1
|
Lafferty RA, Flatt PR, Irwin N. NPYR modulation: Potential for the next major advance in obesity and type 2 diabetes management? Peptides 2024; 179:171256. [PMID: 38825012 DOI: 10.1016/j.peptides.2024.171256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
The approval of the glucagon-like peptide 1 (GLP-1) mimetics semaglutide and liraglutide for management of obesity, independent of type 2 diabetes (T2DM), has initiated a resurgence of interest in gut-hormone derived peptide therapies for the management of metabolic diseases, but side-effect profile is a concern for these medicines. However, the recent approval of tirzepatide for obesity and T2DM, a glucose-dependent insulinotropic polypeptide (GIP), GLP-1 receptor co-agonist peptide therapy, may provide a somewhat more tolerable option. Despite this, an increasing number of non-incretin alternative peptides are in development for obesity, and it stands to reason that other hormones will take to the limelight in the coming years, such as peptides from the neuropeptide Y family. This narrative review outlines the therapeutic promise of the neuropeptide Y family of peptides, comprising of the 36 amino acid polypeptides neuropeptide Y (NPY), peptide tyrosine-tyrosine (PYY) and pancreatic polypeptide (PP), as well as their derivatives. This family of peptides exerts a number of metabolically relevant effects such as appetite regulation and can influence pancreatic beta-cell survival. Although some of these actions still require full translation to the human setting, potential therapeutic application in obesity and type 2 diabetes is conceivable. However, like GLP-1 and GIP, the endogenous NPY, PYY and PP peptide forms are subject to rapid in vivo degradation and inactivation by the serine peptidase, dipeptidyl-peptidase 4 (DPP-4), and hence require structural modification to prolong circulating half-life. Numerous protective modification strategies are discussed in this regard herein, alongside related impact on biological activity profile and therapeutic promise.
Collapse
Affiliation(s)
- Ryan A Lafferty
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK.
| | - Peter R Flatt
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Nigel Irwin
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| |
Collapse
|
2
|
Dasmahapatra AK, Tchounwou PB. Evaluation of pancreatic δ- cells as a potential target site of graphene oxide toxicity in Japanese medaka (Oryzias latipes) fish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114649. [PMID: 36806823 PMCID: PMC10032203 DOI: 10.1016/j.ecoenv.2023.114649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/23/2023] [Accepted: 02/11/2023] [Indexed: 06/18/2023]
Abstract
In continuation to our previous investigations on graphene oxide (GO) as an endocrine disrupting chemical (EDC), in the present experiment, we have investigated endocrine pancreas of Japanese medaka adults focusing on δ-cells in the islet organs as an endpoint. Breeding pairs of adult male and female fish were exposed to 0 mg/L (control) or 20 mg/L GO by continuous immersion (IMR) for 96 h, or to 0 µg/g or 100 µg/g GO by a single intraperitoneal (IP) administration and depurated 21 days in a GO-free environment. Histological investigations indicated that the endocrine cells are concentrated in one large principal islet, and several small secondary islets scattered within the mesentery near the liver and intestine. The cells of the islet organ are in various shapes with basophilic nuclei and eosinophilic cytoplasm. Immunohistochemical evaluation using rabbit polyclonal antisomatostatin antibody indicated that immunoreactivity is localized either at the periphery or at the central region in principal islets, and throughout the secondary islets, and found to be enhanced in fish exposed to GO than controls. The soma of δ-cells exhibits neuron-like morphology and have filopodia like processes. Cell sorting as non-communicating δ-cells (NCDC), communicating cells (CC), and non- δ-cells (NDC) indicated that within an islet organ, the population of NDCC is found to be the least and NDC is the highest. Our data further indicated that GO-induced impairments in the islet organs of medaka pancreas are inconsistent and could be affected by the exposure roots as well as the sex of the fish.
Collapse
Affiliation(s)
- Asok K Dasmahapatra
- RCMI Center for Environmental Health, Jackson State University, Jackson, MS 39217, USA; Department of BioMolecular Sciences, Environmental Toxicology Division, University of Mississippi, University, MS 38677, USA
| | - Paul B Tchounwou
- RCMI Center for Environmental Health, Jackson State University, Jackson, MS 39217, USA.
| |
Collapse
|
3
|
Khan D, Moffett RC, Flatt PR, Tarasov AI. Classical and non-classical islet peptides in the control of β-cell function. Peptides 2022; 150:170715. [PMID: 34958851 DOI: 10.1016/j.peptides.2021.170715] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/25/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022]
Abstract
The dual role of the pancreas as both an endocrine and exocrine gland is vital for food digestion and control of nutrient metabolism. The exocrine pancreas secretes enzymes into the small intestine aiding digestion of sugars and fats, whereas the endocrine pancreas secretes a cocktail of hormones into the blood, which is responsible for blood glucose control and regulation of carbohydrate, protein and fat metabolism. Classical islet hormones, insulin, glucagon, pancreatic polypeptide and somatostatin, interact in an autocrine and paracrine manner, to fine-tube the islet function and insulin secretion to the needs of the body. Recently pancreatic islets have been reported to express a number of non-classical peptide hormones involved in metabolic signalling, whose major production site was believed to reside outside pancreas, e.g. in the small intestine. We highlight the key non-classical islet peptides, and consider their involvement, together with established islet hormones, in regulation of stimulus-secretion coupling as well as proliferation, survival and transdifferentiation of β-cells. We furthermore focus on the paracrine interaction between classical and non-classical islet hormones in the maintenance of β-cell function. Understanding the functional relationships between these islet peptides might help to develop novel, more efficient treatments for diabetes and related metabolic disorders.
Collapse
Affiliation(s)
- Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| | - R Charlotte Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Andrei I Tarasov
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
4
|
Fukaishi T, Nakagawa Y, Fukunaka A, Sato T, Hara A, Nakao K, Saito M, Kohno K, Miyatsuka T, Tamaki M, Matsuhisa M, Matsuoka TA, Yamada T, Watada H, Fujitani Y. Characterisation of Ppy-lineage cells clarifies the functional heterogeneity of pancreatic beta cells in mice. Diabetologia 2021; 64:2803-2816. [PMID: 34498099 PMCID: PMC8563568 DOI: 10.1007/s00125-021-05560-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/28/2021] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS Pancreatic polypeptide (PP) cells, which secrete PP (encoded by the Ppy gene), are a minor population of pancreatic endocrine cells. Although it has been reported that the loss of beta cell identity might be associated with beta-to-PP cell-fate conversion, at present, little is known regarding the characteristics of Ppy-lineage cells. METHODS We used Ppy-Cre driver mice and a PP-specific monoclonal antibody to investigate the association between Ppy-lineage cells and beta cells. The molecular profiles of endocrine cells were investigated by single-cell transcriptome analysis and the glucose responsiveness of beta cells was assessed by Ca2+ imaging. Diabetic conditions were experimentally induced in mice by either streptozotocin or diphtheria toxin. RESULTS Ppy-lineage cells were found to contribute to the four major types of endocrine cells, including beta cells. Ppy-lineage beta cells are a minor subpopulation, accounting for 12-15% of total beta cells, and are mostly (81.2%) localised at the islet periphery. Unbiased single-cell analysis with a Ppy-lineage tracer demonstrated that beta cells are composed of seven clusters, which are categorised into two groups (i.e. Ppy-lineage and non-Ppy-lineage beta cells). These subpopulations of beta cells demonstrated distinct characteristics regarding their functionality and gene expression profiles. Ppy-lineage beta cells had a reduced glucose-stimulated Ca2+ signalling response and were increased in number in experimental diabetes models. CONCLUSIONS/INTERPRETATION Our results indicate that an unexpected degree of beta cell heterogeneity is defined by Ppy gene activation, providing valuable insight into the homeostatic regulation of pancreatic islets and future therapeutic strategies against diabetes. DATA AVAILABILITY The single-cell RNA sequence (scRNA-seq) analysis datasets generated in this study have been deposited in the Gene Expression Omnibus (GEO) under the accession number GSE166164 ( www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE166164 ).
Collapse
Affiliation(s)
- Takahiro Fukaishi
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuko Nakagawa
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Ayako Fukunaka
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Takashi Sato
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Akemi Hara
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Physiology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Keiko Nakao
- Department of Physiology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Michiko Saito
- Institute for Research Initiatives, Nara Institute of Science and Technology (NAIST), Nara, Japan
- Bio-science Research Center, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kenji Kohno
- Institute for Research Initiatives, Nara Institute of Science and Technology (NAIST), Nara, Japan
| | - Takeshi Miyatsuka
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Motoyuki Tamaki
- Diabetes Therapeutics and Research Center, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Munehide Matsuhisa
- Diabetes Therapeutics and Research Center, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Taka-Aki Matsuoka
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tetsuya Yamada
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hirotaka Watada
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshio Fujitani
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan.
| |
Collapse
|
5
|
Yang CH, Onda DA, Oakhill JS, Scott JW, Galic S, Loh K. Regulation of Pancreatic β-Cell Function by the NPY System. Endocrinology 2021; 162:6213414. [PMID: 33824978 DOI: 10.1210/endocr/bqab070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Indexed: 01/24/2023]
Abstract
The neuropeptide Y (NPY) system has been recognized as one of the most critical molecules in the regulation of energy homeostasis and glucose metabolism. Abnormal levels of NPY have been shown to contribute to the development of metabolic disorders including obesity, cardiovascular diseases, and diabetes. NPY centrally promotes feeding and reduces energy expenditure, while the other family members, peptide YY (PYY) and pancreatic polypeptide (PP), mediate satiety. New evidence has uncovered additional functions for these peptides that go beyond energy expenditure and appetite regulation, indicating a more extensive function in controlling other physiological functions. In this review, we will discuss the role of the NPY system in the regulation of pancreatic β-cell function and its therapeutic implications for diabetes.
Collapse
Affiliation(s)
- Chieh-Hsin Yang
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Danise-Ann Onda
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Jonathan S Oakhill
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC 3000, Australia
| | - John W Scott
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC 3000, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010, Australia
| | - Sandra Galic
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| | - Kim Loh
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
6
|
Perez-Frances M, van Gurp L, Abate MV, Cigliola V, Furuyama K, Bru-Tari E, Oropeza D, Carreaux T, Fujitani Y, Thorel F, Herrera PL. Pancreatic Ppy-expressing γ-cells display mixed phenotypic traits and the adaptive plasticity to engage insulin production. Nat Commun 2021; 12:4458. [PMID: 34294685 PMCID: PMC8298494 DOI: 10.1038/s41467-021-24788-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 07/08/2021] [Indexed: 02/06/2023] Open
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 β-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 β-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.
Collapse
Affiliation(s)
- Marta Perez-Frances
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Léon van Gurp
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Maria Valentina Abate
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Valentina Cigliola
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Regeneration Next, Duke University, Durham, NC, USA
| | - Kenichiro Furuyama
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Eva Bru-Tari
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Daniel Oropeza
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Taïna Carreaux
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Yoshio Fujitani
- Lab. of Developmental Biology & Metabolism, Institute for Molecular & Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Fabrizio Thorel
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pedro L Herrera
- Department of Genetic Medicine & Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW Studies have identified several effects of bile acids (BAs) in glucose homeostasis, energy expenditure, and body weight control, through receptor-dependent and independent mechanisms. BAs are produced from cholesterol and characterized by their structures, which result from enzymes in the liver and the gut microbiota. The aim of this review is to characterize the effects of BA structure and composition on diabetes. RECENT FINDINGS The hydroxyl groups of BAs interact with binding pockets of receptors and enzymes that affect glucose homeostasis. Human and animal studies show that BA composition is associated with insulin resistance and food intake regulation. The hydroxylation of BAs and BA composition contributes to glucose regulation. Modulation of BA composition has the potential to improve glucose metabolism.
Collapse
Affiliation(s)
- Sei Higuchi
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
- Russ Berrie Pavilion, Room 315, 1150 St. Nicholas Ave., New York, NY, 10032, USA.
| |
Collapse
|
8
|
Russell R, Carnese PP, Hennings TG, Walker EM, Russ HA, Liu JS, Giacometti S, Stein R, Hebrok M. Loss of the transcription factor MAFB limits β-cell derivation from human PSCs. Nat Commun 2020; 11:2742. [PMID: 32488111 PMCID: PMC7265500 DOI: 10.1038/s41467-020-16550-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
Next generation sequencing studies have highlighted discrepancies in β-cells which exist between mice and men. Numerous reports have identified MAF BZIP Transcription Factor B (MAFB) to be present in human β-cells postnatally, while its expression is restricted to embryonic and neo-natal β-cells in mice. Using CRISPR/Cas9-mediated gene editing, coupled with endocrine cell differentiation strategies, we dissect the contribution of MAFB to β-cell development and function specifically in humans. Here we report that MAFB knockout hPSCs have normal pancreatic differentiation capacity up to the progenitor stage, but favor somatostatin- and pancreatic polypeptide–positive cells at the expense of insulin- and glucagon-producing cells during endocrine cell development. Our results describe a requirement for MAFB late in the human pancreatic developmental program and identify it as a distinguishing transcription factor within islet cell subtype specification. We propose that hPSCs represent a powerful tool to model human pancreatic endocrine development and associated disease pathophysiology. The MAF bZIP transcription factor B (MAFB) is present in postnatal human beta cells but its role is unclear. Here, the authors show that MAFB regulates endocrine pancreatic cell fate specification.
Collapse
Affiliation(s)
- Ronan Russell
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Phichitpol P Carnese
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Thomas G Hennings
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Emily M Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Holger A Russ
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA.,Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Jennifer S Liu
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Simone Giacometti
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Matthias Hebrok
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
9
|
Chen X, Zhang J, Zhou Z. Targeting Islets: Metabolic Surgery Is More than a Bariatric Surgery. Obes Surg 2019; 29:3001-3009. [DOI: 10.1007/s11695-019-03979-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
10
|
Hara A, Nakagawa Y, Nakao K, Tamaki M, Ikemoto T, Shimada M, Matsuhisa M, Mizukami H, Maruyama N, Watada H, Fujitani Y. Development of monoclonal mouse antibodies that specifically recognize pancreatic polypeptide. Endocr J 2019; 66:459-468. [PMID: 30842364 DOI: 10.1507/endocrj.ej18-0441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Pancreatic polypeptide (PP) is a 36-amino acid peptide encoded by the Ppy gene, which is produced by a small population of cells located in the periphery of the islets of Langerhans. Owing to the high amino acid sequence similarity among neuropeptide Y family members, antibodies against PP that are currently available are not convincingly specific to PP. Here we report the development of mouse monoclonal antibodies that specifically bind to PP. We generated Ppy knockout (Ppy-KO) mice in which the Ppy-coding region was replaced by Cre recombinase. The Ppy-KO mice were immunized with mouse PP peptide, and stable hybridoma cell lines producing anti-PP antibodies were isolated. Firstly, positive clones were selected in an enzyme-linked immunosorbent assay for reactivity with PP coupled to bovine serum albumin. During the screening, hybridoma clones producing antibodies that cross-react to the peptide YY (PYY) were excluded. In the second screening, hybridoma clones in which their culture media produce no signal in Ppy-KO islets but detect specific cells in the peripheral region of wild-type islets, were selected. Further studies demonstrated that the selected monoclonal antibody (23-2D3) specifically recognizes PP-producing cells, not only in mouse, but also in human and rat islets. The monoclonal antibodies with high binding specificity for PP developed in this study will be fundamental for future studies towards elucidating the expression profiles and the physiological roles of PP.
Collapse
Affiliation(s)
- Akemi Hara
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Center for Therapeutic Innovation in Diabetes, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yuko Nakagawa
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
| | - Keiko Nakao
- Department of Physiology, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan
| | - Motoyuki Tamaki
- Diabetes Therapeutics and Research Center, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Tetsuya Ikemoto
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Mitsuo Shimada
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima 770-8503, Japan
| | - Munehide Matsuhisa
- Diabetes Therapeutics and Research Center, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Aomori 036-8562, Japan
| | | | - Hirotaka Watada
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Center for Therapeutic Innovation in Diabetes, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yoshio Fujitani
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Center for Therapeutic Innovation in Diabetes, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
| |
Collapse
|
11
|
Jørgensen MC, de Lichtenberg KH, Collin CA, Klinck R, Ekberg JH, Engelstoft MS, Lickert H, Serup P. Neurog3-dependent pancreas dysgenesis causes ectopic pancreas in Hes1 mutant mice. Development 2018; 145:dev.163568. [PMID: 30093553 DOI: 10.1242/dev.163568] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022]
Abstract
Mutations in Hes1, a target gene of the Notch signalling pathway, lead to ectopic pancreas by a poorly described mechanism. Here, we use genetic inactivation of Hes1 combined with lineage tracing and live imaging to reveal an endodermal requirement for Hes1, and show that ectopic pancreas tissue is derived from the dorsal pancreas primordium. RNA-seq analysis of sorted E10.5 Hes1+/+ and Hes1-/- Pdx1-GFP+ cells suggested that upregulation of endocrine lineage genes in Hes1-/- embryos was the major defect and, accordingly, early pancreas morphogenesis was normalized, and the ectopic pancreas phenotype suppressed, in Hes1-/-Neurog3-/- embryos. In Mib1 mutants, we found a near total depletion of dorsal progenitors, which was replaced by an anterior Gcg+ extension. Together, our results demonstrate that aberrant morphogenesis is the cause of ectopic pancreas and that a part of the endocrine differentiation program is mechanistically involved in the dysgenesis. Our results suggest that the ratio of endocrine lineage to progenitor cells is important for morphogenesis and that a strong endocrinogenic phenotype without complete progenitor depletion, as seen in Hes1 mutants, provokes an extreme dysgenesis that causes ectopic pancreas.
Collapse
Affiliation(s)
- Mette C Jørgensen
- NNF Center for Stem Cell Biology (DanStem), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Kristian H de Lichtenberg
- NNF Center for Stem Cell Biology (DanStem), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Caitlin A Collin
- NNF Center for Stem Cell Biology (DanStem), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Rasmus Klinck
- Novo Nordisk A/S, Department of User Research and Communication, Brennum Park 1, DK-3400 Hillerød, Denmark
| | - Jeppe H Ekberg
- NNF Center for Basic Metabolic Research, Section for Metabolic Receptology, Laboratory for Molecular Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Maja S Engelstoft
- NNF Center for Basic Metabolic Research, Section for Metabolic Receptology, Laboratory for Molecular Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Heiko Lickert
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Palle Serup
- NNF Center for Stem Cell Biology (DanStem), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
12
|
Guida C, McCulloch LJ, Godazgar M, Stephen SD, Baker C, Basco D, Dong J, Chen D, Clark A, Ramracheya RD. Sitagliptin and Roux-en-Y gastric bypass modulate insulin secretion via regulation of intra-islet PYY. Diabetes Obes Metab 2018; 20:571-581. [PMID: 28892258 PMCID: PMC5836881 DOI: 10.1111/dom.13113] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/28/2017] [Accepted: 09/05/2017] [Indexed: 12/14/2022]
Abstract
AIMS The gut hormone peptide tyrosine tyrosine (PYY) is critical for maintaining islet integrity and restoring islet function following Roux-en-Y gastric bypass (RYGB). The expression of PYY and its receptors (NPYRs) in islets has been documented but not fully characterized. Modulation of islet PYY by the proteolytic enzyme dipeptidyl peptidase IV (DPP-IV) has not been investigated and the impact of DPP-IV inhibition on islet PYY function remains unexplored. Here we have addressed these gaps and their effects on glucose-stimulated insulin secretion (GSIS). We have also investigated changes in pancreatic PYY in diabetes and following RYGB. METHODS Immunohistochemistry and gene expression analysis were used to assess PYY, NPYRs and DPP-IV expression in rodent and human islets. DPP-IV activity inhibition was achieved by sitagliptin. Secretion studies were used to test PYY and the effects of sitagliptin on insulin release, and the involvement of GLP-1. Radioimmunoassays were used to measure hormone content in islets. RESULTS PYY and DPP-IV localized in different cell types in islets while NPYR expression was confined to the beta-cells. Chronic PYY application enhanced GSIS in rodent and diabetic human islets. DPP-IV inhibition by sitagliptin potentiated GSIS; this was mediated by locally-produced PYY, and not GLP-1. Pancreatic PYY was markedly reduced in diabetes. RYGB strongly increased islet PYY content, but did not lead to full restoration of pancreatic GLP-1 levels. CONCLUSION Local regulation of pancreatic PYY, rather than GLP-1, by DPP-IV inhibition or RYGB can directly modulate the insulin secretory response to glucose, indicating a novel role of pancreatic PYY in diabetes and weight-loss surgery.
Collapse
Affiliation(s)
- Claudia Guida
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Laura J. McCulloch
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Mahdieh Godazgar
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Sam D. Stephen
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Charlotte Baker
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Davide Basco
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | | | - Duan Chen
- Department of Clinical and Molecular MedicineNorwegian University of Science and TechnologyTrondheimNorway
| | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| | - Reshma D Ramracheya
- Oxford Centre for Diabetes, Endocrinology and MetabolismChurchill Hospital, Oxford UniversityOxfordUK
| |
Collapse
|
13
|
Franklin ZJ, Tsakmaki A, Fonseca Pedro P, King AJ, Huang GC, Amjad S, Persaud SJ, Bewick GA. Islet neuropeptide Y receptors are functionally conserved and novel targets for the preservation of beta-cell mass. Diabetes Obes Metab 2018; 20:599-609. [PMID: 28940946 DOI: 10.1111/dom.13119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/13/2017] [Accepted: 09/16/2017] [Indexed: 12/27/2022]
Abstract
AIMS Two unmet therapeutic strategies for diabetes treatment are prevention of beta-cell death and stimulation of beta-cell replication. Our aim was to characterize the role of neuropeptide Y receptors in the control of beta-cell mass. MATERIALS AND METHODS We used endogenous and selective agonists of the NPY receptor system to explore its role in the prevention of beta-cell apoptosis and proliferation in islets isolated from both mouse and human donors. We further explored the intra-cellular signalling cascades involved, using chemical inhibitors of key signalling pathways. As proof of principle we designed a long-acting analogue of [Leu31 Pro34 ]-NPY, an agonist of the islet-expressed Y receptors, to determine if targeting this system could preserve beta-cell mass in vivo. RESULTS Our data reveal that NPY Y1, 4 and 5 receptor activation engages a generalized and powerful anti-apoptotic pathway that protects mouse and human islets from damage. These anti-apoptotic effects were dependent on stimulating a Gαi-PLC-PKC signalling cascade, which prevented cytokine-induced NFkB signalling. NPY receptor activation functionally protected islets by restoring glucose responsiveness following chemically induced injury in both species. NPY receptor activation attenuated beta-cell apoptosis, preserved functional beta-cell mass and attenuated the hyperglycaemic phenotype in a low-dose streptozotocin model of diabetes. CONCLUSION Taken together, our observations identify the islet Y receptors as promising targets for the preservation of beta-cell mass. As such, targeting these receptors could help to maintain beta-cell mass in both type 1 and type 2 diabetes, and may also be useful for improving islet transplantation outcomes.
Collapse
Affiliation(s)
- Zara J Franklin
- Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Anastasia Tsakmaki
- Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | | | - Aileen J King
- Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Guo Cai Huang
- Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Sakeena Amjad
- Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Shanta J Persaud
- Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Gavin A Bewick
- Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| |
Collapse
|
14
|
Qiu WL, Zhang YW, Feng Y, Li LC, Yang L, Xu CR. Deciphering Pancreatic Islet β Cell and α Cell Maturation Pathways and Characteristic Features at the Single-Cell Level. Cell Metab 2017; 25:1194-1205.e4. [PMID: 28467935 DOI: 10.1016/j.cmet.2017.04.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/03/2017] [Accepted: 04/05/2017] [Indexed: 02/02/2023]
Abstract
Pancreatic β and α cells play essential roles in maintaining glucose homeostasis. However, the mechanisms by which these distinct cell populations are generated, expand, and mature during pancreas development remain unclear. In this study, we addressed this critical question by performing a single-cell transcriptomic analysis of mouse β and α cells sorted from fetal to adult stages. We discovered that β and α cells use different regulatory strategies for their maturation and that cell proliferation peaks at different developmental times. However, the quiescent and proliferative cells in both the β lineage and α lineage are synchronous in their maturation states. The heterogeneity of juvenile β cells reflects distinct cell-cycling phases, origins, and maturation states, whereas adult β cells are relatively homogeneous at the transcriptomic level. These analyses provide not only a high-resolution roadmap for islet lineage development but also insights into the mechanisms of cellular heterogeneity, cell number expansion, and maturation of both β and α cells.
Collapse
Affiliation(s)
- Wei-Lin Qiu
- Ministry of Education Key Laboratory of Cell Proliferation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; PKU-Tsinghua-NIBS Graduate Program, Peking University, Beijing 100871, China
| | - Yu-Wei Zhang
- Ministry of Education Key Laboratory of Cell Proliferation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Ye Feng
- Ministry of Education Key Laboratory of Cell Proliferation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; PKU-Tsinghua-NIBS Graduate Program, Peking University, Beijing 100871, China
| | - Lin-Chen Li
- Ministry of Education Key Laboratory of Cell Proliferation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Liu Yang
- Ministry of Education Key Laboratory of Cell Proliferation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Cheng-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
15
|
Stanescu DE, Yu R, Won KJ, Stoffers DA. Single cell transcriptomic profiling of mouse pancreatic progenitors. Physiol Genomics 2016; 49:105-114. [PMID: 28011883 DOI: 10.1152/physiolgenomics.00114.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 12/20/2022] Open
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.
Collapse
Affiliation(s)
- Diana E Stanescu
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Reynold Yu
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Kyoung-Jae Won
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Doris A Stoffers
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; .,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
16
|
Khan D, Vasu S, Moffett RC, Irwin N, Flatt PR. Islet distribution of Peptide YY and its regulatory role in primary mouse islets and immortalised rodent and human beta-cell function and survival. Mol Cell Endocrinol 2016; 436:102-13. [PMID: 27465830 DOI: 10.1016/j.mce.2016.07.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022]
Abstract
Recent evidence suggests that the classic gut peptide, Peptide YY (PYY), could play a fundamental role in endocrine pancreatic function. In the present study expression of PYY and its NPY receptors on mouse islets and immortalised rodent and human beta-cells was examined together with the effects of both major circulating forms of PYY, namely PYY(1-36) and PYY(3-36), on beta-cell function, murine islet adaptions to insulin deficiency/resistance, as well as direct effects on cultured beta-cell proliferation and apoptosis. In vivo administration of PYY(3-36), but not PYY(1-36), markedly (p < 0.05) decreased food intake in overnight fasted mice. Neither form of PYY affected glucose disposal or insulin secretion following an i.p. glucose challenge. However, in vitro, PYY(1-36) and PYY(3-36) inhibited (p < 0.05 to p < 0.001) glucose, alanine and GLP-1 stimulated insulin secretion from immortalised rodent and human beta-cells, as well as isolated mouse islets, by impeding alterations in membrane potential, [Ca(2+)]i and elevations of cAMP. Mice treated with multiple low dose streptozotocin presented with severe (p < 0.01) loss of beta-cell mass accompanied by notable increases (p < 0.001) in alpha and PP cell numbers. In contrast, hydrocortisone-induced insulin resistance increased islet number (p < 0.01) and beta-cell mass (p < 0.001). PYY expression was consistently observed in alpha-, PP- and delta-, but not beta-cells. Streptozotocin decreased islet PYY co-localisation with PP (p < 0.05) and somatostatin (p < 0.001), whilst hydrocortisone increased PYY co-localisation with glucagon (p < 0.05) in mice. More detailed in vitro investigations revealed that both forms of PYY augmented (p < 0.05 to p < 0.01) immortalised human and rodent beta-cell proliferation and protected against streptozotocin-induced cytotoxicity, to a similar or superior extent as the well characterised beta-cell proliferative and anti-apoptotic agent GLP-1. Taken together, these data highlight the significance and potential offered by modulation of pancreatic islet NPY receptor signalling pathways for preservation of beta-cell mass in diabetes.
Collapse
Affiliation(s)
- Dawood Khan
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Srividya Vasu
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - R Charlotte Moffett
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK.
| | - Peter R Flatt
- SAAD Centre for Pharmacy and Diabetes, University of Ulster, Coleraine, Northern Ireland, UK
| |
Collapse
|
17
|
Mundinger TO, Taborsky GJ. Early sympathetic islet neuropathy in autoimmune diabetes: lessons learned and opportunities for investigation. Diabetologia 2016; 59:2058-67. [PMID: 27342407 PMCID: PMC6214182 DOI: 10.1007/s00125-016-4026-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 05/23/2016] [Indexed: 12/13/2022]
Abstract
This review outlines the current state of knowledge regarding a unique neural defect of the pancreatic islet in autoimmune diabetes, one that we have termed early sympathetic islet neuropathy (eSIN). We begin with the findings that a majority of islet sympathetic nerves are lost near the onset of type 1, but not type 2, diabetes and that this nerve loss is restricted to the islet. We discuss later work demonstrating that while the loss of islet sympathetic nerves and the loss of islet beta cells in type 1 diabetes both require infiltration of the islet by lymphocytes, their respective mechanisms of tissue destruction differ. Uniquely, eSIN requires the activation of a specific neurotrophin receptor and we propose two possible pathways for activation of this receptor during the immune attack on the islet. We also outline what is known about the functional consequences of eSIN, focusing on impairment of sympathetically mediated glucagon secretion and its application to the clinical problem of insulin-induced hypoglycaemia. Finally, we offer our view on the important remaining questions regarding this unique neural defect.
Collapse
Affiliation(s)
- Thomas O Mundinger
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, 98105, USA.
- Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA.
| | - Gerald J Taborsky
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, 98105, USA
- Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| |
Collapse
|
18
|
Otsuka T, Tsukahara T, Takeda H. Development of the pancreas in medaka, Oryzias latipes, from embryo to adult. Dev Growth Differ 2015; 57:557-69. [PMID: 26435359 DOI: 10.1111/dgd.12237] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 07/17/2015] [Accepted: 07/19/2015] [Indexed: 12/17/2022]
Abstract
To address conserved and unique features of fish pancreas development, we performed extensive analyses of pancreatic development in medaka embryos and adults using pdx1- and ptf1a-transgenic medaka, in situ hybridization and immunohistochemistry. The markers used in these analyses included pdx1, nkx6.1, nkx6.2, nkx2.2, Islet1, insulin, Somatostatin, glucagon, ptf1a, ela3l, trypsin, and amylase. The double transgenic (Tg) fish produced in the present study visualizes the development of endocrine (pdx1+) and exocrine (ptf1a+) parts simultaneously in living fishes. Like other vertebrates, the medaka pancreas develops as two (dorsal and ventral) buds in the anterior gut tube, which soon fuse into a single anlagen. The double Tg fish demonstrates that the differential property between the two buds is already established at the initial phase of bud development as indicated by strong pdx1 expression in the dorsal one. This Tg fish also allowed us to examine the gross morphology and the structure of adult pancreas and revealed unique characters of medaka pancreas such as broad and multiple connections with the gut tube along the anterior-posterior axis.
Collapse
Affiliation(s)
- Takayoshi Otsuka
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tatsuya Tsukahara
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroyuki Takeda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,JST, CREST, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| |
Collapse
|
19
|
Shi YC, Loh K, Bensellam M, Lee K, Zhai L, Lau J, Cantley J, Luzuriaga J, Laybutt DR, Herzog H. Pancreatic PYY Is Critical in the Control of Insulin Secretion and Glucose Homeostasis in Female Mice. Endocrinology 2015; 156:3122-36. [PMID: 26125465 DOI: 10.1210/en.2015-1168] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Insulin secretion is tightly controlled through coordinated actions of a number of systemic and local factors. Peptide YY (PYY) is expressed in α-cells of the islet, but its role in control of islet function such as insulin release is not clear. In this study, we generated a transgenic mouse model (Pyy(tg/+)/Rip-Cre) overexpressing the Pyy gene under the control of the rat insulin 2 gene promoter and assessed the impact of islet-released PYY on β-cell function, insulin release, and glucose homeostasis in mice. Our results show that up-regulation of PYY in islet β-cells leads to an increase in serum insulin levels as well as improved glucose tolerance. Interestingly, PYY-overproducing mice show increased lean mass and reduced fat mass with no significant changes in food intake or body weight. Energy expenditure is also increased accompanied by increased respiratory exchange ratio. Mechanistically, the enhanced insulin levels and improved glucose tolerance are primarily due to increased β-cell mass and secretion. This is associated with alterations in the expression of genes important for β-cell proliferation and function as well as the maintenance of the β-cell phenotype. Taken together, these data demonstrate that pancreatic islet-derived PYY plays an important role in controlling glucose homeostasis through the modulation of β-cell mass and function.
Collapse
Affiliation(s)
- Yan-Chuan Shi
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| | - Kim Loh
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| | - Mohammed Bensellam
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| | - Kailun Lee
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| | - Lei Zhai
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| | - Jackie Lau
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| | - James Cantley
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| | - Jude Luzuriaga
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| | - D Ross Laybutt
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| | - Herbert Herzog
- Neuroscience (Y.-C.S., K.Lo., K.Le., L.Z., J.La., H.H.) and Diabetes and Metabolism (M.B., J.C., J.Lu., D.R.L.) Divisions, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst NSW 2010, Sydney, Australia; Faculty of Medicine (Y.-C.S., K.Lo., J.C., D.R.L., H.H.), UNSW Australia, Sydney, NSW, 2052 Australia; and Department of Physiology, Anatomy and Genetics (J.C.), University of Oxford, Oxford, OX1 3QX United Kingdom
| |
Collapse
|
20
|
Farzi A, Reichmann F, Holzer P. The homeostatic role of neuropeptide Y in immune function and its impact on mood and behaviour. Acta Physiol (Oxf) 2015; 213:603-27. [PMID: 25545642 DOI: 10.1111/apha.12445] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/10/2014] [Accepted: 12/21/2014] [Indexed: 12/18/2022]
Abstract
Neuropeptide Y (NPY), one of the most abundant peptides in the nervous system, exerts its effects via five receptor types, termed Y1, Y2, Y4, Y5 and Y6. NPY's pleiotropic functions comprise the regulation of brain activity, mood, stress coping, ingestion, digestion, metabolism, vascular and immune function. Nerve-derived NPY directly affects immune cells while NPY also acts as a paracrine and autocrine immune mediator, because immune cells themselves are capable of producing and releasing NPY. NPY is able to induce immune activation or suppression, depending on a myriad of factors such as the Y receptors activated and cell types involved. There is an intricate relationship between psychological stress, mood disorders and the immune system. While stress represents a risk factor for the development of mood disorders, it exhibits diverse actions on the immune system as well. Conversely, inflammation is regarded as an internal stressor and is increasingly recognized to contribute to the pathogenesis of mood and metabolic disorders. Intriguingly, the cerebral NPY system has been found to protect against distinct disturbances in response to immune challenge, attenuating the sickness response and preventing the development of depression. Thus, NPY plays an important homeostatic role in balancing disturbances of physiological systems caused by peripheral immune challenge. This implication is particularly evident in the brain in which NPY counteracts the negative impact of immune challenge on mood, emotional processing and stress resilience. NPY thus acts as a unique signalling molecule in the interaction of the immune system with the brain in health and disease.
Collapse
Affiliation(s)
- A. Farzi
- Research Unit of Translational Neurogastroenterology; Institute of Experimental and Clinical Pharmacology; Medical University of Graz; Graz Austria
| | - F. Reichmann
- Research Unit of Translational Neurogastroenterology; Institute of Experimental and Clinical Pharmacology; Medical University of Graz; Graz Austria
| | - P. Holzer
- Research Unit of Translational Neurogastroenterology; Institute of Experimental and Clinical Pharmacology; Medical University of Graz; Graz Austria
| |
Collapse
|
21
|
Aragón F, Karaca M, Novials A, Maldonado R, Maechler P, Rubí B. Pancreatic polypeptide regulates glucagon release through PPYR1 receptors expressed in mouse and human alpha-cells. Biochim Biophys Acta Gen Subj 2014; 1850:343-51. [PMID: 25445712 DOI: 10.1016/j.bbagen.2014.11.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 11/01/2014] [Accepted: 11/04/2014] [Indexed: 12/24/2022]
Abstract
BACKGROUND Plasma levels of pancreatic polypeptide (PP) rise upon food intake. Although other pancreatic islet hormones, such as insulin and glucagon, have been extensively investigated, PP secretion and actions are still poorly understood. METHODS The release of PP upon glucose stimulation and the effects of PP on glucagon and insulin secretion were analyzed in isolated pancreatic islets. Expression of PP receptor (PPYR1) was investigated by immunoblotting, quantitative RT-PCR on sorted pancreatic islet cells, and immunohistochemistry. RESULTS In isolated mouse pancreatic islets, glucose stimulation increased PP release, while insulin secretion was up and glucagon release was down. Direct exposure of islets to PP inhibited glucagon release. In mouse islets, PPYR1 protein was observed by immunoblotting and quantitative RT-PCR revealed PPYR1 expression in the FACS-enriched glucagon alpha-cell fraction. Immunohistochemistry on pancreatic sections showed the presence of PPYR1 in alpha-cells of both mouse and human islets, while the receptor was absent in other islet cell types and exocrine pancreas. CONCLUSIONS Glucose stimulates PP secretion and PP inhibits glucagon release in mouse pancreatic islets. PP receptors are present in alpha-cells of mouse and human pancreatic islets. GENERAL SIGNIFICANCE These data demonstrate glucose-regulated secretion of PP and its effects on glucagon release through PPYR1 receptors expressed by alpha-cells.
Collapse
Affiliation(s)
- F Aragón
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - M Karaca
- Department of Cell Physiology and Metabolism, Geneva University Medical Center, Geneva, Switzerland
| | - A Novials
- Diabetes Research Laboratory. IDIBAPS (Institut Investigacions Biomèdiques August Pi i Sunyer), CIBERDEM, Barcelona, Spain
| | - R Maldonado
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - P Maechler
- Department of Cell Physiology and Metabolism, Geneva University Medical Center, Geneva, Switzerland.
| | - B Rubí
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain.
| |
Collapse
|
22
|
Persaud SJ, Bewick GA. Peptide YY: more than just an appetite regulator. Diabetologia 2014; 57:1762-9. [PMID: 24917132 DOI: 10.1007/s00125-014-3292-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 05/08/2014] [Indexed: 12/13/2022]
Abstract
Replenishment of beta cell mass is a key aim of novel therapeutic interventions for diabetes, and the implementation of new strategies will be aided by understanding the mechanisms employed to regulate beta cell mass under normal physiological conditions. We have recently identified a new role for the gut hormone peptide YY (PYY) and the neuropeptide Y (NPY) receptor systems in the control of beta cell survival. PYY is perhaps best known for its role in regulating appetite and body weight, but its production by islet cells, the presence of NPY receptors on islets and the demonstration that Y1 activation causes proliferation of beta cells and protects them from apoptosis, suggest a role for this peptide in modulating beta cell mass. This review introduces PYY and its potential role in glucose homeostasis, then focuses on evidence supporting the concept that PYY and NPY receptors are exciting new targets for the preservation of beta cells.
Collapse
Affiliation(s)
- Shanta J Persaud
- Division of Diabetes & Nutritional Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | | |
Collapse
|
23
|
Ozeki J, Choi M, Endo-Umeda K, Sakurai K, Amano S, Makishima M. Enhanced transcription of pancreatic peptide YY by 1α-hydroxyvitamin D3 administration in streptozotocin-induced diabetic mice. Neuropeptides 2013; 47:329-32. [PMID: 23899497 DOI: 10.1016/j.npep.2013.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 06/29/2013] [Accepted: 07/05/2013] [Indexed: 12/17/2022]
Abstract
Peptide YY (PYY) is a peptide hormone secreted from L cells in the intestine in response to food intake that regulates appetite and gastrointestinal function. PYY is also produced in the pancreatic islets. The vitamin D receptor (VDR) is a nuclear receptor for the active form of vitamin D3 that regulates numerous physiological processes. VDR is expressed in the pancreatic islets and pharmacological VDR activation increases PYY expression in mouse peripheral islet cells. Although VDR is present in insulin-producing β cells as well as non-β cells, the role of β cell VDR in Pyy transcription remains unknown. We treated mice with streptozotocin to ablate β cells in the pancreas. Pancreatic Vdr mRNA expression was decreased in streptozotocin-induced diabetic mice. Interestingly, streptozotocin-treated mice exhibited increased basal Pyy expression and 1α-hydroxyvitamin D3 treatment further increased expression. Moreover, 1α-hydroxyvitamin D3 increased mRNA expression of pancreatic polypeptide and decreased that of neuropeptide Y in streptozotocin-induced diabetic mice but not in control mice. 1α-Hydroxyvitamin D3 slightly increased mRNA expression of insulin but transcript levels were nearly undetectable in the pancreas of streptozotocin-treated mice. Thus, VDR in non-β islet cells is involved in Pyy expression in the mouse pancreas. The findings from this β cell ablation study suggest a hormone transcription regulatory network composed of β cells and non-β cells.
Collapse
Affiliation(s)
- Jun Ozeki
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan; Division of Breast and Endocrine Surgery, Department of Surgery, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | | | | | | | | | | |
Collapse
|
24
|
Guo L, Inada A, Aguayo-Mazzucato C, Hollister-Lock J, Fujitani Y, Weir GC, Wright CV, Sharma A, Bonner-Weir S. PDX1 in ducts is not required for postnatal formation of β-cells but is necessary for their subsequent maturation. Diabetes 2013; 62:3459-68. [PMID: 23775765 PMCID: PMC3781453 DOI: 10.2337/db12-1833] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Pancreatic duodenal homeobox-1 (Pdx1), a transcription factor required for pancreatic development and maintenance of β-cell function, was assessed for a possible role in postnatal β-cell formation from progenitors in the pancreatic ducts by selectively deleting Pdx1 from the ducts. Carbonic anhydrase II (CAII)(Cre);Pdx1(Fl) mice were euglycemic for the first 2 postnatal weeks but showed moderate hyperglycemia from 3 to 7 weeks of age. By 10 weeks, they had near-normal morning fed glucose levels but showed severely impaired glucose tolerance and insulin secretion. Yet the loss of Pdx1 did not result in decreased islet and β-cell mass at 4 and 10 weeks of age. Within the same pancreas, there was a mixed population of islets, with PDX1 and MAFA protein expression normal in some cells and severely diminished in others. Even at 10 weeks, islets expressed immaturity markers. Thus, we conclude that Pdx1 is not necessary for the postnatal formation of β-cells but is essential for their full maturation to glucose-responsive β-cells.
Collapse
Affiliation(s)
- Lili Guo
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Akari Inada
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Diabetes and Genes, Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Cristina Aguayo-Mazzucato
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jennifer Hollister-Lock
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Yoshio Fujitani
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Gordon C. Weir
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Christopher V.E. Wright
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Arun Sharma
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Susan Bonner-Weir
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Susan Bonner-Weir,
| |
Collapse
|
25
|
Afelik S, Jensen J. Notch signaling in the pancreas: patterning and cell fate specification. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:531-44. [DOI: 10.1002/wdev.99] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
26
|
Choi M, Ozeki J, Hashizume M, Kato S, Ishihara H, Makishima M. Vitamin D receptor activation induces peptide YY transcription in pancreatic islets. Endocrinology 2012; 153:5188-99. [PMID: 22962257 DOI: 10.1210/en.2012-1396] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Peptide YY (PYY) is a peptide hormone secreted from L cells in the intestine after food intake and regulates appetite and intestinal function. PYY is also expressed in the pancreas, but the mechanisms of regulation of pancreatic PYY expression have not been elucidated. The vitamin D receptor (VDR) is a nuclear receptor for the active form of vitamin D(3) and regulates numerous physiological processes. Because VDR is expressed in the pancreas, we investigated the role of pancreatic VDR activation and found that Pyy is a VDR target gene in the mouse pancreas. Treatment of mice with 1α-hydroxyvitamin D(3) increased plasma PYY levels. VDR activation increased mRNA and protein expression of PYY in the pancreatic islets of mice and pancreatic endocrine cell lines but did not change intestinal PYY expression. 1α-Hydroxyvitamin D(3)-dependent induction of pancreatic and plasma PYY was abolished in VDR-null mice. We identified a functional vitamin D-responsive element in the mouse Pyy promoter using chromatin immunoprecipitation assay, EMSA, and luciferase promoter assay. Thus, Pyy is a tissue-specific VDR target gene. The pancreatic VDR-PYY pathway may mediate a regulatory function of vitamin D in the neuroendocrine system.
Collapse
Affiliation(s)
- Mihwa Choi
- Division of Biochemistry, Department of Medicine, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
27
|
Notch signaling differentially regulates the cell fate of early endocrine precursor cells and their maturing descendants in the mouse pancreas and intestine. Dev Biol 2012; 371:156-69. [PMID: 22964416 DOI: 10.1016/j.ydbio.2012.08.023] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 06/27/2012] [Accepted: 08/15/2012] [Indexed: 11/20/2022]
Abstract
Notch signaling inhibits differentiation of endocrine cells in the pancreas and intestine. In a number of cases, the observed inhibition occurred with Notch activation in multipotential cells, prior to the initiation of endocrine differentiation. It has not been established how direct activation of Notch in endocrine precursor cells affects their subsequent cell fate. Using conditional activation of Notch in cells expressing Neurogenin3 or NeuroD1, we examined the effects of Notch in both organs, on cell fate of early endocrine precursors and maturing endocrine-restricted cells, respectively. Notch did not preclude the differentiation of a limited number of endocrine cells in either organ when activated in Ngn3(+) precursor cells. In addition, in the pancreas most Ngn3(+) cells adopted a duct but not acinar cell fate; whereas in intestinal Ngn3(+) cells, Notch favored enterocyte and goblet cell fates, while selecting against endocrine and Paneth cell differentiation. A small fraction of NeuroD1(+) cells in the pancreas retain plasticity to respond to Notch, giving rise to intraislet ductules as well as cells with no detectable pancreatic lineage markers that appear to have limited ultrastructural features of both endocrine and duct cells. These results suggest that Notch directly regulates cell fate decisions in multipotential early endocrine precursor cells. Some maturing endocrine-restricted NeuroD1(+) cells in the pancreas switch to the duct lineage in response to Notch, indicating previously unappreciated plasticity at such a late stage of endocrine differentiation.
Collapse
|
28
|
Sam AH, Gunner DJ, King A, Persaud SJ, Brooks L, Hostomska K, Ford HE, Liu B, Ghatei MA, Bloom SR, Bewick GA. Selective ablation of peptide YY cells in adult mice reveals their role in beta cell survival. Gastroenterology 2012; 143:459-68. [PMID: 22562022 DOI: 10.1053/j.gastro.2012.04.047] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 03/31/2012] [Accepted: 04/24/2012] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS In the pancreas, peptide YY (PYY) is expressed by a subpopulation of nonbeta cells in the islets of Langerhans. We investigated the function of these cells in the pancreas of adult mice. METHODS We generated mice in which administration of diphtheria toxin (DT) led to specific ablation of PYY-expressing cells. We investigated the effects of loss of PYY cells on glucose homeostasis. RESULTS Loss of PYY cells in adult mice resulted in severe hyperglycemia, which was associated with significant loss of pancreatic insulin and disruption of islet morphology. In vitro administration of DT to isolated islets significantly reduced numbers of PYY-expressing cells and levels of insulin. Administration of either pancreatic polypeptide (a strong agonist of the receptor Y(4)) or PYY(3-36) (a selective agonist of the receptor Y(2)) did not restore loss of pancreatic insulin following administration of DT. However, a long-acting PYY analogue reduced the loss of insulin, and administration of this analogue reduced the hyperglycemia and insulin loss induced by streptozotocin in mice. CONCLUSIONS PYY appears to regulate beta cell function and survival via the receptor Y(1/2). These findings might be developed to treat and prevent loss of beta cells in patients with diabetes mellitus.
Collapse
|
29
|
Carmosino M, Mazzone A, Laforenza U, Gastaldi G, Svelto M, Valenti G. Altered expression of aquaporin 4 and H+/K+-ATPase in the stomachs of peptide YY (PYY) transgenic mice. Biol Cell 2012; 97:735-42. [PMID: 15898955 DOI: 10.1042/bc20040138] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND INFORMATION The hormone PYY (peptide YY), synthesized by endocrine cells in the pancreas, ileum, colon and stomach has widespread inhibitory effects on gastrointestinal and pancreatic fluid secretion. Transgenic mice expressing a viral oncoprotein under the control of the PYY gene 5'-flanking region develop well-differentiated colonic endocrine tumours producing mainly PYY and enteroglucagon. In the present study, we investigated the expression of AQP4 (aquaporin 4) water channel and H(+)/K(+)-ATPase in stomachs from both control and transgenic mice. RESULTS Semi-quantitative RT (reverse transcriptase)-PCR showed an increase in the AQP4 transcript compared with control mice. Quantitative Western-blot analysis of stomachs from control and transgenic mice confirmed a significant increase in the 30 kDa AQP4 protein in transgenic mice. In control mice, AQP4 is specifically expressed in the basolateral membrane of gastric parietal cells, located in the basal region of the fundic glands. This particular location suggests that parietal cells in the base region of gastric pits might have a major role in water transport when compared with the more superficial parietal cells. Interestingly, immunofluorescence studies on transgenic mice revealed that the quantitative increase of AQP4 expression was actually due to an increase in the number of AQP4-expressing epithelial cells rather than to a higher expression of AQP4 in parietal cells. In fact, immunofluorescence experiments using the specific antibody raised against the AE2 isoform of Cl(-)/HCO3- exchanger specifically expressed in parietal cells confirmed that the number of parietal cells was comparable in both PYY and control stomachs. Moreover, in transgenic mice, a parallel significant decrease in the expression of H(+)/K(+)-ATPase was observed, as revealed by RT-PCR, quantitative immunoblotting and immunofluorescence. CONCLUSIONS In the present study, we demonstrate that the sustained inhibition of gastric secretion due to tumours producing PYY/enteroglucagon in transgenic mice is associated with an increase in AQP4 expression and a down-regulation of H(+)/K(+)-ATPase in parietal cells that acquire the characteristics of basal parietal cells. The absence of H2 receptors-mediated signalling due to the inhibition of histamine release from ECL (enterochromaffin-like) cells by PYY may be in part responsible for the observed increase in the number of parietal cells expressing AQP4.
Collapse
Affiliation(s)
- M Carmosino
- Dipartimento di Fisiologia Generale ed Ambientale, Via Amendola 165/A, 70126, Bari, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Multiple bioactive peptides are produced from proglucagon encoded by glucagon gene (Gcg). Glucagon is produced in islet α-cells through processing by prohormone convertase 2 (Pcsk2) and exerts its action through the glucagon receptor (Gcgr). Although it is difficult to produce a genetic model that harbours isolated glucagon deficiency without affecting the production of other peptides derived from proglucagon, three different animal models that harbour deficiencies in glucagon signalling have been generated by gene targeting strategy. Although both Pcsk2(-/-) and Gcgr(-/-) mice display lower blood glucose levels, homozygous glucagon-GFP knock-in mice (Gcg(gfp/gfp) ) display normoglycaemia despite complete glucagon deficiency. In Gcg(gfp/gfp) mice, the metabolic impact of glucagon deficiency is probably ameliorated by lower plasma insulin levels and glucagon-independent mechanisms that maintain gluconeogenesis. As both Pcsk2(-/-) and Gcgr(-/-) mice exhibit increased production of glucagon-like peptide-1 (GLP-1), which is absent in Gcg(gfp/gfp), GLP-1 is the likely cause of the difference in metabolic impact of glucagon deficiency in these animal models. Although all the three models display islet 'α'-cell hyperplasia, the mechanisms involved remain to be elucidated. Studies using Pcsk2(-/-), Gcgr(-/-) and Gcg(gfp/gfp) mice, especially in combination with α-cell ablation models such as pancreas-specific aristaless-related homeobox (ARX) knockout mice, should further clarify the physiological and pathological roles of glucagon in the regulation of metabolism and the control of islet cell differentiation and proliferation.
Collapse
Affiliation(s)
- Y Hayashi
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
| |
Collapse
|
31
|
Kordowich S, Collombat P, Mansouri A, Serup P. Arx and Nkx2.2 compound deficiency redirects pancreatic alpha- and beta-cell differentiation to a somatostatin/ghrelin co-expressing cell lineage. BMC DEVELOPMENTAL BIOLOGY 2011; 11:52. [PMID: 21880149 PMCID: PMC3179930 DOI: 10.1186/1471-213x-11-52] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 08/31/2011] [Indexed: 12/20/2022]
Abstract
Background Nkx2.2 and Arx represent key transcription factors implicated in the specification of islet cell subtypes during pancreas development. Mice deficient for Arx do not develop any alpha-cells whereas beta- and delta-cells are found in considerably higher numbers. In Nkx2.2 mutant animals, alpha- and beta-cell development is severely impaired whereas a ghrelin-expressing cell population is found augmented. Notably, Arx transcription is clearly enhanced in Nkx2.2-deficient pancreata. Hence in order to precise the functional link between both factors we performed a comparative analysis of Nkx2.2/Arx single- and double-mutants but also of Pax6-deficient animals. Results We show that most of the ghrelin+ cells emerging in pancreata of Nkx2.2- and Pax6-deficient mice, express the alpha-cell specifier Arx, but also additional beta-cell related genes. In Nkx2.2-deficient mice, Arx directly co-localizes with iAPP, PC1/3 and Pdx1 suggesting an Nkx2.2-dependent control of Arx in committed beta-cells. The combined loss of Nkx2.2 and Arx likewise results in the formation of a hyperplastic ghrelin+ cell population at the expense of mature alpha- and beta-cells. Surprisingly, such Nkx2.2-/-Arx- ghrelin+ cells also express the somatostatin hormone. Conclusions Our data indicate that Nkx2.2 acts by reinforcing the transcriptional networks initiated by Pax4 and Arx in early committed beta- and alpha-cell, respectively. Our analysis also suggests that one of the coupled functions of Nkx2.2 and Pax4 is to counteract Arx gene activity in early committed beta-cells.
Collapse
Affiliation(s)
- Simon Kordowich
- Department of Molecular Cell Biology, Max-Planck Institute for Biophysical Chemistry, Am Fassberg, Göttingen, Germany
| | | | | | | |
Collapse
|
32
|
Kordowich S, Mansouri A, Collombat P. Reprogramming into pancreatic endocrine cells based on developmental cues. Mol Cell Endocrinol 2010; 323:62-9. [PMID: 20025937 DOI: 10.1016/j.mce.2009.12.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Due to the increasing prevalence of type 1 diabetes and the complications arising from actual therapies, alternative treatments need to be established. In order to compensate the beta-cell deficiency associated with type 1 diabetes, current researches focus on new strategies to generate insulin-producing beta cells for transplantation purpose, including the differentiation of stem or progenitor cells, as well as the transdifferentiation of dispensable mature cell types. However, to successfully force any cell to adopt a functional beta-cell fate or phenotype, a better understanding of the molecular mechanisms underlying the genesis of these in vivo is required. The present short review summarizes the hitherto known functions and interplays of several key factors involved in the differentiation of the endocrine cell lineages during pancreas morphogenesis, as well as there potential in generating beta cells. Furthermore, an emphasize is made on beta-cell regeneration and the determinants implicated.
Collapse
Affiliation(s)
- Simon Kordowich
- Max-Planck Institute for Biophysical Chemistry, Department of Molecular Cell Biology, Am Fassberg, D-37077 Göttingen, Germany
| | | | | |
Collapse
|
33
|
Kordowich S, Mansouri A, Collombat P. Reprogramming into pancreatic endocrine cells based on developmental cues. Mol Cell Endocrinol 2010; 315:11-8. [PMID: 19897012 PMCID: PMC2814956 DOI: 10.1016/j.mce.2009.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 09/14/2009] [Accepted: 10/24/2009] [Indexed: 01/30/2023]
Abstract
Due to the increasing prevalence of type 1 diabetes and the complications arising from actual therapies, alternative treatments need to be established. In order to compensate the beta-cell deficiency associated with type 1 diabetes, current research focuses on new strategies to generate insulin-producing beta-cells for transplantation purpose, including the differentiation of stem or progenitor cells, as well as the transdifferentiation of dispensable mature cell types. However, to successfully force specific cells to adopt a functional beta-cell fate or phenotype, a better understanding of the molecular mechanisms underlying beta-cell genesis is required. The present short review summarizes the hitherto known functions and interplays of several key factors involved in the development of the different endocrine cell lineages during pancreas morphogenesis, as well as their potential to direct the generation of beta-cells. Furthermore, an emphasis is made on beta-cell regeneration and the determinants implicated.
Collapse
Affiliation(s)
- Simon Kordowich
- Max-Planck Institute for Biophysical Chemistry, Department of Molecular Cell Biology, Am Fassberg, D-37077 Göttingen, Germany
| | | | | |
Collapse
|
34
|
Li Z, Korzh V, Gong Z. DTA-mediated targeted ablation revealed differential interdependence of endocrine cell lineages in early development of zebrafish pancreas. Differentiation 2009; 78:241-52. [DOI: 10.1016/j.diff.2009.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 04/23/2009] [Accepted: 05/30/2009] [Indexed: 11/26/2022]
|
35
|
Umeda T, Kanatani A, Iwaasa H. Cloning and characterization of rabbit neuropeptide Y receptor subtypes. Peptides 2009; 30:1441-7. [PMID: 19481128 DOI: 10.1016/j.peptides.2009.05.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 05/19/2009] [Accepted: 05/19/2009] [Indexed: 11/18/2022]
Abstract
Neuropeptide Y (NPY), peptide YY (PYY) and pancreatic polypeptide (PP) are structurally related peptides that have numerous functions in both neural and endocrine signaling. These effects are mediated by the NPY receptor family and five members of this family have been cloned in mammals. To better characterize these receptor subtypes, we cloned and expressed the Y1, Y2, Y4 and Y5 receptor subtypes from the rabbit. Comparison of these sequences with human orthologs revealed that the Y1, Y2 and Y5 receptors have generally strong amino-acid sequence conservation, with 91-96% identity, while Y4 receptor showed relatively weak similarity with 82% identity, as with other species. Particularly in the transmembrane regions, Y1, Y2, and Y5 receptor subtypes showed remarkable conservation, with 98-99% amino acid identity. Competitive binding studies by NPY-family peptides and analogs showed that Y1, Y2 and Y5 receptors had similar pharmacological profiles between the respective rabbit and human receptor subtypes. Interestingly, all the tested peptides had a greater affinity for rabbit Y4 receptor than human Y4 receptor. These results suggest that rabbit and human Y1, Y2 and Y5 receptor subtypes are well conserved, whereas Y4 receptors are less well conserved.
Collapse
Affiliation(s)
- Tatsuya Umeda
- Tsukuba Research Institute, Banyu Pharmaceutical Co., Ltd., 3 Okubo, Tsukuba, Ibaraki 300-2611, Japan.
| | | | | |
Collapse
|
36
|
Vincent RK, Odorico JS. Reduced serum concentration is permissive for increased in vitro endocrine differentiation from murine embryonic stem cells. Differentiation 2009; 78:24-34. [PMID: 19446949 DOI: 10.1016/j.diff.2009.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Accepted: 03/15/2009] [Indexed: 10/20/2022]
Abstract
Embryonic stem cells (ESCs) have been shown to be capable of differentiating into pancreatic progenitors and insulin-producing cells in vitro. However, before ESC derivatives can be used in clinical settings, efficient selective differentiation needs to be achieved. Essential to improving ESC differentiation to islet endocrine cells is an understanding of the influences of extrinsic signals and transcription factors on cell specification. Herein, we investigate the influence of serum-supplemented growth conditions on the differentiation of murine ESCs to endocrine lineages in the context of over-expression of two pancreatic transcription factors, Pdx1 and Ngn3. To study the effect of different serum formulations and concentrations on the ability of murine ESCs to differentiate into endocrine cells in vitro, cells were grown into embryoid bodies and then differentiated in various serum replacement (SR), fetal calf serum (FCS) and serum-free conditions. Using immunohistochemistry and quantitative real-time RT-PCR (QPCR), we found that, of the conditions tested, 1% SR differentiation medium resulted in the highest levels of insulin-1 mRNA and significantly increased the total number of insulin-expressing cells. Applying this knowledge to cell lines in which Pdx1 or Ngn3 transgene expression could be induced by exposure to doxycycline we differentiated TetPDX1 and TetNgn3 ESCs under conditions of either 10% FCS or 1% SR medium. In the presence of 10% serum, induced expression of either Pdx1 or Ngn3 in differentiating ESCs resulted in modest increases in hormone transcripts and cell counts. However, changing the serum formulation from 10% FCS to 1% SR significantly enhanced the number of insulin+/C-peptide+ cells in parallel with increased insulin-1 transcript levels in both inducible cell lines. In summary, these data demonstrate that induced expression of key pancreatic transcription factors in combination with low serum/SR concentrations increases endocrine cell differentiation from murine ESCs.
Collapse
Affiliation(s)
- Robert K Vincent
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | | |
Collapse
|
37
|
Villasenor A, Chong DC, Cleaver O. Biphasic Ngn3 expression in the developing pancreas. Dev Dyn 2009; 237:3270-9. [PMID: 18924236 DOI: 10.1002/dvdy.21740] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Ngn3 is a bHLH transcription factor critical for the specification of endocrine cells in the pancreatic Islets of Langerhans. Previous studies in mouse embryos have reported transient expression of Ngn3 in scattered cells within the developing pancreatic epithelium during midgestation (Schwitzgebel et al. [2000] Development 127:3533-3542). Specifically, these Ngn3-expressing cells have been shown to be progenitor cells fated to give rise to islet endocrine cells (Gradwohl et al. [2000] Proc Natl Acad Sci USA 97:1607-1611). Here, we characterize the expression of Ngn3 transcripts and protein throughout pancreatic development. Interestingly, we identify and define a dramatic and previously unnoticed gap in developmental Ngn3 expression. We show that both Ngn3 transcript and protein expression occur in two distinct temporal waves, the first occurring early from approximately E8.5 to E11.0, and the second initiating at approximately E12.0. Strikingly, this observed biphasic expression correlates with the "first" and "second" transitions, which encompass two distinct waves of embryonic endocrine differentiation. In addition, our studies demonstrate that Ngn3 transcripts are markedly more widespread in the pancreatic epithelium than NGN3 protein, indicating that post-transcriptional regulation is likely to play a critical role during endocrine differentiation.
Collapse
Affiliation(s)
- Alethia Villasenor
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9148, USA
| | | | | |
Collapse
|
38
|
Li Z, Wen C, Peng J, Korzh V, Gong Z. Generation of living color transgenic zebrafish to trace somatostatin-expressing cells and endocrine pancreas organization. Differentiation 2009; 77:128-34. [DOI: 10.1016/j.diff.2008.09.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2008] [Revised: 08/10/2008] [Accepted: 08/11/2008] [Indexed: 11/30/2022]
|
39
|
Gittes GK. Developmental biology of the pancreas: a comprehensive review. Dev Biol 2008; 326:4-35. [PMID: 19013144 DOI: 10.1016/j.ydbio.2008.10.024] [Citation(s) in RCA: 315] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2008] [Revised: 10/09/2008] [Accepted: 10/13/2008] [Indexed: 02/06/2023]
Abstract
Pancreatic development represents a fascinating process in which two morphologically distinct tissue types must derive from one simple epithelium. These two tissue types, exocrine (including acinar cells, centro-acinar cells, and ducts) and endocrine cells serve disparate functions, and have entirely different morphology. In addition, the endocrine tissue must become disconnected from the epithelial lining during its development. The pancreatic development field has exploded in recent years, and numerous published reviews have dealt specifically with only recent findings, or specifically with certain aspects of pancreatic development. Here I wish to present a more comprehensive review of all aspects of pancreatic development, though still there is not a room for discussion of stem cell differentiation to pancreas, nor for discussion of post-natal regeneration phenomena, two important fields closely related to pancreatic development.
Collapse
Affiliation(s)
- George K Gittes
- Children's Hospital of Pittsburgh and the University of Pittsburgh School of Medicine, Department of Pediatric Surgery, 3705 Fifth Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
40
|
Gustavsen CR, Pillay N, Heller RS. An immunohistochemical study of the endocrine pancreas of the African ice rat, Otomys sloggetti robertsi. Acta Histochem 2008; 110:294-301. [PMID: 18406449 DOI: 10.1016/j.acthis.2007.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 10/29/2007] [Accepted: 11/02/2007] [Indexed: 12/22/2022]
Abstract
The African ice rat, Otomys sloggetti robertsi, is a member of the subfamily Otomyinae, in the superfamily of Muroidea, to which all rodents belong. Very little is known about this unique family of rodents. The study reported here examines the endocrine pancreas of this species using immunohistochemical techniques. The islets of Langerhans were scattered in the exocrine pancreas and tended to be quite small. Scattered single endocrine cells (mostly immunoreactive for insulin) were found in the exocrine pancreas and were not generally associated with ducts (as marked by pan-cytokeratin labeling). The normal islet architecture of insulin in the center and glucagon, somatostatin (SS) and pancreatic polypeptide (PP) in the rim was observed, but the islets tended to have 2-3 layers of glucagon immunoreactive cells. Examining for rarer endocrine cell types, we found that cocaine amphetamine regulated transcript (CART) immunoreactive cells were co-localized with SS; and peptide YY (PYY) immunoreactive cells could be found that were singly immunoreactive or co-localized with either PP or glucagon. Ghrelin cells were not found. MafA co-localized only with the insulin cells, while MafB, which localizes to the glucagon cells, also showed a low level of immunoreactivity in most insulin immunoreactive cells. The Nkx family of transcription factors (Nkx6.1 and 2.2) and PDX-1 were all detected in the pancreas in a similar manner to that seen in mouse and rat. In conclusion, the endocrine pancreas of the African ice rat is quite similar to that of other studied rodents, but these animals have more glucagon and SS cells than rat (Rattus) or mouse (Mus) species.
Collapse
|
41
|
Abstract
Since adipose tissue was shown to be more than a storage organ, the many cytokines it produces have been identified, along with their roles in energy homeostasis, appetite, and insulin resistance. Concurrently, numerous gut hormones with a diversity of effects have been discovered. They include, amongst many others, peptide YY, ghrelin and oxyntomodulin. As these peptides have been investigated, the potential for their use as novel anti-obesity and antidiabetic therapies has been realized. In this chapter we describe the actions of four of the peptides that have been proposed as the basis for promising new therapies for diabetes: leptin, adiponectin, obestatin and peptide YY. They each have an effect on appetite and, directly or indirectly, on glucose metabolism. We synthesize available data for these peptides and consider the therapeutic potential of each.
Collapse
Affiliation(s)
- Tom Billyard
- Warwick Medical School, University of Warwick, Clifford Bridge Road, University Hospital, Coventry CV2 2DX, UK
| | | | | |
Collapse
|
42
|
Jørgensen MC, Ahnfelt-Rønne J, Hald J, Madsen OD, Serup P, Hecksher-Sørensen J. An illustrated review of early pancreas development in the mouse. Endocr Rev 2007; 28:685-705. [PMID: 17881611 DOI: 10.1210/er.2007-0016] [Citation(s) in RCA: 258] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pancreas morphogenesis and cell differentiation are highly conserved among vertebrates during fetal development. The pancreas develops through simple budlike structures on the primitive gut tube to a highly branched organ containing many specialized cell types. This review presents an overview of key molecular components and important signaling sources illustrated by an extensive three-dimensional (3D) imaging of the developing mouse pancreas at single cell resolution. The 3D documentation covers the time window between embryonic days 8.5 and 14.5 in which all the pancreatic cell types become specified and therefore includes gene expression patterns of pancreatic endocrine hormones, exocrine gene products, and essential transcription factors. The 3D perspective provides valuable insight into how a complex organ like the pancreas is formed and a perception of ventral and dorsal pancreatic growth that is otherwise difficult to uncover. We further discuss how this global analysis of the developing pancreas confirms and extends previous studies, and we envisage that this type of analysis can be instrumental for evaluating mutant phenotypes in the future.
Collapse
Affiliation(s)
- Mette Christine Jørgensen
- Hagedorn Research Institute, Department of Developmental Biology, Niels Steensens Vej 6, DK-2820 Gentofte, Denmark.
| | | | | | | | | | | |
Collapse
|
43
|
Collombat P, Hecksher-Sørensen J, Krull J, Berger J, Riedel D, Herrera PL, Serup P, Mansouri A. Embryonic endocrine pancreas and mature beta cells acquire alpha and PP cell phenotypes upon Arx misexpression. J Clin Invest 2007; 117:961-70. [PMID: 17404619 PMCID: PMC1839241 DOI: 10.1172/jci29115] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Accepted: 01/02/2007] [Indexed: 12/16/2022] Open
Abstract
Aristaless-related homeobox (Arx) was recently demonstrated to be involved in pancreatic alpha cell fate specification while simultaneously repressing the beta and delta cell lineages. To establish whether Arx is not only necessary, but also sufficient to instruct the alpha cell fate in endocrine progenitors, we used a gain-of-function approach to generate mice conditionally misexpressing this factor. Mice with forced Arx expression in the embryonic pancreas or in developing islet cells developed a dramatic hyperglycemia and eventually died. Further analysis demonstrated a drastic loss of beta and delta cells. Concurrently, a remarkable increase in the number of cells displaying alpha cell or, strikingly, pancreatic polypeptide (PP) cell features was observed. Notably, the ectopic expression of Arx induced in embryonic or adult beta cells led to a loss of the beta cell phenotype and a concomitant increase in a number of cells with alpha or PP cell characteristics. Combining quantitative real-time PCR and lineage-tracing experiments, we demonstrate that, in adult mice, the misexpression of Arx, rather than its overexpression, promotes a conversion of beta cells into glucagon- or PP-producing cells in vivo. These results provide important insights into the complex mechanisms underlying proper pancreatic endocrine cell allocation and cell identity acquisition.
Collapse
Affiliation(s)
- Patrick Collombat
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Developmental Biology, Hagedorn Research Institute, Gentofte, Denmark.
Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
Electron Microscopy Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Jacob Hecksher-Sørensen
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Developmental Biology, Hagedorn Research Institute, Gentofte, Denmark.
Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
Electron Microscopy Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Jens Krull
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Developmental Biology, Hagedorn Research Institute, Gentofte, Denmark.
Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
Electron Microscopy Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Joachim Berger
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Developmental Biology, Hagedorn Research Institute, Gentofte, Denmark.
Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
Electron Microscopy Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Dietmar Riedel
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Developmental Biology, Hagedorn Research Institute, Gentofte, Denmark.
Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
Electron Microscopy Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Pedro L. Herrera
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Developmental Biology, Hagedorn Research Institute, Gentofte, Denmark.
Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
Electron Microscopy Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Palle Serup
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Developmental Biology, Hagedorn Research Institute, Gentofte, Denmark.
Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
Electron Microscopy Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Ahmed Mansouri
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Developmental Biology, Hagedorn Research Institute, Gentofte, Denmark.
Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.
Electron Microscopy Department, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.
Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
44
|
Johansson KA, Dursun U, Jordan N, Gu G, Beermann F, Gradwohl G, Grapin-Botton A. Temporal control of neurogenin3 activity in pancreas progenitors reveals competence windows for the generation of different endocrine cell types. Dev Cell 2007; 12:457-65. [PMID: 17336910 DOI: 10.1016/j.devcel.2007.02.010] [Citation(s) in RCA: 242] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2006] [Revised: 01/05/2007] [Accepted: 02/12/2007] [Indexed: 01/15/2023]
Abstract
All pancreatic endocrine cells, producing glucagon, insulin, somatostatin, or PP, differentiate from Pdx1+ progenitors that transiently express Neurogenin3. To understand whether the competence of pancreatic progenitors changes over time, we generated transgenic mice expressing a tamoxifen-inducible Ngn3 fusion protein under the control of the pdx1 promoter and backcrossed the transgene into the ngn3(-/-) background, devoid of endogenous endocrine cells. Early activation of Ngn3-ER(TM) almost exclusively induced glucagon+ cells, while depleting the pool of pancreas progenitors. As from E11.5, Pdx1+ progenitors became competent to differentiate into insulin+ and PP+ cells. Somatostatin+ cells were generated from E14.5, while the competence to make glucagon+ cells was dramatically decreased. Hence, pancreas progenitors, similar to retinal or cortical progenitors, go through competence states that each allow the generation of a subset of cell types. We further show that the progenitors acquire competence to generate late-born cells in a mechanism that is intrinsic to the epithelium.
Collapse
Affiliation(s)
- Kerstin A Johansson
- Swiss Institute for Experimental Cancer Research, 155 ch des Boveresses, 1066 Epalinges, Switzerland
| | | | | | | | | | | | | |
Collapse
|
45
|
Cerf ME, Williams K, Chapman CS, Louw J. Compromised beta-cell development and beta-cell dysfunction in weanling offspring from dams maintained on a high-fat diet during gestation. Pancreas 2007; 34:347-53. [PMID: 17414058 DOI: 10.1097/mpa.0b013e31802ee9ae] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES Reported here are the effects of a high-fat diet (HFD) fed to dams during pregnancy on the weight, beta- and alpha-cell development, and beta-cell function of their weanling offspring. METHODS Offspring were obtained from dams maintained on an HFD for the first, second, or third week of gestation or throughout gestation and then on a standard laboratory diet for the duration of lactation. Weanling weights and circulating glucose and insulin concentrations were measured on postnatal day 21, after which pancreata were excised and snap-frozen for quantitative polymerase chain reaction of glucokinase (GK) or processed for immunohistochemical examination and image analysis (beta- and alpha-cell volume, number, and size, and GK immunoreactivity). RESULTS All of the weanlings had low body weights and were hypoinsulinemic. In weanlings maintained on an HFD for either the first, second, or third week of gestation, hyperglycemia and a reduction in beta-cell volume and number, in beta- and alpha-cell size, and in both GK messenger RNA expression and immunoreactivity were observed. The development of beta and alpha cells was normal in weanlings maintained on an HFD throughout gestation. CONCLUSIONS Maintenance of dams on an HFD for any single week of gestation results in weanling offspring with an impairment in beta-cell development and function.
Collapse
Affiliation(s)
- Marlon E Cerf
- Diabetes Discovery Platform, Medical Research Council, Tygerberg, South Africa.
| | | | | | | |
Collapse
|
46
|
Yuzuriha H, Inui A, Asakawa A, Ueno N, Kasuga M, Meguid MM, Miyazaki JI, Ninomiya M, Herzog H, Fujimiya M. Gastrointestinal hormones (anorexigenic peptide YY and orexigenic ghrelin) influence neural tube development. FASEB J 2007; 21:2108-12. [PMID: 17400914 DOI: 10.1096/fj.06-7621com] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Gastrointestinal (GI) hormones play an important role in GI secretion, motility, and eating behaviors. It was recently suggested that GI hormones may have a trophic role in GI tract. Here we demonstrate that two principal GI hormones, anorexigenic peptide YY (PYY) and orexigenic ghrelin, affect neural tube development. Chronic administration into the pregnant mice or transgenic overexpression of PYY led to a neural tube defect (NTD) in the embryos that was blocked by ghrelin. PYY Y1 receptor antagonist prevented the occurrence of NTD induced not only by PYY but also by vitamin A, a well-known teratogen in humans and animals. Y1 receptor deficiency also engendered NTDs, indicating the need to maintain normal Y1 receptor signaling. The present study is the first linking GI hormones to the leading cause of infant mortality and provides a novel insight for neurogenesis in which materno-fetal communication through GI hormones appears to be important.
Collapse
Affiliation(s)
- Hideki Yuzuriha
- Division of Diabetes, Digestive and Kidney Diseases, Department of Clinical Molecular Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cerf ME, Muller CJ, Du Toit DF, Louw J, Wolfe-Coote SA. Hyperglycaemia and reduced glucokinase expression in weanling offspring from dams maintained on a high-fat diet. Br J Nutr 2007; 95:391-6. [PMID: 16469158 DOI: 10.1079/bjn20051632] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
High-fat feeding reduces the expression of GLUT-2 and the glycolytic enzyme glucokinase (GK). The transcription factor, pancreatic duodenal homeobox-1 (Pdx-1), is important for β-cell maintenance. The aim of the present study was to determine, in weanling Wistar rats, the effect of a maternal high-fat diet (HFD) during defined periods of gestation and lactation, on body weight, circulating glucose and insulin concentrations, and the expression of GLUT-2, GK and Pdx-1. At postnatal day 21, weights were recorded and glucose and insulin concentrations were measured. The expression levels for mRNA were quantified by LightCycler PCR. Pancreatic sections, immunostained for GLUT-2, GK or Pdx-1, were assessed by image analysis. Weanlings from dams fed an HFD throughout gestation were lighter, with heavier weanlings produced from dams fed an HFD throughout gestation and lactation. Both these groups of weanlings were normoglycaemic, all the others being hyperglycaemic. Hypoinsulinaemia was evident in weanlings from dams fed an HFD throughout gestation only and also for either the first week of lactation or throughout lactation. GLUT-2 mRNA expression was reduced and GLUT-2 immunoreactivity was increased in most of the weanlings. GK mRNA expression and immunoreactivity was reduced in most of the offspring. Pdx-1 mRNA expression was increased in weanlings from dams fed an HFD throughout both gestation and lactation and reduced in those from dams only fed a lactational HFD. Normal Pdx-1 immunoreactivity was found in all of the weanlings. A maternal HFD induces hyperglycaemia in weanlings concomitant with reduced GK expression which may compromise β-cell function.
Collapse
Affiliation(s)
- Marlon E Cerf
- Diabetes Research Group, Medical Research Council, Tygerberg, South Africa.
| | | | | | | | | |
Collapse
|
48
|
Boey D, Sainsbury A, Herzog H. The role of peptide YY in regulating glucose homeostasis. Peptides 2007; 28:390-5. [PMID: 17210210 DOI: 10.1016/j.peptides.2006.07.031] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 07/30/2006] [Indexed: 01/17/2023]
Abstract
The gut-derived hormone peptide YY (PYY) is most commonly known for its effect on satiety, decreasing food intake and body weight in animals and humans. However, PYY is also involved in a wide range of digestive functions including regulating insulin secretion and glucose homeostasis. Over the last few years, there have been several interesting clinical and animal studies investigating the role of PYY in glucose homeostasis. This review aims to present an updated summary of findings over the last few decades highlighting the role of PYY in regulating insulin output and insulin sensitivity, and the potential mechanisms involved.
Collapse
Affiliation(s)
- Dana Boey
- Neuroscience Research Program, Garvan Institute of Medical Research, St. Vincent's Hospital, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia.
| | | | | |
Collapse
|
49
|
Gromada J, Franklin I, Wollheim CB. Alpha-cells of the endocrine pancreas: 35 years of research but the enigma remains. Endocr Rev 2007; 28:84-116. [PMID: 17261637 DOI: 10.1210/er.2006-0007] [Citation(s) in RCA: 433] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Glucagon, a hormone secreted from the alpha-cells of the endocrine pancreas, is critical for blood glucose homeostasis. It is the major counterpart to insulin and is released during hypoglycemia to induce hepatic glucose output. The control of glucagon secretion is multifactorial and involves direct effects of nutrients on alpha-cell stimulus-secretion coupling as well as paracrine regulation by insulin and zinc and other factors secreted from neighboring beta- and delta-cells within the islet of Langerhans. Glucagon secretion is also regulated by circulating hormones and the autonomic nervous system. In this review, we describe the components of the alpha-cell stimulus secretion coupling and how nutrient metabolism in the alpha-cell leads to changes in glucagon secretion. The islet cell composition and organization are described in different species and serve as a basis for understanding how the numerous paracrine, hormonal, and nervous signals fine-tune glucagon secretion under different physiological conditions. We also highlight the pathophysiology of the alpha-cell and how hyperglucagonemia represents an important component of the metabolic abnormalities associated with diabetes mellitus. Therapeutic inhibition of glucagon action in patients with type 2 diabetes remains an exciting prospect.
Collapse
Affiliation(s)
- Jesper Gromada
- Novartis Institutes for BioMedical Research, 100 Technology Square, Cambridge, Massachusetts 02139, USA.
| | | | | |
Collapse
|
50
|
Understanding the extrinsic and intrinsic signals involved in pancreas and β-cell development: from endoderm to β cells. Curr Opin Organ Transplant 2007; 12:40-48. [PMID: 27792088 DOI: 10.1097/mot.0b013e3280129669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW To summarize recent progress in understanding of the extrinsic and intrinsic signals directing pancreas development from early endoderm. RECENT FINDINGS The pancreatic mesoderm was shown not only to play a permissive role in pancreas determination but also to control endocrine commitment and maturation through the interplay between Notch and fibroblast growth factor signaling. The requirement of Wnt (wingless-type)/β-catenin signaling in the expansion of the acinar cell lineage, and the spatial-temporal specificity of PDX1 (pancreatic and duodenal homeobox) activity, which is needed for proper acinar development, were also demonstrated. A novel factor, IA1 (insulinoma-associated 1), was identified as an endocrine marker downstream of Ngn3 (neurogenin); MAFB (musculo-aponeurotic fibrosarcoma) was shown to be a marker of α-cell and β-cell precursors, and ARX (aristaless-related homeobox), a marker of α-cell progenitors, was revealed to directly antagonize PAX4 (paired homeobox) in determining α-cell and β-cell lineages. SUMMARY Cell fate specification results from combined effects of extrinsic and intrinsic regulators and sensitivity of target cells to them, which vary depending on the precise stage of cell commitment or differentiation. Knowledge of the hierarchy of the different factors influencing pancreas development will aid in developing new cell therapies to treat diabetes.
Collapse
|