Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Development ; 149(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35708349

ABSTRACT

Pancreatic and duodenal homeobox 1 (PDX1) is crucial for pancreas organogenesis, yet the dynamic changes in PDX1 binding in human or mouse developing pancreas have not been examined. To address this knowledge gap, we performed PDX1 ChIP-seq and single-cell RNA-seq using fetal human pancreata. We integrated our datasets with published datasets and revealed the dynamics of PDX1 binding and potential cell lineage-specific PDX1-bound genes in the pancreas from fetal to adult stages. We identified a core set of developmentally conserved PDX1-bound genes that reveal the broad multifaceted role of PDX1 in pancreas development. Despite the well-known dramatic changes in PDX1 function and expression, we found that PDX1-bound genes are largely conserved from embryonic to adult stages. This points towards a dual role of PDX1 in regulating the expression of its targets at different ages, dependent on other functionally congruent or directly interacting partners. We also showed that PDX1 binding is largely conserved in mouse pancreas. Together, our study reveals PDX1 targets in the developing pancreas in vivo and provides an essential resource for future studies on pancreas development.


Subject(s)
Genes, Homeobox , Homeodomain Proteins , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Pancreas , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptome/genetics
2.
Endocrinology ; 162(9)2021 09 01.
Article in English | MEDLINE | ID: mdl-34134142

ABSTRACT

Transitional hypoglycemia in normal newborns occurs in the first 3 days of life and has clinical features consistent with hyperinsulinism. We found a lower threshold for glucose-stimulated insulin secretion from freshly isolated embryonic day (E) 22 rat islets, which persisted into the first postnatal days. The threshold reached the adult level by postnatal day (P) 14. Culturing P14 islets also decreased the glucose threshold. Freshly isolated P1 rat islets had a lower threshold for insulin secretion in response to 2-aminobicyclo-(2, 2, 1)-heptane-2-carboxylic acid, a nonmetabolizable leucine analog, and diminished insulin release in response to tolbutamide, an inhibitor of ß-cell KATP channels. These findings suggested that decreased KATP channel function could be responsible for the lower glucose threshold for insulin secretion. Single-cell transcriptomic analysis did not reveal a lower expression of KATP subunit genes in E22 compared with P14 ß cells. The investigation of electrophysiological characteristics of dispersed ß cells showed that early neonatal and cultured cells had fewer functional KATP channels per unit membrane area. Our findings suggest that decreased surface density of KATP channels may contribute to the observed differences in glucose threshold for insulin release.


Subject(s)
Glucose/pharmacology , Insulin Secretion/drug effects , Islets of Langerhans/drug effects , KATP Channels/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Amino Acids, Cyclic/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Embryo, Mammalian , Female , Glucose/metabolism , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , KATP Channels/agonists , KATP Channels/genetics , KATP Channels/metabolism , Potassium Chloride/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley
3.
Mol Metab ; 25: 95-106, 2019 07.
Article in English | MEDLINE | ID: mdl-31023625

ABSTRACT

OBJECTIVE: In type 2 diabetes (T2D), oxidative stress contributes to the dysfunction and loss of pancreatic ß cells. A highly conserved feature of the cellular response to stress is the regulation of mRNA translation; however, the genes regulated at the level of translation are often overlooked due to the convenience of RNA sequencing technologies. Our goal is to investigate translational regulation in ß cells as a means to uncover novel factors and pathways pertinent to cellular adaptation and survival during T2D-associated conditions. METHODS: Translating ribosome affinity purification (TRAP) followed by RNA-seq or RT-qPCR was used to identify changes in the ribosome occupancy of mRNAs in Min6 cells. Gene depletion studies used lentiviral delivery of shRNAs to primary mouse islets or CRISPR-Cas9 to Min6 cells. Oxidative stress and apoptosis were measured in primary islets using cell-permeable dyes with fluorescence readouts of oxidation and activated cleaved caspase-3 and-7, respectively. Gene expression was assessed by RNA-seq, RT-qPCR, and western blot. ChIP-qPCR was used to determine chromatin enrichment. RESULTS: TRAP-seq in a PDX1-deficiency model of ß cell dysfunction uncovered a cohort of genes regulated at the level of mRNA translation, including the transcription factor JUND. Using a panel of diabetes-associated stressors, JUND was found to be upregulated in mouse islets cultured with high concentrations of glucose and free fatty acid, but not after treatment with hydrogen peroxide or thapsigargin. This induction of JUND could be attributed to increased mRNA translation. JUND was also upregulated in islets from diabetic db/db mice and in human islets treated with high glucose and free fatty acid. Depletion of JUND in primary islets reduced oxidative stress and apoptosis in ß cells during metabolic stress. Transcriptome assessment identified a cohort of genes, including pro-oxidant and pro-inflammatory genes, regulated by JUND that are commonly dysregulated in models of ß cell dysfunction, consistent with a maladaptive role for JUND in islets. CONCLUSIONS: A translation-centric approach uncovered JUND as a stress-responsive factor in ß cells that contributes to redox imbalance and apoptosis during pathophysiologically relevant stress.


Subject(s)
Insulin-Secreting Cells/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Stress, Physiological/physiology , Animals , Apoptosis , CRISPR-Cas Systems , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Diabetes Mellitus, Type 2/metabolism , Fatty Acids , Gene Expression Regulation , Glucose/metabolism , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , RNA, Messenger/metabolism , Trans-Activators/genetics , Transcription Factors
4.
Nucleic Acids Res ; 46(21): e124, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30102368

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) is a powerful tool to study heterogeneity and dynamic changes in cell populations. Clustering scRNA-seq is essential in identifying new cell types and studying their characteristics. We develop CellBIC (single Cell BImodal Clustering) to cluster scRNA-seq data based on modality in the gene expression distribution. Compared with classical bottom-up approaches that rely on a distance metric, CellBIC performs hierarchical clustering in a top-down manner. CellBIC outperformed the bottom-up hierarchical clustering approach and other recently developed clustering algorithms while maintaining the hierarchical structure of cells. Importantly, CellBIC identifies type 2 diabetes and age specific ß cell signatures characterized by SIX3 and CDH2, respectively.


Subject(s)
Computational Biology/methods , Pancreas/cytology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Aging/genetics , Aging/pathology , Algorithms , Antigens, CD/genetics , Cadherins/genetics , Cluster Analysis , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Gene Expression Profiling/methods , Gene Expression Profiling/statistics & numerical data , Genetic Markers , Humans , Islets of Langerhans/physiology , Pancreas/physiology , Sequence Analysis, RNA/statistics & numerical data , Single-Cell Analysis/statistics & numerical data
5.
Stem Cell Reports ; 8(3): 589-604, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28196690

ABSTRACT

Induced pluripotent stem cells were created from a pancreas agenesis patient with a mutation in GATA6. Using genome-editing technology, additional stem cell lines with mutations in both GATA6 alleles were generated and demonstrated a severe block in definitive endoderm induction, which could be rescued by re-expression of several different GATA family members. Using the endodermal progenitor stem cell culture system to bypass the developmental block at the endoderm stage, cell lines with mutations in one or both GATA6 alleles could be differentiated into ß-like cells but with reduced efficiency. Use of suboptimal doses of retinoic acid during pancreas specification revealed a more severe phenotype, more closely mimicking the patient's disease. GATA6 mutant ß-like cells fail to secrete insulin upon glucose stimulation and demonstrate defective insulin processing. These data show that GATA6 plays a critical role in endoderm and pancreas specification and ß-like cell functionality in humans.


Subject(s)
Endoderm/metabolism , GATA6 Transcription Factor/genetics , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Pancreas/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Endoderm/drug effects , Endoderm/embryology , GATA6 Transcription Factor/metabolism , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Models, Biological , Multigene Family , Mutation , Pancreas/embryology , Phenotype , Tretinoin/pharmacology
6.
Physiol Genomics ; 49(2): 105-114, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28011883

ABSTRACT

The heterogeneity of the developing pancreatic epithelium and low abundance of endocrine progenitors limit the information derived from traditional expression studies. To identify genes that characterize early developmental tissues composed of multiple progenitor lineages, we applied single-cell RNA-Seq to embryonic day (e)13.5 mouse pancreata and performed integrative analysis with single cell data from mature pancreas. We identified subpopulations expressing macrophage or endothelial markers and new pancreatic progenitor markers. We also identified potential α-cell precursors expressing glucagon (Gcg) among the e13.5 pancreatic cells. Despite their high Gcg expression levels, these cells shared greater transcriptomic similarity with other e13.5 cells than with adult α-cells, indicating their immaturity. Comparative analysis identified the sodium-dependent neutral amino acid transporter, Slc38a5, as a characteristic gene expressed in α-cell precursors but not mature cells. By immunofluorescence analysis, we observed SLC38A5 expression in pancreatic progenitors, including in a subset of NEUROG3+ endocrine progenitors and MAFB+ cells and in all GCG+ cells. Expression declined in α-cells during late gestation and was absent in the adult islet. Our results suggest SLC38A5 as an early marker of α-cell lineage commitment.


Subject(s)
Gene Expression Profiling , Pancreas/cytology , Single-Cell Analysis , Stem Cells/metabolism , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Cell Differentiation/genetics , Cluster Analysis , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Islets of Langerhans/cytology , Mice , Models, Biological , Stem Cells/cytology
7.
Cell Rep ; 15(12): 2637-2650, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27292642

ABSTRACT

Pdx1 and Oc1 are co-expressed in multipotent pancreatic progenitors and regulate the pro-endocrine gene Neurog3. Their expression diverges in later organogenesis, with Oc1 absent from hormone+ cells and Pdx1 maintained in mature ß cells. In a classical genetic test for cooperative functional interactions, we derived mice with combined Pdx1 and Oc1 heterozygosity. Endocrine development in double-heterozygous pancreata was normal at embryonic day (E)13.5, but defects in specification and differentiation were apparent at E15.5, the height of the second wave of differentiation. Pancreata from double heterozygotes showed alterations in the expression of genes crucial for ß-cell development and function, decreased numbers and altered allocation of Neurog3-expressing endocrine progenitors, and defective endocrine differentiation. Defects in islet gene expression and ß-cell function persisted in double heterozygous neonates. These results suggest that Oc1 and Pdx1 cooperate prior to their divergence, in pancreatic progenitors, to allow for proper differentiation and functional maturation of ß cells.


Subject(s)
Cell Differentiation , Hepatocyte Nuclear Factor 6/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Trans-Activators/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Dosage , Gene Expression Regulation, Developmental , Gene Ontology , Gene Regulatory Networks , Glucose/metabolism , Heterozygote , Homeostasis/genetics , Mice , Multigene Family , Nerve Tissue Proteins/metabolism , Weaning
8.
Pediatr Diabetes ; 16(1): 67-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24433315

ABSTRACT

Heterozygous mutations in GATA6 have been linked to pancreatic agenesis and cardiac malformations. The aim of this study was to describe a new mutation in GATA6 in an infant with pancreatic agenesis, associated with truncus arteriosus and absent gallbladder. Clinical data were obtained from chart review. Gene sequencing was performed on genomic DNA. The patient was a female infant diagnosed shortly after birth with a severe cardiac malformation, absent gallbladder, anomalous hepatic blood flow, unilateral hydronephrosis and hydroureter, neonatal diabetes, and pancreatic exocrine insufficiency. Despite prolonged intensive management care, she died at 3 months of age because of cardiac complications. Analysis of her genomic DNA revealed a novel missense mutation of GATA6. The novel mutation described in this case extends the list of GATA6 mutations causing pancreatic agenesis and cardiac malformations.


Subject(s)
GATA6 Transcription Factor/genetics , Mutation, Missense , Pancreas/abnormalities , Pancreatic Diseases/congenital , Diabetes Mellitus/congenital , Diabetes Mellitus/genetics , Digestive System Abnormalities/complications , Digestive System Abnormalities/genetics , Fatal Outcome , Female , Gallbladder/abnormalities , HEK293 Cells/pathology , Heart Defects, Congenital/complications , Heart Defects, Congenital/genetics , Humans , Infant , Pancreatic Diseases/complications , Pancreatic Diseases/genetics , Urogenital Abnormalities/complications , Urogenital Abnormalities/genetics
9.
J Pediatr Endocrinol Metab ; 27(11-12): 1237-41, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25153212

ABSTRACT

Donohue syndrome (DS) is a severe form of congenital insulin resistance due to mutation(s) in the insulin receptor (INSR) gene. Given the similarities between insulin and insulin-like growth factor 1 (IGF-1) receptors, recombinant human IGF-1 (rhIGF-1) has been used to treat severe insulin resistance due to INSR mutation(s). Traditional subcutaneous therapy may be limited by the shortened IGF-1 half-life in these patients. We report the case of a female with molecularly confirmed DS treated with continuous rhIGF-1 therapy via an insulin pump. With treatment, the patient's hemoglobin A1c decreased from 9.8% to 8.8%, and her weight increased by 0.8 kg. Development of an ovarian tumor complicated her course, but it was unclear whether this was related to rhIGF-1 therapy. Limited treatment options exist for patients with DS. The use of continuous rhIGF-1 via an insulin pump may be a viable option, although further experience is needed to establish safety and efficacy.


Subject(s)
Donohue Syndrome/drug therapy , Infusion Pumps , Insulin-Like Growth Factor I/administration & dosage , Recombinant Proteins/administration & dosage , Antigens, CD/genetics , Donohue Syndrome/genetics , Female , Humans , Infant, Newborn , Injections, Subcutaneous , Insulin-Like Growth Factor I/pharmacology , Mutation/genetics , Prognosis , Receptor, Insulin/genetics , Recombinant Proteins/pharmacology
17.
Endocrinol Metab Clin North Am ; 41(4): 679-94, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23099264

ABSTRACT

Type 1 diabetes is one of the most common chronic diseases of childhood and adolescence. Multiple registries have assessed its epidemiology and have noted a steady increase in incidence of the disease. This article addresses the epidemiology of type 1 diabetes in children aged 0 to 19 years, by reviewing the available, current data from both US and international registries. The prevalence and incidence data by race, ethnicity, age of onset, sex, season of onset, and temporal trends of the disease are presented. Multiple risk factors have been implicated for the increasing incidence in type 1 diabetes, and these genetic and environmental risk factors are discussed.


Subject(s)
Diabetes Mellitus, Type 1/epidemiology , Registries/statistics & numerical data , Adolescent , Age Factors , Age of Onset , Child , Child, Preschool , Female , Humans , Incidence , Infant , Male , Prevalence , Risk Factors , Sex Factors , Young Adult
18.
J Clin Endocrinol Metab ; 97(10): E2026-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22802087

ABSTRACT

CONTEXT: Inactivating mutations in HNF1A and HNF4A cause the maturity-onset diabetes of youth (MODY)-3 and MODY1 forms of monogenic diabetes, respectively. Children carrying HNF4A (MODY1) mutations can present in early infancy with macrosomia and diazoxide-responsive hyperinsulinism. OBJECTIVE: Our objective was to describe three novel cases of hyperinsulinism associated with MODY1 and MODY3 mutations. RESEARCH DESIGN AND METHODS: Clinical data were obtained from chart review. Gene sequencing was performed on genomic DNA. RESULTS: Case 1 was diagnosed at 20 months with persistent hyperinsulinemic hypoglycemia and was found to have a novel MODY3 HNF1A mutation, carried by her father who had diabetes. Case 2 was diagnosed with diazoxide-responsive hyperinsulinism at 3 months of age and had complete resolution of hyperinsulinism by 4 yr. She was found to have a novel MODY3 HNF1A missense mutation, also carried by her father. Case 3 presented as a newborn with diazoxide-responsive hyperinsulinism and later developed renal Fanconi syndrome, hypophosphatemic rickets, and hepatic glycogenosis. Although the latter's features suggested Fanconi-Bickel syndrome, sequencing of the SLC2A2 gene was normal. The patient was found to have a known MODY1 mutation in HNF4A. In all cases, the hyperinsulinism improved with age. CONCLUSIONS: The first two cases demonstrate that mutations in HNF1A (MODY3) can cause hyperinsulinism early in life and diabetes later, similar to the phenotype recently reported for HNF4A (MODY1) mutations. Case 3 indicates that the effects of HNF4A mutations in infancy may extend beyond pancreatic ß-cells to produce a disorder similar to glucose transporter 2 deficiency involving both liver glycogen metabolism and renal tubular transport.


Subject(s)
Congenital Hyperinsulinism/genetics , Diabetes Mellitus, Type 2/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 4/genetics , Age of Onset , Child , Child, Preschool , Female , Humans , Hypoglycemia/genetics , Mutation, Missense , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...