Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Biochim Biophys Acta Mol Cell Res ; 1869(5): 119235, 2022 05.
Article in English | MEDLINE | ID: mdl-35151663

ABSTRACT

Glucose homeostasis is maintained by hormones secreted from different types of pancreatic islets and its dysregulation can result in diseases including diabetes mellitus. The secretion of hormones from pancreatic islets is highly complex and tightly controlled by G protein-coupled receptors (GPCRs). Moreover, GPCR signaling may play a role in enhancing islet cell replication and proliferation. Thus, targeting GPCRs offers a promising strategy for regulating the functionality of pancreatic islets. Here, available RNAseq datasets from human and mouse islets were used to identify the GPCR expression profile and the impact of GPCR signaling for normal islet functionality is discussed.


Subject(s)
Islets of Langerhans/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/growth & development , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Transcriptome
2.
Nat Commun ; 12(1): 4458, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294685

ABSTRACT

The cellular identity of pancreatic polypeptide (Ppy)-expressing γ-cells, one of the rarest pancreatic islet cell-type, remains elusive. Within islets, glucagon and somatostatin, released respectively from α- and δ-cells, modulate the secretion of insulin by ß-cells. Dysregulation of insulin production raises blood glucose levels, leading to diabetes onset. Here, we present the genetic signature of human and mouse γ-cells. Using different approaches, we identified a set of genes and pathways defining their functional identity. We found that the γ-cell population is heterogeneous, with subsets of cells producing another hormone in addition to Ppy. These bihormonal cells share identity markers typical of the other islet cell-types. In mice, Ppy gene inactivation or conditional γ-cell ablation did not alter glycemia nor body weight. Interestingly, upon ß-cell injury induction, γ-cells exhibited gene expression changes and some of them engaged insulin production, like α- and δ-cells. In conclusion, we provide a comprehensive characterization of γ-cells and highlight their plasticity and therapeutic potential.


Subject(s)
Insulin/biosynthesis , Pancreatic Polypeptide-Secreting Cells/metabolism , Pancreatic Polypeptide/metabolism , Protein Precursors/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Cell Lineage/genetics , Female , Gene Knock-In Techniques , Humans , Insulin-Secreting Cells/classification , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Transgenic , Pancreas/cytology , Pancreas/embryology , Pancreas/growth & development , Pancreatic Polypeptide/deficiency , Pancreatic Polypeptide/genetics , Pancreatic Polypeptide-Secreting Cells/classification , Pancreatic Polypeptide-Secreting Cells/cytology , Pregnancy , RNA-Seq
3.
Nat Genet ; 53(4): 455-466, 2021 04.
Article in English | MEDLINE | ID: mdl-33795864

ABSTRACT

Single-nucleus assay for transposase-accessible chromatin using sequencing (snATAC-seq) creates new opportunities to dissect cell type-specific mechanisms of complex diseases. Since pancreatic islets are central to type 2 diabetes (T2D), we profiled 15,298 islet cells by using combinatorial barcoding snATAC-seq and identified 12 clusters, including multiple alpha, beta and delta cell states. We cataloged 228,873 accessible chromatin sites and identified transcription factors underlying lineage- and state-specific regulation. We observed state-specific enrichment of fasting glucose and T2D genome-wide association studies for beta cells and enrichment for other endocrine cell types. At T2D signals localized to islet-accessible chromatin, we prioritized variants with predicted regulatory function and co-accessibility with target genes. A causal T2D variant rs231361 at the KCNQ1 locus had predicted effects on a beta cell enhancer co-accessible with INS and genome editing in embryonic stem cell-derived beta cells affected INS levels. Together our findings demonstrate the power of single-cell epigenomics for interpreting complex disease genetics.


Subject(s)
Chromatin/chemistry , Diabetes Mellitus, Type 2/genetics , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , KCNQ1 Potassium Channel/genetics , Pancreatic Polypeptide-Secreting Cells/metabolism , Somatostatin-Secreting Cells/metabolism , Blood Glucose/metabolism , Cell Differentiation , Chromatin/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Epigenomics , Fasting , Gene Expression Profiling , Genome-Wide Association Study , Glucagon-Secreting Cells/pathology , High-Throughput Nucleotide Sequencing , Human Embryonic Stem Cells/cytology , Humans , Insulin-Secreting Cells/pathology , KCNQ1 Potassium Channel/metabolism , Multigene Family , Pancreatic Polypeptide-Secreting Cells/pathology , Polymorphism, Genetic , Single-Cell Analysis , Somatostatin-Secreting Cells/pathology , Transcription Factors/classification , Transcription Factors/genetics , Transcription Factors/metabolism
4.
PLoS One ; 15(3): e0230627, 2020.
Article in English | MEDLINE | ID: mdl-32208453

ABSTRACT

The gene encoding eukaryotic initiation factor 5A (EIF5A) is found in diabetes-susceptibility loci in mouse and human. eIF5A is the only protein known to contain hypusine (hydroxyputrescine lysine), a polyamine-derived amino acid formed post-translationally in a reaction catalyzed by deoxyhypusine synthase (DHPS). Previous studies showed pharmacologic blockade of DHPS in type 1 diabetic NOD mice and type 2 diabetic db/db mice improved glucose tolerance and preserved beta cell mass, which suggests that hypusinated eIF5A (eIF5AHyp) may play a role in diabetes pathogenesis by direct action on the beta cells and/or altering the adaptive or innate immune responses. To translate these findings to human, we examined tissue from individuals with and without type 1 and type 2 diabetes to determine the expression of eIF5AHyp. We detected eIF5AHyp in beta cells, exocrine cells and immune cells; however, there was also unexpected enrichment of eIF5AHyp in pancreatic polypeptide-expressing PP cells. Interestingly, the presence of eIF5AHyp co-expressing PP cells was not enhanced with disease. These data identify new aspects of eIF5A biology and highlight the need to examine human tissue to understand disease.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/pathology , Lysine/analogs & derivatives , Pancreas/metabolism , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Spleen/metabolism , Adult , Animals , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Middle Aged , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/metabolism , Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Young Adult , Eukaryotic Translation Initiation Factor 5A
5.
Nature ; 567(7746): 43-48, 2019 03.
Article in English | MEDLINE | ID: mdl-30760930

ABSTRACT

Cell-identity switches, in which terminally differentiated cells are converted into different cell types when stressed, represent a widespread regenerative strategy in animals, yet they are poorly documented in mammals. In mice, some glucagon-producing pancreatic α-cells and somatostatin-producing δ-cells become insulin-expressing cells after the ablation of insulin-secreting ß-cells, thus promoting diabetes recovery. Whether human islets also display this plasticity, especially in diabetic conditions, remains unknown. Here we show that islet non-ß-cells, namely α-cells and pancreatic polypeptide (PPY)-producing γ-cells, obtained from deceased non-diabetic or diabetic human donors, can be lineage-traced and reprogrammed by the transcription factors PDX1 and MAFA to produce and secrete insulin in response to glucose. When transplanted into diabetic mice, converted human α-cells reverse diabetes and continue to produce insulin even after six months. Notably, insulin-producing α-cells maintain expression of α-cell markers, as seen by deep transcriptomic and proteomic characterization. These observations provide conceptual evidence and a molecular framework for a mechanistic understanding of in situ cell plasticity as a treatment for diabetes and other degenerative diseases.


Subject(s)
Diabetes Mellitus/pathology , Diabetes Mellitus/therapy , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/pathology , Animals , Biomarkers/analysis , Cell Lineage/drug effects , Cellular Reprogramming/drug effects , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Disease Models, Animal , Female , Glucagon/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/transplantation , Glucose/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Male , Mice , Organ Specificity/drug effects , Pancreatic Polypeptide/metabolism , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/drug effects , Pancreatic Polypeptide-Secreting Cells/metabolism , Proteomics , Sequence Analysis, RNA , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptome , Transduction, Genetic
6.
Cell Metab ; 28(4): 557-572.e6, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30017352

ABSTRACT

The gut microbiota is essential for the normal function of the gut immune system, and microbiota alterations are associated with autoimmune disorders. However, how the gut microbiota prevents autoimmunity in distant organs remains poorly defined. Here we reveal that gut microbiota conditioned innate lymphoid cells (ILCs) induce the expression of mouse ß-defensin 14 (mBD14) by pancreatic endocrine cells, preventing autoimmune diabetes in the non-obese diabetic (NOD) mice. MBD14 stimulates, via Toll-like receptor 2, interleukin-4 (IL-4)-secreting B cells that induce regulatory macrophages, which in turn induce protective regulatory T cells. The gut microbiota-derived molecules, aryl hydrocarbon receptor (AHR) ligands and butyrate, promote IL-22 secretion by pancreatic ILCs, which induce expression of mBD14 by endocrine cells. Dysbiotic microbiota and low-affinity AHR allele explain the defective pancreatic expression of mBD14 observed in NOD mice. Our study reveals a yet unidentified crosstalk between ILCs and endocrine cells in the pancreas that is essential for the prevention of autoimmune diabetes development.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Gastrointestinal Microbiome/immunology , Insulin-Secreting Cells/metabolism , Lymphocytes/metabolism , Pancreatic Polypeptide-Secreting Cells/metabolism , beta-Defensins/metabolism , Animals , B-Lymphocytes, Regulatory/metabolism , Female , Humans , Immunity, Innate , Interleukins/metabolism , Islets of Langerhans/metabolism , Kaplan-Meier Estimate , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Statistics, Nonparametric , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 2/metabolism , Interleukin-22
7.
Diabetes Obes Metab ; 19 Suppl 1: 124-136, 2017 09.
Article in English | MEDLINE | ID: mdl-28880471

ABSTRACT

The progressive loss of pancreatic ß-cell mass that occurs in both type 1 and type 2 diabetes is a primary factor driving efforts to identify strategies for effectively increasing, enhancing or restoring ß-cell mass. While factors that seem to influence ß-cell proliferation in specific contexts have been described, reliable stimulation of human ß-cell proliferation has remained a challenge. Importantly, ß-cells exist in the context of a complex, integrated pancreatic islet microenvironment where they interact with other endocrine cells, vascular endothelial cells, extracellular matrix, neuronal projections and islet macrophages. This review highlights different components of the pancreatic microenvironment, and reviews what is known about how signaling that occurs between ß-cells and these other components influences ß-cell proliferation. Future efforts to further define the role of the pancreatic islet microenvironment on ß-cell proliferation may lead to the development of successful approaches to increase or restore ß-cell mass in diabetes.


Subject(s)
Cell Communication , Cell Proliferation , Cellular Microenvironment , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Models, Biological , Animals , Apoptosis , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/immunology , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Islets of Langerhans/blood supply , Islets of Langerhans/innervation , Islets of Langerhans/pathology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/immunology , Pancreatic Polypeptide-Secreting Cells/metabolism , Pancreatic Polypeptide-Secreting Cells/pathology , Somatostatin-Secreting Cells/cytology , Somatostatin-Secreting Cells/immunology , Somatostatin-Secreting Cells/metabolism , Somatostatin-Secreting Cells/pathology , Species Specificity
8.
Pancreas ; 46(6): 820-824, 2017 07.
Article in English | MEDLINE | ID: mdl-28609372

ABSTRACT

A partial pancreaticogastrodudenectomy was performed on a 66-year old man with type 2 diabetes mellitus because of an invasive, moderately differentiated adenocarcinoma in the head of the pancreas. In the adjacent grossly normal tissue of the uncinate process, there was a massive proliferation of pancreatic polypeptide (PP) cells confined to this region and showed invasive pattern. Strikingly, in the heaped area of his duodenum, there was a strikingly large number of PP, glucagon, a few insulin cells in a mini-islet-like patterns composed of glucagon and insulin cells. Among the etiological factors, the possible long-lasting effects of the GLP-1 analog, with which the patient was treated, are discussed. This is the first report in the literature of both the coexistence of a pancreatic adenocarcinoma and invasive PPoma and the occurrence of PP and insulin cells in human duodenal mucosa.


Subject(s)
Adenocarcinoma/complications , Cell Proliferation/drug effects , Diabetes Mellitus, Type 2/drug therapy , Duodenum/drug effects , Hypoglycemic Agents/adverse effects , Liraglutide/adverse effects , Pancreas/drug effects , Pancreatic Neoplasms/complications , Pancreatic Polypeptide-Secreting Cells/drug effects , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Aged , Chemotherapy, Adjuvant , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Duodenum/metabolism , Duodenum/pathology , Duodenum/surgery , Fatal Outcome , Humans , Hyperplasia , Immunohistochemistry , Male , Pancreas/metabolism , Pancreas/pathology , Pancreas/surgery , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Pancreatic Polypeptide-Secreting Cells/metabolism , Pancreaticoduodenectomy , Risk Factors , Time Factors , Treatment Outcome
9.
Diabetes Obes Metab ; 19(9): 1267-1275, 2017 09.
Article in English | MEDLINE | ID: mdl-28345790

ABSTRACT

AIMS: Ghrelin is a gastric-derived hormone that stimulates growth hormone (GH) secretion and has a multi-faceted role in the regulation of energy homeostasis, including glucose metabolism. Circulating ghrelin concentrations are modulated in response to nutritional status, but responses to ghrelin in altered metabolic states are poorly understood. We investigated the metabolic effects of ghrelin in obesity and early after Roux-en-Y gastric bypass (RYGB). MATERIALS AND METHODS: We assessed central and peripheral metabolic responses to acyl ghrelin infusion (1 pmol kg-1 min-1 ) in healthy, lean subjects (n = 9) and non-diabetic, obese subjects (n = 9) before and 2 weeks after RYGB. Central responses were assessed by GH and pancreatic polypeptide (surrogate for vagal activity) secretion. Peripheral responses were assessed by hepatic and skeletal muscle insulin sensitivity during a hyperinsulinaemic-euglycaemic clamp. RESULTS: Ghrelin-stimulated GH secretion was attenuated in obese subjects, but was restored by RYGB to a response similar to that of lean subjects. The heightened pancreatic polypeptide response to ghrelin infusion in the obese was attenuated after RYGB. Hepatic glucose production and hepatic insulin sensitivity were not altered by ghrelin infusion in RYGB subjects. Skeletal muscle insulin sensitivity was impaired to a similar degree in lean, obese and post-RYGB individuals in response to ghrelin infusion. CONCLUSIONS: These data suggest that obesity is characterized by abnormal central, but not peripheral, responsiveness to ghrelin that can be restored early after RYGB before significant weight loss. Further work is necessary to fully elucidate the role of ghrelin in the metabolic changes that occur in obesity and following RYGB.


Subject(s)
Anti-Obesity Agents/therapeutic use , Gastric Bypass , Ghrelin/therapeutic use , Human Growth Hormone/agonists , Insulin Resistance , Obesity, Morbid/drug therapy , Obesity, Morbid/surgery , Acylation , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/chemistry , Cohort Studies , Combined Modality Therapy/adverse effects , Cross-Over Studies , Energy Metabolism/drug effects , Ghrelin/administration & dosage , Ghrelin/adverse effects , Ghrelin/chemistry , Gluconeogenesis/drug effects , Glucose Clamp Technique , Human Growth Hormone/blood , Human Growth Hormone/metabolism , Humans , Infusions, Intravenous , Liver/drug effects , Liver/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Obesity, Morbid/blood , Obesity, Morbid/metabolism , Pancreatic Polypeptide/agonists , Pancreatic Polypeptide/blood , Pancreatic Polypeptide/metabolism , Pancreatic Polypeptide-Secreting Cells/drug effects , Pancreatic Polypeptide-Secreting Cells/metabolism , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Postoperative Care , Preoperative Care , Protein Precursors/agonists , Protein Precursors/blood , Protein Precursors/metabolism , Single-Blind Method
10.
Diabetes Obes Metab ; 18 Suppl 1: 10-22, 2016 09.
Article in English | MEDLINE | ID: mdl-27615127

ABSTRACT

During embryonic development, endocrine cells of the pancreas are specified from multipotent progenitors. The transcription factor Neurogenin 3 (NEUROG3) is critical for this development and it has been shown that all endocrine cells of the pancreas arise from endocrine progenitors expressing NEUROG3. A thorough understanding of the role of NEUROG3 during development, directed differentiation of pluripotent stem cells and in models of cellular reprogramming, will guide future efforts directed at finding novel sources of ß-cells for cell replacement therapies. In this article, we review the expression and function of NEUROG3 in both mouse and human and present the further characterization of a monoclonal antibody directed against NEUROG3. This antibody has been previously been used for detection of both mouse and human NEUROG3. However, our results suggest that the epitope recognized by this antibody is specific to mouse NEUROG3. Thus, we have also generated a monoclonal antibody specifically recognizing human NEUROG3 and present the characterization of this antibody here. Together, these antibodies will provide useful tools for future studies of NEUROG3 expression, and the data presented in this article suggest that recently described expression patterns of NEUROG3 in human foetal and adult pancreas should be re-examined.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Islets of Langerhans/cytology , Nerve Tissue Proteins/genetics , Animals , Antibodies, Monoclonal , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Cellular Reprogramming , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Humans , Immunohistochemistry , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/metabolism , Somatostatin-Secreting Cells/cytology , Somatostatin-Secreting Cells/metabolism
11.
PLoS One ; 11(6): e0157138, 2016.
Article in English | MEDLINE | ID: mdl-27270601

ABSTRACT

The transcription factor Glis-similar 3 (Glis3) has been implicated in the development of neonatal, type 1 and type 2 diabetes. In this study, we examined the spatiotemporal expression of Glis3 protein during embryonic and neonatal pancreas development as well as its function in PP cells. To obtain greater insights into the functions of Glis3 in pancreas development, we examined the spatiotemporal expression of Glis3 protein in a knockin mouse strain expressing a Glis3-EGFP fusion protein. Immunohistochemistry showed that Glis3-EGFP was not detectable during early pancreatic development (E11.5 and E12.5) and at E13.5 and 15.5 was not expressed in Ptf1a+ cells in the tip domains indicating that Glis3 is not expressed in multipotent pancreatic progenitors. Glis3 was first detectable at E13.5 in the nucleus of bipotent progenitors in the trunk domains, where it co-localized with Sox9, Hnf6, and Pdx1. It remained expressed in preductal and Ngn3+ endocrine progenitors and at later stages becomes restricted to the nucleus of pancreatic beta and PP cells as well as ductal cells. Glis3-deficiency greatly reduced, whereas exogenous Glis3, induced Ppy expression, as reported for insulin. Collectively, our study demonstrates that Glis3 protein exhibits a temporal and cell type-specific pattern of expression during embryonic and neonatal pancreas development that is consistent with a regulatory role for Glis3 in promoting endocrine progenitor generation, regulating insulin and Ppy expression in beta and PP cells, respectively, and duct morphogenesis.


Subject(s)
Insulin-Secreting Cells/metabolism , Pancreas/growth & development , Pancreatic Ducts/metabolism , Pancreatic Polypeptide-Secreting Cells/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , HEK293 Cells , Humans , Mice , Pancreas/cytology , Pancreatic Polypeptide/metabolism
12.
J Endocrinol ; 229(2): 123-32, 2016 05.
Article in English | MEDLINE | ID: mdl-26931137

ABSTRACT

The aim of this study was to evaluate the location of PP and δ cells in relation to the vascularization within human pancreatic islets. To this end, pancreas sections were analysed by immunofluorescence using antibodies against endocrine islet and endothelial cells. Staining in different islet areas corresponding to islet cells adjacent or not to peripheral or central vascular channels was quantified by computerized morphometry. As results, α, PP and δ cells were preferentially found adjacent to vessels. In contrast to α cells, which were evenly distributed between islet periphery and intraislet vascular channels, PP and δ cells had asymmetric and opposite distributions: PP staining was higher and somatostatin staining was lower in the islet periphery than in the area around intraislet vascular channels. Additionally, frequencies of PP and δ cells were negatively correlated in the islets. No difference was observed between islets from the head and the tail of the pancreas, and from type 2 diabetic and non-diabetic donors. In conclusion, the distribution of δ cells differs from that of PP cells in human islets, suggesting that vessels at the periphery and at the centre of islets drain different hormonal cocktails.


Subject(s)
Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/metabolism , Somatostatin-Secreting Cells/cytology , Somatostatin-Secreting Cells/metabolism , Adolescent , Adult , Aged , Fluorescent Antibody Technique , Humans , Middle Aged , Pancreatic Polypeptide/metabolism , Somatostatin/metabolism , Tissue Distribution , Young Adult
13.
PLoS One ; 10(12): e0144597, 2015.
Article in English | MEDLINE | ID: mdl-26658466

ABSTRACT

The transcription factor Pax6 is an important regulator of development and cell differentiation in various organs. Thus, Pax6 was shown to promote neural development in the cerebral cortex and spinal cord, and to control pancreatic endocrine cell genesis. However, the role of Pax6 in distinct endocrine cells of the adult pancreas has not been addressed. We report the conditional inactivation of Pax6 in insulin and glucagon producing cells of the adult mouse pancreas. In the absence of Pax6, beta- and alpha-cells lose their molecular maturation characteristics. Our findings provide strong evidence that Pax6 is responsible for the maturation of beta-, and alpha-cells, but not of delta-, and PP-cells. Moreover, lineage-tracing experiments demonstrate that Pax6-deficient beta- and alpha-cells are shunted towards ghrelin marked cells, sustaining the idea that ghrelin may represent a marker for endocrine cell maturation.


Subject(s)
Eye Proteins/genetics , Ghrelin/genetics , Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/genetics , Insulin-Secreting Cells/metabolism , Paired Box Transcription Factors/genetics , Pancreatic Polypeptide-Secreting Cells/metabolism , Repressor Proteins/genetics , Somatostatin-Secreting Cells/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Differentiation , Cell Lineage/drug effects , Cell Lineage/genetics , Crosses, Genetic , Eye Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Ghrelin/metabolism , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/drug effects , Homeodomain Proteins/metabolism , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Integrases/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Knockout , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/drug effects , Repressor Proteins/metabolism , Signal Transduction , Somatostatin-Secreting Cells/cytology , Somatostatin-Secreting Cells/drug effects , Tamoxifen/pharmacology
14.
Tissue Cell ; 46(6): 535-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25458814

ABSTRACT

tIn this study, we investigated the presence of ovoid or ellipsoidal amylin-immunoreactive cells of the pancreatic islets of the black-spotted frog Rana (Pelophylax) nigromaculata. Using double immunofluorescent staining, all amylin-immunoreactive cells were shown to be immuno-negative for insulin, glucagon, and somatostatin, and they were often observed in peripheral regions of clusters of insulin-immunoreactive cells. Under immunoelectron microscopy, amylin-immunoreactive signals were detected on the secretory granules in a specific type of endocrine cells. From our results, we conclude that the amylin-immunoreactive cells correspond to X cells among the 4 distinct types of endocrine cells (B, A/PP, D, and X) previously identified in the frog. Amylin secreted from X cells may regulate the hormone secretion from A/PP cells and/or B cells through a paracrine mechanism.


Subject(s)
Insulin/metabolism , Islet Amyloid Polypeptide/metabolism , Pancreas/metabolism , Animals , Connective Tissue/metabolism , Glucagon , Glucagon-Secreting Cells/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/immunology , Pancreas/cytology , Pancreatic Polypeptide-Secreting Cells/metabolism , Ranidae
15.
Pancreas ; 43(4): 648-50, 2014 May.
Article in English | MEDLINE | ID: mdl-24713672

ABSTRACT

A 74-year-old man with recurrent duodenal ulcers underwent somatostatin receptor scintigraphy (SRS) in suspicion of gastrinoma. A 2-cm area of focal uptake was visualized within the pancreatic head. Serum chromogranin A levels were elevated, but serum gastrin levels and the secretin test were normal. Computed tomography and endoscopic ultrasonography were not conclusive. After partial duodenopancreatectomy, pathological examination failed to reveal any neuroendocrine tumor. Instead, the dorsal portion of the pancreatic head was found to be densely populated by pancreatic polypeptide cell-rich islets. This area correlated with the site of tracer uptake seen on SRS. Thus, pancreatic polypeptide cell-rich islets in elderly patients should be kept in mind when interpreting SRS results to avoid unnecessary major pancreatic resections.


Subject(s)
Diagnostic Errors , Neuroendocrine Tumors/diagnostic imaging , Octreotide/analogs & derivatives , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Polypeptide-Secreting Cells/diagnostic imaging , Radiopharmaceuticals , Receptors, Somatostatin/metabolism , Tomography, Emission-Computed, Single-Photon , Aged , Biopsy , Humans , Male , Multimodal Imaging/methods , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/surgery , Pancreatectomy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/surgery , Pancreatic Polypeptide-Secreting Cells/metabolism , Predictive Value of Tests , Tomography, X-Ray Computed , Unnecessary Procedures
16.
Regul Pept ; 187: 42-50, 2013 Nov 10.
Article in English | MEDLINE | ID: mdl-24183983

ABSTRACT

Xenin-25 (Xen) is a 25-amino acid neurotensin-related peptide that activates neurotensin receptor-1 (NTSR1). We previously showed that Xen increases the effect of glucose-dependent insulinotropic polypeptide (GIP) on insulin release 1) in hyperglycemic mice via a cholinergic relay in the periphery independent from the central nervous system and 2) in humans with normal or impaired glucose tolerance, but not type 2 diabetes mellitus (T2DM). Since this blunted response to Xen defines a novel defect in T2DM, it is important to understand how Xen regulates islet physiology. On separate visits, subjects received intravenous graded glucose infusions with vehicle, GIP, Xen, or GIP plus Xen. The pancreatic polypeptide response was used as an indirect measure of cholinergic input to islets. The graded glucose infusion itself had little effect on the pancreatic polypeptide response whereas administration of Xen equally increased the pancreatic polypeptide response in humans with normal glucose tolerance, impaired glucose tolerance, and T2DM. The pancreatic polypeptide response to Xen was similarly amplified by GIP in all 3 groups. Antibody staining of human pancreas showed that NTSR1 is not detectable on islet endocrine cells, sympathetic neurons, blood vessels, or endothelial cells but is expressed at high levels on PGP9.5-positive axons in the exocrine tissue and at low levels on ductal epithelial cells. PGP9.5 positive nerve fibers contacting beta cells in the islet periphery were also observed. Thus, a neural relay, potentially involving muscarinic acetylcholine receptors, indirectly increases the effects of Xen on pancreatic polypeptide release in humans.


Subject(s)
Diabetes Mellitus, Type 2/blood , Gastric Inhibitory Polypeptide/pharmacology , Neurotensin/pharmacology , Pancreas/innervation , Pancreatic Polypeptide/metabolism , Adult , Blood Glucose , Case-Control Studies , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Female , Humans , Male , Middle Aged , Pancreas/drug effects , Pancreas/metabolism , Pancreatic Polypeptide/blood , Pancreatic Polypeptide-Secreting Cells/metabolism , Receptors, Neurotensin/metabolism
17.
PLoS One ; 8(8): e72213, 2013.
Article in English | MEDLINE | ID: mdl-23977255

ABSTRACT

Mutations in the human homolog of the Vhlh gene [encoding the von-Hippel Lindau (VHL) protein] lead to tumor development. In mice, depletion of Vhlh in pancreatic ß-cells causes perturbed glucose homeostasis, but the role of this gene in other pancreatic cells is poorly understood. To investigate the function of VHL/HIF pathway in pancreatic cells, we inactivated Vhlh in the pancreatic epithelium as well as in the endocrine and exocrine lineages. Our results show that embryonic depletion of Vhlh within the pancreatic epithelium causes postnatal lethality due to severe hypoglycemia. The hypoglycemia is recapitulated in mice with endocrine-specific removal of Vhlh, while animals with loss of Vhlh predominantly in the exocrine compartment survive to adulthood with no overt defects in glucose metabolism. Mice with hypoglycemia display diminished insulin release in response to elevated glucose. Significantly, the glucagon response is impaired both in vivo (circulating glucagon levels) as well as in an in vitro secretion assay in isolated islets. Hypoxia also impairs glucagon secretion in a glucagon-expressing cell line in culture. Our results reveal a novel role for the hypoxia/HIF pathway in islet hormone secretion and maintenance of the fine balance that allows for the establishment of normoglycemia.


Subject(s)
Endocrine System/metabolism , Glucagon-Secreting Cells/metabolism , Hypoxia-Inducible Factor 1/genetics , Hypoxia/genetics , Somatostatin-Secreting Cells/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Cells, Cultured , Embryo, Mammalian , Endocrine System/pathology , Gene Deletion , Gene Expression Regulation, Developmental , Glucagon/metabolism , Glucagon-Secreting Cells/pathology , Glucose/metabolism , Homeostasis/genetics , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Mice, Knockout , Pancreatic Polypeptide-Secreting Cells/metabolism , Pancreatic Polypeptide-Secreting Cells/pathology , Signal Transduction , Somatostatin-Secreting Cells/pathology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
18.
PLoS One ; 8(1): e55501, 2013.
Article in English | MEDLINE | ID: mdl-23383206

ABSTRACT

The pancreatic islet is mainly composed of beta-, alpha- and delta-cells with small numbers of pancreatic polypeptide (PP) and epsilon cells. It is known that there is a region in the head of the pancreas that is rich in PP-cells. In the present study, we examined the distribution of PP-cells, and assessed the influence of the PP-cell rich region to quantify the total islet mass. Pancreatic tissues were collected from donors with no history of diabetes or pancreatic diseases (n = 12). A stereological approach with a computer-assisted large-scale analysis of whole pancreatic sections was applied to quantify the entire distribution of endocrine cells within a given section. The initial whole pancreas analysis showed that a PP-cell rich region was largely restricted to the uncinate process with a clear boundary. The distinct distribution of PP-cells includes irregularly shaped clusters composed solely of PP-cells. Furthermore, in the PP-cell rich region, beta- and alpha-cell mass is significantly reduced compared to surrounding PP-cell poor regions. The results suggest that the analysis of the head region should distinguish the PP-cell rich region, which is best examined separately. This study presents an important implication for the regional selection and interpretation of the results.


Subject(s)
Pancreas/cytology , Pancreatic Polypeptide-Secreting Cells/cytology , Glucagon-Secreting Cells/cytology , Humans , Immunohistochemistry , Insulin-Secreting Cells/cytology , Islets of Langerhans , Pancreas/anatomy & histology , Pancreas/metabolism , Pancreatic Polypeptide-Secreting Cells/metabolism
19.
Pancreas ; 39(6): 836-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20182388

ABSTRACT

OBJECTIVES: Somatostatin inhibits hormone release through 5 G protein-coupled somatostatin receptors (sst1-sst5). However, the role of somatostatin in islet physiology is not fully known. The immunoreactivity to sst1 to sst5 in normal human endocrine pancreas has been described. The present study reports the expression of sst1 to sst5 in human pancreatic islets with type 2 diabetes mellitus. METHODS: Pancreatic autopsy specimens from individuals with type 2 diabetes mellitus and matched controls were double immunostained to demonstrate sst1 to sst5 in the major islet cell types. RESULTS: Most apparent differences in type 2 diabetic islets were the lack of sst1 and sst4 in glucagon cells and sst1-3 and 4 in somatostatin cells, whereas minor changes were demonstrated in insulin cells. The pancreatic polypeptide cells showed a reversed staining pattern in diabetic islets compared with the controls. CONCLUSIONS: In type 2 diabetes mellitus, the sst pattern differed from that of the controls in somatostatin, pancreatic polypeptide, and glucagon cells, to a minor extent in insulin cells. It is unclear whether the changes in sst patterns are primarily due to the diabetes or secondary to metabolic disturbances. However, this study may be the basis for further functional studies to evaluate the role of sst1 to sst5 in the diabetic state.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , Receptors, Somatostatin/metabolism , Aged , Aged, 80 and over , Autopsy , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Immunohistochemistry , Male , Pancreatic Polypeptide-Secreting Cells/metabolism , Protein Isoforms/metabolism , Somatostatin-Secreting Cells/metabolism
20.
Eur J Histochem ; 53(2): 81-5, 2009.
Article in English | MEDLINE | ID: mdl-19683981

ABSTRACT

We used immunofluorescence double staining method to investigate the cellular localization of glucagon and pancreatic polypeptide (PP) in rat pancreatic islets. The results showed that both A-cells (glucagon-secreting cells) and PP-cells (PP-secreting cells) were located in the periphery of the islets. However, A-cells and PP-cells had a different regional distribution. Most of A-cells were located in the splenic lobe but a few of them were in the duodenal lobe of the pancreas. In contrast, the majority of PP-cells were found in the duodenal lobe and a few of them were in the splenic lobe of the pancreas. Furthermore, we found that 67.74% A-cells had PP immunoreactivity, 70.92% PP-cells contained glucagon immunoreactivity with immunofluorescence double staining. Our data support the concept of a common precursor stem cell for pancreatic hormone-producing cells.


Subject(s)
Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pancreatic Polypeptide-Secreting Cells/metabolism , Pancreatic Polypeptide/metabolism , Animals , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...