Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Diabetes ; 71(3): 367-375, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35196393

RESUMEN

Secretion of insulin from pancreatic ß-cells is complex, but physiological glucose-dependent secretion is dominated by electrical activity, in turn controlled by ATP-sensitive potassium (KATP) channel activity. Accordingly, loss-of-function mutations of the KATP channel Kir6.2 (KCNJ11) or SUR1 (ABCC8) subunit increase electrical excitability and secretion, resulting in congenital hyperinsulinism (CHI), whereas gain-of-function mutations cause underexcitability and undersecretion, resulting in neonatal diabetes mellitus (NDM). Thus, diazoxide, which activates KATP channels, and sulfonylureas, which inhibit KATP channels, have dramatically improved therapies for CHI and NDM, respectively. However, key findings do not fit within this simple paradigm: mice with complete absence of ß-cell KATP activity are not hyperinsulinemic; instead, they are paradoxically glucose intolerant and prone to diabetes, as are older human CHI patients. Critically, despite these advances, there has been little insight into any role of KATP channel activity changes in the development of type 2 diabetes (T2D). Intriguingly, the CHI progression from hypersecretion to undersecretion actually mirrors the classical response to insulin resistance in the progression of T2D. In seeking to explain the progression of CHI, multiple lines of evidence lead us to propose that underlying mechanisms are also similar and that development of T2D may involve loss of KATP activity.


Asunto(s)
Hiperinsulinismo Congénito/genética , Hiperinsulinismo Congénito/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Canales KATP/fisiología , Animales , Glucemia , Calcio/farmacología , Humanos , Resistencia a la Insulina , Secreción de Insulina/genética , Secreción de Insulina/fisiología , Canales KATP/genética , Ratones , Ratones Noqueados , Mutación , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/fisiología , Receptores de Sulfonilureas/genética , Receptores de Sulfonilureas/fisiología
2.
Diabetologia ; 64(3): 630-640, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33404684

RESUMEN

AIMS/HYPOTHESIS: Congenital hyperinsulinism caused by mutations in the KATP-channel-encoding genes (KATPHI) is a potentially life-threatening disorder of the pancreatic beta cells. No optimal medical treatment is available for patients with diazoxide-unresponsive diffuse KATPHI. Therefore, we aimed to create a model of KATPHI using patient induced pluripotent stem cell (iPSC)-derived islets. METHODS: We derived iPSCs from a patient carrying a homozygous ABCC8V187D mutation, which inactivates the sulfonylurea receptor 1 (SUR1) subunit of the KATP-channel. CRISPR-Cas9 mutation-corrected iPSCs were used as controls. Both were differentiated to stem cell-derived islet-like clusters (SC-islets) and implanted into NOD-SCID gamma mice. RESULTS: SUR1-mutant and -corrected iPSC lines both differentiated towards the endocrine lineage, but SUR1-mutant stem cells generated 32% more beta-like cells (SC-beta cells) (64.6% vs 49.0%, p = 0.02) and 26% fewer alpha-like cells (16.1% vs 21.8% p = 0.01). SUR1-mutant SC-beta cells were 61% more proliferative (1.23% vs 0.76%, p = 0.006), and this phenotype could be induced in SUR1-corrected cells with pharmacological KATP-channel inactivation. The SUR1-mutant SC-islets secreted 3.2-fold more insulin in low glucose conditions (0.0174% vs 0.0054%/min, p = 0.0021) and did not respond to KATP-channel-acting drugs in vitro. Mice carrying grafts of SUR1-mutant SC-islets presented with 38% lower fasting blood glucose (4.8 vs 7.7 mmol/l, p = 0.009) and their grafts failed to efficiently shut down insulin secretion during induced hypoglycaemia. Explanted SUR1-mutant grafts displayed an increase in SC-beta cell proportion and SC-beta cell nucleomegaly, which was independent of proliferation. CONCLUSIONS/INTERPRETATION: We have created a model recapitulating the known pathophysiology of KATPHI both in vitro and in vivo. We have also identified a novel role for KATP-channel activity during human islet development. This model will enable further studies for the improved understanding and clinical management of KATPHI without the need for primary patient tissue.


Asunto(s)
Hiperinsulinismo Congénito/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Islotes Pancreáticos/metabolismo , Receptores de Sulfonilureas/metabolismo , Animales , Diferenciación Celular , Línea Celular , Proliferación Celular , Hiperinsulinismo Congénito/genética , Hiperinsulinismo Congénito/patología , Hiperinsulinismo Congénito/fisiopatología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/trasplante , Secreción de Insulina , Islotes Pancreáticos/patología , Islotes Pancreáticos/fisiopatología , Trasplante de Islotes Pancreáticos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Mutación , Fenotipo , Receptores de Sulfonilureas/genética
3.
Pan Afr Med J ; 35: 53, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32537058

RESUMEN

Neonatal hypoglycemia (NH) is one of the most common abnormalities encountered in the newborn. Hypoglycemia continues to be an important cause of morbidity in neonates and children. Prompt diagnosis and management of the underlying hypoglycemia disorder is critical for preventing brain damage and improving outcomes. Congenital hyperinsulinism (CHI) is the most common and severe cause of persistent hypoglycemia in neonates and children, it represents a group of clinically, genetically and morphologically heterogeneous disorders characterised by dysregulation of insulin secretion from pancreatic ß-cells. It is extremely important to recognize this condition early and institute appropriate management to prevent significant brain injury leading to complications like epilepsy, cerebral palsy and neurological impairment. Histologically, CHI is divided mainly into two types focal and diffuse disease. The diffuse form is inherited in an autosomal recessive (or dominant) manner whereas the focal form is sporadic in inheritance and is localized to a small region of the pancreas. Recent discoveries of the genetic causes of CHI have improved our understanding of the pathophysiology, but its management is complex and requires the integration of clinical, biochemical, molecular, and imaging findings to establish the appropriate treatment according to the subtype. Here we present a case of sever congenital hyperinsulinism in a girl admitted for lethargy, irritability and general seizures accompanied with profound hypoglycemia, in spite of aggressive medical treatment, she died because of sever congenital hyperinsulinism diazoxide unresponsive.


Asunto(s)
Hiperinsulinismo Congénito/diagnóstico , Convulsiones/etiología , Hiperinsulinismo Congénito/fisiopatología , Diazóxido/administración & dosificación , Resultado Fatal , Femenino , Humanos , Recién Nacido , Índice de Severidad de la Enfermedad
4.
Artículo en Inglés, Español | MEDLINE | ID: mdl-32448747

RESUMEN

BACKGROUND: Congenital hyperinsulinism (CHI) is a neuroendocrine disease with focal or diffuse abnormalities in pancreas. While drug-resistant diffuse forms require near-total pancreatectomy or prolonged pharmacotherapy, focal CHI may be treated by targeted surgical resection. We evaluated the usefulness of 18F-DOPA PET/CT to identify the focal pancreatic form. SUBJECTS AND METHODS: Nineteen children (11 boys, 8 girls, aged 2-54 months) with clinical signs of neonatal CHI and positive genetic examinations were enrolled in the study. After i.v. administration of 18F-DOPA, early PET and late PET/CT acquisition covering one-bed length over thoraco-abdominal region were performed. Both acquisitions were done in dynamic mode to allow exclusion of frames with motion artefacts. Standardized uptake values were adjusted to bodyweight (SUVbw). The finding was considered as focal when the ratio of SUVbwmax between the suspicious region and the rest of pancreas was greater than 1.2. RESULTS: Focal forms were recorded in 10/19 children and 4 of them underwent surgical resection with complete recovery. Focal uptake was significantly higher than the uptake in the normal pancreatic tissue (p=0.0059). Focal and diffuse forms of CHI did not differ significantly in normal pancreatic tissue uptake. We found no advantage in the measurement of SUVbwmean ratio compared to SUVbwmax ratio (p=0.50). CONCLUSION: 18F-DOPA PET/CT is a useful tool for the localization of focal CHI and planning of surgical treatment.


Asunto(s)
Hiperinsulinismo Congénito/diagnóstico por imagen , Dihidroxifenilalanina/análogos & derivados , Radioisótopos de Flúor , Pancreatectomía/métodos , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Radiofármacos , Cirugía Asistida por Computador , Preescolar , Terapia Combinada , Hiperinsulinismo Congénito/tratamiento farmacológico , Hiperinsulinismo Congénito/fisiopatología , Hiperinsulinismo Congénito/cirugía , Diazóxido/uso terapéutico , Dihidroxifenilalanina/farmacocinética , Femenino , Radioisótopos de Flúor/farmacocinética , Humanos , Lactante , Insulinoma/complicaciones , Insulinoma/cirugía , Masculino , Células Neuroendocrinas/metabolismo , Octreótido/uso terapéutico , Páncreas/fisiopatología , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/cirugía , Tomografía de Emisión de Positrones , Radiofármacos/farmacocinética , Estudios Retrospectivos , Sirolimus/uso terapéutico , Receptores de Sulfonilureas/genética , Distribución Tisular
5.
Eur J Pediatr ; 179(1): 39-50, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31840185

RESUMEN

Hypertrophic cardiomyopathy (HCM) in neonates is a rare and heterogeneous disorder which is characterized by hypertrophy of heart with histological and functional disruption of the myocardial structure/composition. The prognosis of HCM depends on the underlying diagnosis. In this review, we emphasize the importance to consider hyperinsulinism in the differential diagnosis of HCM, as hyperinsulinism is widely associated with cardiac hypertrophy (CH) which cannot be distinguished from HCM on echocardiographic examination. We supply an overview of the incidence and treatment strategies of neonatal CH in a broad spectrum of hyperinsulinemic diseases. Reviewing the literature, we found that CH is reported in 13 to 44% of infants of diabetic mothers, in approximately 40% of infants with congenital hyperinsulinism, in 61% of infants with leprechaunism and in 48 to 61% of the patients with congenital generalized lipodystrophy. The correct diagnosis is of importance since there is a large variation in prognoses and there are various strategies to treat CH in hyperinsulinemic diseases.Conclusion: The relationship between CH and hyperinsulism has implications for clinical practice as it might help to establish the correct diagnosis in neonates with cardiac hypertrophy which has both prognostic and therapeutic consequences. In addition, CH should be recognized as a potential comorbidity which might necessitate treatment in all neonates with known hyperinsulinism.What is Known:• Hyperinsulinism is currently not acknowledged as a cause of hypertrophic cardiomyopathy (HCM) in textbooks and recent Pediatric Cardiomyopathy Registry publications.What is New:• This article presents an overview of the literature of hyperinsulinism in neonates and infants showing that hyperinsulinism is associated with cardiac hypertrophy (CH) in a broad range of hyperinsulinemic diseases.• As CH cannot be distinguished from HCM on echocardiographic examination, we emphasize the importance to consider hyperinsulinism in the differential diagnosis of HCM/CH as establishing the correct diagnosis has both prognostic and therapeutic consequences.


Asunto(s)
Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/etiología , Hiperinsulinismo Congénito/complicaciones , Cardiomiopatía Hipertrófica/terapia , Hiperinsulinismo Congénito/diagnóstico , Hiperinsulinismo Congénito/fisiopatología , Diagnóstico Diferencial , Femenino , Humanos , Recién Nacido , Embarazo , Diagnóstico Prenatal , Pronóstico , Factores de Riesgo
6.
Clin Endocrinol (Oxf) ; 91(6): 770-775, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31520536

RESUMEN

OBJECTIVE: Diazoxide is first-line treatment for hyperinsulinaemic hypoglycaemia (HH) but diazoxide-induced pulmonary hypertension (PH) can occur. We aim to characterize the incidence and risk factors of diazoxide-induced PH in a large HH cohort to provide recommendations for anticipating and preventing PH in diazoxide-treated patients with HH. DESIGN AND PATIENTS: Retrospective cohort study involving four UK regional HH centres; review of case notes of HH patients on diazoxide. MEASUREMENTS: The diagnosis of PH was based on clinical and echocardiography evidence. Patient and treatment-related risk factors were analysed for association. RESULTS: Thirteen (6 men) of 177 HH diazoxide-treated patients developed PH, an incidence of 7%. In the PH group, HH was diagnosed at median (range) of 9 (1,180) days, with diazoxide commenced 4 (0,76) days from diagnosis and reaching a maximum dose of 7 (2.5,20) mg/kg/d. The majority (8 of 13 patients) developed PH within 2 weeks of diazoxide. Complete diazoxide withdrawal, but not dose reduction, led to PH resolution at 41 (3,959) days. In three patients, PH continued beyond 12 months. Risk factors for the development of PH included the presence of congenital heart disease (CHD) (P = .008), and total fluid volume exceeding 130 mL/kg/d in the immediate 24 hours preceding diazoxide (P = .019). CONCLUSION: Pulmonary hypertension can occur in 7% of diazoxide-treated HH patients. Risk factors include the presence of congenital heart disease and fluid overload. Recommendations include echocardiography and fluid restriction to 130 mL/kg/d prior to diazoxide treatment and immediate discontinuation of diazoxide if PH develops.


Asunto(s)
Hiperinsulinismo Congénito/tratamiento farmacológico , Hiperinsulinismo Congénito/fisiopatología , Diazóxido/efectos adversos , Diazóxido/uso terapéutico , Hipertensión Pulmonar/inducido químicamente , Hipoglucemia/fisiopatología , Hiperinsulinismo Congénito/genética , Ecocardiografía , Femenino , Edad Gestacional , Humanos , Hipertensión Pulmonar/genética , Hipoglucemia/genética , Masculino , Canales de Potasio de Rectificación Interna/genética , Estudios Retrospectivos , Factores de Riesgo , Receptores de Sulfonilureas/genética , Reino Unido
7.
Am J Med Genet C Semin Med Genet ; 181(4): 682-692, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31414570

RESUMEN

Congenital hyperinsulinism (HI) is the most frequent cause of persistent hypoglycemia in infants and children. Delays in diagnosis and initiation of appropriate treatment contribute to a high risk of neurocognitive impairment. HI represents a heterogeneous group of disorders characterized by dysregulated insulin secretion by the pancreatic beta cells, which in utero, may result in somatic overgrowth. There are at least nine known monogenic forms of HI as well as several syndromic forms. Molecular diagnosis allows for prediction of responsiveness to medical treatment and likelihood of surgically-curable focal hyperinsulinism. Timely genetic mutation analysis has thus become standard of care. However, despite significant advances in our understanding of the molecular basis of this disorder, the number of patients without an identified genetic diagnosis remains high, suggesting that there are likely additional genetic loci that have yet to be discovered.


Asunto(s)
Hiperinsulinismo Congénito/genética , Hiperinsulinismo Congénito/fisiopatología , Niño , Hiperinsulinismo Congénito/metabolismo , Epigénesis Genética , Pruebas Genéticas , Humanos , Lactante , Mutación , Philadelphia , Síndrome
8.
Saudi Med J ; 40(7): 669-674, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31287126

RESUMEN

OBJECTIVES: To report the genotype-phenotype characteristics, demographic features and clinical outcome of Omani patients with congenital hyperinsulinism (CHI). Methods: We retrospectively analyzed the clinical, biochemical, genotypical, phenotypical characteristics and outcomes of  children with CHI who were presented to the pediatric endocrine team in the Royal Hospital, Muscat, Oman between January 2007 and December 2016. Results: Analysis of 25 patients with CHI genetically revealed homozygous mutation in ABCC8 in 23 (92%) patients and 2 patients (8%) with compound heterozygous mutation in ABCC8. Fifteen (60%) patients underwent subtotal pancreatectomy as medical therapy failed and 2 (8%) patients showed response to medical therapy. Three patients expired during the neonatal period, 2 had cardiomyopathy and sepsis, and one had sepsis and severe metabolic acidosis. Out of the 15 patients who underwent pancreatectomy, 6 developed diabetes mellitus, 6 continued to have hypoglycemia and required medical therapy and one had pancreatic exocrine dysfunction post-pancreatectomy, following up with gastroenterology clinic and was placed on pancreatic enzyme supplements, while 2 patients continued to have hypoglycemia and both had abdominal MRI and 18-F-fluoro-L-DOPA positron emission tomography scan (PET-scan), that showed  persistent of the disease and started on medical therapy. Conclusion:  Mutation in ABCC8 is the most common cause of CHI and reflects the early age of presentation. There is a need for early diagnosis and appropriate therapeutic strategy.


Asunto(s)
Hiperinsulinismo Congénito/metabolismo , Hipoglucemia/metabolismo , Apnea/etiología , Apnea/fisiopatología , Preescolar , Hiperinsulinismo Congénito/genética , Hiperinsulinismo Congénito/fisiopatología , Hiperinsulinismo Congénito/terapia , Diabetes Mellitus/tratamiento farmacológico , Terapia de Reemplazo Enzimático , Insuficiencia Pancreática Exocrina/tratamiento farmacológico , Femenino , Fármacos Gastrointestinales/uso terapéutico , Heterocigoto , Homocigoto , Humanos , Hipoglucemia/complicaciones , Hipoglucemia/fisiopatología , Lactante , Recién Nacido , Letargia/etiología , Letargia/fisiopatología , Masculino , Mutación , Octreótido/uso terapéutico , Omán , Pancreatectomía , Péptidos Cíclicos/uso terapéutico , Complicaciones Posoperatorias/tratamiento farmacológico , Estudios Retrospectivos , Convulsiones/etiología , Convulsiones/fisiopatología , Sirolimus/uso terapéutico , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico , Receptores de Sulfonilureas/genética , Resultado del Tratamiento
9.
Eur J Pediatr ; 178(8): 1151-1160, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31243576

RESUMEN

Hyperinsulinaemic hypoglycaemia (HH) is a major cause of hypoglycaemia in the neonatal period, infancy and childhood. It is caused by unsuppressed insulin secretion in the setting of hypoglycaemia and carries a high risk of significant neurological sequelae, such as cognitive impairment. Genetic mutations have been implicated in the pathogenesis of the condition. Other causes include intra-uterine growth retardation, perinatal asphyxia, maternal diabetes mellitus and syndromes, such as Beckwith-Wiedemann. Based on the aetiology, the clinical presentation can range from absence of symptoms to the typical adrenergic symptoms and coma and even death. The diagnosis is based on biochemical findings and the gold-standard imaging technique is 18F-DOPA PET/CT scanning. Treatment options involve medications, such as diazoxide, nifedipine, glucagon and octreotide, as well as surgery. Novel treatment, such as long-acting octreotide, lanreotide and sirolimus, may be used as an alternative to pancreatectomy. Potential future medical treatments include exendin, a GLP-1 receptor antagonist, and glucagon infusion via a pump.Conclusion: Advances in the fields of genetic testing, imaging techniques and medical treatment are beginning to provide novel insights into earlier detection, less invasive treatment approaches and fewer complications associated with the complex entity of hyperinsulinaemic hypoglycaemia. What is Known: • HH is caused by dysregulated insulin release from the ß cell due to genetic mutations and carries a risk for complications, such as neurocognitive impairment. 18F-DOPA PET/CT scanning is presented as the gold-standard imaging technique currently in children with hyperinsulinaemic hypoglycaemia. • Clinical presentation is heterogeneous and treatment options include medical therapy and pancreatectomy. What is New: • 18F-DOPA PET/CT is indicated in suspected focal CHI due to paternal transmitted mutations in ABCC8 or KCNJ11. • Novel treatment options have been introduced, such as long-acting octreotide, lanreotide, sirolimus and selective nonpeptide somatostatin receptor subtype 5 (SSTR5) agonists. Future medical treatments include exendin, a GLP-1 antagonist, and glucagon infusion via a pump. However, all these options are off-label at present.


Asunto(s)
Hiperinsulinismo Congénito , Niño , Preescolar , Terapia Combinada , Hiperinsulinismo Congénito/diagnóstico , Hiperinsulinismo Congénito/etiología , Hiperinsulinismo Congénito/fisiopatología , Hiperinsulinismo Congénito/terapia , Marcadores Genéticos , Pruebas Genéticas , Humanos , Lactante , Recién Nacido
10.
Paediatr Drugs ; 21(3): 123-136, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31218604

RESUMEN

Congenital hyperinsulinism (CHI) is the most common cause of persistent hypoglycmia in neonatles and children. The inappropriate secretion of insulin by the pancreatic ß-cells produces recurrent hypoglycemia, which can lead to severe and permanent brain damage. CHI results from mutations in different genes that play a role in the insulin secretion pathway, and each differs in their responsiveness to medical treatment. Currently, the only available approved treatment for hyperinsulinism is diazoxide. Patients unresponsive to diazoxide may benefit from specialized evaluation including genetic testing and 18F-DOPA PET to identify those with focal forms of CHI. The focal forms can be cured by selective pancreatectomy, but the management of diazoxide-unresponsive diffuse CHI is a real therapeutic challenge. Current off-label therapies include intravenous glucagon, octreotide and long-acting somatostatin analogs; however, they are often insufficient, and a 98% pancreatectomy or continuous feeds may be required. For the first time in over 40 years, new drugs are being developed, but none have made it to market yet. In this review, we will discuss current on-label and off-label drugs and review the currently available data on the novel drugs under development.


Asunto(s)
Hiperinsulinismo Congénito/complicaciones , Hipoglucemia/tratamiento farmacológico , Niño , Hiperinsulinismo Congénito/tratamiento farmacológico , Hiperinsulinismo Congénito/patología , Hiperinsulinismo Congénito/fisiopatología , Humanos
12.
Genet Med ; 21(1): 233-242, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29907798

RESUMEN

PURPOSE: Describe the clinical and molecular findings of patients with Kabuki syndrome (KS) who present with hypoglycemia due to congenital hyperinsulinism (HI), and assess the incidence of KS in patients with HI. METHODS: We documented the clinical features and molecular diagnoses of 9 infants with persistent HI and KS via a combination of sequencing and copy-number profiling methodologies. Subsequently, we retrospectively evaluated 100 infants with HI lacking a genetic diagnosis, for causative variants in KS genes. RESULTS: Molecular diagnoses of KS were established by identification of pathogenic variants in KMT2D (n = 5) and KDM6A (n = 4). Among the 100 infants with HI of unknown genetic etiology, a KS diagnosis was uncovered in one patient. CONCLUSIONS: The incidence of HI among patients with KS may be higher than previously reported, and KS may account for as much as 1% of patients diagnosed with HI. As the recognition of dysmorphic features associated with KS is challenging in the neonatal period, we propose KS should be considered in the differential diagnosis of HI. Since HI in patients with KS is well managed medically, a timely recognition of hyperinsulinemic episodes will improve outcomes, and prevent aggravation of the preexisting mild to moderate intellectual disability in KS.


Asunto(s)
Anomalías Múltiples/genética , Hiperinsulinismo Congénito/genética , Proteínas de Unión al ADN/genética , Cara/anomalías , Enfermedades Hematológicas/genética , Histona Demetilasas/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Enfermedades Vestibulares/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/fisiopatología , Preescolar , Hiperinsulinismo Congénito/complicaciones , Hiperinsulinismo Congénito/diagnóstico , Hiperinsulinismo Congénito/fisiopatología , Cara/fisiopatología , Femenino , Predisposición Genética a la Enfermedad , Enfermedades Hematológicas/complicaciones , Enfermedades Hematológicas/diagnóstico , Enfermedades Hematológicas/fisiopatología , Humanos , Lactante , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Masculino , Mutación , Patología Molecular , Estudios Retrospectivos , Enfermedades Vestibulares/complicaciones , Enfermedades Vestibulares/diagnóstico , Enfermedades Vestibulares/fisiopatología
13.
Diabet Med ; 36(1): 9-21, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30246418

RESUMEN

Congenital hyperinsulinism is a rare disease, but is the most frequent cause of persistent and severe hypoglycaemia in early childhood. Hypoglycaemia caused by excessive and dysregulated insulin secretion (hyperinsulinism) from disordered pancreatic ß cells can often lead to irreversible brain damage with lifelong neurodisability. Although congenital hyperinsulinism has a genetic cause in a significant proportion (40%) of children, often being the result of mutations in the genes encoding the KATP channel (ABCC8 and KCNJ11), not all children have severe and persistent forms of the disease. In approximately half of those without a genetic mutation, hyperinsulinism may resolve, although timescales are unpredictable. From a histopathology perspective, congenital hyperinsulinism is broadly grouped into diffuse and focal forms, with surgical lesionectomy being the preferred choice of treatment in the latter. In contrast, in diffuse congenital hyperinsulinism, medical treatment is the best option if conservative management is safe and effective. In such cases, children receiving treatment with drugs, such as diazoxide and octreotide, should be monitored for side effects and for signs of reduction in disease severity. If hypoglycaemia is not safely managed by medical therapy, subtotal pancreatectomy may be required; however, persistent hypoglycaemia may continue after surgery and diabetes is an inevitable consequence in later life. It is important to recognize the negative cognitive impact of early-life hypoglycaemia which affects half of all children with congenital hyperinsulinism. Treatment options should be individualized to the child/young person with congenital hyperinsulinism, with full discussion regarding efficacy, side effects, outcomes and later life impact.


Asunto(s)
Antihipertensivos/uso terapéutico , Hiperinsulinismo Congénito/complicaciones , Hiperinsulinismo Congénito/terapia , Diazóxido/uso terapéutico , Fármacos Gastrointestinales/uso terapéutico , Glucagón/uso terapéutico , Hipoglucemia/etiología , Hipoglucemia/terapia , Antihipertensivos/efectos adversos , Hiperinsulinismo Congénito/fisiopatología , Diazóxido/efectos adversos , Humanos , Hipoglucemia/fisiopatología , Pancreatectomía/métodos , Canales de Potasio de Rectificación Interna/genética , Medicina de Precisión , Receptores de Sulfonilureas/efectos de los fármacos , Receptores de Sulfonilureas/genética , Resultado del Tratamiento
14.
Horm Res Paediatr ; 91(3): 216-220, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30092575

RESUMEN

INTRODUCTION: Post-prandial hyperinsulinaemic hypoglycaemia (PPHH) is a recognized complication of various gastric surgeries in children, but rarely reported after oesophageal atresia repair. We report 2 children diagnosed with PPHH after oesophageal surgery and the challenges of their management. Case 1: A 2-year-old boy diagnosed with oesophageal atresia at birth was surgically repaired requiring 6 oesophageal dilatations in the first year of life. At 11 months of age, he manifested hypoglycaemic seizures and investigations confirmed PPHH. Acarbose and diazoxide trials failed. He was managed with 17-h continuous gastrostomy feeds. Currently, he is 28 months old with euglycaemia on daytime bolus gastrostomy feeds and overnight 12-h continuous gastrostomy feeds. Case 2: A 6-month-old girl diagnosed with Wolf-Hirschhorn syndrome and tracheo-oesophageal fistula was surgically repaired, requiring monthly oesophageal dilatations. At 5 months of age, she was reported to have hypoglycaemia and PPHH was confirmed. She responded to diazoxide and continuous nasogastric tube feeds, but developed pulmonary hypertension pos-sibly diazoxide-induced. Subsequently, diazoxide was stopped and normoglycaemia was secured via 20-h continuous gastrostomy feeds. CONCLUSION: PPHH may be an underdiagnosed complication in children undergoing surgery for oesophageal atresia. These children must be monitored closely for symptoms of hypoglycaemia and if there are concerns must be screened for possible PPHH. Our cases demonstrate that continuous feeding regimens might be the only therapeutic option, until PPHH gradually lessens in intensity over time.


Asunto(s)
Hiperinsulinismo Congénito , Nutrición Enteral , Atresia Esofágica , Gastrostomía , Complicaciones Posoperatorias , Periodo Posprandial , Preescolar , Hiperinsulinismo Congénito/diagnóstico , Hiperinsulinismo Congénito/etiología , Hiperinsulinismo Congénito/fisiopatología , Atresia Esofágica/fisiopatología , Atresia Esofágica/cirugía , Femenino , Humanos , Lactante , Masculino , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/fisiopatología
15.
Pediatr Diabetes ; 19(2): 259-264, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28597971

RESUMEN

OBJECTIVES: To define the ranges of biochemical markers during hypoglycemia for the diagnosis of congenital hyperinsulinism (CHI), using high sensitivity insulin assays. SUBJECTS: A total of 298 patients with CHI and 58 control patients with non-hyperinsulinemic hypoglycemia, who were diagnosed after 2007. METHODS: The levels of biochemical markers (glucose, insulin, ß-hydroxybutyrate [BHB], free fatty acids [FFA], lactate, ammonia) at the time of hypoglycemia were analyzed along with the maximal glucose infusion rate (GIR) to maintain euglycemia and clinical outcomes. RESULTS: Median levels of blood glucose in patients with CHI and in controls were 30 and 46 mg/dL, while insulin levels were 9.90 and undetectable (<.5) µU/mL, respectively. Similarly, median levels of BHB were 17.5 and 3745 µmol/L, and those of FFA were 270.5 and 2660 µmol/L, respectively. For patients after 5 months, cutoffs of insulin >1.25 µU/mL, BHB < 2000 µmol/L, and FFA < 1248 µmol/L predicted CHI with sensitivities of 97.5, 96.2, and 95.2% and specificities of 84.2, 89.3, and 92.3%, respectively. Maximal GIR in the CHI groups tended to decrease with age. In addition, decreased gestational age, low birth weight, and elevated lactate at hypoglycemia were significantly more common in patients who were off treatment within 100 days without pancreatectomy. CONCLUSIONS: After introduction of high-sensitive assays, the diagnostic value of insulin was improved, allowing for more efficient cutoffs to be set for diagnosis of CHI. Premature birth, low birth weight and elevated lactate might be helpful in predicting early remission of hypoglycemia.


Asunto(s)
Hiperinsulinismo Congénito/diagnóstico , Hiperamonemia/etiología , Hiperlactatemia/etiología , Hipoglucemia/etiología , Ácido 3-Hidroxibutírico/sangre , Biomarcadores/sangre , Niño , Preescolar , Hiperinsulinismo Congénito/sangre , Hiperinsulinismo Congénito/etiología , Hiperinsulinismo Congénito/fisiopatología , Ácidos Grasos no Esterificados/sangre , Femenino , Encuestas Epidemiológicas , Hospitales Generales , Humanos , Lactante , Recién Nacido de Bajo Peso , Recién Nacido , Japón , Masculino , Nacimiento Prematuro/fisiopatología , Derivación y Consulta , Remisión Espontánea , Sensibilidad y Especificidad
16.
Horm Res Paediatr ; 89(1): 1-6, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29151084

RESUMEN

BACKGROUND: Congenital hyperinsulinism (CHI) is hallmarked by persistent hypoketotic hypoglycemia in infancy. In the majority of all patients, CHI is caused by mutations in the KATP channel genes ABCC8 and KCNJ11, but other genes in the insulin-regulatory pathway have also been described. Repeated episodes of hypoglycemia include an increased risk of seizures and intellectual disability. So far, controlled psychometric studies on cognitive, motor, speech, and social-emotional outcome of CHI patients are missing. Until now, neurodevelopmental long-term outcome in CHI patients has only been measured by questionnaires, self-, parental-, or caregiver-administered instruments. METHODS: This is a prospective study of 60 patients (median age 3.3 years, range 3 months to 57 years): 48 with a diffuse, 9 with a focal, and 3 with an atypical histology. Neurodevelopmental outcome was assessed using standardized psychological tests and questionnaires. RESULTS: 28 of 60 patients showed developmental delay (46.7%). 9 of 57 patients had cognitive deficits (15.8%), 7 of 26 patients had speech problems (26.9%), and 17 of 44 patients had motor problems (38.6%). In 5 of 53 patients, social-emotional problems were reported. Outcome and the underlying genetic defect were not correlated. CONCLUSIONS: Motor problems seem to be prominent in CHI patients. Despite a high incidence of developmental delay, a permanent cognitive defect was only detectable in 9 of 58 patients.


Asunto(s)
Cognición , Disfunción Cognitiva , Hiperinsulinismo Congénito , Trastornos Motores , Trastornos del Habla , Adolescente , Adulto , Niño , Preescolar , Disfunción Cognitiva/epidemiología , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , Hiperinsulinismo Congénito/epidemiología , Hiperinsulinismo Congénito/fisiopatología , Hiperinsulinismo Congénito/psicología , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Trastornos Motores/epidemiología , Trastornos Motores/fisiopatología , Trastornos Motores/psicología , Estudios Prospectivos , Trastornos del Habla/epidemiología , Trastornos del Habla/fisiopatología , Trastornos del Habla/psicología
17.
Ann N Y Acad Sci ; 1411(1): 65-82, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29044608

RESUMEN

Pancreatic beta cells sense changes in nutrients during the cycles of fasting and feeding and release insulin accordingly to maintain glucose homeostasis. Abnormal beta cell nutrient sensing resulting from gene mutations leads to hypoglycemia or diabetes. Glucokinase (GCK) plays a key role in beta cell glucose sensing. As one form of congenital hyperinsulinism (CHI), activating mutations of GCK result in a decreased threshold for glucose-stimulated insulin secretion and hypoglycemia. In contrast, inactivating mutations of GCK result in diabetes, including a mild form (MODY2) and a severe form (permanent neonatal diabetes mellitus (PNDM)). Mutations of beta cell ion channels involved in insulin secretion regulation also alter glucose sensing. Activating or inactivating mutations of ATP-dependent potassium (KATP ) channel genes result in severe but completely opposite clinical phenotypes, including PNDM and CHI. Mutations of the other ion channels, including voltage-gated potassium channels (Kv 7.1) and voltage-gated calcium channels, also lead to abnormal glucose sensing and CHI. Furthermore, amino acids can stimulate insulin secretion in a glucose-independent manner in some forms of CHI, including activating mutations of the glutamate dehydrogenase gene, HDAH deficiency, and inactivating mutations of KATP channel genes. These genetic defects have provided insight into a better understanding of the complicated nature of beta cell fuel-sensing mechanisms.


Asunto(s)
Hiperinsulinismo Congénito/fisiopatología , Diabetes Mellitus/fisiopatología , Glucoquinasa/fisiología , Canales Iónicos/fisiología , Islotes Pancreáticos/fisiología , Nutrientes/farmacocinética , 3-Hidroxiacil-CoA Deshidrogenasas/deficiencia , Aminoácidos/farmacocinética , Animales , Glucemia/metabolismo , Hiperinsulinismo Congénito/genética , Diabetes Mellitus/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Ácidos Grasos/metabolismo , Glucoquinasa/deficiencia , Glucoquinasa/genética , Glucosa/farmacocinética , Humanos , Insulina/fisiología , Canales Iónicos/genética , Errores Innatos del Metabolismo/fisiopatología , Ratones , Ratones Noqueados , Mutación , Oxidación-Reducción
18.
J Pediatr Endocrinol Metab ; 30(12): 1311-1315, 2017 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-29127764

RESUMEN

BACKGROUND: Congenital hyperinsulinism (CHI) is frequently caused by mutations in one of the KATP channel subunits encoded by the genes ABCC8 and KCNJ11. The effect of simultaneous mutations in both of these genes on the pancreatic ß-cell function is not known and patients with CHI carrying both ABCC8 and KCNJ11 mutations have not yet been reported. We questioned if a combination of heterozygous mutations in the ABCC8 and KCNJ11 genes could also lead to ß-cell dysfunction presenting as CHI. METHODS: As a model, we used a patient with transient CHI that paternally inherited novel heterozygous mutations in ABCC8 (p.Tyr1293Asp) and KCNJ11 (p.Arg50Trp) genes. The pathogenic effects on the pancreatic ß-cells function were examined in an in vitro functional study using radioactive rubidium efflux assay. RESULTS: We showed that the activation of the mutated KATP channels by diazoxide was decreased by 60.9% in the channels with the heterozygous combination of both mutations compared to the wild type channels. This could indicate the pathogenic effect on the pancreatic ß-cell function leading to CHI although conclusive evidence is needed to be added. CONCLUSIONS: Our findings may widen the spectrum of genetic causes of CHI and suggest a novel pathogenic mechanism of CHI that must however, be further investigated.


Asunto(s)
Hiperinsulinismo Congénito/genética , Canales de Potasio de Rectificación Interna/genética , Receptores de Sulfonilureas/genética , Adulto , Anciano , Niño , Preescolar , Hiperinsulinismo Congénito/fisiopatología , Epistasis Genética/fisiología , Familia , Heterocigoto , Humanos , Recién Nacido , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Masculino , Persona de Mediana Edad , Herencia Multifactorial/genética , Mutación , Linaje
19.
Pediatr Diabetes ; 18(7): 532-539, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27681997

RESUMEN

BACKGROUND: There are few reports pertaining to Asian patients with neonatal diabetes mellitus (NDM) caused by activating mutations in the ATP-sensitive potassium channel genes (KATP-NDM). OBJECTIVES: To elucidate the characteristics of Japanese patients with KATP-NDM. METHODS: By the amplification and direct sequencing of all exons and exon-intron boundaries of the KCNJ11 and ABCC8 genes, 25 patients with KATP-NDM were identified from a total of 70 patients with NDM. Clinical data were collected from the medical charts. RESULTS: Sixteen patients had mutations in KCNJ11 and nine in ABCC8. Eight novel mutations were identified; two in KCNJ11 (V64M, R201G) and six in ABCC8 (R216C, G832C, F1176L, A1263V, I196N, T229N). Interestingly, V64M caused DEND (developmental delay, epilepsy, neonatal diabetes) syndrome in our patient, while mutation of the same residue (V64G) had been reported to cause congenital hyperinsulinism. Mutations in ABCC8 were associated with TNDM (4/9) or isolated PNDM (5/9), whereas those in KCNJ11 were associated with more severe phenotypes, including DEND (3/16), iDEND (intermediate DEND, 4/16), or isolated PNDM (6/16). Switching from insulin to glibenclamide monotherapy was successful in 87.5% of the patients. Neurological improvement was observed in two patients, one with DEND (T293N) and one with iDEND (R50P) syndrome. Three others with iDEND mutations (R201C, G53D, and V59M) remained neurologically normal at 5, 1, and 4 years of age, respectively, with early introduction of sulfonylurea. CONCLUSION: Overall, clinical presentation of KATP-NDM in Japanese patients was similar to those of other populations. Early introduction of sulfonylurea appeared beneficial in ameliorating neurological symptoms.


Asunto(s)
Diabetes Mellitus/genética , Predisposición Genética a la Enfermedad , Mutación , Canales de Potasio de Rectificación Interna/genética , Receptores de Sulfonilureas/genética , Sustitución de Aminoácidos , Hiperinsulinismo Congénito/sangre , Hiperinsulinismo Congénito/tratamiento farmacológico , Hiperinsulinismo Congénito/genética , Hiperinsulinismo Congénito/fisiopatología , Análisis Mutacional de ADN , Diabetes Mellitus/sangre , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/fisiopatología , Monitoreo de Drogas , Resistencia a Medicamentos , Epilepsia/sangre , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Epilepsia/fisiopatología , Femenino , Estudios de Asociación Genética , Gliburida/uso terapéutico , Humanos , Hiperglucemia/prevención & control , Hipoglucemia/prevención & control , Hipoglucemiantes/uso terapéutico , Lactante , Enfermedades del Recién Nacido/sangre , Enfermedades del Recién Nacido/tratamiento farmacológico , Enfermedades del Recién Nacido/genética , Enfermedades del Recién Nacido/fisiopatología , Insulina/uso terapéutico , Japón , Masculino , Canales de Potasio de Rectificación Interna/química , Trastornos Psicomotores/sangre , Trastornos Psicomotores/tratamiento farmacológico , Trastornos Psicomotores/genética , Trastornos Psicomotores/fisiopatología , Índice de Severidad de la Enfermedad , Receptores de Sulfonilureas/química
20.
Indian Pediatr ; 53(10): 912-913, 2016 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-27771675

RESUMEN

BACKGROUND: Hyperinsulinemia is the commonest cause of persistent hypoglycemia in infancy. Inactivating mutations in the genes ABCC8 and KCNJ11 are the commonest cause. Mutation in the HADH gene, which encodes the short-chain-L-3-hydroxyacyl-CoA dehydrogenase, is a rare cause. CASE CHARACTERISTICS: Two Indian sisters who presented with hyperinsulinemic hypoglycemia of infancy. OBSERVATION/INTERVENTION: A novel homozygous missense mutation in the HADH gene was identified in both the sisters, while the parents were found to be heterozygous carriers. OUTCOME: Establishment of molecular diagnosis, optimization of therapy and counseling of parents regarding risk of recurrence in future pregnancy. MESSAGE: HADH mutations are rare causes of hypoglycemia and can be mitigated with diazoxide and appropriate dietary therapy if identified early.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/genética , Hiperinsulinismo Congénito , Preescolar , Hiperinsulinismo Congénito/diagnóstico , Hiperinsulinismo Congénito/genética , Hiperinsulinismo Congénito/fisiopatología , Femenino , Humanos , Lactante , Mutación/genética , Hermanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...