGROUND: Hypoketotic hypoglycaemia with suppressed plasma fatty acids and detectable insulin suggests congenital hyperinsulinism (CHI). Severe hypoketotic hypoglycaemia mimicking hyperinsulinism but without detectable insulin has recently been described in syndromic individuals with mosaic genetic activation of post-receptor insulin signalling. We set out to expand understanding of this entity focusing on metabolic phenotypes. METHODS: Metabolic profiling, candidate gene and exome sequencing were performed in six infants with hypoketotic, hypoinsulinaemic hypoglycaemia, with or without syndromic features. Additional signalling studies were carried out in dermal fibroblasts from two individuals. RESULTS: Two infants had no syndromic features. One was mistakenly diagnosed with CHI. One had mild features of megalencephaly-capillary malformation-polymicrogyria (MCAP) syndrome, one had non-specific macrosomia, and two had complex syndromes. All required intensive treatment to maintain euglycaemia, with CHI-directed therapies being ineffective. Pathogenic PIK3CA variants were found in two individuals - de novo germline c.323G>A (p.Arg108His) in one non-syndromic infant and postzygotic mosaic c.2740G>A (p.Gly914Arg) in the infant with MCAP. No causal variants were proven in the other individuals despite extensive investigation, although rare variants in mTORC components were identified in one. No increased PI3K signalling in fibroblasts of two individuals was seen. CONCLUSIONS: We expand the spectrum of PI3K-related hypoinsulinaemic hypoketotic hypoglycaemia. We demonstrate that pathogenic germline variants activating post-insulin-receptor signalling may cause non-syndromic hypoinsulinaemic hypoketotic hypoglycaemia closely resembling CHI. This distinct biochemical footprint should be sought and differentiated from CHI in infantile hypoglycaemia. To facilitate adoption of this differential diagnosis, we propose the term "pseudohyperinsulinism".


Subject(s)
Congenital Hyperinsulinism , Proto-Oncogene Proteins c-akt , Infant , Humans , Proto-Oncogene Proteins c-akt/genetics , Insulin , Congenital Hyperinsulinism/genetics , Phosphatidylinositol 3-Kinases/metabolism
20.
Front Endocrinol (Lausanne) ; 14: 1283907, 2023.
Article in English | MEDLINE | ID: mdl-38033998

ABSTRACT

Objective: Congenital hyperinsulinism (CHI) is a group of clinically and genetically heterogeneous disorders characterized by dysregulated insulin secretion. The aim of the study was to elucidate genetic etiologies of Taiwanese children with the most severe diazoxide-unresponsive CHI and analyze their genotype-phenotype correlations. Methods: We combined Sanger with whole exome sequencing (WES) to analyze CHI-related genes. The allele frequency of the most common variant was estimated by single-nucleotide polymorphism haplotype analysis. The functional effects of the ATP-sensitive potassium (KATP) channel variants were assessed using patch clamp recording and Western blot. Results: Nine of 13 (69%) patients with ten different pathogenic variants (7 in ABCC8, 2 in KCNJ11 and 1 in GCK) were identified by the combined sequencing. The variant ABCC8 p.T1042QfsX75 identified in three probands was located in a specific haplotype. Functional study revealed the human SUR1 (hSUR1)-L366F KATP channels failed to respond to intracellular MgADP and diazoxide while hSUR1-R797Q and hSUR1-R1393C KATP channels were defective in trafficking. One patient had a de novo dominant mutation in the GCK gene (p.I211F), and WES revealed mosaicism of this variant from another patient. Conclusion: Pathogenic variants in KATP channels are the most common underlying cause of diazoxide-unresponsive CHI in the Taiwanese cohort. The p.T1042QfsX75 variant in the ABCC8 gene is highly suggestive of a founder effect. The I211F mutation in the GCK gene and three rare SUR1 variants associated with defective gating (p.L366F) or traffic (p.R797Q and p.R1393C) KATP channels are also associated with the diazoxide-unresponsive phenotype.


Subject(s)
Congenital Hyperinsulinism , Potassium Channels, Inwardly Rectifying , Humans , Child , Diazoxide/therapeutic use , Potassium Channels, Inwardly Rectifying/genetics , Sulfonylurea Receptors/genetics , Congenital Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/genetics , Genetic Association Studies , Adenosine Triphosphate
SELECTION OF CITATIONS
SEARCH DETAIL
...