Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
AACE Clin Case Rep ; 9(4): 101-103, 2023.
Article in English | MEDLINE | ID: mdl-37520758

ABSTRACT

Background: Identifying cases of diabetes caused by single gene mutations between the more common type 1 diabetes (T1D) and type 2 diabetes (T2D) is a difficult but important task. We report the diagnosis of ATP-binding cassette transporter sub-family C member 8 (ABCC8)-related monogenic diabetes in a 35-year-old woman with a protective human leukocyte antigen (HLA) allele who was originally diagnosed with T1D at 18 years of age. Case Report: Patient A presented with polyuria, polydipsia, and hypertension at the age of 18 years and was found to have a blood glucose > 500 mg/dL (70-199 mg/dL) and an HbA1C (hemoglobin A1C) >14% (4%-5.6%). She had an unmeasurable C-peptide but no urine ketones. She was diagnosed with T1D and started on insulin therapy. Antibody testing was negative. She required low doses of insulin and later had persistence of low but detectable C-peptide. At the age of 35 years, she was found to have a protective HLA allele, and genetic testing revealed a pathogenic mutation in the ABCC8 gene. The patient was then successfully transitioned to sulfonylurea therapy. Discussion: Monogenic diabetes diagnosed in adolescence typically presents with mild to moderate hyperglycemia, positive family history and, in some cases, other organ findings or dysfunction. The patient in this report presented with very high blood glucose, prompting the diagnosis of T1D. When she was found to have a protective HLA allele, further investigation revealed the mutation in the sulfonylurea receptor gene, ABCC8. Conclusion: Patients suspected of having T1D but with atypical clinical characteristics such as negative autoantibodies, low insulin requirements, and persistence of C-peptide should undergo genetic testing for monogenic diabetes.

2.
Sci Data ; 10(1): 323, 2023 05 26.
Article in English | MEDLINE | ID: mdl-37237059

ABSTRACT

The Network for Pancreatic Organ donors with Diabetes (nPOD) is the largest biorepository of human pancreata and associated immune organs from donors with type 1 diabetes (T1D), maturity-onset diabetes of the young (MODY), cystic fibrosis-related diabetes (CFRD), type 2 diabetes (T2D), gestational diabetes, islet autoantibody positivity (AAb+), and without diabetes. nPOD recovers, processes, analyzes, and distributes high-quality biospecimens, collected using optimized standard operating procedures, and associated de-identified data/metadata to researchers around the world. Herein describes the release of high-parameter genotyping data from this collection. 372 donors were genotyped using a custom precision medicine single nucleotide polymorphism (SNP) microarray. Data were technically validated using published algorithms to evaluate donor relatedness, ancestry, imputed HLA, and T1D genetic risk score. Additionally, 207 donors were assessed for rare known and novel coding region variants via whole exome sequencing (WES). These data are publicly-available to enable genotype-specific sample requests and the study of novel genotype:phenotype associations, aiding in the mission of nPOD to enhance understanding of diabetes pathogenesis to promote the development of novel therapies.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Tissue Donors , Humans , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Genomics , Pancreas
3.
Diabetes Care ; 44(8): 1816-1825, 2021 08.
Article in English | MEDLINE | ID: mdl-34172489

ABSTRACT

OBJECTIVE: Multiple genome-wide association studies have identified a strong genetic linkage between the SKAP2 locus and type 1 diabetes (T1D), but how this leads to disease remains obscure. Here, we characterized the functional consequence of a novel SKAP2 coding mutation in a patient with T1D to gain further insight into how this impacts immune tolerance. RESEARCH DESIGN AND METHODS: We identified a 24-year-old individual with T1D and other autoimmune and inflammatory conditions. The proband and first-degree relatives were recruited for whole-exome sequencing. Functional studies of the protein variant were performed using a cell line and primary myeloid immune cells collected from family members. RESULTS: Sequencing identified a de novo SKAP2 variant (c.457G>A, p.Gly153Arg) in the proband. Assays using monocyte-derived macrophages from the individual revealed enhanced activity of integrin pathways and a migratory phenotype in the absence of chemokine stimulation, consistent with SKAP2 p.Gly153Arg being constitutively active. The p.Gly153Arg variant, located in the well-conserved lipid-binding loop, induced similar phenotypes when expressed in a human macrophage cell line. SKAP2 p.Gly153Arg is a gain-of-function, pathogenic mutation that disrupts myeloid immune cell function, likely resulting in a break in immune tolerance and T1D. CONCLUSIONS: SKAP2 plays a key role in myeloid cell activation and migration. This particular mutation in a patient with T1D and multiple autoimmune conditions implicates a role for activating SKAP2 variants in autoimmune T1D.


Subject(s)
Autoimmune Diseases , Diabetes Mellitus, Type 1 , Intracellular Signaling Peptides and Proteins , Adult , Diabetes Mellitus, Type 1/genetics , Genome-Wide Association Study , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Phenotype , Young Adult
4.
Sci Transl Med ; 12(541)2020 04 29.
Article in English | MEDLINE | ID: mdl-32350130

ABSTRACT

Pregnancy imposes a substantial metabolic burden on women through weight gain and insulin resistance. Lactation reduces the risk of maternal postpartum diabetes, but the mechanisms underlying this benefit are unknown. Here, we identified long-term beneficial effects of lactation on ß cell function, which last for years after the cessation of lactation. We analyzed metabolic phenotypes including ß cell characteristics in lactating and non-lactating humans and mice. Lactating and non-lactating women showed comparable glucose tolerance at 2 months after delivery, but after a mean of 3.6 years, glucose tolerance in lactated women had improved compared to non-lactated women. In humans, the disposition index, a measure of insulin secretory function of ß cells considering the degree of insulin sensitivity, was higher in lactated women at 3.6 years after delivery. In mice, lactation improved glucose tolerance and increased ß cell mass at 3 weeks after delivery. Amelioration of glucose tolerance and insulin secretion were maintained up to 4 months after delivery in lactated mice. During lactation, prolactin induced serotonin production in ß cells. Secreted serotonin stimulated ß cell proliferation through serotonin receptor 2B in an autocrine and paracrine manner. In addition, intracellular serotonin acted as an antioxidant to mitigate oxidative stress and improved ß cell survival. Together, our results suggest that serotonin mediates the long-term beneficial effects of lactation on female metabolic health by increasing ß cell proliferation and reducing oxidative stress in ß cells.


Subject(s)
Insulin-Secreting Cells , Lactation , Animals , Blood Glucose , Breast Feeding , Female , Humans , Insulin , Mice , Serotonin
6.
Diabetes ; 69(2): 205-214, 2020 02.
Article in English | MEDLINE | ID: mdl-31806625

ABSTRACT

A sufficient ß-cell mass is crucial for preventing diabetes, and perinatal ß-cell proliferation is important in determining the adult ß-cell mass. However, it is not yet known how perinatal ß-cell proliferation is regulated. Here, we report that serotonin regulates ß-cell proliferation through serotonin receptor 2B (HTR2B) in an autocrine/paracrine manner during the perinatal period. In ß-cell-specific Tph1 knockout (Tph1 ßKO) mice, perinatal ß-cell proliferation was reduced along with the loss of serotonin production in ß-cells. Adult Tph1 ßKO mice exhibited glucose intolerance with decreased ß-cell mass. Disruption of Htr2b in ß-cells also resulted in decreased perinatal ß-cell proliferation and glucose intolerance in adulthood. Growth hormone (GH) was found to induce serotonin production in ß-cells through activation of STAT5 during the perinatal period. Thus, our results indicate that GH-GH receptor-STAT5-serotonin-HTR2B signaling plays a critical role in determining the ß-cell mass by regulating perinatal ß-cell proliferation, and defects in this pathway affect metabolic phenotypes in adults.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/physiology , Serotonin/metabolism , Animals , Animals, Newborn , Cell Proliferation , Female , Growth Hormone/metabolism , Humans , Infant , Mice , Mice, Knockout , Pregnancy , Propafenone/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
7.
JCI Insight ; 5(1)2020 01 16.
Article in English | MEDLINE | ID: mdl-31805014

ABSTRACT

Biallelic mutations of the gene encoding the transcription factor NEUROG3 are associated with a rare disorder that presents in neonates as generalized malabsorption - due to a complete absence of enteroendocrine cells - followed, in early childhood or beyond, by insulin-dependent diabetes mellitus (IDDM). The commonly delayed onset of IDDM suggests a differential requirement for NEUROG3 in endocrine cell generation in the human pancreas versus the intestine. However, previously identified human mutations were hypomorphic and, hence, may have had residual function in pancreas. We report 2 patients with biallelic functionally null variants of the NEUROG3 gene who nonetheless did not present with IDDM during infancy but instead developed permanent IDDM during middle childhood ages. The variants showed no evidence of function in traditional promoter-based assays of NEUROG3 function and also failed to exhibit function in a variety of potentially novel in vitro and in vivo molecular assays designed to discern residual NEUROG3 function. These findings imply that, unlike in mice, pancreatic endocrine cell generation in humans is not entirely dependent on NEUROG3 expression and, hence, suggest the presence of unidentified redundant in vivo pathways in human pancreas capable of yielding ß cell mass sufficient to maintain euglycemia until early childhood.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Diabetes Mellitus/genetics , Genetic Predisposition to Disease , Loss of Function Mutation , Nerve Tissue Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Child , Diabetes Mellitus, Type 1 , Enteroendocrine Cells/metabolism , Female , Gene Expression Regulation , Helix-Loop-Helix Motifs/genetics , Humans , Insulin-Secreting Cells/metabolism , Islets of Langerhans , Malabsorption Syndromes , Male , Nerve Tissue Proteins/metabolism , Pancreas , Promoter Regions, Genetic
8.
J Clin Invest ; 129(11): 4676-4681, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31369399

ABSTRACT

While improvements in genetic analysis have greatly enhanced our understanding of the mechanisms behind pancreatitis, it continues to afflict many families for whom the hereditary factors remain unknown. Recent evaluation of a patient with a strong family history of pancreatitis sparked us to reexamine a large kindred originally reported over 50 years ago with an autosomal dominant inheritance pattern of chronic pancreatitis, diabetes and pancreatic adenocarcinoma. Whole exome sequencing analysis identified a rare missense mutation in the gene encoding pancreas-specific protease Elastase 3B (CELA3B) that cosegregates with disease. Studies of the mutant protein in vitro, in cell lines and in CRISPR-Cas9 engineered mice indicate that this mutation causes translational upregulation of CELA3B, which upon secretion and activation by trypsin leads to uncontrolled proteolysis and recurrent pancreatitis. Although lesions in several other pancreatitic proteases have been previously linked to hereditary pancreatitis, this is the first known instance of a mutation in CELA3B and a defect in translational control contributing to this disease.


Subject(s)
Adenocarcinoma/genetics , Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease , Mutation , Neoplasm Proteins/genetics , Pancreatic Elastase/genetics , Pancreatic Neoplasms/genetics , Pancreatitis/genetics , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Animals , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Diseases, Inborn/enzymology , Genetic Diseases, Inborn/pathology , Humans , Mice , Neoplasm Proteins/metabolism , Pancreatic Elastase/biosynthesis , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Pancreatitis/enzymology , Pancreatitis/pathology , Up-Regulation , Exome Sequencing , Pancreatic Neoplasms
9.
Environ Sci Technol ; 53(3): 1078-1086, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30620879

ABSTRACT

Although unknown 25 years ago, natural arsenic contamination of groundwater affects over 50 countries and up to 200 million people. The economic viability was analyzed and modeled of eighty-eight community-based arsenic mitigation systems existing for up to 20 years in India and Bangladesh. The performances of three community-based arsenic mitigation systems that are ethnically different and separated across two different countries were monitored closely for 24 months of self-sustainable, long-term operation at WHO standards through local, paid caretakers. Based on data from the use of hybrid ion exchange materials (HIX-Nano) and the broad set of field operations, Monte Carlo simulations were used to explore the conditions required for self-sustainable operation and job creation in low-income communities (<$2/day/capita). The results from field data and cost modeling provided clear evidence of economic growth and job creation for systems managed by villagers' committee through collection of monthly tariffs. Ethnicity and religion did not have perceptible impacts on day-to-day operations or cumulative long-term revenue. The cost of the treatment technology (i.e., HIX-Nano) had minimal impact on the operational profitability, while number of customers and water delivery significantly affected profitability. Local employment generation with income significantly higher than poverty level was the most enduring outcome and led to enhanced sustainability.


Subject(s)
Arsenic , Water Pollutants, Chemical , Bangladesh , Developing Countries , India , Small Business , Water Supply
10.
Mol Cancer Ther ; 17(12): 2702-2709, 2018 12.
Article in English | MEDLINE | ID: mdl-30254185

ABSTRACT

Patients with pancreatic neuroendocrine tumors (PNET) commonly develop advanced disease and require systemic therapy. However, treatment options remain limited, in part, because experimental models that reliably emulate PNET disease are lacking. We therefore developed a patient-derived xenograft model of PNET (PDX-PNET), which we then used to evaluate two mTOR inhibitor drugs: FDA-approved everolimus and the investigational new drug sapanisertib. PDX-PNETs maintained a PNET morphology and PNET-specific gene expression signature with serial passage. PDX-PNETs also harbored mutations in genes previously associated with PNETs (such as MEN1 and PTEN), displayed activation of the mTOR pathway, and could be detected by Gallium-68 DOTATATE PET-CT. Treatment of PDX-PNETs with either everolimus or sapanisertib strongly inhibited growth. As seen in patients, some PDX-PNETs developed resistance to everolimus. However, sapanisertib, a more potent inhibitor of the mTOR pathway, caused tumor shrinkage in most everolimus-resistant tumors. Our PDX-PNET model is the first available, validated PDX model for PNET, and preclinical data from the use of this model suggest that sapanisertib may be an effective new treatment option for patients with PNET or everolimus-resistant PNET.


Subject(s)
Benzoxazoles/therapeutic use , Drug Resistance, Neoplasm , Everolimus/therapeutic use , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Mice, Nude , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/pathology , Organometallic Compounds/chemistry , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Positron Emission Tomography Computed Tomography , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
11.
Endocrinology ; 159(9): 3321-3330, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30059978

ABSTRACT

Insulin production by the pancreatic ß cell is critical for the glucose homeostasis of the whole organism. Although the transcription factors required for insulin production are known, the upstream pathways that control insulin production are less clear. To further elucidate this regulatory network, we created a genetic interaction map of insulin production by performing ∼20,000 pairwise RNA interference knockdowns of insulin promoter regulators. Our map correctly predicted known physical complexes in the electron transport chain and a role for Spry2 in the unfolded protein response. To further validate our map, we used it to predict the function of an unannotated gene encoding a 37-kDa protein with no identifiable domains we have termed mitochondrial fission factor interactor (Mfi). We have shown that Mfi is a binding partner of the mitochondrial fission factor and that Mfi inhibits dynamin-like protein 1 recruitment to mitochondria. Our data provide a resource to understand the regulatory network of insulin promoter activity.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Animals , Cell Line , Dynamins , GTP Phosphohydrolases , Gene Regulatory Networks , Humans , Insulin/genetics , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins , Mitochondrial Proteins/metabolism , Promoter Regions, Genetic/genetics , Unfolded Protein Response
12.
Physiol Rev ; 98(3): 1143-1167, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29717931

ABSTRACT

Diabetes mellitus results from disturbed glucose homeostasis due to an absolute (type 1) or relative (type 2) deficiency of insulin, a peptide hormone almost exclusively produced by the beta cells of the endocrine pancreas in a tightly regulated manner. Current therapy only delays disease progression through insulin injection and/or oral medications that increase insulin secretion or sensitivity, decrease hepatic glucose production, or promote glucosuria. These drugs have turned diabetes into a chronic disease as they do not solve the underlying beta cell defects or entirely prevent the long-term complications of hyperglycemia. Beta cell replacement through islet transplantation is a more physiological therapeutic alternative but is severely hampered by donor shortage and immune rejection. A curative strategy should combine newer approaches to immunomodulation with beta cell replacement. Success of this approach depends on the development of practical methods for generating beta cells, either in vitro or in situ through beta cell replication or beta cell differentiation. This review provides an overview of human beta cell generation.


Subject(s)
Cell Culture Techniques , Insulin-Secreting Cells/physiology , Regeneration , Animals , Homeostasis , Humans , Insulin-Secreting Cells/transplantation
13.
Diabetologia ; 61(1): 168-181, 2018 01.
Article in English | MEDLINE | ID: mdl-28852792

ABSTRACT

AIMS/HYPOTHESIS: Identification of a pancreatic neuro-insular network in mice suggests that a similar integration of islets and nerves may be present in the human pancreas. To characterise the neuro-insular network and the intra-pancreatic ganglia in a clinically related setting, we examined human pancreases in health and with fatty infiltration via 3-dimensional (3D) histology and compared the human pancreatic microenvironment with its counterpart in mice. METHODS: Human pancreatic specimens from individuals with normal BMI, high BMI (≥ 25) and type 2 diabetes were used to investigate the neuro-insular network. Transparent specimens were prepared by tissue clearing for transmitted light and deep-tissue fluorescence imaging to simultaneously visualise infiltrated adipocytes, islets and neurovascular networks. RESULTS: High-definition images of human islets reveal that both the sympathetic and parasympathetic nerves enter the islet core and reside in the immediate microenvironment of islet cells. Around the islets, the neuro-insular network is visualised with 3D histology to identify the intra-pancreatic ganglia (peri-lobular and intra-parenchymal ganglia) and the islet-ganglionic association. In humans, but not in mice, pancreatic fatty infiltration (BMI dependent) features adipocytes infiltrating into the parenchyma and accumulating in the peri-lobular space, in which the peri-lobular ganglia also reside. We identified the formation of adipose-ganglionic complexes in the peri-lobular space and enlargement of ganglia around adipocytes. In the specimen from the individual with type 2 diabetes, an increase in the number of nerve projections from the intra-parenchymal ganglia is associated with severe fatty infiltration. CONCLUSIONS/INTERPRETATION: We present new perspectives of human pancreas and islet innervation via 3D histology. Our results strongly suggest that fatty infiltration in the human pancreas creates a neurotrophic microenvironment and promotes remodelling of pancreatic innervation.


Subject(s)
Pancreas/metabolism , Adipocytes/metabolism , Animals , Body Mass Index , Diabetes Mellitus, Type 2/metabolism , Humans , Islets of Langerhans/metabolism , Mice , Obesity/metabolism , Sympathetic Nervous System/metabolism
14.
J Med Food ; 21(3): 274-281, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29068758

ABSTRACT

During maturation, pancreatic islets achieve their full capacity to secrete insulin in response to glucose, undergo morphological changes in which alpha-cells decrease and beta-cell mass increases, and they acquire the normal alpha- and beta-cell proportion changes that are important for islet functions later in life. In rodents, the first week of postweaning is critical for islet maturation. Multiple studies have documented the detrimental effects of several conditions on pancreatic maturation; however, few studies have addressed the use of pharmacological agents to enhance islet maturation. Biotin might have a potential action on islet maturation. Pharmacological concentrations of biotin have been found to modify islet morphology and function. In a previous study, we found that mice fed a biotin-supplemented diet for 8 weeks after weaning showed an increase in basal and glucose stimulated insulin secretion, enlarged islet size, and modified islet structure. In the present study, we investigated the effect of biotin on maturation features during the first week postweaning. Female BALB/cAnN Hsd mice were fed a control or a biotin-supplemented diet for 1 week after weaning. Compared with the control, biotin-supplemented mice showed an increase in pancreatic islet number and area in addition to an augmented proportion of beta-cells in the islet. These effects were related to an increase in beta-cell proliferation. No differences were found in insulin secretion, blood glucose concentrations, or serum insulin levels. These results indicate that biotin supplementation is capable of affecting beta-cell proliferation and might be a therapeutic agent for establishing strategies for regenerative medicine.


Subject(s)
Biotin/administration & dosage , Cell Differentiation , Cell Proliferation , Dietary Supplements , Insulin-Secreting Cells/cytology , Islets of Langerhans/growth & development , Vitamin B Complex/administration & dosage , Animals , Apoptosis , Biotin/adverse effects , Biotin/metabolism , Biotin/therapeutic use , Blood Glucose/analysis , Cell Count , Dietary Supplements/adverse effects , Female , Insulin/blood , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice, Inbred BALB C , Organ Size , Osmolar Concentration , Prediabetic State/prevention & control , Random Allocation , Tissue Culture Techniques , Vitamin B Complex/adverse effects , Vitamin B Complex/metabolism , Vitamin B Complex/therapeutic use , Weaning
15.
Biochem Biophys Res Commun ; 495(2): 1986-1991, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29223399

ABSTRACT

Genetically modified mice have been widely used in the field of ß-cell research. However, analysis of results gathered using genetically modified organisms should be interpreted carefully as the results may be confounded by several factors. Here, we showed the ectopic serotonin (5-HT) production in ß-cells of RIP-CreMgn, MIP-GFP, and MIP-Cre/ERT mice. These mice contained a human growth hormone (hGH) cassette to enhance transgene expression and hGH expression and Stat5 phosphorylation were detected in pancreatic islets of these mice. The expression level of tryptophan hydroxylase 1 (Tph1) was upregulated in pancreatic islets of transgenic mice with an hGH cassette but not in transgenic mice without an hGH cassette. Ectopic 5-HT production was not observed in ß-cell-specific prolactin receptor (Prlr) knockout mice or Stat5 knockout mice crossed with RIP-CreMgn. We further confirmed that 5-HT production in ß-cells of several transgenic mice was induced by hGH expression followed by the activation of the Prlr-Stat5-Tph1 pathway. These findings indicate that results obtained using transgenic mice containing the hGH cassette should be interpreted with care.


Subject(s)
B-Lymphocytes/metabolism , Human Growth Hormone/genetics , Human Growth Hormone/metabolism , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Serotonin/genetics , Serotonin/metabolism , Animals , Mice , Mice, Inbred C57BL
16.
Channels (Austin) ; 11(6): 636-647, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29087246

ABSTRACT

The ATP-sensitive potassium channel (KATP) functions as a metabo-electric transducer in regulating insulin secretion from pancreatic ß-cells. The pancreatic KATP channel is composed of a pore-forming inwardly-rectifying potassium channel, Kir6.2, and a regulatory subunit, sulphonylurea receptor 1 (SUR1). Loss-of-function mutations in either subunit often lead to the development of persistent hyperinsulinemic hypoglycemia of infancy (PHHI). PHHI is a rare genetic disease and most patients present with immediate onset within the first few days after birth. In this study, we report an unusual form of PHHI, in which the index patient developed hyperinsulinemic hypoglycemia after 1 year of age. The patient failed to respond to routine medication for PHHI and underwent a complete pancreatectomy. Genotyping of the index patient and his immediate family members showed that the patient and other family members with hypoglycemic episodes carried a heterozygous novel mutation in KCNJ11 (C83T), which encodes Kir6.2 (A28V). Electrophysiological and cell biological experiments revealed that A28V hKir6.2 is a dominant-negative, loss-of-function mutation and that KATP channels carrying this mutation failed to reach the cell surface. De novo protein structure prediction indicated that this A28V mutation reoriented the ER retention motif located at the C-terminal of the hKir6.2, and this result may explain the trafficking defect caused by this point mutation. Our study is the first report of a novel form of late-onset PHHI that is caused by a dominant mutation in KCNJ11 and exhibits a defect in proper surface expression of Kir6.2.


Subject(s)
Congenital Hyperinsulinism/metabolism , Mutation , Potassium Channels, Inwardly Rectifying/genetics , Humans , Potassium Channels, Inwardly Rectifying/metabolism
17.
Nutrition ; 43-44: 47-53, 2017.
Article in English | MEDLINE | ID: mdl-28935144

ABSTRACT

OBJECTIVE: Despite increasing evidence that pharmacologic concentrations of biotin modify glucose metabolism, to our knowledge there have not been any studies addressing the effects of biotin supplementation on glucagon production and secretion, considering glucagon is one of the major hormones in maintaining glucose homeostasis. The aim of this study was to investigate the effects of dietary biotin supplementation on glucagon expression, secretion, and action. METHODS: Male BALB/cAnN Hsd mice were fed a control or a biotin-supplemented diet (1.76 or 97.7 mg biotin/kg diet) for 8 wk postweaning. Glucagon gene mRNA expression was measured by the real-time polymerase chain reaction. Glucagon secretion was assessed in isolated islets and by glucagon concentration in plasma. Glucagon action was evaluated by glucagon tolerance tests, phosphoenolpyruvate carboxykinase (Pck1) mRNA expression, and glycogen degradation. RESULTS: Compared with the control group, glucagon mRNA and secretion were increased from the islets of the biotin-supplemented group. Fasting plasma glucagon levels were higher, but no differences between the groups were observed in nonfasting glucagon levels. Despite the elevated fasting glucagon levels, no differences were found in fasting blood glucose concentrations, fasting/fasting-refeeding glucagon tolerance tests, glycogen content and degradation, or mRNA expression of the hepatic gluconeogenic rate-limiting enzyme, Pck1. CONCLUSIONS: These results demonstrated that dietary biotin supplementation increased glucagon expression and secretion without affecting fasting blood glucose concentrations or glucagon tolerance and provided new insights into the effect of biotin supplementation on glucagon production and action.


Subject(s)
Biotin/administration & dosage , Glucagon/metabolism , Glucagon/pharmacology , Animals , Diet , Dietary Supplements , Gene Expression/drug effects , Glucagon/genetics , Gluconeogenesis/drug effects , Glycogen/metabolism , Islets of Langerhans/chemistry , Islets of Langerhans/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , RNA, Messenger/analysis
18.
Dev Cell ; 41(2): 129-142.e6, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28441528

ABSTRACT

During pancreatic development, proliferating pancreatic progenitors activate the proendocrine transcription factor neurogenin 3 (NEUROG3), exit the cell cycle, and differentiate into islet cells. The mechanisms that direct robust NEUROG3 expression within a subset of progenitor cells control the size of the endocrine population. Here we demonstrate that NEUROG3 is phosphorylated within the nucleus on serine 183, which catalyzes its hyperphosphorylation and proteosomal degradation. During progression through the progenitor cell cycle, NEUROG3 phosphorylation is driven by the actions of cyclin-dependent kinases 2 and 4/6 at G1/S cell-cycle checkpoint. Using models of mouse and human pancreas development, we show that lengthening of the G1 phase of the pancreatic progenitor cell cycle is essential for proper induction of NEUROG3 and initiation of endocrine cell differentiation. In sum, these studies demonstrate that progenitor cell-cycle G1 lengthening, through its actions on stabilization of NEUROG3, is an essential variable in normal endocrine cell genesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/physiology , Cell Differentiation/physiology , Nerve Tissue Proteins/metabolism , Pancreas/cytology , Pancreas/metabolism , Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Endocrine Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Humans , Islets of Langerhans/cytology , Mice , Nerve Tissue Proteins/genetics , Phosphorylation/physiology , Stem Cells/metabolism
19.
Diabetes ; 66(6): 1703-1712, 2017 06.
Article in English | MEDLINE | ID: mdl-28246293

ABSTRACT

Insulin production by the pancreatic ß-cell is required for normal glucose homeostasis. While key transcription factors that bind to the insulin promoter are known, relatively little is known about the upstream regulators of insulin transcription. Using a whole-genome RNA interference screen, we uncovered 26 novel regulators of insulin transcription that regulate diverse processes including oxidative phosphorylation, vesicle traffic, and the unfolded protein response (UPR). We focused on Spry2-a gene implicated in human type 2 diabetes by genome-wide association studies but without a clear connection to glucose homeostasis. We showed that Spry2 is a novel UPR target and its upregulation is dependent on PERK. Knockdown of Spry2 resulted in reduced expression of Serca2, reduced endoplasmic reticulum calcium levels, and induction of the UPR. Spry2 deletion in the adult mouse ß-cell caused hyperglycemia and hypoinsulinemia. Our study greatly expands the compendium of insulin promoter regulators and demonstrates a novel ß-cell link between Spry2 and human diabetes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation/genetics , Insulin-Secreting Cells/metabolism , Insulin/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Unfolded Protein Response/genetics , Animals , Annexin A5/metabolism , Blotting, Western , Calcium/metabolism , Cell Line , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum/metabolism , Gene Knockdown Techniques , Genome-Wide Association Study , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin/metabolism , Mice , Protein Serine-Threonine Kinases , RNA Interference , Real-Time Polymerase Chain Reaction , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , eIF-2 Kinase/metabolism
20.
Cell Stem Cell ; 18(4): 431-3, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058933

ABSTRACT

Widespread application of ß cell replacement to obtain insulin independence in people with type 1 diabetes requires an unlimited source of insulin-producing cells and the ability to block the pathological immune response. Recent reports in Nature Medicine and Nature Biotechnology present a creative bioengineering strategy for achieving these goals.


Subject(s)
Bioengineering/trends , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/immunology , Insulin/immunology , Biotechnology , Diabetes Mellitus, Type 1/pathology , Humans , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...