151
|
Cras-Méneur C, Elghazi L, Fort P, Bernal-Mizrachi E. Noninvasive in vivo imaging of embryonic β-cell development in the anterior chamber of the eye. Islets 2016; 8:35-47. [PMID: 26950054 PMCID: PMC4878273 DOI: 10.1080/19382014.2016.1148236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The fetal environment plays a decisive role in modifying the risk for developing diabetes later in life. Developing novel methodology for noninvasive imaging of β-cell development in vivo under the controlled physiological conditions of the host can serve to understand how this environment affects β-cell growth and differentiation. A number of culture models have been designed for pancreatic rudiment but none match the complexity of the in utero or even normal physiological environment. Speier et al. recently developed a platform of noninvasive in vivo imaging of pancreatic islets using the anterior chamber of the eye where islets get vascularized, grow and respond to physiological changes. The same methodology was adapted for the study of pancreatic development. E13.0, still undifferentiated rudiments with fluorescent lineage tracing were implanted in the AC of the eye, allowing the longitudinal study of their growth and differentiation. Within 48 h the anlages get vascularized and grow but their mesenchyme displays a selective growth advantage. The resulting imbalance leads to alteration in the differentiation pattern of the progenitors. Reducing the mesenchyme to its bare minimum before implantation allows the restoration of a proper balance and a development that mimics the normal pancreatic development. These groundbreaking observations demonstrate that the anterior chamber of the eye provides a good system for noninvasive in vivo fluorescence imaging of the developing pancreas under the physiology of the host and can have important implications for designing strategies to prevent or reverse the deleterious effects of hyperglycemia on altering β-cell function later in life.
Collapse
Affiliation(s)
- Corentin Cras-Méneur
- Internal Medicine Department, Division of Metabolism, Endocrinology and Diabetes, University of Michigan in Ann Arbor, Ann Arbor, Michigan, USA
| | - Lynda Elghazi
- Internal Medicine Department, Division of Metabolism, Endocrinology and Diabetes, University of Michigan in Ann Arbor, Ann Arbor, Michigan, USA
| | - Patrice Fort
- Ophthalmology Department, University of Michigan in Ann Arbor, Ann Arbor, Michigan, USA
| | - Ernesto Bernal-Mizrachi
- Internal Medicine Department, Division of Metabolism, Endocrinology and Diabetes, University of Michigan in Ann Arbor, Ann Arbor, Michigan, USA
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
152
|
Brown ML, Andrzejewski D, Burnside A, Schneyer AL. Activin Enhances α- to β-Cell Transdifferentiation as a Source For β-Cells In Male FSTL3 Knockout Mice. Endocrinology 2016; 157:1043-54. [PMID: 26727106 DOI: 10.1210/en.2015-1793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Diabetes results from inadequate β-cell number and/or function to control serum glucose concentrations so that replacement of lost β-cells could become a viable therapy for diabetes. In addition to embryonic stem cell sources for new β-cells, evidence for transdifferentiation/reprogramming of non-β-cells to functional β-cells is accumulating. In addition, de-differentiation of β-cells observed in diabetes and their subsequent conversion to α-cells raises the possibility that adult islet cell fate is malleable and controlled by local hormonal and/or environmental cues. We previously demonstrated that inactivation of the activin antagonist, follistatin-like 3 (FSTL3) resulted in β-cell expansion and improved glucose homeostasis in the absence of β-cell proliferation. We recently reported that activin directly suppressed expression of critical α-cell genes while increasing expression of β-cell genes, supporting the hypothesis that activin is one of the local hormones controlling islet cell fate and that increased activin signaling accelerates α- to β-cell transdifferentiation. We tested this hypothesis using Gluc-Cre/yellow fluorescent protein (YFP) α-cell lineage tracing technology combined with FSTL3 knockout (KO) mice to label α-cells with YFP. Flow cytometry was used to quantify unlabeled and labeled α- and β-cells. We found that Ins+/YFP+ cells were significantly increased in FSTL3 KO mice compared with wild type littermates. Labeled Ins+/YFP+ cells increased significantly with age in FSTL3 KO mice but not wild type littermates. Sorting results were substantiated by counting fluorescently labeled cells in pancreatic sections. Activin treatment of isolated islets significantly increased the number of YFP+/Ins+ cells. These results suggest that α- to β-cell transdifferentiation is influenced by activin signaling and may contribute substantially to β-cell mass.
Collapse
Affiliation(s)
- Melissa L Brown
- Departments of Nutrition (M.L.B.) and Veterinary and Animal Science (D.A., A.B., A.L.S.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Danielle Andrzejewski
- Departments of Nutrition (M.L.B.) and Veterinary and Animal Science (D.A., A.B., A.L.S.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Amy Burnside
- Departments of Nutrition (M.L.B.) and Veterinary and Animal Science (D.A., A.B., A.L.S.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Alan L Schneyer
- Departments of Nutrition (M.L.B.) and Veterinary and Animal Science (D.A., A.B., A.L.S.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
153
|
Paul L, Walker EM, Drosos Y, Cyphert HA, Neale G, Stein R, South J, Grosveld G, Herrera PL, Sosa-Pineda B. Lack of Prox1 Downregulation Disrupts the Expansion and Maturation of Postnatal Murine β-Cells. Diabetes 2016; 65:687-98. [PMID: 26631740 PMCID: PMC4764148 DOI: 10.2337/db15-0713] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 11/20/2015] [Indexed: 12/18/2022]
Abstract
Transcription factor expression fluctuates during β-cell ontogeny, and disruptions in this pattern can affect the development or function of those cells. Here we uncovered that murine endocrine pancreatic progenitors express high levels of the homeodomain transcription factor Prox1, whereas both immature and mature β-cells scarcely express this protein. We also investigated if sustained Prox1 expression is incompatible with β-cell development or maintenance using transgenic mouse approaches. We discovered that Prox1 upregulation in mature β-cells has no functional consequences; in contrast, Prox1 overexpression in immature β-cells promotes acute fasting hyperglycemia. Using a combination of immunostaining and quantitative and comparative gene expression analyses, we determined that Prox1 upregulation reduces proliferation, impairs maturation, and enables apoptosis in postnatal β-cells. Also, we uncovered substantial deficiency in β-cells that overexpress Prox1 of the key regulator of β-cell maturation MafA, several MafA downstream targets required for glucose-stimulated insulin secretion, and genes encoding important components of FGF signaling. Moreover, knocking down PROX1 in human EndoC-βH1 β-cells caused increased expression of many of these same gene products. These and other results in our study indicate that reducing the expression of Prox1 is beneficial for the expansion and maturation of postnatal β-cells.
Collapse
Affiliation(s)
- Leena Paul
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN
| | - Emily M Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical School, Nashville, TN
| | - Yiannis Drosos
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN
| | - Holly A Cyphert
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical School, Nashville, TN
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics & Biotechnology, St. Jude Children's Research Hospital, Memphis, TN
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical School, Nashville, TN
| | - Jack South
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN
| | - Gerard Grosveld
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN
| | - Pedro L Herrera
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Beatriz Sosa-Pineda
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
154
|
Kim HS, Lee MK. β-Cell regeneration through the transdifferentiation of pancreatic cells: Pancreatic progenitor cells in the pancreas. J Diabetes Investig 2016; 7:286-96. [PMID: 27330712 PMCID: PMC4847880 DOI: 10.1111/jdi.12475] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/27/2015] [Accepted: 01/04/2016] [Indexed: 12/17/2022] Open
Abstract
Pancreatic progenitor cell research has been in the spotlight, as these cells have the potential to replace pancreatic β‐cells for the treatment of type 1 and 2 diabetic patients with the absence or reduction of pancreatic β‐cells. During the past few decades, the successful treatment of diabetes through transplantation of the whole pancreas or isolated islets has nearly been achieved. However, novel sources of pancreatic islets or insulin‐producing cells are required to provide sufficient amounts of donor tissues. To overcome this limitation, the use of pancreatic progenitor cells is gaining more attention. In particular, pancreatic exocrine cells, such as duct epithelial cells and acinar cells, are attractive candidates for β‐cell regeneration because of their differentiation potential and pancreatic lineage characteristics. It has been assumed that β‐cell neogenesis from pancreatic progenitor cells could occur in pancreatic ducts in the postnatal stage. Several studies have shown that insulin‐producing cells can arise in the duct tissue of the adult pancreas. Acinar cells also might have the potential to differentiate into insulin‐producing cells. The present review summarizes recent progress in research on the transdifferentiation of pancreatic exocrine cells into insulin‐producing cells, especially duct and acinar cells.
Collapse
Affiliation(s)
- Hyo-Sup Kim
- Division of Endocrinology and Metabolism Department of Medicine Sungkyunkwan University School of Medicine Samsung Biomedical Research Institute Samsung Medical Center Seoul Korea
| | - Moon-Kyu Lee
- Division of Endocrinology and Metabolism Department of Medicine Sungkyunkwan University School of Medicine Samsung Biomedical Research Institute Samsung Medical Center Seoul Korea
| |
Collapse
|
155
|
Hasni Ebou M, Singh-Estivalet A, Launay JM, Callebert J, Tronche F, Ferré P, Gautier JF, Guillemain G, Bréant B, Blondeau B, Riveline JP. Glucocorticoids Inhibit Basal and Hormone-Induced Serotonin Synthesis in Pancreatic Beta Cells. PLoS One 2016; 11:e0149343. [PMID: 26901633 PMCID: PMC4763453 DOI: 10.1371/journal.pone.0149343] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/29/2016] [Indexed: 12/31/2022] Open
Abstract
Diabetes is a major complication of chronic Glucocorticoids (GCs) treatment. GCs induce insulin resistance and also inhibit insulin secretion from pancreatic beta cells. Yet, a full understanding of this negative regulation remains to be deciphered. In the present study, we investigated whether GCs could inhibit serotonin synthesis in beta cell since this neurotransmitter has been shown to be involved in the regulation of insulin secretion. To this aim, serotonin synthesis was evaluated in vitro after treatment with GCs of either islets from CD1 mice or MIN6 cells, a beta-cell line. We also explored the effect of GCs on the stimulation of serotonin synthesis by several hormones such as prolactin and GLP 1. We finally studied this regulation in islet in two in vivo models: mice treated with GCs and with liraglutide, a GLP1 analog, and mice deleted for the glucocorticoid receptor in the pancreas. We showed in isolated islets and MIN6 cells that GCs decreased expression and activity of the two key enzymes of serotonin synthesis, Tryptophan Hydroxylase 1 (Tph1) and 2 (Tph2), leading to reduced serotonin contents. GCs also blocked the induction of serotonin synthesis by prolactin or by a previously unknown serotonin activator, the GLP-1 analog exendin-4. In vivo, activation of the Glucagon-like-Peptide-1 receptor with liraglutide during 4 weeks increased islet serotonin contents and GCs treatment prevented this increase. Finally, islets from mice deleted for the GR in the pancreas displayed an increased expression of Tph1 and Tph2 and a strong increased serotonin content per islet. In conclusion, our results demonstrate an original inhibition of serotonin synthesis by GCs, both in basal condition and after stimulation by prolactin or activators of the GLP-1 receptor. This regulation may contribute to the deleterious effects of GCs on beta cells.
Collapse
Affiliation(s)
- Moina Hasni Ebou
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Amrit Singh-Estivalet
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Jean-Marie Launay
- INSERM U942, Assistance Publique–Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Service de Biochimie, Paris, France
| | - Jacques Callebert
- INSERM U942, Assistance Publique–Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Service de Biochimie, Paris, France
| | - François Tronche
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- CNRS UMR INSERM 952-CNRS 7224, Paris, France
| | - Pascal Ferré
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Jean-François Gautier
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Department of Diabetes and Endocrinology, Hôpital Lariboisière, AP-HP, Paris, France
- Université Paris Diderot, Paris, France
| | - Ghislaine Guillemain
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Bernadette Bréant
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
| | - Bertrand Blondeau
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- * E-mail:
| | - Jean-Pierre Riveline
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC, Univ Paris 06, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Department of Diabetes and Endocrinology, Hôpital Lariboisière, AP-HP, Paris, France
| |
Collapse
|
156
|
Ganic E, Singh T, Luan C, Fadista J, Johansson JK, Cyphert HA, Bennet H, Storm P, Prost G, Ahlenius H, Renström E, Stein R, Groop L, Fex M, Artner I. MafA-Controlled Nicotinic Receptor Expression Is Essential for Insulin Secretion and Is Impaired in Patients with Type 2 Diabetes. Cell Rep 2016; 14:1991-2002. [PMID: 26904947 PMCID: PMC5918632 DOI: 10.1016/j.celrep.2016.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/18/2015] [Accepted: 01/22/2016] [Indexed: 11/23/2022] Open
Abstract
Monoamine and acetylcholine neurotransmitters from the autonomic nervous system (ANS) regulate insulin secretion in pancreatic islets. The molecular mechanisms controlling neurotransmitter signaling in islet β cells and their impact on diabetes development are only partially understood. Using a glucose-intolerant, MafA-deficient mouse model, we demonstrate that MAFA controls ANS-mediated insulin secretion by activating the transcription of nicotinic (ChrnB2 and ChrnB4) and adrenergic (Adra2A) receptor genes, which are integral parts of acetylcholine-and monoamine-signaling pathways. We show that acetylcholine-mediated insulin secretion requires nicotinic signaling and that nicotinic receptor expression is positively correlated with insulin secretion and glycemic control in human donor islets. Moreover, polymorphisms spanning MAFA-binding regions within the human CHRNB4 gene are associated with type 2 diabetes. Our data show that MAFA transcriptional activity is required for establishing β cell sensitivity to neurotransmitter signaling and identify nicotinic signaling as a modulator of insulin secretion impaired in type 2 diabetes.
Collapse
MESH Headings
- Animals
- Autonomic Nervous System/metabolism
- Binding Sites
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Female
- Gene Expression Regulation
- Glucose Tolerance Test
- Humans
- Insulin/genetics
- Insulin/metabolism
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Maf Transcription Factors, Large/genetics
- Maf Transcription Factors, Large/metabolism
- Mice
- Mice, Knockout
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Polymorphism, Genetic
- Protein Binding
- Receptors, Adrenergic, alpha-2/genetics
- Receptors, Adrenergic, alpha-2/metabolism
- Receptors, Nicotinic/genetics
- Receptors, Nicotinic/metabolism
- Signal Transduction
- Transcription, Genetic
Collapse
Affiliation(s)
- Elvira Ganic
- Stem Cell Center, Lund University, Klinikgatan 26, Lund 22184, Sweden; Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Tania Singh
- Stem Cell Center, Lund University, Klinikgatan 26, Lund 22184, Sweden; Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Cheng Luan
- Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - João Fadista
- Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Jenny K Johansson
- Stem Cell Center, Lund University, Klinikgatan 26, Lund 22184, Sweden; Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Holly Ann Cyphert
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Hedvig Bennet
- Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Petter Storm
- Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Gaëlle Prost
- Stem Cell Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Henrik Ahlenius
- Stem Cell Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Erik Renström
- Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Leif Groop
- Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Malin Fex
- Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden
| | - Isabella Artner
- Stem Cell Center, Lund University, Klinikgatan 26, Lund 22184, Sweden; Lund University Diabetes Center, Lund University, Klinikgatan 26, Lund 22184, Sweden.
| |
Collapse
|
157
|
Knockin of Cre Gene at Ins2 Locus Reveals No Cre Activity in Mouse Hypothalamic Neurons. Sci Rep 2016; 6:20438. [PMID: 26830324 PMCID: PMC4735843 DOI: 10.1038/srep20438] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/04/2016] [Indexed: 12/20/2022] Open
Abstract
The recombination efficiency and cell specificity of Cre driver lines are critical for exploring pancreatic β cell biology with the Cre/LoxP approach. Some commonly used Cre lines are based on the short Ins2 promoter fragment and show recombination activity in hypothalamic neurons; however, whether this stems from endogenous Ins2 promoter activity remains controversial. In this study, we generated Ins2-Cre knockin mice with a targeted insertion of IRES-Cre at the Ins2 locus and demonstrated with a cell lineage tracing study that the Ins2 gene is not transcriptionally active in the hypothalamus. The Ins2-Cre driver line displayed robust Cre expression and activity in pancreatic β cells without significant alterations in insulin expression. In the brain, Cre activity was mainly restricted to the choroid plexus, without significant recombination detected in the hippocampus or hypothalamus by the LacZ or fluorescent tdTomato reporters. Furthermore, Ins2-Cre mice exhibited normal glucose tolerance and insulin secretion upon glucose stimulation in vivo. In conclusion, this Ins2-Cre driver line allowed high-fidelity detection of endogenous Ins2 promoter activity in vivo, and the negative activity in the hypothalamus demonstrated that this system is a promising alternative tool for studying β cell biology.
Collapse
|
158
|
Kofent J, Spagnoli FM. Xenopus as a model system for studying pancreatic development and diabetes. Semin Cell Dev Biol 2016; 51:106-16. [PMID: 26806634 DOI: 10.1016/j.semcdb.2016.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/06/2016] [Indexed: 02/07/2023]
Abstract
Diabetes is a chronic disease caused by the loss or dysfunction of the insulin-producing β-cells in the pancreas. To date, much of our knowledge about β-cells in humans comes from studying rare monogenic forms of diabetes. Importantly, the majority of mutations so far associated to monogenic diabetes are in genes that exert a regulatory role in pancreatic development and/or β-cell function. Thus, the identification and study of novel mutations open an unprecedented window into human pancreatic development. In this review, we summarize major advances in the genetic dissection of different types of monogenic diabetes and the insights gained from a developmental perspective. We highlight future challenges to bridge the gap between the fast accumulation of genetic data through next-generation sequencing and the need of functional insights into disease mechanisms. Lastly, we discuss the relevance and advantages of studying candidate gene variants in vivo using the Xenopus as model system.
Collapse
Affiliation(s)
- Julia Kofent
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, D-13125 Berlin, Germany
| | - Francesca M Spagnoli
- Max Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, D-13125 Berlin, Germany.
| |
Collapse
|
159
|
Abstract
A wealth of data and comprehensive reviews exist on pancreas development in mammals, primarily mice, and other vertebrates. By contrast, human pancreatic development has been less comprehensively reviewed. Here, we draw together those studies conducted directly in human embryonic and fetal tissue to provide an overview of what is known about human pancreatic development. We discuss the relevance of this work to manufacturing insulin-secreting β-cells from pluripotent stem cells and to different aspects of diabetes, especially permanent neonatal diabetes, and its underlying causes.
Collapse
Affiliation(s)
- Rachel E Jennings
- Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Oxford Rd, Manchester M13 9PT, UK Endocrinology Department, Central Manchester University Hospitals NHS Foundation Trust, Grafton St, Manchester M13 9WU, UK
| | - Andrew A Berry
- Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Oxford Rd, Manchester M13 9PT, UK
| | - James P Strutt
- Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Oxford Rd, Manchester M13 9PT, UK
| | - David T Gerrard
- Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Oxford Rd, Manchester M13 9PT, UK Bioinformatics Unit, Faculty of Life Science, Manchester Academic Health Science Centre, University of Manchester, Oxford Rd, Manchester M13 9PT, UK
| | - Neil A Hanley
- Centre for Endocrinology and Diabetes, Institute of Human Development, Faculty of Medical & Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Oxford Rd, Manchester M13 9PT, UK Endocrinology Department, Central Manchester University Hospitals NHS Foundation Trust, Grafton St, Manchester M13 9WU, UK
| |
Collapse
|
160
|
Abstract
PTEN plays an important role in diabetes pathogenesis not only as a key negative regulator of the PI3K/Akt pathway required for insulin action, but also via its role in other cell processes required to maintain metabolic homeostasis. We describe the generation of tissue-specific PTEN knockout mice and models of both type 1 and type 2 diabetes, which we have found useful for the study of diabetes pathogenesis. We also outline common methods suitable for the characterization of glucose homeostasis in rodent models, including techniques to measure beta cell function and insulin sensitivity.
Collapse
Affiliation(s)
- Cynthia T Luk
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada, M5G 2C4
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada, M5G 2C4
| | - Stephanie A Schroer
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada, M5G 2C4
| | - Minna Woo
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada, M5G 2C4.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada, M5G 2C4.
- Division of Endocrinology, Department of Medicine, University Health Network, University of Toronto, Toronto, ON, Canada, M5G 2C4.
- Toronto General Research Institute, 101 College Street, MaRS Centre/TMDT, Room 10-363, Toronto, ON, Canada, M5G 1L7.
| |
Collapse
|
161
|
Abstract
Lineage tracing studies have revealed that transcription factors play a cardinal role in pancreatic development, differentiation and function. Three transitions define pancreatic organogenesis, differentiation and maturation. In the primary transition, when pancreatic organogenesis is initiated, there is active proliferation of pancreatic progenitor cells. During the secondary transition, defined by differentiation, there is growth, branching, differentiation and pancreatic cell lineage allocation. The tertiary transition is characterized by differentiated pancreatic cells that undergo further remodeling, including apoptosis, replication and neogenesis thereby establishing a mature organ. Transcription factors function at multiple levels and may regulate one another and auto-regulate. The interaction between extrinsic signals from non-pancreatic tissues and intrinsic transcription factors form a complex gene regulatory network ultimately culminating in the different cell lineages and tissue types in the developing pancreas. Mutations in these transcription factors clinically manifest as subtypes of diabetes mellitus. Current treatment for diabetes is not curative and thus, developmental biologists and stem cell researchers are utilizing knowledge of normal pancreatic development to explore novel therapeutic alternatives. This review summarizes current knowledge of transcription factors involved in pancreatic development and β-cell differentiation in rodents.
Collapse
Affiliation(s)
- Reshmi Dassaye
- a Discipline of Pharmaceutical Sciences; Nelson R. Mandela School of Medicine, University of KwaZulu-Natal , Durban , South Africa
| | - Strini Naidoo
- a Discipline of Pharmaceutical Sciences; Nelson R. Mandela School of Medicine, University of KwaZulu-Natal , Durban , South Africa
| | - Marlon E Cerf
- b Diabetes Discovery Platform, South African Medical Research Council , Cape Town , South Africa
| |
Collapse
|
162
|
Li XY, Zhai WJ, Teng CB. Notch Signaling in Pancreatic Development. Int J Mol Sci 2015; 17:ijms17010048. [PMID: 26729103 PMCID: PMC4730293 DOI: 10.3390/ijms17010048] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/22/2015] [Accepted: 12/24/2015] [Indexed: 12/12/2022] Open
Abstract
The Notch signaling pathway plays a significant role in embryonic cell fate determination and adult tissue homeostasis. Various studies have demonstrated the deep involvement of Notch signaling in the development of the pancreas and the lateral inhibition of Notch signaling in pancreatic progenitor differentiation and maintenance. The targeted inactivation of the Notch pathway components promotes premature differentiation of the endocrine pancreas. However, there is still the contrary opinion that Notch signaling specifies the endocrine lineage. Here, we review the current knowledge of the Notch signaling pathway in pancreatic development and its crosstalk with the Wingless and INT-1 (Wnt) and fibroblast growth factor (FGF) pathways.
Collapse
Affiliation(s)
- Xu-Yan Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
- College of Life Sciences, Agriculture and Forestry, Qiqihar University, Qiqihar 161006, China.
| | - Wen-Jun Zhai
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Chun-Bo Teng
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| |
Collapse
|
163
|
Alejandro EU, Bozadjieva N, Kumusoglu D, Abdulhamid S, Levine H, Haataja L, Vadrevu S, Satin LS, Arvan P, Bernal-Mizrachi E. Disruption of O-linked N-Acetylglucosamine Signaling Induces ER Stress and β Cell Failure. Cell Rep 2015; 13:2527-2538. [PMID: 26673325 PMCID: PMC4839001 DOI: 10.1016/j.celrep.2015.11.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/22/2015] [Accepted: 11/03/2015] [Indexed: 11/30/2022] Open
Abstract
Nutrient levels dictate the activity of O-linked N-acetylglucosamine transferase (OGT) to regulate O-GlcNAcylation, a post-translational modification mechanism to "fine-tune" intracellular signaling and metabolic status. However, the requirement of O-GlcNAcylation for maintaining glucose homeostasis by regulating pancreatic β cell mass and function is unclear. Here, we reveal that mice lacking β cell OGT (βOGT-KO) develop diabetes and β cell failure. βOGT-KO mice demonstrated increased ER stress and distended ER architecture, and these changes ultimately caused the loss of β cell mass due to ER-stress-induced apoptosis and decreased proliferation. Akt1/2 signaling was also dampened in βOGT-KO islets. The mechanistic role of these processes was demonstrated by rescuing the phenotype of βOGT-KO mice with concomitant Chop gene deletion or genetic reconstitution of Akt2. These findings identify OGT as a regulator of β cell mass and function and provide a direct link between O-GlcNAcylation and β cell survival by regulation of ER stress responses and modulation of Akt1/2 signaling.
Collapse
Affiliation(s)
- Emilyn U Alejandro
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109-0678, USA
| | - Nadejda Bozadjieva
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109-0678, USA
| | - Doga Kumusoglu
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109-0678, USA
| | - Sarah Abdulhamid
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109-0678, USA
| | - Hannah Levine
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109-0678, USA
| | - Leena Haataja
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109-0678, USA
| | - Suryakiran Vadrevu
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109-0678, USA
| | - Leslie S Satin
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109-0678, USA
| | - Peter Arvan
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109-0678, USA
| | - Ernesto Bernal-Mizrachi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI 48109-0678, USA; VA Ann Arbor Healthcare System, Ann Arbor, MI 48109-0678, USA.
| |
Collapse
|
164
|
Ahmad Z, Rafeeq M, Collombat P, Mansouri A. Pax6 Inactivation in the Adult Pancreas Reveals Ghrelin as Endocrine Cell Maturation Marker. PLoS One 2015; 10:e0144597. [PMID: 26658466 PMCID: PMC4676685 DOI: 10.1371/journal.pone.0144597] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/20/2015] [Indexed: 11/18/2022] Open
Abstract
The transcription factor Pax6 is an important regulator of development and cell differentiation in various organs. Thus, Pax6 was shown to promote neural development in the cerebral cortex and spinal cord, and to control pancreatic endocrine cell genesis. However, the role of Pax6 in distinct endocrine cells of the adult pancreas has not been addressed. We report the conditional inactivation of Pax6 in insulin and glucagon producing cells of the adult mouse pancreas. In the absence of Pax6, beta- and alpha-cells lose their molecular maturation characteristics. Our findings provide strong evidence that Pax6 is responsible for the maturation of beta-, and alpha-cells, but not of delta-, and PP-cells. Moreover, lineage-tracing experiments demonstrate that Pax6-deficient beta- and alpha-cells are shunted towards ghrelin marked cells, sustaining the idea that ghrelin may represent a marker for endocrine cell maturation.
Collapse
Affiliation(s)
- Zeeshan Ahmad
- Max Planck Institute for Biophysical Chemistry, Department of Molecular Developmental Biology, RG Molecular Cell Differentiation, Goettingen, Germany
- * E-mail: (AM); (ZA)
| | - Maria Rafeeq
- Max Planck Institute for Biophysical Chemistry, Department of Molecular Developmental Biology, RG Molecular Cell Differentiation, Goettingen, Germany
| | - Patrick Collombat
- Université de Nice Sophia Antipolis, Nice, France
- Inserm U1091, IBV, Diabetes Genetics Team, Nice, France
- JDRF, New York, NY, United States of America
- Genome and Stem Cell Center, GENKOK, Erciyes University, Kayseri, Turkey
| | - Ahmed Mansouri
- Max Planck Institute for Biophysical Chemistry, Department of Molecular Developmental Biology, RG Molecular Cell Differentiation, Goettingen, Germany
- JDRF, New York, NY, United States of America
- University of Goettingen, Department of Clinical Neurophysiology, Goettingen, Germany
- * E-mail: (AM); (ZA)
| |
Collapse
|
165
|
Fukaya M, Brorsson CA, Meyerovich K, Catrysse L, Delaroche D, Vanzela EC, Ortis F, Beyaert R, Nielsen LB, Andersen ML, Mortensen HB, Pociot F, van Loo G, Størling J, Cardozo AK. A20 Inhibits β-Cell Apoptosis by Multiple Mechanisms and Predicts Residual β-Cell Function in Type 1 Diabetes. Mol Endocrinol 2015; 30:48-61. [PMID: 26652732 DOI: 10.1210/me.2015-1176] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Activation of the transcription factor nuclear factor kappa B (NFkB) contributes to β-cell death in type 1 diabetes (T1D). Genome-wide association studies have identified the gene TNF-induced protein 3 (TNFAIP3), encoding for the zinc finger protein A20, as a susceptibility locus for T1D. A20 restricts NF-κB signaling and has strong antiapoptotic activities in β-cells. Although the role of A20 on NF-κB inhibition is well characterized, its other antiapoptotic functions are largely unknown. By studying INS-1E cells and rat dispersed islet cells knocked down or overexpressing A20 and islets isolated from the β-cell-specific A20 knockout mice, we presently demonstrate that A20 has broader effects in β-cells that are not restricted to inhibition of NF-κB. These involves, suppression of the proapoptotic mitogen-activated protein kinase c-Jun N-terminal kinase (JNK), activation of survival signaling via v-akt murine thymoma viral oncogene homolog (Akt) and consequently inhibition of the intrinsic apoptotic pathway. Finally, in a cohort of T1D children, we observed that the risk allele of the rs2327832 single nucleotide polymorphism of TNFAIP3 predicted lower C-peptide and higher hemoglobin A1c (HbA1c) levels 12 months after disease onset, indicating reduced residual β-cell function and impaired glycemic control. In conclusion, our results indicate a critical role for A20 in the regulation of β-cell survival and unveil novel mechanisms by which A20 controls β-cell fate. Moreover, we identify the single nucleotide polymorphism rs2327832 of TNFAIP3 as a possible prognostic marker for diabetes outcome in children with T1D.
Collapse
Affiliation(s)
- Makiko Fukaya
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Caroline A Brorsson
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Kira Meyerovich
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Leen Catrysse
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Diane Delaroche
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Emerielle C Vanzela
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Fernanda Ortis
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Rudi Beyaert
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Lotte B Nielsen
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Marie L Andersen
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Henrik B Mortensen
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Flemming Pociot
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Geert van Loo
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Joachim Størling
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| | - Alessandra K Cardozo
- Université Libre de Bruxelles Center for Diabetes Research (M.F., K.M., D.D., E.C.V., A.K.C.), Free University Brussels, 1070 Brussels, Belgium; Copenhagen Diabetes Research Center (C.A.B., L.B.N., M.L.A., H.B.M., F.P., J.S.), Department of Pediatrics E, Copenhagen University Hospital Herlev, DL-2730 Herlev, Denmark; Inflammation Research Center (L.C., R.B., G.v.L.), Vlaams Instituut voor Biotechnologie, 9052 Gent, Belgium; Department of Biomedical Molecular Biology (L.C., R.B., G.v.L.), 9052 Gent University, Gent, Belgium; Laboratory of Endocrine Pancreas and Metabolism (E.C.V.), Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-970 Campinas, Brazil; and Department of Development and Cellular Biology (F.O.), Institute of Biomedical Sciences, 05508-900 São Paulo University, São Paulo, Brazil
| |
Collapse
|
166
|
Abstract
Although similar, mouse and human pancreatic development and beta cell physiology have significant differences. For this reason, mouse models present shortcomings that can obscure the understanding of human diabetes pathology. Progress in the field of human pluripotent stem cell (hPSC) differentiation now makes it possible to derive unlimited numbers of human beta cells in vitro. This constitutes an invaluable approach to gain insight into human beta cell development and physiology and to generate improved disease models. Here we summarize the main differences in terms of development and physiology of the pancreatic endocrine cells between mouse and human, and describe the recent progress in modeling diabetes using hPSC. We highlight the need of developing more physiological hPSC-derived beta cell models and anticipate the future prospects of these approaches.
Collapse
Affiliation(s)
- Diego Balboa
- University of Helsinki, Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Center, Finland
| | - Timo Otonkoski
- University of Helsinki, Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Center, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Finland.
| |
Collapse
|
167
|
Abstract
The alpha cells that co-occupy the islets in association with beta cells have been long recognized as the source of glucagon, a hyperglycemia-producing and diabetogenic hormone. Although the mechanisms that control the functions of alpha cells, glucagon secretion, and the role of glucagon in diabetes have remained somewhat enigmatic over the fifty years since their discovery, seminal findings during the past few years have moved alpha cells into the spotlight of scientific discovery. These findings obtained largely from studies in mice are: Alpha cells have the capacity to trans-differentiate into insulin-producing beta cells. Alpha cells contain a GLP-1 generating system that produces GLP-1 locally for paracrine actions within the islets that likely promotes beta cell growth and survival and maintains beta cell mass. Impairment of glucagon signaling both prevents the occurrence of diabetes in conditions of the near absence of insulin and expands alpha cell mass. Alpha cells appear to serve as helper cells or guardians of beta cells to ensure their health and well-being. Of potential relevance to the possibility of promoting the transformation of alpha to beta cells is the observation that impairment of glucagon signaling leads to a marked increase in alpha cell mass in the islets. Such alpha cell hyperplasia provides an increased supply of alpha cells for their transdifferentiation into new beta cells. In this review we discuss these recent discoveries from the perspective of their potential relevance to the treatment of diabetes.
Collapse
Affiliation(s)
- Violeta Stanojevic
- Laboratory of Molecular Endocrinology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Joel F Habener
- Laboratory of Molecular Endocrinology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
168
|
Tauscher S, Nakagawa H, Völker K, Gaßner B, Pröhl S, Kuhn M. Role of atrial natriuretic peptide (ANP) in the regulation of insulin secretion and vitality of pancreatic ß cells. BMC Pharmacol Toxicol 2015. [PMCID: PMC4565191 DOI: 10.1186/2050-6511-16-s1-a92] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
169
|
Oropeza D, Jouvet N, Budry L, Campbell JE, Bouyakdan K, Lacombe J, Perron G, Bergeron V, Neuman JC, Brar HK, Fenske RJ, Meunier C, Sczelecki S, Kimple ME, Drucker DJ, Screaton RA, Poitout V, Ferron M, Alquier T, Estall JL. Phenotypic Characterization of MIP-CreERT1Lphi Mice With Transgene-Driven Islet Expression of Human Growth Hormone. Diabetes 2015; 64:3798-807. [PMID: 26153246 PMCID: PMC4613972 DOI: 10.2337/db15-0272] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/24/2015] [Indexed: 01/17/2023]
Abstract
There is growing concern over confounding artifacts associated with β-cell-specific Cre-recombinase transgenic models, raising questions about their general usefulness in research. The inducible β-cell-specific transgenic (MIP-CreERT(1Lphi)) mouse was designed to circumvent many of these issues, and we investigated whether this tool effectively addressed concerns of ectopic expression and disruption of glucose metabolism. Recombinase activity was absent from the central nervous system using a reporter line and high-resolution microscopy. Despite increased pancreatic insulin content, MIP-CreERT mice on a chow diet exhibited normal ambient glycemia, glucose tolerance and insulin sensitivity, and appropriate insulin secretion in response to glucose in vivo and in vitro. However, MIP-CreERT mice on different genetic backgrounds were protected from high-fat/ streptozotocin (STZ)-induced hyperglycemia that was accompanied by increased insulin content and islet density. Ectopic human growth hormone (hGH) was highly expressed in MIP-CreERT islets independent of tamoxifen administration. Circulating insulin levels remained similar to wild-type controls, whereas STZ-associated increases in α-cell number and serum glucagon were significantly blunted in MIP-CreERT(1Lphi) mice, possibly due to paracrine effects of hGH-induced serotonin expression. These studies reveal important new insight into the strengths and limitations of the MIP-CreERT mouse line for β-cell research.
Collapse
Affiliation(s)
- Daniel Oropeza
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Nathalie Jouvet
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Lionel Budry
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Jonathan E Campbell
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Khalil Bouyakdan
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada Département de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Julie Lacombe
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada
| | - Gabrielle Perron
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Valerie Bergeron
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada Département de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Joshua C Neuman
- Department of Medicine and Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Harpreet K Brar
- Department of Medicine and Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Rachel J Fenske
- Department of Medicine and Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Clemence Meunier
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada
| | - Sarah Sczelecki
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Michelle E Kimple
- Department of Medicine and Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Robert A Screaton
- Department of Cellular and Molecular Medicine, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada Département de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Mathieu Ferron
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada Division of Experimental Medicine, McGill University, Montreal, QC, Canada Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Thierry Alquier
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada Département de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Jennifer L Estall
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada Division of Experimental Medicine, McGill University, Montreal, QC, Canada Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada Département de Médecine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
170
|
Greggio C, De Franceschi F, Grapin-Botton A. Concise reviews: In vitro-produced pancreas organogenesis models in three dimensions: self-organization from few stem cells or progenitors. Stem Cells 2015; 33:8-14. [PMID: 25185771 DOI: 10.1002/stem.1828] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/14/2014] [Indexed: 01/10/2023]
Abstract
Three-dimensional models of organ biogenesis have recently flourished. They promote a balance between stem/progenitor cell expansion and differentiation without the constraints of flat tissue culture vessels, allowing for autonomous self-organization of cells. Such models allow the formation of miniature organs in a dish and are emerging for the pancreas, starting from embryonic progenitors and adult cells. This review focuses on the currently available systems and how these allow new types of questions to be addressed. We discuss the expected advancements including their potential to study human pancreas development and function as well as to develop diabetes models and therapeutic cells.
Collapse
Affiliation(s)
- Chiara Greggio
- Ecole Polytechnique Fédérale de Lausanne, Life Sciences, Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland; Département de Physiologie, Université de Lausanne, Rue du Bugnon 7, Lausanne, Switzerland
| | | | | |
Collapse
|
171
|
Mastracci TL, Robertson MA, Mirmira RG, Anderson RM. Polyamine biosynthesis is critical for growth and differentiation of the pancreas. Sci Rep 2015; 5:13269. [PMID: 26299433 PMCID: PMC4547391 DOI: 10.1038/srep13269] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/30/2015] [Indexed: 02/03/2023] Open
Abstract
The pancreas, in most studied vertebrates, is a compound organ with both exocrine and endocrine functions. The exocrine compartment makes and secretes digestive enzymes, while the endocrine compartment, organized into islets of Langerhans, produces hormones that regulate blood glucose. High concentrations of polyamines, which are aliphatic amines, are reported in exocrine and endocrine cells, with insulin-producing β cells showing the highest concentrations. We utilized zebrafish as a model organism, together with pharmacological inhibition or genetic manipulation, to determine how polyamine biosynthesis functions in pancreatic organogenesis. We identified that inhibition of polyamine biosynthesis reduces exocrine pancreas and β cell mass, and that these reductions are at the level of differentiation. Moreover, we demonstrate that inhibition of ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine biosynthesis, phenocopies inhibition or knockdown of the enzyme deoxyhypusine synthase (DHS). These data identify that the pancreatic requirement for polyamine biosynthesis is largely mediated through a requirement for spermidine for the downstream posttranslational modification of eIF5A by its enzymatic activator DHS, which in turn impacts mRNA translation. Altogether, we have uncovered a role for polyamine biosynthesis in pancreatic organogenesis and identified that it may be possible to exploit polyamine biosynthesis to manipulate pancreatic cell differentiation.
Collapse
Affiliation(s)
- Teresa L Mastracci
- Department of Pediatrics, Indiana University School of Medicine, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| | - Morgan A Robertson
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics, Indiana University School of Medicine, USA.,Department of Physiology, Indiana University School of Medicine, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| | - Ryan M Anderson
- Department of Pediatrics, Indiana University School of Medicine, USA.,Department of Physiology, Indiana University School of Medicine, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, USA
| |
Collapse
|
172
|
Pax4 acts as a key player in pancreas development and plasticity. Semin Cell Dev Biol 2015; 44:107-14. [DOI: 10.1016/j.semcdb.2015.08.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 08/05/2015] [Accepted: 08/22/2015] [Indexed: 11/19/2022]
|
173
|
Jiang FX, Mishina Y, Baten A, Morahan G, Harrison LC. Transcriptome of pancreas-specific Bmpr1a-deleted islets links to TPH1-5-HT axis. Biol Open 2015; 4:1016-23. [PMID: 26187948 PMCID: PMC4542282 DOI: 10.1242/bio.011858] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling is crucial for the development and function of numerous organs, but its role on the function of pancreatic islets is not completely clear. To explore this question, we applied the high throughput transcriptomic analyses on the islets isolated from mice with a pancreas-specific deletion of the gene, Bmpr1a, encoding the type 1a BMP receptor. Consistently, these pBmpr1aKO mice had impaired glucose homeostasis at 3 months, and were more severely affected at 12 months of age. These had lower fasting blood insulin concentrations, with reduced expression of several key regulators of β-cell function. Importantly, transcriptomic profiling of 3-month pBmpr1aKO islets and bioinformatic analyses revealed abnormal expression of 203 metabolic genes. Critically among these, the tryptophan hydroxylase 1 gene (Tph1), encoding the rate-limiting enzyme for the production of 5-hydroxytryptamine (5-HT) was the highest over-expressed one. 5-HT is an important regulator of insulin secretion from β cells. Treatment with excess 5-HT inhibited this secretion. Thus our transcriptomic analysis links two highly conserved molecular pathways the BMP signaling and the TPH1–5-HT axis on glucose homeostasis.
Collapse
Affiliation(s)
- Fang-Xu Jiang
- The Walter & Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Akma Baten
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Grant Morahan
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Leonard C Harrison
- The Walter & Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| |
Collapse
|
174
|
Solomou A, Meur G, Bellomo E, Hodson DJ, Tomas A, Li SM, Philippe E, Herrera PL, Magnan C, Rutter GA. The Zinc Transporter Slc30a8/ZnT8 Is Required in a Subpopulation of Pancreatic α-Cells for Hypoglycemia-induced Glucagon Secretion. J Biol Chem 2015; 290:21432-42. [PMID: 26178371 PMCID: PMC4571871 DOI: 10.1074/jbc.m115.645291] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Indexed: 12/02/2022] Open
Abstract
SLC30A8 encodes a zinc transporter ZnT8 largely restricted to pancreatic islet β- and α-cells, and responsible for zinc accumulation into secretory granules. Although common SLC30A8 variants, believed to reduce ZnT8 activity, increase type 2 diabetes risk in humans, rare inactivating mutations are protective. To investigate the role of Slc30a8 in the control of glucagon secretion, Slc30a8 was inactivated selectively in α-cells by crossing mice with alleles floxed at exon 1 to animals expressing Cre recombinase under the pre-proglucagon promoter. Further crossing to Rosa26:tdRFP mice, and sorting of RFP+: glucagon+ cells from KO mice, revealed recombination in ∼30% of α-cells, of which ∼50% were ZnT8-negative (14 ± 1.8% of all α-cells). Although glucose and insulin tolerance were normal, female αZnT8KO mice required lower glucose infusion rates during hypoglycemic clamps and displayed enhanced glucagon release (p < 0.001) versus WT mice. Correspondingly, islets isolated from αZnT8KO mice secreted more glucagon at 1 mm glucose, but not 17 mm glucose, than WT controls (n = 5; p = 0.008). Although the expression of other ZnT family members was unchanged, cytoplasmic (n = 4 mice per genotype; p < 0.0001) and granular (n = 3, p < 0.01) free Zn2+ levels were significantly lower in KO α-cells versus control cells. In response to low glucose, the amplitude and frequency of intracellular Ca2+ increases were unchanged in α-cells of αZnT8KO KO mice. ZnT8 is thus important in a subset of α-cells for normal responses to hypoglycemia and acts via Ca2+-independent mechanisms.
Collapse
Affiliation(s)
- Antonia Solomou
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Gargi Meur
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Elisa Bellomo
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - David J Hodson
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Alejandra Tomas
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom, the Department of Cell Biology, Institute of Ophthalmology, University College London, Greater London EC1V 9EL, United Kingdom
| | - Stéphanie Migrenne Li
- the University Paris Diderot-Paris 7, Unit of Functional and Adaptive Biology (BFA) EAC 7059 CNRS, 75013 Paris, France, and
| | - Erwann Philippe
- the University Paris Diderot-Paris 7, Unit of Functional and Adaptive Biology (BFA) EAC 7059 CNRS, 75013 Paris, France, and
| | - Pedro L Herrera
- the Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, 1211 Geneva-4, Switzerland
| | - Christophe Magnan
- the University Paris Diderot-Paris 7, Unit of Functional and Adaptive Biology (BFA) EAC 7059 CNRS, 75013 Paris, France, and
| | - Guy A Rutter
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom,
| |
Collapse
|
175
|
Márquez-Aguirre AL, Canales-Aguirre AA, Padilla-Camberos E, Esquivel-Solis H, Díaz-Martínez NE. Development of the endocrine pancreas and novel strategies for β-cell mass restoration and diabetes therapy. ACTA ACUST UNITED AC 2015; 48:765-76. [PMID: 26176316 PMCID: PMC4568803 DOI: 10.1590/1414-431x20154363] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 03/22/2015] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus represents a serious public health problem owing to its global
prevalence in the last decade. The causes of this metabolic disease include
dysfunction and/or insufficient number of β cells. Existing diabetes mellitus
treatments do not reverse or control the disease. Therefore, β-cell mass restoration
might be a promising treatment. Several restoration approaches have been developed:
inducing the proliferation of remaining insulin-producing cells, de
novo islet formation from pancreatic progenitor cells (neogenesis), and
converting non-β cells within the pancreas to β cells (transdifferentiation) are the
most direct, simple, and least invasive ways to increase β-cell mass. However, their
clinical significance is yet to be determined. Hypothetically, β cells or islet
transplantation methods might be curative strategies for diabetes mellitus; however,
the scarcity of donors limits the clinical application of these approaches. Thus,
alternative cell sources for β-cell replacement could include embryonic stem cells,
induced pluripotent stem cells, and mesenchymal stem cells. However, most
differentiated cells obtained using these techniques are functionally immature and
show poor glucose-stimulated insulin secretion compared with native β cells.
Currently, their clinical use is still hampered by ethical issues and the risk of
tumor development post transplantation. In this review, we briefly summarize the
current knowledge of mouse pancreas organogenesis, morphogenesis, and maturation,
including the molecular mechanisms involved. We then discuss two possible approaches
of β-cell mass restoration for diabetes mellitus therapy: β-cell regeneration and
β-cell replacement. We critically analyze each strategy with respect to the
accessibility of the cells, potential risk to patients, and possible clinical
outcomes.
Collapse
Affiliation(s)
- A L Márquez-Aguirre
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| | - A A Canales-Aguirre
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| | - E Padilla-Camberos
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| | - H Esquivel-Solis
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| | - N E Díaz-Martínez
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| |
Collapse
|
176
|
Ye L, Robertson MA, Hesselson D, Stainier DYR, Anderson RM. Glucagon is essential for alpha cell transdifferentiation and beta cell neogenesis. Development 2015; 142:1407-17. [PMID: 25852199 DOI: 10.1242/dev.117911] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The interconversion of cell lineages via transdifferentiation is an adaptive mode of tissue regeneration and an appealing therapeutic target. However, its clinical exploitation is contingent upon the discovery of contextual regulators of cell fate acquisition and maintenance. In murine models of diabetes, glucagon-secreting alpha cells transdifferentiate into insulin-secreting beta cells following targeted beta cell depletion, regenerating the form and function of the pancreatic islet. However, the molecular triggers of this mode of regeneration are unknown. Here, using lineage-tracing assays in a transgenic zebrafish model of beta cell ablation, we demonstrate conserved plasticity of alpha cells during islet regeneration. In addition, we show that glucagon expression is upregulated after injury. Through gene knockdown and rescue approaches, we also find that peptides derived from the glucagon gene are necessary for alpha-to-beta cell fate switching. Importantly, whereas beta cell neogenesis was stimulated by glucose, alpha-to-beta cell conversion was not, suggesting that transdifferentiation is not mediated by glucagon/GLP-1 control of hepatic glucose production. Overall, this study supports the hypothesis that alpha cells are an endogenous reservoir of potential new beta cells. It further reveals that glucagon plays an important role in maintaining endocrine cell homeostasis through feedback mechanisms that govern cell fate stability.
Collapse
Affiliation(s)
- Lihua Ye
- Wells Center for Pediatric Research and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2053, Indianapolis, IN 46202, USA
| | - Morgan A Robertson
- Wells Center for Pediatric Research and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2053, Indianapolis, IN 46202, USA
| | - Daniel Hesselson
- Department of Biochemistry and Biophysics, 1550 4th Street Rock Hall Room 381, University of California San Francisco, San Francisco, CA 94143, USA
| | - Didier Y R Stainier
- Department of Biochemistry and Biophysics, 1550 4th Street Rock Hall Room 381, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ryan M Anderson
- Wells Center for Pediatric Research and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Van Nuys Medical Sciences Building MS2053, Indianapolis, IN 46202, USA
| |
Collapse
|
177
|
Estall JL, Screaton RA. To Be(ta Cell) or Not to Be(ta cell): New Mouse Models for Studying Gene Function in the Pancreatic β-Cell. Endocrinology 2015; 156:2365-7. [PMID: 26091426 DOI: 10.1210/en.2015-1418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A challenge in the pancreatic β-cell field has been to identify a promoter fragment that is active only in the β-cell compartment and inactive in other regions, such as the hypothalamic region of the brain. The presence of Cre recombinase alone in some models may also affect glucoregulation, confounding interpretation of gene function in the β-cell. A paper presented within describes the development and characterization of 2 new transgenic mice expressing Cre recombinase under the mouse insulin1 promoter that are useful for β-cell-specific gene ablation: the first is constitutive and coexpresses DsRed (Ins1-Cre-DsRed); the second allows β-cell-specific expression of the reverse tetracycline-controlled transactivator, which can be used for drug-dependent expression of a target gene of interest for overexpression studies. These novel models show robust specificity and efficiency and will be valuable tools for functional studies of gene action in β-cells, potentially alleviating current issues associated with previously available mouse lines.
Collapse
Affiliation(s)
- Jennifer L Estall
- Institut de Recherches Cliniques de Montréal (J.L.E.), Montreal, Quebec, Canada H2W 1R7; and Department of Biochemistry (R.A.S.), University of Toronto and Sunnybrook Research Institute, Toronto, Canada M4N 3M5
| | - Robert A Screaton
- Institut de Recherches Cliniques de Montréal (J.L.E.), Montreal, Quebec, Canada H2W 1R7; and Department of Biochemistry (R.A.S.), University of Toronto and Sunnybrook Research Institute, Toronto, Canada M4N 3M5
| |
Collapse
|
178
|
Soedling H, Hodson DJ, Adrianssens AE, Gribble FM, Reimann F, Trapp S, Rutter GA. Limited impact on glucose homeostasis of leptin receptor deletion from insulin- or proglucagon-expressing cells. Mol Metab 2015; 4:619-30. [PMID: 26413468 PMCID: PMC4563029 DOI: 10.1016/j.molmet.2015.06.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/12/2015] [Indexed: 01/07/2023] Open
Abstract
AIMS/HYPOTHESIS The adipose tissue-derived hormone leptin plays an important role in the maintenance of body weight and glucose homeostasis. Leptin mediates its effects by interaction with leptin receptors (LepRb), which are highly expressed in the hypothalamus and other brain centres, and at lower levels in the periphery. Previous studies have used relatively promiscuous or inefficient Cre deleter strains, respectively, to explore the roles of LepR in pancreatic β and α cells. Here, we use two newly-developed Cre lines to explore the role of leptin signalling in insulin and proglucagon-expressing cells. METHODS Leptin receptor expression was measured in isolated mouse islets and highly-purified islet cells by RNASeq and quantitative RT-PCR. Mice lacking leptin signalling in pancreatic β, or in α and other proglucagon-expressing cells, were generated using Ins1Cre- or iGluCre-mediated recombination respectively of flox'd leptin receptor alleles. In vivo glucose homeostasis, changes in body weight, pancreatic histology and hormone secretion from isolated islets were assessed using standard techniques. RESULTS Leptin receptor mRNA levels were at or below the level of detection in wild-type adult mouse isolated islets and purified cells, and leptin signalling to Stat3 phosphorylation was undetectable. Whereas male mice further deleted for leptin receptors in β cells exhibited no abnormalities in glucose tolerance up to 16 weeks of age, females transiently displayed improved glucose tolerance at 8 weeks (11.2 ± 3.2% decrease in area under curve; p < 0.05), and improved (39.0 ± 13.0%, P < 0.05) glucose-stimulated insulin secretion in vitro. No differences were seen between genotypes in body weight, fasting glucose or β/α cell ratio. Deletion of LepR from α-cells, a minority of β cells, and a subset of proglucagon-expressing cells in the brain, exerted no effects on body weight, glucose or insulin tolerance, nor on pancreatic hormone secretion assessed in vivo and in vitro. CONCLUSIONS/INTERPRETATION The use here of a highly selective Cre recombinase indicates that leptin signalling plays a relatively minor, age- and sex-dependent role in the control of β cell function in the mouse. No in vivo role for leptin receptors on α cells, nor in other proglucagon-expressing cells, was detected in this study.
Collapse
Affiliation(s)
- Helen Soedling
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, du Cane Road, London W12 0NN, UK
| | - David J Hodson
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, du Cane Road, London W12 0NN, UK
| | | | - Fiona M Gribble
- University of Cambridge Metabolic Research Laboratories, Cambridge, UK
| | - Frank Reimann
- University of Cambridge Metabolic Research Laboratories, Cambridge, UK
| | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, du Cane Road, London W12 0NN, UK
| |
Collapse
|
179
|
Mohan R, Mao Y, Zhang S, Zhang YW, Xu CR, Gradwohl G, Tang X. Differentially Expressed MicroRNA-483 Confers Distinct Functions in Pancreatic β- and α-Cells. J Biol Chem 2015; 290:19955-66. [PMID: 26109062 DOI: 10.1074/jbc.m115.650705] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Indexed: 01/08/2023] Open
Abstract
Insulin secreted from pancreatic β-cells and glucagon secreted from pancreatic α-cells are the two major hormones working in the pancreas in an opposing manner to regulate and maintain a normal glucose homeostasis. How microRNAs (miRNAs), a population of non-coding RNAs so far demonstrated to be differentially expressed in various types of cells, regulate gene expression in pancreatic β-cells and its closely associated α-cells is not completely clear. In this study, miRNA profiling was performed and compared between pancreatic β-cells and their partner α-cells. One novel miRNA, miR-483, was identified for its highly differential expression in pancreatic β-cells when compared to its expression in α-cells. Overexpression of miR-483 in β-cells increased insulin transcription and secretion by targeting SOCS3, a member of suppressor of cytokine signaling family. In contrast, overexpression of miR-483 decreased glucagon transcription and secretion in α-cells. Moreover, overexpressed miR-483 protected against proinflammatory cytokine-induced apoptosis in β-cells. This correlates with a higher expression level of miR-483 and the expanded β-cell mass observed in the islets of prediabetic db/db mice. Together, our data suggest that miR-483 has opposite effects in α- and β-cells by targeting SOCS3, and the imbalance of miR-483 and its targets may play a crucial role in diabetes pathogenesis.
Collapse
Affiliation(s)
- Ramkumar Mohan
- From the Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931
| | - Yiping Mao
- From the Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931
| | - Shungang Zhang
- From the Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931
| | - Yu-Wei Zhang
- the College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China, and
| | - Cheng-Ran Xu
- the College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China, and
| | - Gérard Gradwohl
- the Institute of Genetics and Molecular and Cellular Biology, Department of Development and Stem cells, University of Strasbourg, 67404 Illkirch, France
| | - Xiaoqing Tang
- From the Department of Biological Sciences, Michigan Technological University, Houghton, Michigan 49931,
| |
Collapse
|
180
|
Dhawan S, Tschen SI, Zeng C, Guo T, Hebrok M, Matveyenko A, Bhushan A. DNA methylation directs functional maturation of pancreatic β cells. J Clin Invest 2015; 125:2851-60. [PMID: 26098213 DOI: 10.1172/jci79956] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 04/23/2015] [Indexed: 12/24/2022] Open
Abstract
Pancreatic β cells secrete insulin in response to postprandial increases in glucose levels to prevent hyperglycemia and inhibit insulin secretion under fasting conditions to protect against hypoglycemia. β cells lack this functional capability at birth and acquire glucose-stimulated insulin secretion (GSIS) during neonatal life. Here, we have shown that during postnatal life, the de novo DNA methyltransferase DNMT3A initiates a metabolic program by repressing key genes, thereby enabling the coupling of insulin secretion to glucose levels. In a murine model, β cell-specific deletion of Dnmt3a prevented the metabolic switch, resulting in loss of GSIS. DNMT3A bound to the promoters of the genes encoding hexokinase 1 (HK1) and lactate dehydrogenase A (LDHA) - both of which regulate the metabolic switch - and knockdown of these two key DNMT3A targets restored the GSIS response in islets from animals with β cell-specific Dnmt3a deletion. Furthermore, DNA methylation-mediated repression of glucose-secretion decoupling genes to modulate GSIS was conserved in human β cells. Together, our results reveal a role for DNA methylation to direct the acquisition of pancreatic β cell function.
Collapse
|
181
|
Urocortin3 mediates somatostatin-dependent negative feedback control of insulin secretion. Nat Med 2015; 21:769-76. [PMID: 26076035 PMCID: PMC4496282 DOI: 10.1038/nm.3872] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/30/2015] [Indexed: 12/15/2022]
Abstract
The peptide hormone urocortin3 (Ucn3) is abundantly expressed by mature beta cells, yet its physiological role is unknown. Here we demonstrate that Ucn3 is stored and co-released with insulin and potentiates glucose-stimulated somatostatin secretion via cognate receptors on delta cells. Further, we found that islets lacking endogenous Ucn3 have fewer delta cells, reduced somatostatin content, impaired somatostatin secretion, and exaggerated insulin release, and that these defects are rectified by treatment with synthetic Ucn3 in vitro. Our observations indicate that the paracrine actions of Ucn3 activate a negative feedback loop that promotes somatostatin release to ensure the timely reduction of insulin secretion upon normalization of plasma glucose. Moreover, Ucn3 is markedly depleted from beta cells in mouse and macaque models of diabetes and in human diabetic islets. This suggests that Ucn3 is a key contributor to stable glycemic control, whose reduction during diabetes aggravates glycemic volatility and contributes to the pathophysiology of this disease.
Collapse
|
182
|
Heinrich C, Spagnoli FM, Berninger B. In vivo reprogramming for tissue repair. Nat Cell Biol 2015; 17:204-11. [PMID: 25720960 DOI: 10.1038/ncb3108] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vital organs such as the pancreas and the brain lack the capacity for effective regeneration. To overcome this limitation, an emerging strategy consists of converting resident tissue-specific cells into the cell types that are lost due to disease by a process called in vivo lineage reprogramming. Here we discuss recent breakthroughs in regenerating pancreatic β-cells and neurons from various cell types, and highlight fundamental challenges that need to be overcome for the translation of in vivo lineage reprogramming into therapy.
Collapse
Affiliation(s)
- Christophe Heinrich
- INSERM U836, F-38000 Grenoble, France and Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000 Grenoble, France
| | | | - Benedikt Berninger
- Institute of Physiological Chemistry and the Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, D-55128 Mainz, Germany, and Physiological Genomics, Institute of Physiology, Ludwig Maximilians University Munich, D-80336 Munich, Germany
| |
Collapse
|
183
|
Nair G, Hebrok M. Islet formation in mice and men: lessons for the generation of functional insulin-producing β-cells from human pluripotent stem cells. Curr Opin Genet Dev 2015; 32:171-80. [PMID: 25909383 DOI: 10.1016/j.gde.2015.03.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 02/24/2015] [Accepted: 03/11/2015] [Indexed: 12/23/2022]
Abstract
The Islets of Langerhans are crucial 'micro-organs' embedded in the glandular exocrine pancreas that regulate nutrient metabolism. They not only synthesize, but also secrete endocrine hormones in a modulated fashion in response to physiologic metabolic demand. These highly sophisticated structures with intricate organization of multiple cell types, namely endocrine, vascular, neuronal and mesenchymal cells, have evolved to perform this task to perfection over time. Not surprisingly, islet architecture and function are dissimilar between humans and typically studied model organisms, such as rodents and zebrafish. Further, recent findings also suggest noteworthy differences in human islet development from that in mouse, including delayed appearance and gradual resolution of key differentiation markers, a single-phase of endocrine differentiation, and prenatal association of developing islets with neurovascular milieu. In light of these findings, it is imperative that a systematic study is undertaken to compare islet development between human and mouse. Illuminating inter-species differences in islet development will likely be critical in furthering our pursuit to generate an unlimited supply of truly functional and fully mature β-cells from human pluripotent stem cell (hPSC) sources for therapeutic purposes.
Collapse
Affiliation(s)
- Gopika Nair
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
184
|
Dadi PK, Luo B, Vierra NC, Jacobson DA. TASK-1 Potassium Channels Limit Pancreatic α-Cell Calcium Influx and Glucagon Secretion. Mol Endocrinol 2015; 29:777-87. [PMID: 25849724 DOI: 10.1210/me.2014-1321] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glucose regulation of pancreatic α-cell Ca(2+) entry through voltage-dependent Ca(2+) channels is essential for normal glucagon secretion and becomes defective during the pathogenesis of diabetes mellitus. The 2-pore domain K(+) channel, TWIK-related acid-sensitive K(+) channel 1 (TASK-1), is an important modulator of membrane voltage and Ca(2+) entry. However, its role in α-cells has not been determined. Therefore, we addressed how TASK-1 channels regulate α-cell electrical activity, Ca(2+) entry, and glucagon secretion. We find that TASK-1 channels expressed in human and rodent α-cells are blocked by the TASK-1 channel inhibitor A1899. Alpha-cell 2-pore domain K(+) currents were also significantly reduced after ablation of mouse α-cell TASK-1 channels. Inhibition of TASK-1 channels with A1899 caused plasma membrane potential depolarization in both human and mouse α-cells, which resulted in increased electrical excitability. Moreover, ablation of α-cell TASK-1 channels increased α-cell electrical excitability under elevated glucose (11 mM) conditions compared with control α-cells. This resulted in significantly elevated α-cell Ca(2+) influx when TASK-1 channels were inhibited in the presence of high glucose (14 mM). However, there was an insignificant change in α-cell Ca(2+) influx after TASK-1 inhibition in low glucose (1 mM). Glucagon secretion from mouse and human islets was also elevated specifically in high (11 mM) glucose after acute TASK-1 inhibition. Interestingly, mice deficient for α-cell TASK-1 showed improvements in both glucose inhibition of glucagon secretion and glucose tolerance, which resulted from the chronic loss of α-cell TASK-1 currents. Therefore, these data suggest an important role for TASK-1 channels in limiting α-cell excitability and glucagon secretion during glucose stimulation.
Collapse
Affiliation(s)
- Prasanna K Dadi
- Department of Molecular Physiology and Biophysics (P.K.D., N.C.V., D.A.J.), Vanderbilt University, Nashville, Tennessee 37232-0615; and University of Oklahoma College of Medicine (B.L.), Oklahoma City, Oklahoma 73104-5042
| | | | | | | |
Collapse
|
185
|
Nostro MC, Sarangi F, Yang C, Holland A, Elefanty AG, Stanley EG, Greiner DL, Keller G. Efficient generation of NKX6-1+ pancreatic progenitors from multiple human pluripotent stem cell lines. Stem Cell Reports 2015; 4:591-604. [PMID: 25843049 PMCID: PMC4400642 DOI: 10.1016/j.stemcr.2015.02.017] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 12/18/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) represent a renewable source of pancreatic beta cells for both basic research and therapeutic applications. Given this outstanding potential, significant efforts have been made to identify the signaling pathways that regulate pancreatic development in hPSC differentiation cultures. In this study, we demonstrate that the combination of epidermal growth factor (EGF) and nicotinamide signaling induces the generation of NKX6-1+ progenitors from all hPSC lines tested. Furthermore, we show that the size of the NKX6-1+ population is regulated by the duration of treatment with retinoic acid, fibroblast growth factor 10 (FGF10), and inhibitors of bone morphogenetic protein (BMP) and hedgehog signaling pathways. When transplanted into NOD scid gamma (NSG) recipients, these progenitors differentiate to give rise to exocrine and endocrine cells, including monohormonal insulin+ cells. Together, these findings provide an efficient and reproducible strategy for generating highly enriched populations of hPSC-derived beta cell progenitors for studies aimed at further characterizing their developmental potential in vivo and deciphering the pathways that regulate their maturation in vitro. EGF and nicotinamide induce NKX6-1+ progenitors from hPSC-derived endoderm NKX6-1+ progenitor generation can be controlled by the duration of stage 3 treatment The generation of polyhormonal cells is dependent on hedgehog signaling inhibition NKX6-1+ progenitors give rise to ductal, acinar, and endocrine cells in vivo
Collapse
Affiliation(s)
- M Cristina Nostro
- McEwen Centre for Regenerative Medicine, Toronto, ON M5G 1L7, Canada; Toronto General Research Institute, Department of Experimental Therapeutics, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Farida Sarangi
- McEwen Centre for Regenerative Medicine, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Chaoxing Yang
- Department of Molecular Medicine and Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Andrew Holland
- Department of Anatomy and Cell Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Andrew G Elefanty
- Department of Anatomy and Cell Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia; Murdoch Childrens Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, VIC 3052, Australia
| | - Edouard G Stanley
- Department of Anatomy and Cell Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia; Murdoch Childrens Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, VIC 3052, Australia
| | - Dale L Greiner
- Department of Molecular Medicine and Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Gordon Keller
- McEwen Centre for Regenerative Medicine, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
186
|
Sun G, da Silva Xavier G, Gorman T, Priest C, Solomou A, Hodson DJ, Foretz M, Viollet B, Herrera PL, Parker H, Reimann F, Gribble FM, Migrenne S, Magnan C, Marley A, Rutter GA. LKB1 and AMPKα1 are required in pancreatic alpha cells for the normal regulation of glucagon secretion and responses to hypoglycemia. Mol Metab 2015; 4:277-86. [PMID: 25830091 PMCID: PMC4354920 DOI: 10.1016/j.molmet.2015.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 01/14/2015] [Accepted: 01/17/2015] [Indexed: 10/24/2022] Open
Abstract
AIMS/HYPOTHESIS Glucagon release from pancreatic alpha cells is required for normal glucose homoeostasis and is dysregulated in both Type 1 and Type 2 diabetes. The tumour suppressor LKB1 (STK11) and the downstream kinase AMP-activated protein kinase (AMPK), modulate cellular metabolism and growth, and AMPK is an important target of the anti-hyperglycaemic agent metformin. While LKB1 and AMPK have emerged recently as regulators of beta cell mass and insulin secretion, the role of these enzymes in the control of glucagon production in vivo is unclear. METHODS Here, we ablated LKB1 (αLKB1KO), or the catalytic alpha subunits of AMPK (αAMPKdKO, -α1KO, -α2KO), selectively in ∼45% of alpha cells in mice by deleting the corresponding flox'd alleles with a preproglucagon promoter (PPG) Cre. RESULTS Blood glucose levels in male αLKB1KO mice were lower during intraperitoneal glucose, aminoimidazole carboxamide ribonucleotide (AICAR) or arginine tolerance tests, and glucose infusion rates were increased in hypoglycemic clamps (p < 0.01). αLKB1KO mice also displayed impaired hypoglycemia-induced glucagon release. Glucose infusion rates were also elevated (p < 0.001) in αAMPKα1 null mice, and hypoglycemia-induced plasma glucagon increases tended to be lower (p = 0.06). Glucagon secretion from isolated islets was sensitized to the inhibitory action of glucose in αLKB1KO, αAMPKdKO, and -α1KO, but not -α2KO islets. CONCLUSIONS/INTERPRETATION An LKB1-dependent signalling cassette, involving but not restricted to AMPKα1, is required in pancreatic alpha cells for the control of glucagon release by glucose.
Collapse
Affiliation(s)
- Gao Sun
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, UK
| | - Gabriela da Silva Xavier
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, UK
| | | | | | - Antonia Solomou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, UK
| | - David J. Hodson
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, UK
| | - Marc Foretz
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris cité, Paris, France
| | - Benoit Viollet
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris cité, Paris, France
| | - Pedro-Luis Herrera
- Department of Genetic Medicine & Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Helen Parker
- Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, UK
| | - Frank Reimann
- Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, UK
| | - Fiona M. Gribble
- Cambridge Institute for Medical Research, Addenbrooke's Hospital, Cambridge, UK
| | - Stephanie Migrenne
- University Paris Diderot-Paris 7-Unit of Functional and Adaptive Biology (BFA) EAC 7059C NRS, France
| | - Christophe Magnan
- University Paris Diderot-Paris 7-Unit of Functional and Adaptive Biology (BFA) EAC 7059C NRS, France
| | | | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
187
|
van der Meulen T, Huising MO. Role of transcription factors in the transdifferentiation of pancreatic islet cells. J Mol Endocrinol 2015; 54:R103-17. [PMID: 25791577 PMCID: PMC4373662 DOI: 10.1530/jme-14-0290] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The α and β cells act in concert to maintain blood glucose. The α cells release glucagon in response to low levels of glucose to stimulate glycogenolysis in the liver. In contrast, β cells release insulin in response to elevated levels of glucose to stimulate peripheral glucose disposal. Despite these opposing roles in glucose homeostasis, α and β cells are derived from a common progenitor and share many proteins important for glucose sensing and hormone secretion. Results from recent work have underlined these similarities between the two cell types by revealing that β-to-α as well as α-to-β transdifferentiation can take place under certain experimental circumstances. These exciting findings highlight unexpected plasticity of adult islets and offer hope of novel therapeutic paths to replenish β cells in diabetes. In this review, we focus on the transcription factor networks that establish and maintain pancreatic endocrine cell identity and how they may be perturbed to facilitate transdifferentiation.
Collapse
Affiliation(s)
- Talitha van der Meulen
- Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA
| | - Mark O Huising
- Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA
| |
Collapse
|
188
|
Thorens B, Tarussio D, Maestro MA, Rovira M, Heikkilä E, Ferrer J. Ins1(Cre) knock-in mice for beta cell-specific gene recombination. Diabetologia 2015; 58:558-65. [PMID: 25500700 PMCID: PMC4320308 DOI: 10.1007/s00125-014-3468-5] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/14/2014] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS Pancreatic beta cells play a central role in the control of glucose homeostasis by secreting insulin to stimulate glucose uptake by peripheral tissues. Understanding the molecular mechanisms that control beta cell function and plasticity has critical implications for the pathophysiology and therapy of major forms of diabetes. Selective gene inactivation in pancreatic beta cells, using the Cre-lox system, is a powerful approach to assess the role of particular genes in beta cells and their impact on whole body glucose homeostasis. Several Cre recombinase (Cre) deleter mice have been established to allow inactivation of genes in beta cells, but many show non-specific recombination in other cell types, often in the brain. METHODS We describe the generation of Ins1(Cre) and Ins1(CreERT2) mice in which the Cre or Cre-oestrogen receptor fusion protein (CreERT2) recombinases have been introduced at the initiation codon of the Ins1 gene. RESULTS We show that Ins1(Cre) mice induce efficient and selective recombination of floxed genes in beta cells from the time of birth, with no recombination in the central nervous system. These mice have normal body weight and glucose homeostasis. Furthermore, we show that tamoxifen treatment of adult Ins1(CreERT2) mice crossed with Rosa26-tdTomato mice induces efficient recombination in beta cells. CONCLUSIONS/INTERPRETATION These two strains of deleter mice are useful new resources to investigate the molecular physiology of pancreatic beta cells.
Collapse
Affiliation(s)
- Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Genopode Building, CH-1015 Lausanne, Switzerland
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, Genopode Building, CH-1015 Lausanne, Switzerland
| | - Miguel Angel Maestro
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain, http://www.ciberdem.org/
| | - Meritxell Rovira
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain, http://www.ciberdem.org/
| | - Eija Heikkilä
- Center for Integrative Genomics, University of Lausanne, Genopode Building, CH-1015 Lausanne, Switzerland
| | - Jorge Ferrer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain, http://www.ciberdem.org/
- Department of Medicine, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, W12 0NN UK
| |
Collapse
|
189
|
Tetteh PW, Farin HF, Clevers H. Plasticity within stem cell hierarchies in mammalian epithelia. Trends Cell Biol 2015; 25:100-8. [DOI: 10.1016/j.tcb.2014.09.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/08/2014] [Accepted: 09/12/2014] [Indexed: 12/20/2022]
|
190
|
Sangan CB, Jover R, Heimberg H, Tosh D. In vitro reprogramming of pancreatic alpha cells towards a beta cell phenotype following ectopic HNF4α expression. Mol Cell Endocrinol 2015; 399:50-9. [PMID: 25224487 DOI: 10.1016/j.mce.2014.09.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 08/21/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022]
Abstract
There is currently a shortage of organ donors available for pancreatic beta cell transplantation into diabetic patients. An alternative source of beta cells is pre-existing pancreatic cells. While we know that beta cells can arise directly from alpha cells during pancreatic regeneration we do not understand the molecular basis for the switch in phenotype. The aim of the present study was to investigate if hepatocyte nuclear factor 4 alpha (HNF4α), a transcription factor essential for a normal beta cell phenotype, could induce the reprogramming of alpha cells towards potential beta cells. We utilised an in vitro model of pancreatic alpha cells, the murine αTC1-9 cell line. We initially characterised the αTC1-9 cell line before and following adenovirus-mediated ectopic expression of HNF4α. We analysed the phenotype at transcript and protein level and assessed its glucose-responsiveness. Ectopic HNF4α expression in the αTC1-9 cell line induced a change in morphology (1.7-fold increase in size), suppressed glucagon expression, induced key beta cell-specific markers (insulin, C-peptide, glucokinase, GLUT2 and Pax4) and pancreatic polypeptide (PP) and enabled the cells to secrete insulin in a glucose-regulated manner. In conclusion, HNF4α reprograms alpha cells to beta-like cells.
Collapse
Affiliation(s)
| | - Ramiro Jover
- Experimental Hepatology Unit. Hosp. La Fe & Dep. Biochemistry, University of Valencia. CIBERehd, Spain
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - David Tosh
- Centre for Regenerative Medicine, University of Bath, Bath, UK.
| |
Collapse
|
191
|
Li F, Su Y, Cheng Y, Jiang X, Peng Y, Li Y, Lu J, Gu Y, Zhang C, Cao Y, Wang W, Ning G. Conditional deletion of Men1 in the pancreatic β-cell leads to glucagon-expressing tumor development. Endocrinology 2015; 156:48-57. [PMID: 25343275 DOI: 10.1210/en.2014-1433] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The tumor suppressor menin is recognized as a key regulator of β-cell proliferation. To induce tumorigenesis within the pancreatic β-cells, floxed alleles of Men1 were selectively ablated using Cre-recombinase driven by the insulin promoter. Despite the β-cell specificity of the RipCre, glucagon-expressing tumors as well as insulinomas developed in old mutant mice. These glucagon-expressing tumor cells were menin deficient and expressed the mature α-cell-specific transcription factors Brain-specific homeobox POU domain protein 4 (Brn4) and v-maf musculoaponeurotic fibrosarcoma oncogene family, protein B (MafB). Moreover, the inactivation of β-cell-specific transcription factors was observed in mutant β-cells. Our work shows that Men1 ablation in the pancreatic β-cells leads to the inactivation of specific transcription factors, resulting in glucagon-expressing tumor development, which sheds light on the mechanisms of islet tumorigenesis.
Collapse
Affiliation(s)
- Feng Li
- Department of Endocrinology and Metabolism (F.L., Y.S., Y.Ch., X.J., Y.P., Y.L., J.L., Y.G., Y.Ca., W.W., G.N.), Shanghai Clinical Center for Endocrine and Metabolic Diseases and Shanghai Institute of Endocrinology and Metabolism, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, and Laboratory of Endocrinology and Metabolism (G.N.), Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and Laboratoire Génétique Moléculaire, Signalisation et Cancer (C.Z.), Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5201, Faculté de Médecine, Université Claude Bernard Lyon, Centre Leon-Berard, Lyon69366, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Gong GC, Fan WZ, Li DZ, Tian X, Chen SJ, Fu YC, Xu WC, Wei CJ. Increased Specific Labeling of INS-1 Pancreatic Beta-Cell by Using RIP-Driven Cre Mutants with Reduced Activity. PLoS One 2015; 10:e0129092. [PMID: 26046525 PMCID: PMC4457865 DOI: 10.1371/journal.pone.0129092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/06/2015] [Indexed: 02/05/2023] Open
Abstract
Ectopically expressed Cre recombinase in extrapancreatic tissues in RIP-Cre mice has been well documented. The objective of this study was to find a simple solution that allows for improved beta-cell specific targeting. To this end, the RIP-Cre and reporter CMV-loxP-DsRed-loxP-EGFP expression cassettes were configurated into a one-plasmid and two-plasmid systems, which labeled approximately 80% insulin-positive INS-1 cells after 48 h transfection. However, off-target labeling was robustly found in more than 15% insulin-negative Ad293 cells. When an IRES element was inserted in front of Cre to reduce the translation efficiency, the ratio of recombination between INS-1 and Ad293 cells increased 3-4-fold. Further, a series of Cre mutants were generated by site-directed mutagenesis. When one of the mutants, Cre(H289P) in both configurations, was used in the experiment, the percentage of recombination dropped to background levels in a number of insulin-negative cell lines, but decreased only slightly in INS-1 cells. Consistently, DNA substrate digestion assay showed that the enzymatic activity of Cre(H289P) was reduced by 30-fold as compared to that of wild-type. In this study, we reported the generation of constructs containing RIP and Cre mutants, which enabled enhanced beta-cell specific labeling in vitro. These tools could be invaluable for beta-cell targeting and to the study of islet development.
Collapse
Affiliation(s)
- Gen-cheng Gong
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Wen-zhu Fan
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Di-zheng Li
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Xiong Tian
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Shao-jun Chen
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
| | - Yu-cai Fu
- Laboratory of Cell Senescence, Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Wen-can Xu
- Department of Endocrinology, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Chi-ju Wei
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong, 515063, China
- * E-mail:
| |
Collapse
|
193
|
Wang L, Luk CT, Cai EP, Schroer SA, Allister EM, Shi SY, Wheeler MB, Gaisano HY, Woo M. PTEN deletion in pancreatic α-cells protects against high-fat diet-induced hyperglucagonemia and insulin resistance. Diabetes 2015; 64:147-57. [PMID: 25092678 DOI: 10.2337/db13-1715] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
An aberrant increase in circulating catabolic hormone glucagon contributes to type 2 diabetes pathogenesis. However, mechanisms regulating glucagon secretion and α-cell mass are not well understood. In this study, we aimed to demonstrate that phosphatidylinositol 3-kinase (PI3K) signaling is an important regulator of α-cell function. Mice with deletion of PTEN, a negative regulator of this pathway, in α-cells show reduced circulating glucagon levels and attenuated l-arginine-stimulated glucagon secretion both in vivo and in vitro. This hypoglucagonemic state is maintained after high-fat-diet feeding, leading to reduced expression of hepatic glycogenolytic and gluconeogenic genes. These beneficial effects protected high-fat diet-fed mice against hyperglycemia and insulin resistance. The data demonstrate an inhibitory role of PI3K signaling on α-cell function and provide experimental evidence for enhancing α-cell PI3K signaling for diabetes treatment.
Collapse
Affiliation(s)
- Linyuan Wang
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Cynthia T Luk
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Erica P Cai
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Stephanie A Schroer
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Emma M Allister
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Sally Y Shi
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Michael B Wheeler
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Herbert Y Gaisano
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Minna Woo
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada Institute of Medical Science, University of Toronto, Toronto, ON, Canada Division of Endocrinology & Metabolism, Department of Medicine, University Health Network, Toronto, ON, Canada
| |
Collapse
|
194
|
Riley KG, Gannon M. Pancreas Development and Regeneration. PRINCIPLES OF DEVELOPMENTAL GENETICS 2015:565-590. [DOI: 10.1016/b978-0-12-405945-0.00031-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
195
|
Teitelman G, Kedees M. Mouse insulin cells expressing an inducible RIPCre transgene are functionally impaired. J Biol Chem 2014; 290:3647-53. [PMID: 25533471 DOI: 10.1074/jbc.m114.615484] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
We used cre-lox technology to test whether the inducible expression of Cre minimize the deleterious effect of the enzyme on beta cell function. We studied mice in which Cre is linked to a modified estrogen receptor (ER), and its expression is controlled by the rat insulin promoter (RIP). Following the injection of tamoxifen (TM), CreER- migrates to the nucleus and promotes the appearance of a reporter protein, enhanced yellow fluorescent protein (EYFP), in cells. Immunocytochemical analysis indicated that 46.6 ± 2.1% insulin cells of adult RIPCreER- EYFP expressed EYFP. RIPCreER-EYFP (+TM) mice were normoglycemic throughout the study, and their glucose tolerance test results were similar to control CD-1 mice. However, an extended exposure to reagents that stimulate insulin synthesis was detrimental to the survival of IN+EYFP+cells. The administration of an inhibitor of the enzyme dipeptidyl-peptidase (DPP4i), which prevents the cleavage of glucagon-like peptide (GLP-1), to adult RIPCreER-EYFP mice lead to a decrease in the percentage of IN+EYFP+ to 17.5 ± 1.73 and a significant increase in apoptotic cells in islets. Similarly, a 2-week administration of the GLP-1 analog exendin 4 (ex-4) induced an almost complete ablation of IN+ expressing a different reporter protein and a significant decrease in the beta cell mass and rate of beta cell proliferation. Since normal beta cells do not die when induced to increase insulin synthesis, our observations indicate that insulin cells expressing an inducible RIPCre transgene are functionally deficient. Studies employing these mice should carefully consider the pitfalls of the Cre-Lox technique.
Collapse
Affiliation(s)
- Gladys Teitelman
- From the Department of Cell Biology, SUNY-Downstate Medical Center, Brooklyn, New York 11203
| | - Mamdouh Kedees
- From the Department of Cell Biology, SUNY-Downstate Medical Center, Brooklyn, New York 11203
| |
Collapse
|
196
|
Targeting β-catenin signaling for therapeutic intervention in MEN1-deficient pancreatic neuroendocrine tumours. Nat Commun 2014; 5:5809. [PMID: 25517963 PMCID: PMC4284642 DOI: 10.1038/ncomms6809] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 11/10/2014] [Indexed: 02/08/2023] Open
Abstract
Inactivating MEN1 mutations are the most common genetic defects present in sporadic and inherited pancreatic neuroendocrine tumours (PNETs). The lack of interventional therapies prompts us to explore the therapeutic approach of targeting β-catenin signalling in MEN1-mutant PNETs. Here we show the MEN1-encoded scaffold protein menin regulates phosphorylation of β-catenin. β-catenin signalling is activated in MEN1-mutant human and mouse PNETs. Conditional knockout of β-catenin suppresses the tumorigenesis and growth of Men1-deficient PNETs, and significantly prolongs the survival time in mice. Suppression of β-catenin signalling by genetic ablation or a molecular antagonist inhibits the expression of proproliferative genes in menin-null PNETs and potently improves hyperinsulinemia and hypoglycemia in mice. Blockade of β-catenin has no adverse effect on physiological function of pancreatic β-cells. Our data demonstrate that β-catenin signalling is an effective therapeutic target for MEN1-mutant PNETs. Our findings may contribute to individualized and combined medication treatment for PNETs. MEN1 gene encodes menin, a nuclear scaffold protein that regulates transcription and is often inactivated in pancreatic neuroendocrine tumours (PNETs). Here Jiang et al. show that MEN1-driven PNET development involves activation of β-catenin, and that β-catenin deletion ameliorates the disease.
Collapse
|
197
|
De Angelis MT, Russo F, D'Angelo F, Federico A, Gemei M, Del Vecchio L, Ceccarelli M, De Felice M, Falco G. Novel pancreas organogenesis markers refine the pancreatic differentiation roadmap of embryonic stem cells. Stem Cell Rev Rep 2014; 10:269-79. [PMID: 24390927 DOI: 10.1007/s12015-013-9489-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The generation of pancreatic endocrine and exocrine functional precursors from embryonic stem cells (ESCs) is an intriguing opportunity to address cell therapy challenges. The main goal of cellular regeneration is to derive, in vitro, pancreatic progenitor cells (PPCs) that retain the capacity to differentiate following the in vivo developmental ontogeny. In our work, we aim to refine the pancreatic in vitro cellular transitions, through the identification of the intrinsic factors that mark the pancreas budding process at embryonic stage 10.5 (E10.5), in which pancreas precursor specification predominantly occurs. We identified a cohort of genes (Bex1, Nepn, Pcbd1, Prdxdd1, Rnf160, Slc2a1, and Tff3) that marked the pancreas budding genesis, and above all signaled ESC differentiation transitions during pancreatic lineage commitment. Noticeably, we demonstrated that the expression of Nepn marked a naïve pancreatic cellular state that resembled PPC-like specification. Our data considerably improve the comprehension of pancreatic cellular ontogeny, which could be critical for implementing pluripotent stem cells programming and reprogramming toward pancreatic lineage commitment.
Collapse
|
198
|
Brouwers B, de Faudeur G, Osipovich AB, Goyvaerts L, Lemaire K, Boesmans L, Cauwelier EJG, Granvik M, Pruniau VPEG, Van Lommel L, Van Schoors J, Stancill JS, Smolders I, Goffin V, Binart N, in't Veld P, Declercq J, Magnuson MA, Creemers JWM, Schuit F, Schraenen A. Impaired islet function in commonly used transgenic mouse lines due to human growth hormone minigene expression. Cell Metab 2014; 20:979-90. [PMID: 25470546 PMCID: PMC5674787 DOI: 10.1016/j.cmet.2014.11.004] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/29/2014] [Accepted: 11/04/2014] [Indexed: 11/15/2022]
Abstract
The human growth hormone (hGH) minigene is frequently used in the derivation of transgenic mouse lines to enhance transgene expression. Although this minigene is present in the transgenes as a secondcistron, and thus not thought to be expressed, we found that three commonly used lines, Pdx1-Cre(Late), RIP-Cre, and MIP-GFP, each expressed significant amounts of hGH in pancreatic islets. Locally secreted hGH binds to prolactin receptors on β cells, activates STAT5 signaling, and induces pregnancy-like changes in gene expression, thereby augmenting pancreatic β cell mass and insulin content. In addition, islets of Pdx1-Cre(Late) mice have lower GLUT2 expression and reduced glucose-induced insulin release and are protected against the β cell toxin streptozotocin. These findings may be important when interpreting results obtained when these and other hGH minigene-containing transgenic mice are used.
Collapse
Affiliation(s)
- Bas Brouwers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Geoffroy de Faudeur
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Anna B Osipovich
- Center for Stem Cell Biology and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lotte Goyvaerts
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Katleen Lemaire
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Leen Boesmans
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Elisa J G Cauwelier
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Mikaela Granvik
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Vincent P E G Pruniau
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Leentje Van Lommel
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Jolien Van Schoors
- Center for Neurosciences, Department of Pharmaceutical Chemistry and Drug Analysis, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Jennifer S Stancill
- Center for Stem Cell Biology and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Ilse Smolders
- Center for Neurosciences, Department of Pharmaceutical Chemistry and Drug Analysis, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Vincent Goffin
- INSERM U845, Research Center Growth and Signaling, PRL/GH Pathophysiology Laboratory, Faculty of Medicine, University Paris Descartes, Sorbonne Paris Cité, Paris 75993, France
| | - Nadine Binart
- INSERM U693, Faculté de Médecine Paris-Sud, University Paris-Sud, Le Kremlin-Bicêtre 94276, France
| | - Peter in't Veld
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Jeroen Declercq
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Mark A Magnuson
- Center for Stem Cell Biology and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - John W M Creemers
- Laboratory for Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium.
| | - Frans Schuit
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium.
| | - Anica Schraenen
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
199
|
Tan G, Elefanty AG, Stanley EG. β-cell regeneration and differentiation: how close are we to the 'holy grail'? J Mol Endocrinol 2014; 53:R119-29. [PMID: 25385843 DOI: 10.1530/jme-14-0188] [Citation(s) in RCA: 8] [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: 12/12/2022]
Abstract
Diabetes can be managed by careful monitoring of blood glucose and timely delivery of exogenous insulin. However, even with fastidious compliance, people with diabetes can suffer from numerous complications including atherosclerosis, retinopathy, neuropathy, and kidney disease. This is because delivery of exogenous insulin coupled with glucose monitoring cannot provide the fine level of glucose control normally provided by endogenous β-cells in the context of intact islets. Moreover, a subset of people with diabetes lack awareness of hypoglycemic events; a status that can have grave consequences. Therefore, much effort has been focused on replacing lost or dysfunctional β-cells with cells derived from other sources. The advent of stem cell biology and cellular reprogramming strategies have provided impetus to this work and raised hopes that a β-cell replacement therapy is on the horizon. In this review, we look at two components that will be required for successful β-cell replacement therapy: a reliable and safe source of β-cells and a mechanism by which such cells can be delivered and protected from host immune destruction. Particular attention is paid to insulin-producing cells derived from pluripotent stem cells because this platform addresses the issue of scale, one of the more significant hurdles associated with potential cell-based therapies. We also review methods for encapsulating transplanted cells, a technique that allows grafts to evade immune attack and survive for a long term in the absence of ongoing immunosuppression. In surveying the literature, we conclude that there are still several substantial hurdles that need to be cleared before a stem cell-based β-cell replacement therapy for diabetes becomes a reality.
Collapse
Affiliation(s)
- Gemma Tan
- Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia
| | - Andrew G Elefanty
- Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia
| | - Edouard G Stanley
- Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia
| |
Collapse
|
200
|
Vázquez P, Robles AM, de Pablo F, Hernández-Sánchez C. Non-neural tyrosine hydroxylase, via modulation of endocrine pancreatic precursors, is required for normal development of beta cells in the mouse pancreas. Diabetologia 2014; 57:2339-47. [PMID: 25082160 PMCID: PMC4181516 DOI: 10.1007/s00125-014-3341-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 07/01/2014] [Indexed: 11/07/2022]
Abstract
AIMS/HYPOTHESIS Apart from transcription factors, little is known about the molecules that modulate the proliferation and differentiation of pancreatic endocrine cells. The early expression of tyrosine hydroxylase (TH) in a subset of glucagon(+) cells led us to investigate whether catecholamines have a role in beta cell development. METHODS We studied the immunohistochemical characteristics of TH-expressing cells in wild-type (Th (+/+) ) mice during early pancreas development, and analysed the endocrine pancreas phenotype of TH-deficient (Th (-/-) ) mice. We also studied the effect of dopamine addition and TH-inhibition on insulin-producing cells in explant cultures. RESULTS In the mouse pancreas at embryonic day (E)12.5-E13.5, the ∼10% of early glucagon(+) cells that co-expressed TH rarely proliferated and did not express the precursor marker neurogenin 3 at E13.5. The number of insulin(+) cells in the Th (-/-) embryonic pancreas was decreased as compared with wild-type embryos at E13.5. While no changes in pancreatic and duodenal homeobox 1 (PDX1)(+)-progenitor cell number were observed between groups at E12.5, the number of neurogenin 3 and NK2 homeobox 2 (NKX2.2)-expressing cells was reduced in Th (-/-) embryonic pancreas, an effect that occurred in parallel with increased expression of the transcriptional repressor Hes1. The potential role of dopamine as a pro-beta cell stimulus was tested by treating pancreas explants with this catecholamine, which resulted in an increase in total insulin content and insulin(+) cells relative to control explants. CONCLUSIONS/INTERPRETATION A non-neural catecholaminergic pathway appears to modulate the pancreatic endocrine precursor and insulin producing cell neogenesis. This finding may have important implications for approaches seeking to promote the generation of beta cells to treat diabetes.
Collapse
Affiliation(s)
- Patricia Vázquez
- 3D (Development, Differentiation, Degeneration) Lab, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (ISCIII), Ministerio de Economía y Competitividad, Spain, http://www.ciberdem.org/
| | - Ana M. Robles
- 3D (Development, Differentiation, Degeneration) Lab, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Flora de Pablo
- 3D (Development, Differentiation, Degeneration) Lab, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (ISCIII), Ministerio de Economía y Competitividad, Spain, http://www.ciberdem.org/
| | - Catalina Hernández-Sánchez
- 3D (Development, Differentiation, Degeneration) Lab, Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (ISCIII), Ministerio de Economía y Competitividad, Spain, http://www.ciberdem.org/
| |
Collapse
|