51
|
Balaji S, Zhou Y, Ganguly A, Opara EC, Soker S. The combined effect of PDX1, epidermal growth factor and poly-L-ornithine on human amnion epithelial cells' differentiation. BMC DEVELOPMENTAL BIOLOGY 2016; 16:8. [PMID: 27068127 PMCID: PMC4828805 DOI: 10.1186/s12861-016-0108-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/01/2016] [Indexed: 12/22/2022]
Abstract
Background It has been suggested that the ectopic expression of PDX1, a dominant pancreatic transcription factor, plays a critical role in the developmental programming of the pancreas even from cells of unrelated tissues such as keratinocytes and amniotic fluid stem cells. In this study we have chosen to drive pancreatic development in human amnion epithelial cells by inducing endogenous PDX1 expression. Further, we have investigated the role of Epidermal Growth Factor (EGF) and Poly-L-Ornithine (PLO) on this differentiation process. Results Human amnion epithelial cells expressed high levels of endogenous PDX1 upon transduction with an adenoviral vector expressing murine Pdx1. Other markers of various stages of pancreatic differentiation such as NKX6.1, SOX17, RFX6, FOXA2, CFTR, NEUROD1, PAX4 and PPY were also expressed upon Pdx1 transduction. Although initial expression of pancreatic progenitor markers was higher in culture conditions lacking EGF, for a sustained and increased expression EGF was required. Culture on PLO further increased the positive impact of EGF. Conclusion Pancreatic marker expression subsequent to mPdx1 transduction suggests that this approach may facilitate the in vitro differentiation of hAECs into cells of the endocrine pancreas. This result may have important implications in diabetes therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12861-016-0108-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shruti Balaji
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, 27101, USA.,Birla Institute of Technology & Science, Pilani K K Birla Goa campus, Zuari Nagar, 403726, Goa, India
| | - Yu Zhou
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, 27101, USA
| | - Anasuya Ganguly
- Birla Institute of Technology & Science, Pilani K K Birla Goa campus, Zuari Nagar, 403726, Goa, India.
| | - Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, 27101, USA.,Virginia Tech-Wake Forest University School of Biomedical Engineering & Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, 27101, USA.,Virginia Tech-Wake Forest University School of Biomedical Engineering & Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| |
Collapse
|
52
|
Xu J, Cui J, Del Campo A, Shin CH. Four and a Half LIM Domains 1b (Fhl1b) Is Essential for Regulating the Liver versus Pancreas Fate Decision and for β-Cell Regeneration. PLoS Genet 2016; 12:e1005831. [PMID: 26845333 PMCID: PMC4741517 DOI: 10.1371/journal.pgen.1005831] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022] Open
Abstract
The liver and pancreas originate from overlapping embryonic regions, and single-cell lineage tracing in zebrafish has shown that Bone morphogenetic protein 2b (Bmp2b) signaling is essential for determining the fate of bipotential hepatopancreatic progenitors towards the liver or pancreas. Despite its pivotal role, the gene regulatory networks functioning downstream of Bmp2b signaling in this process are poorly understood. We have identified four and a half LIM domains 1b (fhl1b), which is primarily expressed in the prospective liver anlage, as a novel target of Bmp2b signaling. fhl1b depletion compromised liver specification and enhanced induction of pancreatic cells from endodermal progenitors. Conversely, overexpression of fhl1b favored liver specification and inhibited induction of pancreatic cells. By single-cell lineage tracing, we showed that fhl1b depletion led lateral endodermal cells, destined to become liver cells, to become pancreatic cells. Reversely, when fhl1b was overexpressed, medially located endodermal cells, fated to differentiate into pancreatic and intestinal cells, contributed to the liver by directly or indirectly modulating the discrete levels of pdx1 expression in endodermal progenitors. Moreover, loss of fhl1b increased the regenerative capacity of β-cells by increasing pdx1 and neurod expression in the hepatopancreatic ductal system. Altogether, these data reveal novel and critical functions of Fhl1b in the hepatic versus pancreatic fate decision and in β-cell regeneration. Lineage-specific multipotent progenitors play crucial roles in embryonic development, regeneration in adult tissues, and diseases such as cancer. Bone morphogenetic protein (Bmp) signaling is critical for regulating the cell fate choice of liver versus pancreas, two essential organs of body metabolism. Through transcriptome profiling of endodermal tissues exposed to increased or decreased Bmp2b signaling, we have discovered the zebrafish gene four and a half LIM domains 1b (fhl1b) as a novel target of Bmp2b signaling. fhl1b is primarily expressed in the prospective liver anlage. Loss- and gain-of-function analyses indicate that Fhl1b suppresses specification of the pancreas and induces the liver. By single-cell lineage tracing, we showed that depletion of fhl1b caused a liver-to-pancreas fate switch, while fhl1b overexpression redirected pancreatic progenitors to become liver cells. At later stages, Fhl1b regulates regeneration of insulin-secreting β-cells by directly or indirectly modulating pdx1 and neurod expression in the hepatopancreatic ductal system. Therefore, our work provides a novel paradigm of how Bmp signaling regulates the hepatic versus pancreatic fate decision and β-cell regeneration through its novel target Fhl1b.
Collapse
Affiliation(s)
- Jin Xu
- School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Jiaxi Cui
- Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Chong Hyun Shin
- School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
53
|
Nivlet L, Herrmann J, Martin DE, Meunier A, Orvain C, Gradwohl G. Expression and functional studies of the GDNF family receptor alpha 3 in the pancreas. J Mol Endocrinol 2016; 56:77-90. [PMID: 26576643 PMCID: PMC5911917 DOI: 10.1530/jme-15-0213] [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] [Accepted: 11/17/2015] [Indexed: 01/11/2023]
Abstract
The generation of therapeutic β-cells from human pluripotent stem cells relies on the identification of growth factors that faithfully mimic pancreatic β-cell development in vitro. In this context, the aim of the study was to determine the expression and function of the glial cell line derived neurotrophic factor receptor alpha 3 (GFRα3) and its ligand artemin (Artn) in islet cell development and function. GFRα3 and Artn expression were characterized by in situ hybridization, immunochemistry, and qRT-PCR. We used GFRα3-deficient mice to study GFRα3 function and generated transgenic mice overexpressing Artn in the embryonic pancreas to study Artn function. We found that GFRα3 is expressed at the surface of a subset of Ngn3-positive endocrine progenitors as well as of embryonic α- and β-cells, while Artn is found in the pancreatic mesenchyme. Adult β-cells lack GFRα3 but α-cells express the receptor. GFRα3 was also found in parasympathetic and sympathetic intra-islet neurons as well as in glial cells in the embryonic and adult pancreas. The loss of GFRα3 or overexpression of Artn has no impact on Ngn3 and islet cell formation and maintenance in the embryo. Islet organization and innervation as well as glucose homeostasis is normal in GFRα3-deficient mice suggesting functional redundancy.
Collapse
Affiliation(s)
- Laure Nivlet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)Institut National de la Santé et de la Recherche Médicale (INSERM) U964, Centre National de Recherche Scientifique (CNRS) UMR 7104, Université de Strasbourg (UdS), 1 Rue Laurent Fries, 67404 Illkirch, France
| | - Joel Herrmann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)Institut National de la Santé et de la Recherche Médicale (INSERM) U964, Centre National de Recherche Scientifique (CNRS) UMR 7104, Université de Strasbourg (UdS), 1 Rue Laurent Fries, 67404 Illkirch, France
| | - Delia Esteban Martin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)Institut National de la Santé et de la Recherche Médicale (INSERM) U964, Centre National de Recherche Scientifique (CNRS) UMR 7104, Université de Strasbourg (UdS), 1 Rue Laurent Fries, 67404 Illkirch, France
| | - Aline Meunier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)Institut National de la Santé et de la Recherche Médicale (INSERM) U964, Centre National de Recherche Scientifique (CNRS) UMR 7104, Université de Strasbourg (UdS), 1 Rue Laurent Fries, 67404 Illkirch, France
| | - Christophe Orvain
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)Institut National de la Santé et de la Recherche Médicale (INSERM) U964, Centre National de Recherche Scientifique (CNRS) UMR 7104, Université de Strasbourg (UdS), 1 Rue Laurent Fries, 67404 Illkirch, France
| | - Gérard Gradwohl
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC)Institut National de la Santé et de la Recherche Médicale (INSERM) U964, Centre National de Recherche Scientifique (CNRS) UMR 7104, Université de Strasbourg (UdS), 1 Rue Laurent Fries, 67404 Illkirch, France
| |
Collapse
|
54
|
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
|
55
|
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
|
56
|
George NM, Boerner BP, Mir SUR, Guinn Z, Sarvetnick NE. Exploiting Expression of Hippo Effector, Yap, for Expansion of Functional Islet Mass. Mol Endocrinol 2015; 29:1594-607. [PMID: 26378466 DOI: 10.1210/me.2014-1375] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Loss of pancreas β-cell function is the precipitating factor in all forms of diabetes. Cell replacement therapies, such as islet transplantation, remain the best hope for a cure; however, widespread implementation of this method is hampered by availability of donor tissue. Thus, strategies that expand functional β-cell mass are crucial for widespread usage in diabetes cell replacement therapy. Here, we investigate the regulation of the Hippo-target protein, Yes-associated protein (Yap), during development of the endocrine pancreas and its function after reactivation in human cadaveric islets. Our results demonstrate that Yap expression is extinguished at the mRNA level after neurogenin-3-dependent specification of the pancreas endocrine lineage, correlating with proliferation decreases in these cells. Interestingly, when a constitutively active form of Yap was expressed in human cadaver islets robust increases in proliferation were noted within insulin-producing β-cells. Importantly, proliferation in these cells occurs without negatively affecting β-cell differentiation or functional status. Finally, we show that the proproliferative mammalian target of rapamycin pathway is activated after Yap expression, providing at least one explanation for the observed increases in β-cell proliferation. Together, these results provide a foundation for manipulating Yap activity as a novel approach to expand functional islet mass for diabetes regenerative therapy.
Collapse
Affiliation(s)
- Nicholas M George
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Brian P Boerner
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Shakeel U R Mir
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Zachary Guinn
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Nora E Sarvetnick
- Holland Regenerative Medicine Program (N.M.G., B.P.B., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha, Nebraska 68198; Department of Surgery (N.M.G., S.U.R.M., Z.G., N.E.S.), University of Nebraska Medical Center, Omaha 68198, Nebraska; and Department of Internal Medicine (B.P.B.), University of Nebraska Medical Center, Omaha, Nebraska 68198
| |
Collapse
|
57
|
Pauerstein PT, Sugiyama T, Stanley SE, McLean GW, Wang J, Martín MG, Kim SK. Dissecting Human Gene Functions Regulating Islet Development With Targeted Gene Transduction. Diabetes 2015; 64:3037-49. [PMID: 25901096 PMCID: PMC4512220 DOI: 10.2337/db15-0042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 04/09/2015] [Indexed: 01/19/2023]
Abstract
During pancreas development, endocrine precursors and their progeny differentiate, migrate, and cluster to form nascent islets. The transcription factor Neurogenin 3 (Neurog3) is required for islet development in mice, but its role in these dynamic morphogenetic steps has been inferred from fixed tissues. Moreover, little is known about the molecular genetic functions of NEUROG3 in human islet development. We developed methods for gene transduction by viral microinjection in the epithelium of cultured Neurog3-null mutant fetal pancreas, permitting genetic complementation in a developmentally relevant context. In addition, we developed methods for quantitative assessment of live-cell phenotypes in single developing islet cells. Delivery of wild-type NEUROG3 rescued islet differentiation, morphogenesis, and live cell deformation, whereas the patient-derived NEUROG3(R107S) allele partially restored indicators of islet development. NEUROG3(P39X), a previously unreported patient allele, failed to restore islet differentiation or morphogenesis and was indistinguishable from negative controls, suggesting that it is a null mutation. Our systems also permitted genetic suppression analysis and revealed that targets of NEUROG3, including NEUROD1 and RFX6, can partially restore islet development in Neurog3-null mutant mouse pancreata. Thus, advances described here permitted unprecedented assessment of gene functions in regulating crucial dynamic aspects of islet development in the fetal pancreas.
Collapse
Affiliation(s)
- Philip T Pauerstein
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Takuya Sugiyama
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Susan E Stanley
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Graeme W McLean
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Jing Wang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| | - Martín G Martín
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
58
|
Kistler WS, Baas D, Lemeille S, Paschaki M, Seguin-Estevez Q, Barras E, Ma W, Duteyrat JL, Morlé L, Durand B, Reith W. RFX2 Is a Major Transcriptional Regulator of Spermiogenesis. PLoS Genet 2015; 11:e1005368. [PMID: 26162102 PMCID: PMC4498915 DOI: 10.1371/journal.pgen.1005368] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 06/17/2015] [Indexed: 11/21/2022] Open
Abstract
Spermatogenesis consists broadly of three phases: proliferation of diploid germ cells, meiosis, and finally extensive differentiation of the haploid cells into effective delivery vehicles for the paternal genome. Despite detailed characterization of many haploid developmental steps leading to sperm, only fragmentary information exists on the control of gene expression underlying these processes. Here we report that the RFX2 transcription factor is a master regulator of genes required for the haploid phase. A targeted mutation of Rfx2 was created in mice. Rfx2-/- mice are perfectly viable but show complete male sterility. Spermatogenesis appears to progress unperturbed through meiosis. However, haploid cells undergo a complete arrest in spermatid development just prior to spermatid elongation. Arrested cells show altered Golgi apparatus organization, leading to a deficit in the generation of a spreading acrosomal cap from proacrosomal vesicles. Arrested cells ultimately merge to form giant multinucleated cells released to the epididymis. Spermatids also completely fail to form the flagellar axoneme. RNA-Seq analysis and ChIP-Seq analysis identified 139 genes directly controlled by RFX2 during spermiogenesis. Gene ontology analysis revealed that genes required for cilium function are specifically enriched in down- and upregulated genes showing that RFX2 allows precise temporal expression of ciliary genes. Several genes required for cell adhesion and cytoskeleton remodeling are also downregulated. Comparison of RFX2-regulated genes with those controlled by other major transcriptional regulators of spermiogenesis showed that each controls independent gene sets. Altogether, these observations show that RFX2 plays a major and specific function in spermiogenesis. Failure of spermatogenesis, which is presumed to often result from genetic defects, is a common cause of male sterility. Although numerous genes associated with defects in male spermatogenesis have been identified, numerous cases of genetic male infertility remain unelucidated. We report here that the transcription factor RFX2 is a master regulator of gene expression programs required for progression through the haploid phase of spermatogenesis. Male RFX2-deficient mice are completely sterile. Spermatogenesis progresses through meiosis, but haploid cells undergo a complete block in development just prior to spermatid elongation. Gene expression profiling and ChIP-Seq analysis revealed that RFX2 controls key pathways implicated in cilium/flagellum formation, as well as genes implicated in microtubule and vesicle associated transport. The set of genes activated by RFX2 in spermatids exhibits virtually no overlap with those controlled by other known transcriptional regulators of spermiogenesis, establishing RFX2 as an essential new player in this developmental process. RFX2-deficient mice should therefore represent a valuable new model for deciphering the regulatory networks that direct sperm formation, and thereby contribute to the identification of causes of human male infertility.
Collapse
Affiliation(s)
- W. Stephen Kistler
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, United States of America
- * E-mail: (WSK); (BD)
| | - Dominique Baas
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR 5534, Université Claude Bernard Lyon-1, Villeurbanne, Lyon, France
| | - Sylvain Lemeille
- Department of Pathology and Immunology, University of Geneva Medical School, CMU, Geneva, Switzerland
| | - Marie Paschaki
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR 5534, Université Claude Bernard Lyon-1, Villeurbanne, Lyon, France
| | - Queralt Seguin-Estevez
- Department of Pathology and Immunology, University of Geneva Medical School, CMU, Geneva, Switzerland
| | - Emmanuèle Barras
- Department of Pathology and Immunology, University of Geneva Medical School, CMU, Geneva, Switzerland
| | - Wenli Ma
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, United States of America
| | - Jean-Luc Duteyrat
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR 5534, Université Claude Bernard Lyon-1, Villeurbanne, Lyon, France
| | - Laurette Morlé
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR 5534, Université Claude Bernard Lyon-1, Villeurbanne, Lyon, France
| | - Bénédicte Durand
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS UMR 5534, Université Claude Bernard Lyon-1, Villeurbanne, Lyon, France
- * E-mail: (WSK); (BD)
| | - Walter Reith
- Department of Pathology and Immunology, University of Geneva Medical School, CMU, Geneva, Switzerland
| |
Collapse
|
59
|
Soria B, Gauthier BR, Martín F, Tejedo JR, Bedoya FJ, Rojas A, Hmadcha A. Using stem cells to produce insulin. Expert Opin Biol Ther 2015; 15:1469-89. [PMID: 26156425 DOI: 10.1517/14712598.2015.1066330] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Tremendous progress has been made in generating insulin-producing cells from pluripotent stem cells. The best outcome of the refined protocols became apparent in the first clinical trial announced by ViaCyte, based on the implantation of pancreatic progenitors that would further mature into functional insulin-producing cells inside the patient's body. AREAS COVERED Several groups, including ours, have contributed to improve strategies to generate insulin-producing cells. Of note, the latest results have gained a substantial amount of interest as a method to create a potentially functional and limitless supply of β-cell to revert diabetes mellitus. This review analyzes the accomplishments that have taken place over the last few decades, summarizes the state-of-art methods for β-cell replacement therapies based on the differentiation of embryonic stem cells into glucose-responsive and insulin-producing cells in a dish and discusses alternative approaches to obtain new sources of insulin-producing cells. EXPERT OPINION Undoubtedly, recent events preface the beginning of a new era in diabetes therapy. However, in our opinion, a number of significant hurdles still stand in the way of clinical application. We believe that the combination of the private and public sectors will accelerate the process of obtaining the desired safe and functional β-cell surrogates.
Collapse
Affiliation(s)
- Bernat Soria
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Benoit R Gauthier
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ;
| | - Franz Martín
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Juan R Tejedo
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Francisco J Bedoya
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Anabel Rojas
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Abdelkrim Hmadcha
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| |
Collapse
|
60
|
Martin D, Kim YH, Sever D, Mao CA, Haefliger JA, Grapin-Botton A. REST represses a subset of the pancreatic endocrine differentiation program. Dev Biol 2015; 405:316-27. [PMID: 26156633 DOI: 10.1016/j.ydbio.2015.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 07/01/2015] [Accepted: 07/02/2015] [Indexed: 12/20/2022]
Abstract
To contribute to devise successful beta-cell differentiation strategies for the cure of Type 1 diabetes we sought to uncover barriers that restrict endocrine fate acquisition by studying the role of the transcriptional repressor REST in the developing pancreas. Rest expression is prevented in neurons and in endocrine cells, which is necessary for their normal function. During development, REST represses a subset of genes in the neuronal differentiation program and Rest is down-regulated as neurons differentiate. Here, we investigate the role of REST in the differentiation of pancreatic endocrine cells, which are molecularly close to neurons. We show that Rest is widely expressed in pancreas progenitors and that it is down-regulated in differentiated endocrine cells. Sustained expression of REST in Pdx1(+) progenitors impairs the differentiation of endocrine-committed Neurog3(+) progenitors, decreases beta and alpha cell mass by E18.5, and triggers diabetes in adulthood. Conditional inactivation of Rest in Pdx1(+) progenitors is not sufficient to trigger endocrine differentiation but up-regulates a subset of differentiation genes. Our results show that the transcriptional repressor REST is active in pancreas progenitors where it gates the activation of part of the beta cell differentiation program.
Collapse
Affiliation(s)
- David Martin
- Swiss Institute for Experimental Cancer Research, Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Station 19, 1015 Lausanne, Switzerland
| | - Yung-Hae Kim
- DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200 Copenhagen N, Denmark
| | - Dror Sever
- DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200 Copenhagen N, Denmark
| | - Chai-An Mao
- Department of Systems Biology, The University of MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jacques-Antoine Haefliger
- Department of Medicine, Laboratory of Experimental Medicine, C/O Department of Physiology, Bugnon 7a, 1005 Lausanne, Switzerland
| | - Anne Grapin-Botton
- Swiss Institute for Experimental Cancer Research, Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Station 19, 1015 Lausanne, Switzerland; DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
61
|
McGrath PS, Watson CL, Ingram C, Helmrath MA, Wells JM. The Basic Helix-Loop-Helix Transcription Factor NEUROG3 Is Required for Development of the Human Endocrine Pancreas. Diabetes 2015; 64:2497-505. [PMID: 25650326 PMCID: PMC4477351 DOI: 10.2337/db14-1412] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 01/20/2015] [Indexed: 12/18/2022]
Abstract
Neurogenin3 (NEUROG3) is a basic helix-loop-helix transcription factor required for development of the endocrine pancreas in mice. In contrast, humans with NEUROG3 mutations are born with endocrine pancreas function, calling into question whether NEUROG3 is required for human endocrine pancreas development. To test this directly, we generated human embryonic stem cell (hESC) lines where both alleles of NEUROG3 were disrupted using CRISPR/Cas9-mediated gene targeting. NEUROG3(-/-) hESC lines efficiently formed pancreatic progenitors but lacked detectible NEUROG3 protein and did not form endocrine cells in vitro. Moreover, NEUROG3(-/-) hESC lines were unable to form mature pancreatic endocrine cells after engraftment of PDX1(+)/NKX6.1(+) pancreatic progenitors into mice. In contrast, a 75-90% knockdown of NEUROG3 caused a reduction, but not a loss, of pancreatic endocrine cell development. We conclude that NEUROG3 is essential for endocrine pancreas development in humans and that as little as 10% NEUROG3 is sufficient for formation of pancreatic endocrine cells.
Collapse
Affiliation(s)
- Patrick S McGrath
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Carey L Watson
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH Department of General Surgery, University of Cincinnati, Cincinnati, OH
| | - Cameron Ingram
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Michael A Helmrath
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH Department of General Surgery, University of Cincinnati, Cincinnati, OH
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
62
|
Martinez-Sanchez A, Nguyen-Tu MS, Rutter GA. DICER Inactivation Identifies Pancreatic β-Cell "Disallowed" Genes Targeted by MicroRNAs. Mol Endocrinol 2015; 29:1067-79. [PMID: 26038943 PMCID: PMC4484783 DOI: 10.1210/me.2015-1059] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pancreatic β-cells are the body's sole source of circulating insulin and essential for the maintenance of blood glucose homeostasis. Levels of up to 66 “disallowed” genes, which are strongly expressed and play housekeeping roles in most other mammalian tissues, are unusually low in β-cells. The molecular mechanisms involved in repressing these genes are largely unknown. Here, we explore the role in gene disallowance of microRNAs (miRNAs), a type of small noncoding RNAs that silence gene expression at the posttranscriptional level and are essential for β-cell development and function. To selectively deplete miRNAs from adult β-cells, the miRNA-processing enzyme DICER was inactivated by deletion of the RNase III domain with a tamoxifen-inducible Pdx1CreER transgene. In this model, β-cell dysfunction was apparent 2 weeks after recombination and preceded a decrease in insulin content and loss of β-cell mass. Of the 14 disallowed genes studied, quantitative RT-quantitative real-time PCR revealed that 6 genes (Fcgrt, Igfbp4, Maf, Oat, Pdgfra, and Slc16a1) were up-regulated (1.4- to 2.1-fold, P < .05) at this early stage. Expression of luciferase constructs bearing the 3′-untranslated regions of the corresponding mRNAs in wild-type or DICER-null β-cells demonstrated that Fcgrt, Oat, and Pdgfra are miRNA direct targets. We thus reveal a role for miRNAs in the regulation of disallowed genes in β-cells and provide evidence for a novel means through which noncoding RNAs control the functional identity of these cells independently of actions on β-cell mass.
Collapse
Affiliation(s)
- Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Marie-Sophie Nguyen-Tu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Guy A Rutter
- 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
|
63
|
Thulé PM, Jia D, Safley S, Gordon K, Barber G, Yi H, Nalli S, Onderci M, Sharma J, Shires J, Weber CJ. Engineered insulin secretion from neuroendocrine cells isolated from human thyroid. World J Surg 2015; 38:1251-61. [PMID: 24549997 DOI: 10.1007/s00268-014-2457-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Insulin-secreting beta-like cells are vulnerable to diabetic autoimmunity. We hypothesized that human thyroid neuroendocrine (NE) cells could be engineered to secrete human insulin, be glucose-responsive, and avoid autoimmunity. METHODS Collagenase-digested thyroid tissue was cultured and subjected to size-based fluorescence-activated cell sorting. Insulin secretion and storage in NE cells transduced with viral vectors carrying an insulin sequence was assessed by enzyme-linked immunosorbent assay (ELISA) and immunogold transmission electron microscopy (TEM). Baseline mRNA expression was assessed by Illumina expression array analysis. Transduction with retrovirus expressing transcription factors PDX1, NGN3, MAFA, or HNF6 altered mRNA expression in a custom polymerase chain reaction (PCR) array. Gastrin-releasing peptide (GRP) in conditioned medium and cell lysates was determined by reverse transcription (RT)-PCR, ELISA, and immunohistochemistry. RESULTS Isolation yielded an average of 2.2 × 10(6) cells/g thyroid tissue, which stained for calcitonin/calcitonin gene-related protein, expressed genes consistent with NE origins, and secreted GRP. Transduced cells secreted 56 % and retained 48 % of total insulin produced. Immunogold TEM revealed insulin in secretory vesicles. PDX1, NGN3, and MAFA overexpression increased expression of genes typical for hepatocytes and beta cells. Overexpression of HNF6 also increased the message of genes critical for glucose sensing. CONCLUSIONS Human thyroid NE cells can produce human insulin, fractions of which are both secreted and retained in secretory granules. Overexpression of HNF6, PDX1, or NGN3 enhances expression of both hepatocyte and beta cell typical mRNAs, including the message of proteins critical for glucose sensing. These data suggest that reimplantation of engineered autologous NE cells may develop as a viable treatment for diabetes mellitus type 1.
Collapse
Affiliation(s)
- Peter M Thulé
- Section Endocrinology & Metabolism, Atlanta VA Medical Center, Decatur, GA, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Piccand J, Strasser P, Hodson DJ, Meunier A, Ye T, Keime C, Birling MC, Rutter GA, Gradwohl G. Rfx6 maintains the functional identity of adult pancreatic β cells. Cell Rep 2014; 9:2219-32. [PMID: 25497096 PMCID: PMC4542305 DOI: 10.1016/j.celrep.2014.11.033] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/27/2014] [Accepted: 11/20/2014] [Indexed: 01/09/2023] Open
Abstract
Increasing evidence suggests that loss of β cell characteristics may cause insulin secretory deficiency in diabetes, but the underlying mechanisms remain unclear. Here, we show that Rfx6, whose mutation leads to neonatal diabetes in humans, is essential to maintain key features of functionally mature β cells in mice. Rfx6 loss in adult β cells leads to glucose intolerance, impaired β cell glucose sensing, and defective insulin secretion. This is associated with reduced expression of core components of the insulin secretion pathway, including glucokinase, the Abcc8/SUR1 subunit of KATP channels and voltage-gated Ca(2+) channels, which are direct targets of Rfx6. Moreover, Rfx6 contributes to the silencing of the vast majority of "disallowed" genes, a group usually specifically repressed in adult β cells, and thus to the maintenance of β cell maturity. These findings raise the possibility that changes in Rfx6 expression or activity may contribute to β cell failure in humans.
Collapse
Affiliation(s)
- Julie Piccand
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale U964, Centre National de Recherche Scientifique UMR7104, Université de Strasbourg, Illkirch 67404, France
| | - Perrine Strasser
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale U964, Centre National de Recherche Scientifique UMR7104, Université de Strasbourg, Illkirch 67404, France
| | - David J Hodson
- Section of Cell Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Hammersmith Hospital, du Cane Road, London W12 0NN, UK
| | - Aline Meunier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale U964, Centre National de Recherche Scientifique UMR7104, Université de Strasbourg, Illkirch 67404, France
| | - Tao Ye
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale U964, Centre National de Recherche Scientifique UMR7104, Université de Strasbourg, Illkirch 67404, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale U964, Centre National de Recherche Scientifique UMR7104, Université de Strasbourg, Illkirch 67404, France
| | | | - Guy A Rutter
- Section of Cell Biology, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Hammersmith Hospital, du Cane Road, London W12 0NN, UK
| | - Gérard Gradwohl
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale U964, Centre National de Recherche Scientifique UMR7104, Université de Strasbourg, Illkirch 67404, France.
| |
Collapse
|
65
|
Chandra V, Albagli-Curiel O, Hastoy B, Piccand J, Randriamampita C, Vaillant E, Cavé H, Busiah K, Froguel P, Vaxillaire M, Rorsman P, Polak M, Scharfmann R. RFX6 Regulates Insulin Secretion by Modulating Ca2+ Homeostasis in Human β Cells. Cell Rep 2014; 9:2206-18. [DOI: 10.1016/j.celrep.2014.11.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/09/2014] [Accepted: 11/06/2014] [Indexed: 10/24/2022] Open
|
66
|
An integrated cell purification and genomics strategy reveals multiple regulators of pancreas development. PLoS Genet 2014; 10:e1004645. [PMID: 25330008 PMCID: PMC4199491 DOI: 10.1371/journal.pgen.1004645] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 08/02/2014] [Indexed: 12/15/2022] Open
Abstract
The regulatory logic underlying global transcriptional programs controlling development of visceral organs like the pancreas remains undiscovered. Here, we profiled gene expression in 12 purified populations of fetal and adult pancreatic epithelial cells representing crucial progenitor cell subsets, and their endocrine or exocrine progeny. Using probabilistic models to decode the general programs organizing gene expression, we identified co-expressed gene sets in cell subsets that revealed patterns and processes governing progenitor cell development, lineage specification, and endocrine cell maturation. Purification of Neurog3 mutant cells and module network analysis linked established regulators such as Neurog3 to unrecognized gene targets and roles in pancreas development. Iterative module network analysis nominated and prioritized transcriptional regulators, including diabetes risk genes. Functional validation of a subset of candidate regulators with corresponding mutant mice revealed that the transcription factors Etv1, Prdm16, Runx1t1 and Bcl11a are essential for pancreas development. Our integrated approach provides a unique framework for identifying regulatory genes and functional gene sets underlying pancreas development and associated diseases such as diabetes mellitus. Discovery of specific pancreas developmental regulators has accelerated in recent years. In contrast, the global regulatory programs controlling pancreas development are poorly understood compared to other organs or tissues like heart or blood. Decoding this regulatory logic may accelerate development of replacement organs from renewable sources like stem cells, but this goal requires identification of regulators and assessment of their functions on a global scale. To address this important challenge for pancreas biology, we combined purification of normal and mutant cells with genome-scale methods to generate and analyze expression profiles from developing pancreas cells. Our work revealed regulatory gene sets governing development of pancreas progenitor cells and their progeny. Our integrative approach nominated multiple pancreas developmental regulators, including suspected risk genes for human diabetes, which we validated by phenotyping mutant mice on a scale not previously reported. Selection of these candidate regulators was unbiased; thus it is remarkable that all were essential for pancreatic islet development. Thus, our studies provide a new heuristic resource for identifying genetic functions underlying pancreas development and diseases like diabetes mellitus.
Collapse
|
67
|
Osipovich AB, Long Q, Manduchi E, Gangula R, Hipkens SB, Schneider J, Okubo T, Stoeckert CJ, Takada S, Magnuson MA. Insm1 promotes endocrine cell differentiation by modulating the expression of a network of genes that includes Neurog3 and Ripply3. Development 2014; 141:2939-49. [PMID: 25053427 PMCID: PMC4197673 DOI: 10.1242/dev.104810] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Insulinoma associated 1 (Insm1) plays an important role in regulating the development of cells in the central and peripheral nervous systems, olfactory epithelium and endocrine pancreas. To better define the role of Insm1 in pancreatic endocrine cell development we generated mice with an Insm1GFPCre reporter allele and used them to study Insm1-expressing and null populations. Endocrine progenitor cells lacking Insm1 were less differentiated and exhibited broad defects in hormone production, cell proliferation and cell migration. Embryos lacking Insm1 contained greater amounts of a non-coding Neurog3 mRNA splice variant and had fewer Neurog3/Insm1 co-expressing progenitor cells, suggesting that Insm1 positively regulates Neurog3. Moreover, endocrine progenitor cells that express either high or low levels of Pdx1, and thus may be biased towards the formation of specific cell lineages, exhibited cell type-specific differences in the genes regulated by Insm1. Analysis of the function of Ripply3, an Insm1-regulated gene enriched in the Pdx1-high cell population, revealed that it negatively regulates the proliferation of early endocrine cells. Taken together, these findings indicate that in developing pancreatic endocrine cells Insm1 promotes the transition from a ductal progenitor to a committed endocrine cell by repressing a progenitor cell program and activating genes essential for RNA splicing, cell migration, controlled cellular proliferation, vasculogenesis, extracellular matrix and hormone secretion.
Collapse
Affiliation(s)
- Anna B Osipovich
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Qiaoming Long
- Department of Animal Science, Cornell University, Ithaca, NY 14850, USA
| | - Elisabetta Manduchi
- Penn Center for Bioinformatics, Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Rama Gangula
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Susan B Hipkens
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Judsen Schneider
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Tadashi Okubo
- Department of Laboratory Animal Science, Kitasato University School of Medicine, Sagamihara, 252-0374, Japan
| | - Christian J Stoeckert
- Penn Center for Bioinformatics, Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Shinji Takada
- Okazaki Institute for Integrative Bioscience, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Mark A Magnuson
- Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
68
|
Xu CR, Li LC, Donahue G, Ying L, Zhang YW, Gadue P, Zaret KS. Dynamics of genomic H3K27me3 domains and role of EZH2 during pancreatic endocrine specification. EMBO J 2014; 33:2157-70. [PMID: 25107471 DOI: 10.15252/embj.201488671] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Endoderm cells undergo sequential fate choices to generate insulin-secreting beta cells. Ezh2 of the PRC2 complex, which generates H3K27me3, modulates the transition from endoderm to pancreas progenitors, but the role of Ezh2 and H3K27me3 in the next transition to endocrine progenitors is unknown. We isolated endoderm cells, pancreas progenitors, and endocrine progenitors from different staged mouse embryos and analyzed H3K27me3 genome-wide. Unlike the decline in H3K27me3 domains reported during embryonic stem cell differentiation in vitro, we find that H3K27me3 domains increase in number during endocrine progenitor development in vivo. Genes that lose the H3K27me3 mark typically encode transcriptional regulators, including those for pro-endocrine fates, whereas genes that acquire the mark typically are involved in cell biology and morphogenesis. Deletion of Ezh2 at the pancreas progenitor stage enhanced the production of endocrine progenitors and beta cells. Inhibition of EZH2 in embryonic pancreas explants and in human embryonic stem cell cultures increased endocrine progenitors in vitro. Our studies reveal distinct dynamics in H3K27me3 targets in vivo and a means to modulate beta cell development from stem cells.
Collapse
Affiliation(s)
- Cheng-Ran Xu
- Institute for Regenerative Medicine, Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA Ministry of Education Key Laboratory of Cell Proliferation, College of Life Sciences Peking-Tsinghua Center for Life Sciences Peking University, Beijing, China
| | - Lin-Chen Li
- Ministry of Education Key Laboratory of Cell Proliferation, College of Life Sciences Peking-Tsinghua Center for Life Sciences Peking University, Beijing, China
| | - Greg Donahue
- Institute for Regenerative Medicine, Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
| | - Lei Ying
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yu-Wei Zhang
- Ministry of Education Key Laboratory of Cell Proliferation, College of Life Sciences Peking-Tsinghua Center for Life Sciences Peking University, Beijing, China
| | - Paul Gadue
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kenneth S Zaret
- Institute for Regenerative Medicine, Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
69
|
Therapeutic implications of novel mutations of the RFX6 gene associated with early-onset diabetes. THE PHARMACOGENOMICS JOURNAL 2014; 15:49-54. [PMID: 25048417 DOI: 10.1038/tpj.2014.37] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/08/2014] [Accepted: 06/04/2014] [Indexed: 11/08/2022]
Abstract
Identification of the genetic defect underlying early-onset diabetes is important for determining the specific diabetes subtype, which would then permit appropriate treatment and accurate assessment of recurrence risk in offspring. Given the extensive genetic and clinical heterogeneity of the disease, high-throughput sequencing might provide additional diagnostic potential when Sanger sequencing is ineffective. Our aim was to develop a targeted next-generation assay able to detect mutations in several genes involved in glucose metabolism. All 13 known MODY genes, genes identified from a genome-wide linkage study or genome-wide association studies as increasing the risk of type 2 diabetes and genes causing diabetes in animal models, were included in the custom panel. We selected a total of 102 genes by performing a targeting re-sequencing in 30 patients negative for mutations in the GCK, HNF1α, HNF4α, HNF1β and IPF1 genes at the Sanger sequencing analysis. Previously unidentified variants in the RFX6 gene were found in three patients and in two of them we also detected rare variants in WFS1 and ABCC8 genes. All patients showed a good therapeutic response to dipeptidyl peptidase-4 (DPP4) inhibitors. Our study reveals that next-generation sequencing provides a highly sensitive method for identification of variants in new causative genes of diabetes. This approach may help in understanding the molecular etiology of diabetes and in providing more personalized treatment for each genetic subtype.
Collapse
|
70
|
Cano DA, Soria B, Martín F, Rojas A. Transcriptional control of mammalian pancreas organogenesis. Cell Mol Life Sci 2014; 71:2383-402. [PMID: 24221136 PMCID: PMC11113897 DOI: 10.1007/s00018-013-1510-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/19/2013] [Accepted: 10/29/2013] [Indexed: 12/12/2022]
Abstract
The field of pancreas development has markedly expanded over the last decade, significantly advancing our understanding of the molecular mechanisms that control pancreas organogenesis. This growth has been fueled, in part, by the need to generate new therapeutic approaches for the treatment of diabetes. The creation of sophisticated genetic tools in mice has been instrumental in this progress. Genetic manipulation involving activation or inactivation of genes within specific cell types has allowed the identification of many transcription factors (TFs) that play critical roles in the organogenesis of the pancreas. Interestingly, many of these TFs act at multiple stages of pancreatic development, and adult organ function or repair. Interaction with other TFs, extrinsic signals, and epigenetic regulation are among the mechanisms by which TFs may play context-dependent roles during pancreas organogenesis. Many of the pancreatic TFs directly regulate each other and their own expression. These combinatorial interactions generate very specific gene regulatory networks that can define the different cell lineages and types in the developing pancreas. Here, we review recent progress made in understanding the role of pancreatic TFs in mouse pancreas formation. We also summarize our current knowledge of human pancreas development and discuss developmental pancreatic TFs that have been associated with human pancreatic diseases.
Collapse
Affiliation(s)
- David A. Cano
- Endocrinology Unit, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, Seville, Spain
| | - Bernat Soria
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Avda. Americo Vespucio s/n., Parque Científico Isla de la Cartuja, 41092 Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Francisco Martín
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Avda. Americo Vespucio s/n., Parque Científico Isla de la Cartuja, 41092 Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Anabel Rojas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Avda. Americo Vespucio s/n., Parque Científico Isla de la Cartuja, 41092 Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| |
Collapse
|
71
|
Abstract
Monogenic diabetes represents a heterogeneous group of disorders resulting from defects in single genes. Defects are categorized primarily into two groups: disruption of β-cell function or a reduction in the number of β-cells. A complex network of transcription factors control pancreas formation, and a dysfunction of regulators high in the hierarchy leads to pancreatic agenesis. Dysfunction among factors further downstream might cause organ hypoplasia, absence of islets of Langerhans or a reduction in the number of β-cells. Many transcription factors have pleiotropic effects, explaining the association of diabetes with other congenital malformations, including cerebellar agenesis and pituitary agenesis. Monogenic diabetes variants are classified conventionally according to age of onset, with neonatal diabetes occurring before the age of 6 months and maturity onset diabetes of the young (MODY) manifesting before the age of 25 years. Recently, certain familial genetic defects were shown to manifest as neonatal diabetes, MODY or even adult onset diabetes. Patients with neonatal diabetes require a thorough genetic work-up in any case, and because extensive phenotypic overlap exists between monogenic, type 2, and type 1 diabetes, genetic analysis will also help improve diagnosis in these cases. Next generation sequencing will facilitate rapid screening, leading to the discovery of digenic and oligogenic diabetes variants, and helping to improve our understanding of the genetics underlying other types of diabetes. An accurate diagnosis remains important, because it might lead to a change in the treatment of affected subjects and influence long-term complications.
Collapse
Affiliation(s)
- Valerie M Schwitzgebel
- Pediatric Endocrine and Diabetes UnitDepartment of Child and Adolescent HealthChildren's University HospitalGenevaSwitzerland
| |
Collapse
|
72
|
Li HJ, Johnston B, Aiello D, Caffrey DR, Giel-Moloney M, Rindi G, Leiter AB. Distinct cellular origins for serotonin-expressing and enterochromaffin-like cells in the gastric corpus. Gastroenterology 2014; 146:754-764.e3. [PMID: 24316261 PMCID: PMC3943955 DOI: 10.1053/j.gastro.2013.11.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 11/12/2013] [Accepted: 11/24/2013] [Indexed: 02/05/2023]
Abstract
BACKGROUND & AIMS The alimentary tract contains a diffuse endocrine system comprising enteroendocrine cells that secrete peptides or biogenic amines to regulate digestion, insulin secretion, food intake, and energy homeostasis. Lineage analysis in the stomach revealed that a significant fraction of endocrine cells in the gastric corpus did not arise from Neurogenin3 (Neurog3)-expressing cells, unlike enteroendocrine cells elsewhere in the digestive tract. We aimed to isolate enriched serotonin-secreting and enterochromaffin-like (ECL) cells from the stomach and to clarify their cellular origin. METHODS We used Neurogenic differentiation 1 (NeuroD1) and Neurog3 lineage analysis and examined the differentiation of serotonin-producing and ECL cells in stomach tissues of NeuroD1-cre;ROSA(tdTom), tryptophan hydroxylase 1 (Tph1)-cyan fluorescent protein (CFP), c-Kit(wsh/wsh), and Neurog3Cre;ROSA(tdTom) mice by immunohistochemistry. We used fluorescence-activated cell sorting to isolate each cell type for gene expression analysis. We also performed RNA sequencing analysis of ECL cells. RESULTS Neither serotonin-secreting nor ECL cells of the corpus arose from cells expressing NeuroD1. Serotonin-secreting cells expressed a number of mast cell genes but not genes associated with endocrine differentiation; they did not develop in c-Kit(wsh/wsh) mice and were labeled with transplanted bone marrow cells. RNA sequencing analysis of ECL cells revealed high expression levels of many genes common to endocrine cells, including transcription factors, hormones, ion channels, and solute transporters but not markers of bone marrow cells. CONCLUSIONS Serotonin-expressing cells of the gastric corpus of mice appear to be bone marrow-derived mucosal mast cells. Gene expression analysis of ECL cells indicated that they are endocrine cells of epithelial origin that do not express the same transcription factors as their intestinal enteroendocrine cell counterparts.
Collapse
Affiliation(s)
- Hui Joyce Li
- Division of Gastroenterology, Department of Medicine, University of
Massachusetts Medical School, Worcester, MA 01605
| | - Brian Johnston
- Division of Gastroenterology, Department of Medicine, University of
Massachusetts Medical School, Worcester, MA 01605
| | - Daniel Aiello
- Department of Medicine, University of Massachusetts Medical School,
Worcester, MA 01605
| | - Daniel R Caffrey
- Department of Medicine, University of Massachusetts Medical School,
Worcester, MA 01605
| | | | | | - Andrew B. Leiter
- Division of Gastroenterology, Department of Medicine, University of
Massachusetts Medical School, Worcester, MA 01605,Department of Medicine, University of Massachusetts Medical School,
Worcester, MA 01605
| |
Collapse
|
73
|
Seeberger KL, Anderson SJ, Ellis CE, Yeung TY, Korbutt GS. Identification and differentiation of PDX1 β-cell progenitors within the human pancreatic epithelium. World J Diabetes 2014; 5:59-68. [PMID: 24567802 PMCID: PMC3932428 DOI: 10.4239/wjd.v5.i1.59] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 11/27/2013] [Accepted: 12/16/2013] [Indexed: 02/05/2023] Open
Abstract
AIM: To minimize the expansion of pancreatic mesenchymal cells in vitro and confirm that β-cell progenitors reside within the pancreatic epithelium.
METHODS: Due to mesenchymal stem cell (MSC) expansion and overgrowth, progenitor cells within the pancreatic epithelium cannot be characterized in vitro, though β-cell dedifferentiation and expansion of MSC intermediates via epithelial-mesenchymal transition (EMT) may generate β-cell progenitors. Pancreatic epithelial cells from endocrine and non-endocrine tissue were expanded and differentiated in a novel pancreatic epithelial expansion medium supplemented with growth factors known to support epithelial cell growth (dexamethasone, epidermal growth factor, 3,5,3’-triiodo-l-thyronine, bovine brain extract). Cells were also infected with a single and dual lentiviral reporter prior to cell differentiation. Enhanced green fluorescent protein was controlled by the rat Insulin 1 promoter and the monomeric red fluorescent protein was controlled by the mouse PDX1 promoter. In combination with lentiviral tracing, cells expanded and differentiated in the pancreatic medium were characterized by flow cytometry (BD fluorescence activated cell sorting), immunostaining and real-time polymerase chain reaction (PCR) (7900HT Fast Realtime PCR System).
RESULTS: In the presence of 10% serum MSCs rapidly expand in vitro while the epithelial cell population declines. The percentage of vimentin+ cells increased from 22% ± 5.83% to 80.43% ± 3.24% (14 d) and 99.00% ± 0.0% (21 d), and the percentage of epithelial cells decreased from 74.71% ± 8.34% to 26.57% ± 9.75% (14 d) and 4.00% ± 1.53% (21 d), P < 0.01 for all time points. Our novel pancreatic epithelial expansion medium preserved the epithelial cell phenotype and minimized epithelial cell dedifferentiation and EMT. Cells expanded in our epithelial medium contained significantly less mesenchymal cells (vimentin+) compared to controls (44.87% ± 4.93% vs 95.67% ± 1.36%; P < 0.01). During cell differentiation lentiviral reporting demonstrated that, PDX1+ and insulin+ cells were localized within adherent epithelial cell aggregates compared to controls. Compared to starting islets differentiated cells had at least two fold higher gene expression of PDX1, insulin, PAX4 and RFX (P < 0.05).
CONCLUSION: PDX1+ cells were confined to adherent epithelial cell aggregates and not vimentin+ cells (mesenchymal), suggesting that EMT is not a mechanism for generating pancreatic progenitor cells.
Collapse
|
74
|
Manojlovic Z, Earwood R, Kato A, Stefanovic B, Kato Y. RFX7 is required for the formation of cilia in the neural tube. Mech Dev 2014; 132:28-37. [PMID: 24530844 DOI: 10.1016/j.mod.2014.02.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 02/04/2014] [Indexed: 12/22/2022]
Abstract
Regulatory Factor X (RFX) transcription factors are important for development and are likely involved in the pathogenesis of serious human diseases including ciliopathies. While seven RFX genes have been identified in vertebrates and several RFX transcription factors have been reported to be regulators of ciliogenesis, the role of RFX7 in development including ciliogenesis is not known. Here we show that RFX7 in Xenopus laevis is expressed in the neural tube, eye, otic vesicles, and somites. Knockdown of RFX7 in Xenopus embryos resulted in a defect of ciliogenesis in the neural tube and failure of neural tube closure. RFX7 controlled the formation of cilia by regulating the expression of RFX4 gene, which has been reported to be required for ciliogenesis in the neural tube. Moreover, ectopic expression of Foxj1, which is a master regulator of motile cilia formation, suppressed the expression of RFX4 but not RFX7. Taken together, RFX7 plays an important role in the process of neural tube closure at the top of the molecular cascade which controls ciliogenesis in the neural tube.
Collapse
Affiliation(s)
- Zarko Manojlovic
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Ryan Earwood
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Akiko Kato
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Branko Stefanovic
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| | - Yoichi Kato
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| |
Collapse
|
75
|
Impact of high-fat feeding on basic helix-loop-helix transcription factors controlling enteroendocrine cell differentiation. Int J Obes (Lond) 2014; 38:1440-8. [PMID: 24480860 DOI: 10.1038/ijo.2014.20] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/03/2013] [Accepted: 01/17/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES Gut hormones secreted by enteroendocrine cells (EECs) play a major role in energy regulation. Differentiation of EEC is controlled by the expression of basic helix-loop-helix (bHLH) transcription factors. High-fat (HF) feeding alters gut hormone levels; however, the impact of HF feeding on bHLH transcription factors in mediating EEC differentiation and subsequent gut hormone secretion and expression is not known. METHODS Outbred Sprague-Dawley rats were maintained on chow or HF diet for 12 weeks. Gene and protein expression of intestinal bHLH transcription factors, combined with immunofluorescence studies, were analyzed for both groups in the small intestine and colon. Gut permeability, intestinal lipid and carbohydrate transporters as well as circulating levels and intestinal protein expression of gut peptides were determined. RESULTS We showed that HF feeding resulted in hyperphagia and increased adiposity. HF-fed animals exhibited decreased expression of bHLH transcription factors controlling EEC differentiation (MATH1, NGN3, NEUROD1) and increased expression of bHLH factors modulating enterocyte expression. Furthermore, HF-fed animals had decreased number of total EECs and L-cells. This was accompanied by increased gut permeability and expression of lipid and carbohydrate transporters, and a decrease in circulating and intestinal gut hormone levels. CONCLUSIONS Taken together, our results demonstrate that HF feeding caused decreased secretory lineage (that is, EECs) differentiation through downregulation of bHLH transcription factors, resulting in reduced EEC number and gut hormone levels. Thus, impaired EEC differentiation pathways by HF feeding may promote hyperphagia and subsequent obesity.
Collapse
|
76
|
Piccand J, Meunier A, Merle C, Jia Z, Barnier JV, Gradwohl G. Pak3 promotes cell cycle exit and differentiation of β-cells in the embryonic pancreas and is necessary to maintain glucose homeostasis in adult mice. Diabetes 2014; 63:203-15. [PMID: 24163148 PMCID: PMC3968432 DOI: 10.2337/db13-0384] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The transcription factor neurogenin3 (Ngn3) triggers islet cell differentiation in the developing pancreas. However, little is known about the molecular mechanisms coupling cell cycle exit and differentiation in Ngn3(+) islet progenitors. We identified a novel effector of Ngn3 endocrinogenic function, the p21 protein-activated kinase Pak3, known to control neuronal differentiation and implicated in X-linked intellectual disability in humans. We show that Pak3 expression is initiated in Ngn3(+) endocrine progenitor cells and next maintained in maturing hormone-expressing cells during pancreas development as well as in adult islet cells. In Pak3-deficient embryos, the proliferation of Ngn3(+) progenitors and β-cells is transiently increased concomitantly with an upregulation of Ccnd1. β-Cell differentiation is impaired at E15.5 but resumes at later stages. Pak3-deficient mice do not develop overt diabetes but are glucose intolerant under high-fat diet (HFD). In the intestine, Pak3 is expressed in enteroendocrine cells but is not necessary for their differentiation. Our results indicate that Pak3 is a novel regulator of β-cell differentiation and function. Pak3 acts downstream of Ngn3 to promote cell cycle exit and differentiation in the embryo by a mechanism that might involve repression of Ccnd1. In the adult, Pak3 is required for the proper control of glucose homeostasis under challenging HFD.
Collapse
Affiliation(s)
- Julie Piccand
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Development and Stem Cells, Institut National de la Santé et de la Recherche Médicale UMR 964, Centre National de Recherche Scientifique, UMR 964, Université de Strasbourg, Illkirch, France
| | - Aline Meunier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Development and Stem Cells, Institut National de la Santé et de la Recherche Médicale UMR 964, Centre National de Recherche Scientifique, UMR 964, Université de Strasbourg, Illkirch, France
| | - Carole Merle
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Development and Stem Cells, Institut National de la Santé et de la Recherche Médicale UMR 964, Centre National de Recherche Scientifique, UMR 964, Université de Strasbourg, Illkirch, France
| | - Zhengping Jia
- Neurosciences and Mental Health, The Hospital for Sick Children, Department of Physiology, University of Toronto, Toronto, Canada
| | - Jean-Vianney Barnier
- Université Paris-Sud, Centre de Neurosciences Paris-Sud, UMR 8195, Orsay, France
- Centre National de Recherche Scientifique, UMR 8195, Orsay, France
| | - Gérard Gradwohl
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Development and Stem Cells, Institut National de la Santé et de la Recherche Médicale UMR 964, Centre National de Recherche Scientifique, UMR 964, Université de Strasbourg, Illkirch, France
- Corresponding author: Gérard Gradwohl,
| |
Collapse
|
77
|
Courtney M, Gjernes E, Druelle N, Ravaud C, Vieira A, Ben-Othman N, Pfeifer A, Avolio F, Leuckx G, Lacas-Gervais S, Burel-Vandenbos F, Ambrosetti D, Hecksher-Sorensen J, Ravassard P, Heimberg H, Mansouri A, Collombat P. The inactivation of Arx in pancreatic α-cells triggers their neogenesis and conversion into functional β-like cells. PLoS Genet 2013; 9:e1003934. [PMID: 24204325 PMCID: PMC3814322 DOI: 10.1371/journal.pgen.1003934] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
Recently, it was demonstrated that pancreatic new-born glucagon-producing cells can regenerate and convert into insulin-producing β-like cells through the ectopic expression of a single gene, Pax4. Here, combining conditional loss-of-function and lineage tracing approaches, we show that the selective inhibition of the Arx gene in α-cells is sufficient to promote the conversion of adult α-cells into β-like cells at any age. Interestingly, this conversion induces the continuous mobilization of duct-lining precursor cells to adopt an endocrine cell fate, the glucagon+ cells thereby generated being subsequently converted into β-like cells upon Arx inhibition. Of interest, through the generation and analysis of Arx and Pax4 conditional double-mutants, we provide evidence that Pax4 is dispensable for these regeneration processes, indicating that Arx represents the main trigger of α-cell-mediated β-like cell neogenesis. Importantly, the loss of Arx in α-cells is sufficient to regenerate a functional β-cell mass and thereby reverse diabetes following toxin-induced β-cell depletion. Our data therefore suggest that strategies aiming at inhibiting the expression of Arx, or its molecular targets/co-factors, may pave new avenues for the treatment of diabetes. Type 1 diabetes is a condition that results from the loss of insulin-producing β-cells. Despite current therapies, diabetic patients are prone to vascular complications. Using the mouse as a model, we previously found that pancreatic glucagon-expressing cells can be regenerated and converted into β-like cells by the forced expression of a single gene, Pax4. Here, we generated transgenic mice allowing both the permanent labeling of α-cells and the inactivation of Arx solely in this cell subtype. Our results indicate that, upon Arx inactivation, α-cells can be continuously regenerated from duct-lining precursors and converted into β-like cells. Importantly, the additional loss of Pax4 does not impact these processes, suggesting that Arx is the main trigger of α-cell-mediated β-like cell neogenesis. Most interestingly, upon chemical induction of diabetes/β-cell loss, while control animals die or remain severely hyperglycemic, a normalization of the glycemia, a clear regeneration of the β-like cell mass, and an extended lifespan are noted in animals with the conditional inactivation of Arx. Our data therefore suggest that strategies aiming at inhibiting the expression of Arx, or its molecular targets/co-factors, may pave new avenues for the treatment of diabetes.
Collapse
Affiliation(s)
- Monica Courtney
- Université de Nice Sophia Antipolis, iBV, UMR 7277, Nice, France ; Inserm, iBV, U1091, Nice, France ; CNRS, iBV, UMR 7277, Nice, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Wang H, Yang Y, Ho G, Lin X, Wu W, Li W, Lin L, Feng X, Huo X, Jiang J, Liu X, Huang T, Wei C, Ma L. Programming of human umbilical cord mesenchymal stem cells in vitro to promote pancreatic gene expression. Mol Med Rep 2013; 8:769-74. [PMID: 23900717 DOI: 10.3892/mmr.2013.1598] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 07/11/2013] [Indexed: 02/05/2023] Open
Abstract
Human umbilical cord mesenchymal stem cells (HUMSCs) are candidates for tissue engineering and may potentially be used for transdifferentiation into pancreatic endocrine cells. The adenoviral vector is effective in transducing genes into stem cells that are refractory to gene delivery by non‑viral approaches. qPCR was used to detect the pancreatic endogenous gene expression of HUMSCs transfected by islet cell-specific transcription factors (TFs). In the present study, using adenoviruses, the mouse TFs, pancreatic and duodenal homeobox 1 (pdx1), V-maf musculoaponeurotic fibrosarcoma oncogene homolog A (mafa) and class B basic helix‑loop‑helix factor neurogenin 3 (ngn3), which are essential for pancreatic cell development, were introduced into HUMSCs to assess the expression of the pancreatic genes, glucagon, pdx1 and nk2 homeobox 2 (nkx2.2). When pdx1, mafa and ngn3 were cotransduced into HUMSCs, the expression of glucagon increased by 21‑fold at days 3 and 7 following transduction, while the endogenous pdx1 gene expression was increased by 15‑fold at day 3 and decreased by 70% at day 7. When mafa and ngn3 were cotransduced into HUMSCs, there was a 5‑fold increase in pdx1 gene expression at day 7, but no activation was observed at day 3. When mafa alone was introduced into HUMSCs, the pdx1 gene expression was elevated by 6‑fold at day 3 and decreased by 3‑fold at day 7. Transduction of ngn3 alone into HUMSCs induced nkx2.2 gene expression at day 3 but the expression levels were decreased at day 7. However, when pdx1 and ngn3 were cotransduced into HUMSCs, the expression levels of glucagon, pdx1 and nks2.2 were all lower than those observed with pdx1 or ngn3 transduction alone. These results suggested that the transduction of pdx1, mafa and ngn3 genes into HUMSCs induced the expression of the pancreatic genes, glucagon, pdx‑1 and nkx2.2, and that the expression was time dependent. In addition, different combinations of the TFs may demonstrate synergistic or antagonistic effects. This data may be beneficial for guiding future studies obtaining mature pancreatic endocrine cells from HUMSCs.
Collapse
Affiliation(s)
- Hongwu Wang
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Reconstituting pancreas development from purified progenitor cells reveals genes essential for islet differentiation. Proc Natl Acad Sci U S A 2013; 110:12691-6. [PMID: 23852729 DOI: 10.1073/pnas.1304507110] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Developmental biology is challenged to reveal the function of numerous candidate genes implicated by recent genome-scale studies as regulators of organ development and diseases. Recapitulating organogenesis from purified progenitor cells that can be genetically manipulated would provide powerful opportunities to dissect such gene functions. Here we describe systems for reconstructing pancreas development, including islet β-cell and α-cell differentiation, from single fetal progenitor cells. A strict requirement for native genetic regulators of in vivo pancreas development, such as Ngn3, Arx, and Pax4, revealed the authenticity of differentiation programs in vitro. Efficient genetic screens permitted by this system revealed that Prdm16 is required for pancreatic islet development in vivo. Discovering the function of genes regulating pancreas development with our system should enrich strategies for regenerating islets for treating diabetes mellitus.
Collapse
|
80
|
Flasse LC, Pirson JL, Stern DG, Von Berg V, Manfroid I, Peers B, Voz ML. Ascl1b and Neurod1, instead of Neurog3, control pancreatic endocrine cell fate in zebrafish. BMC Biol 2013; 11:78. [PMID: 23835295 PMCID: PMC3726459 DOI: 10.1186/1741-7007-11-78] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 06/28/2013] [Indexed: 12/15/2022] Open
Abstract
Background NEUROG3 is a key regulator of pancreatic endocrine cell differentiation in mouse, essential for the generation of all mature hormone producing cells. It is repressed by Notch signaling that prevents pancreatic cell differentiation by maintaining precursors in an undifferentiated state. Results We show that, in zebrafish, neurog3 is not expressed in the pancreas and null neurog3 mutant embryos do not display any apparent endocrine defects. The control of endocrine cell fate is instead fulfilled by two basic helix-loop-helix factors, Ascl1b and Neurod1, that are both repressed by Notch signaling. ascl1b is transiently expressed in the mid-trunk endoderm just after gastrulation and is required for the generation of the first pancreatic endocrine precursor cells. Neurod1 is expressed afterwards in the pancreatic anlagen and pursues the endocrine cell differentiation program initiated by Ascl1b. Their complementary role in endocrine differentiation of the dorsal bud is demonstrated by the loss of all hormone-secreting cells following their simultaneous inactivation. This defect is due to a blockage of the initiation of endocrine cell differentiation. Conclusions This study demonstrates that NEUROG3 is not the unique pancreatic endocrine cell fate determinant in vertebrates. A general survey of endocrine cell fate determinants in the whole digestive system among vertebrates indicates that they all belong to the ARP/ASCL family but not necessarily to the Neurog3 subfamily. The identity of the ARP/ASCL factor involved depends not only on the organ but also on the species. One could, therefore, consider differentiating stem cells into insulin-producing cells without the involvement of NEUROG3 but via another ARP/ASCL factor.
Collapse
Affiliation(s)
- Lydie C Flasse
- Laboratory of zebrafish development and disease models, University of Liege (ULg), Liege 4000, Belgium
| | | | | | | | | | | | | |
Collapse
|
81
|
Liao X, Xue H, Wang YC, Nazor KL, Guo S, Trivedi N, Peterson SE, Liu Y, Loring JF, Laurent LC. Matched miRNA and mRNA signatures from an hESC-based in vitro model of pancreatic differentiation reveal novel regulatory interactions. J Cell Sci 2013; 126:3848-61. [PMID: 23813959 DOI: 10.1242/jcs.123570] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The differentiation of human pluripotent stem cells (hPSCs) to insulin-expressing beta islet-like cells is a promising in vitro model system for studying the molecular signaling pathways underlying beta cell differentiation, as well as a potential source of cells for the treatment of type 1 diabetes. MicroRNAs (miRNAs) are a class of small non-coding RNAs that regulate many biological processes, including cellular differentiation. We studied the miRNA and mRNA expression profiles of hPSCs at five stages of in vitro differentiation along the pancreatic beta cell lineage (definitive endoderm, primitive gut tube, posterior foregut, pancreatic progenitor and hormone-expressing endocrine cells) in the context of samples of primary human fetal pancreas and purified adult islet cells using microarray analysis. Bioinformatic analysis of the resulting data identified a unique miRNA signature in differentiated beta islet cells, and predicted the effects of key miRNAs on mRNA expression. Many of the predicted miRNA-mRNA interactions involved mRNAs known to play key roles in the epithelial-mesenchymal transition process and pancreatic differentiation. We validated a subset of the predictions using qRT-PCR, luciferase reporter assays and western blotting, including the known interaction between miR-200 and ZEB2 (involved in epithelial-mesenchymal transition) and the novel interaction between miR-200 and SOX17 (a key transcription factor in specification of definitive endoderm). In addition, we found that miR-30d and let-7e, two miRNAs induced during differentiation, regulated the expression of RFX6, a transcription factor that directs pancreatic islet formation. These findings suggest that precise control of target mRNA expression by miRNAs ensures proper lineage specification during pancreatic development.
Collapse
Affiliation(s)
- Xiaoyan Liao
- Department of Reproductive Medicine, University of California, San Diego, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Paschaki M, Schneider C, Rhinn M, Thibault-Carpentier C, Dembélé D, Niederreither K, Dollé P. Transcriptomic analysis of murine embryos lacking endogenous retinoic acid signaling. PLoS One 2013; 8:e62274. [PMID: 23638021 PMCID: PMC3634737 DOI: 10.1371/journal.pone.0062274] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 03/19/2013] [Indexed: 11/30/2022] Open
Abstract
Retinoic acid (RA), an active derivative of the liposoluble vitamin A (retinol), acts as an important signaling molecule during embryonic development, regulating phenomenons as diverse as anterior-posterior axial patterning, forebrain and optic vesicle development, specification of hindbrain rhombomeres, pharyngeal arches and second heart field, somitogenesis, and differentiation of spinal cord neurons. This small molecule directly triggers gene activation by binding to nuclear receptors (RARs), switching them from potential repressors to transcriptional activators. The repertoire of RA-regulated genes in embryonic tissues is poorly characterized. We performed a comparative analysis of the transcriptomes of murine wild-type and Retinaldehyde Dehydrogenase 2 null-mutant (Raldh2−/−) embryos — unable to synthesize RA from maternally-derived retinol — using Affymetrix DNA microarrays. Transcriptomic changes were analyzed in two embryonic regions: anterior tissues including forebrain and optic vesicle, and posterior (trunk) tissues, at early stages preceding the appearance of overt phenotypic abnormalities. Several genes expected to be downregulated under RA deficiency appeared in the transcriptome data (e.g. Emx2, Foxg1 anteriorly, Cdx1, Hoxa1, Rarb posteriorly), whereas reverse-transcriptase-PCR and in situ hybridization performed for additional selected genes validated the changes identified through microarray analysis. Altogether, the affected genes belonged to numerous molecular pathways and cellular/organismal functions, demonstrating the pleiotropic nature of RA-dependent events. In both tissue samples, genes upregulated were more numerous than those downregulated, probably due to feedback regulatory loops. Bioinformatic analyses highlighted groups (clusters) of genes displaying similar behaviors in mutant tissues, and biological functions most significantly affected (e.g. mTOR, VEGF, ILK signaling in forebrain tissues; pyrimidine and purine metabolism, calcium signaling, one carbon metabolism in posterior tissues). Overall, these data give an overview of the gene expression changes resulting from embryonic RA deficiency, and provide new candidate genes and pathways that may help understanding retinoid-dependent molecular events.
Collapse
Affiliation(s)
- Marie Paschaki
- Developmental Biology and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique (Unité Mixte de Recherche 7104), Institut National de la Santé et de la Recherche Médicale (Unité 964), Université de Strasbourg, Illkirch-Strasbourg, France
| | - Carole Schneider
- Developmental Biology and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique (Unité Mixte de Recherche 7104), Institut National de la Santé et de la Recherche Médicale (Unité 964), Université de Strasbourg, Illkirch-Strasbourg, France
| | - Muriel Rhinn
- Developmental Biology and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique (Unité Mixte de Recherche 7104), Institut National de la Santé et de la Recherche Médicale (Unité 964), Université de Strasbourg, Illkirch-Strasbourg, France
| | - Christelle Thibault-Carpentier
- Biochips platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique (Unité Mixte de Recherche 7104), Institut National de la Santé et de la Recherche Médicale (Unité 964), Université de Strasbourg, Illkirch-Strasbourg, France
| | - Doulaye Dembélé
- Biochips platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique (Unité Mixte de Recherche 7104), Institut National de la Santé et de la Recherche Médicale (Unité 964), Université de Strasbourg, Illkirch-Strasbourg, France
| | - Karen Niederreither
- Developmental Biology and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique (Unité Mixte de Recherche 7104), Institut National de la Santé et de la Recherche Médicale (Unité 964), Université de Strasbourg, Illkirch-Strasbourg, France
| | - Pascal Dollé
- Developmental Biology and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique (Unité Mixte de Recherche 7104), Institut National de la Santé et de la Recherche Médicale (Unité 964), Université de Strasbourg, Illkirch-Strasbourg, France
- * E-mail:
| |
Collapse
|
83
|
Flasse LC, Stern DG, Pirson JL, Manfroid I, Peers B, Voz ML. The bHLH transcription factor Ascl1a is essential for the specification of the intestinal secretory cells and mediates Notch signaling in the zebrafish intestine. Dev Biol 2013; 376:187-97. [PMID: 23352790 DOI: 10.1016/j.ydbio.2013.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 01/09/2013] [Accepted: 01/11/2013] [Indexed: 11/24/2022]
Abstract
Notch signaling has a fundamental role in stem cell maintenance and in cell fate choice in the intestine of different species. Canonically, Notch signaling represses the expression of transcription factors of the achaete-scute like (ASCL) or atonal related protein (ARP) families. Identifying the ARP/ASCL genes expressed in the gastrointestinal tract is essential to build the regulatory cascade controlling the differentiation of gastrointestinal progenitors into the different intestinal cell types. The expression of the ARP/ASCL factors was analyzed in zebrafish to identify, among all the ARP/ASCL factors found in the zebrafish genome, those expressed in the gastrointestinal tract. ascl1a was found to be the earliest factor detected in the intestine. Loss-of-function analyses using the pia/ascl1a mutant, revealed that ascl1a is crucial for the differentiation of all secretory cells. Furthermore, we identify a battery of transcription factors expressed during secretory cell differentiation and downstream of ascl1a. Finally, we show that the repression of secretory cell fate by Notch signaling is mediated by the inhibition of ascl1a expression. In conclusion, this work identifies Ascl1a as a key regulator of the secretory cell lineage in the zebrafish intestine, playing the same role as Atoh1 in the mouse intestine. This highlights the diversity in the ARP/ASCL family members acting as cell fate determinants downstream from Notch signaling.
Collapse
Affiliation(s)
- Lydie C Flasse
- Unit of Molecular Biology and Genetic Engineering, Giga-Research, University of Liège, 1 avenue de l'Hôpital B34, B-4000 Sart-Tilman (Liège), Belgium
| | | | | | | | | | | |
Collapse
|
84
|
Suzuki K, Harada N, Yamane S, Nakamura Y, Sasaki K, Nasteska D, Joo E, Shibue K, Harada T, Hamasaki A, Toyoda K, Nagashima K, Inagaki N. Transcriptional regulatory factor X6 (Rfx6) increases gastric inhibitory polypeptide (GIP) expression in enteroendocrine K-cells and is involved in GIP hypersecretion in high fat diet-induced obesity. J Biol Chem 2013; 288:1929-38. [PMID: 23192339 PMCID: PMC3548501 DOI: 10.1074/jbc.m112.423137] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 11/28/2012] [Indexed: 11/06/2022] Open
Abstract
Gastric inhibitory polypeptide (GIP) is an incretin released from enteroendocrine K-cells in response to nutrient ingestion. GIP potentiates glucose-stimulated insulin secretion and induces energy accumulation into adipose tissue, resulting in obesity. Plasma GIP levels are reported to be increased in the obese state. However, the molecular mechanisms of GIP secretion and high fat diet (HFD)-induced GIP hypersecretion remain unclear, primarily due to difficulties in separating K-cells from other intestinal epithelial cells in vivo. In this study, GIP-GFP knock-in mice that enable us to visualize K-cells by enhanced GFP were established. Microarray analysis of isolated K-cells from these mice revealed that transcriptional regulatory factor X6 (Rfx6) is expressed exclusively in K-cells. In vitro experiments using the mouse intestinal cell line STC-1 showed that knockdown of Rfx6 decreased mRNA expression, cellular content, and secretion of GIP. Rfx6 bound to the region in the gip promoter that regulates gip promoter activity, and overexpression of Rfx6 increased GIP mRNA expression. HFD induced obesity and GIP hypersecretion in GIP-GFP heterozygous mice in vivo. Immunohistochemical and flow cytometry analysis showed no significant difference in K-cell number between control fat diet-fed (CFD) and HFD-fed mice. However, GIP content in the upper small intestine and GIP mRNA expression in K-cells were significantly increased in HFD-fed mice compared with those in CFD-fed mice. Furthermore, expression levels of Rfx6 mRNA were increased in K-cells of HFD-fed mice. These results suggest that Rfx6 increases GIP expression and content in K-cells and is involved in GIP hypersecretion in HFD-induced obesity.
Collapse
Affiliation(s)
- Kazuyo Suzuki
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Norio Harada
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shunsuke Yamane
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiko Nakamura
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazuki Sasaki
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Daniela Nasteska
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Erina Joo
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kimitaka Shibue
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takanari Harada
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akihiro Hamasaki
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kentaro Toyoda
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazuaki Nagashima
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Nobuya Inagaki
- From the Department of Diabetes and Clinical Nutrition, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
85
|
Singh H, Farouk M, Bose BB, Singh P. Novel genes underlying beta cell survival in metabolic stress. Bioinformation 2013; 9:37-41. [PMID: 23390342 PMCID: PMC3563414 DOI: 10.6026/97320630009037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 12/26/2012] [Indexed: 02/07/2023] Open
Abstract
Relative insulin deficiency, in response to increased metabolic demand (obesity, genetic insulin resistance, pregnancy and aging) lead to Type2 diabetes. Susceptibility of the type 2 diabetes has a genetic basis, as a subset of people with risk factors (obesity, Insulin Resistance, pregnancy), develop Type2 Diabetes. We aimed to identify 'cluster' of overexpressed genes, underlying increased beta cell survival in diabetes resistant C57BL/6J ob/ob mice (compared to diabetes susceptible BTBR ob/ob mice). We used 'consensus' overexpression status to identify 'cluster' of 11 genes consisting of Aldh18a1, Rfc4, Dynlt3, Prom1, H13, Psen1, Ssr4, Dad1, Anpep, Fam111a and Plk1. Information (biological processes, molecular functions, cellular components, protein-protein interactions/associations, gene deletion/knockout/inhibition studies) of all the genes in 'cluster' were collected by text mining using different literature search tools, gene information databases and protein-protein interaction databases. Beta cell specific function of these genes were also inferred using meta analysis tool of Beta Cell Biology Consortium, by studying the expression pattern of these genes in microarray studies related to beta-cell stimulation/injury, pancreas development and growth and cell differentiation. In the 'clusters', 6 genes (Dad1, Psen1, Ssr4, Rfc4, H13, Plk1) have a role in cell survival. Only Psen1 was previously identified to have role in successful beta cell compensation. We advocate these genes to be potentially involved in successful beta cell compensation and prevent T2D in humans, by conferring protection against diabetogenic insults.
Collapse
Affiliation(s)
- Himadri Singh
- Sevayan Medical and Research Centre, Dr BG Bose Lane, Munger, 811201, India
| | - Mohammed Farouk
- Institute of Liver Disease and Transplantation, Global Hospitals, Chennai, India
| | - Barish Baran Bose
- Sevayan Medical and Research Centre, Dr BG Bose Lane, Munger, 811201, India
| | - Prabhakar Singh
- Sevayan Medical and Research Centre, Dr BG Bose Lane, Munger, 811201, India
| |
Collapse
|
86
|
Mansouri A. Development and regeneration in the endocrine pancreas. ISRN ENDOCRINOLOGY 2012; 2012:640956. [PMID: 23326678 PMCID: PMC3544272 DOI: 10.5402/2012/640956] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 12/10/2012] [Indexed: 12/16/2022]
Abstract
The pancreas is composed of two compartments that deliver digestive enzymes and endocrine hormones to control the blood sugar level. The endocrine pancreas consists of functional units organized into cell clusters called islets of Langerhans where insulin-producing cells are found in the core and surrounded by glucagon-, somatostatin-, pancreatic polypeptide-, and ghrelin-producing cells. Diabetes is a devastating disease provoked by the depletion or malfunction of insulin-producing beta-cells in the endocrine pancreas. The side effects of diabetes are multiple, including cardiovascular, neuropathological, and kidney diseases. The analyses of transgenic and knockout mice gave major insights into the molecular mechanisms controlling endocrine pancreas genesis. Moreover, the study of animal models of pancreas injury revealed that the pancreas has the propensity to undergo regeneration and opened new avenues to develop novel therapeutic approaches for the treatment of diabetes. Thus, beside self-replication of preexisting insulin-producing cells, several potential cell sources in the adult pancreas were suggested to contribute to beta-cell regeneration, including acinar, intraislet, and duct epithelia. However, regeneration in the adult endocrine pancreas is still under controversial debate.
Collapse
Affiliation(s)
- Ahmed Mansouri
- Research Group Molecular Cell Differentiation, Department Molecular Cell Biology, Max-Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany ; Department of Clinical Neurophysiology, University of Goettingen, Robert-Koch-Strasse 40, 37075 Goettingen, Germany
| |
Collapse
|
87
|
Sui J, Mehta M, Shi B, Morahan G, Jiang FX. Directed differentiation of embryonic stem cells allows exploration of novel transcription factor genes for pancreas development. Stem Cell Rev Rep 2012; 8:803-12. [PMID: 22278131 DOI: 10.1007/s12015-011-9346-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Embryonic stem cells (ESCs) have been promised as a renewable source for regenerative medicine, including providing a replacement therapy in type 1 diabetes. However, they have not yet been differentiated into functional insulin-secreting β cells. This is due partially to the knowledge gap regarding the transcription factors (TFs) required for pancreas development. We hypothesize that, if directed differentiation in vitro recapitulates the developmental process in vivo, ESCs provide a powerful model to discover novel pancreatic TF genes. Guided by knowledge of their normal development and using RT-PCR and immunochemical analyses, we have established protocols for directed differentiation of mouse ESCs into pancreatic progenitors. Microarray analyses of these differentiating ESC cells at days 0, 4, 8 and 15 confirmed their sequential differentiation. By day 15, we found up-regulation of a group of pancreatic progenitor marker genes including Pdx1, Ptf1a, Nkx6.1, Pax4 and Pax6. Consistently, Pdx1-immunoreactive cells were detected on day 15. Most of these Pdx1(+) cells also expressed Nkx6.1. Bioinformatic analyses of sequential datasets allowed identification of over 20 novel TF genes potentially important for pancreas development. The dynamic expression of representative known and novel genes was confirmed by quantitative real time RT-PCR analysis. This strategy may be modified to study novel regulatory molecules for development of other tissue and organ systems.
Collapse
Affiliation(s)
- Jing Sui
- Centre for Diabetes Research, The Western Australian Institute for Medical Research, University of Western Australia, 50 Murray St (Rear), Perth, WA 6000, Australia
| | | | | | | | | |
Collapse
|
88
|
Freeman TC, Ivens A, Baillie JK, Beraldi D, Barnett MW, Dorward D, Downing A, Fairbairn L, Kapetanovic R, Raza S, Tomoiu A, Alberio R, Wu C, Su AI, Summers KM, Tuggle CK, Archibald AL, Hume DA. A gene expression atlas of the domestic pig. BMC Biol 2012; 10:90. [PMID: 23153189 PMCID: PMC3814290 DOI: 10.1186/1741-7007-10-90] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 10/23/2012] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND This work describes the first genome-wide analysis of the transcriptional landscape of the pig. A new porcine Affymetrix expression array was designed in order to provide comprehensive coverage of the known pig transcriptome. The new array was used to generate a genome-wide expression atlas of pig tissues derived from 62 tissue/cell types. These data were subjected to network correlation analysis and clustering. RESULTS The analysis presented here provides a detailed functional clustering of the pig transcriptome where transcripts are grouped according to their expression pattern, so one can infer the function of an uncharacterized gene from the company it keeps and the locations in which it is expressed. We describe the overall transcriptional signatures present in the tissue atlas, where possible assigning those signatures to specific cell populations or pathways. In particular, we discuss the expression signatures associated with the gastrointestinal tract, an organ that was sampled at 15 sites along its length and whose biology in the pig is similar to human. We identify sets of genes that define specialized cellular compartments and region-specific digestive functions. Finally, we performed a network analysis of the transcription factors expressed in the gastrointestinal tract and demonstrate how they sub-divide into functional groups that may control cellular gastrointestinal development. CONCLUSIONS As an important livestock animal with a physiology that is more similar than mouse to man, we provide a major new resource for understanding gene expression with respect to the known physiology of mammalian tissues and cells. The data and analyses are available on the websites http://biogps.org and http://www.macrophages.com/pig-atlas.
Collapse
Affiliation(s)
- Tom C Freeman
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - Alasdair Ivens
- Fios Genomics Ltd, ETTC, King's Buildings, Edinburgh EH9 3JL UK,Centre for Immunity, Infection and Evolution, University of Edinburgh Ashworth
Labs, King's Buildings, West Mains Road, Edinburgh EH9 3JT
| | - J Kenneth Baillie
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - Dario Beraldi
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK,Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson
way, Cambridge, CB2 0RE, UK
| | - Mark W Barnett
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - David Dorward
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - Alison Downing
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - Lynsey Fairbairn
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - Ronan Kapetanovic
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - Sobia Raza
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - Andru Tomoiu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - Ramiro Alberio
- Division of Animal Sciences, School of Biosciences, University of Nottingham,
Sutton Bonington, Leicestershire LE12 5RD UK
| | - Chunlei Wu
- Department of Molecular and Experimental Medicine, The Scripps Research Institute,
MEM-216, 10550 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Andrew I Su
- Department of Molecular and Experimental Medicine, The Scripps Research Institute,
MEM-216, 10550 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Kim M Summers
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | | | - Alan L Archibald
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| | - David A Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of
Edinburgh, Easter Bush, EH25 9PS, UK
| |
Collapse
|
89
|
Metukuri MR, Zhang P, Basantani MK, Chin C, Stamateris RE, Alonso LC, Takane KK, Gramignoli R, Strom SC, O’Doherty RM, Stewart AF, Vasavada RC, Garcia-Ocaña A, Scott DK. ChREBP mediates glucose-stimulated pancreatic β-cell proliferation. Diabetes 2012; 61:2004-15. [PMID: 22586588 PMCID: PMC3402328 DOI: 10.2337/db11-0802] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Glucose stimulates rodent and human β-cell replication, but the intracellular signaling mechanisms are poorly understood. Carbohydrate response element-binding protein (ChREBP) is a lipogenic glucose-sensing transcription factor with unknown functions in pancreatic β-cells. We tested the hypothesis that ChREBP is required for glucose-stimulated β-cell proliferation. The relative expression of ChREBP was determined in liver and β-cells using quantitative RT-PCR (qRT-PCR), immunoblotting, and immunohistochemistry. Loss- and gain-of-function studies were performed using small interfering RNA and genetic deletion of ChREBP and adenoviral overexpression of ChREBP in rodent and human β-cells. Proliferation was measured by 5-bromo-2'-deoxyuridine incorporation, [(3)H]thymidine incorporation, and fluorescence-activated cell sorter analysis. In addition, the expression of cell cycle regulatory genes was measured by qRT-PCR and immunoblotting. ChREBP expression was comparable with liver in mouse pancreata and in rat and human islets. Depletion of ChREBP decreased glucose-stimulated proliferation in β-cells isolated from ChREBP(-/-) mice, in INS-1-derived 832/13 cells, and in primary rat and human β-cells. Furthermore, depletion of ChREBP decreased the glucose-stimulated expression of cell cycle accelerators. Overexpression of ChREBP amplified glucose-stimulated proliferation in rat and human β-cells, with concomitant increases in cyclin gene expression. In conclusion, ChREBP mediates glucose-stimulated proliferation in pancreatic β-cells.
Collapse
Affiliation(s)
- Mallikarjuna R. Metukuri
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Pili Zhang
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mahesh K. Basantani
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Connie Chin
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rachel E. Stamateris
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Laura C. Alonso
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Karen K. Takane
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Roberto Gramignoli
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Stephen C. Strom
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert M. O’Doherty
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew F. Stewart
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rupangi C. Vasavada
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adolfo Garcia-Ocaña
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Donald K. Scott
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
- Corresponding author: Donald K. Scott,
| |
Collapse
|
90
|
Rodríguez-Trejo A, Ortiz-López MG, Zambrano E, Granados-Silvestre MDLÁ, Méndez C, Blondeau B, Bréant B, Nathanielsz PW, Menjivar M. Developmental programming of neonatal pancreatic β-cells by a maternal low-protein diet in rats involves a switch from proliferation to differentiation. Am J Physiol Endocrinol Metab 2012; 302:E1431-9. [PMID: 22436693 PMCID: PMC3378070 DOI: 10.1152/ajpendo.00619.2011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Maternal low-protein diets (LP) impair pancreatic β-cell development, resulting in later-life failure and susceptibility to type 2 diabetes (T2D). We hypothesized that intrauterine and/or postnatal developmental programming seen in this situation involve altered β-cell structure and relative time course of expression of genes critical to β-cell differentiation and growth. Pregnant Wistar rats were fed either control (C) 20% or restricted (R) 6% protein diets during pregnancy (1st letter) and/or lactation (2nd letter) in four groups: CC, RR, RC, and CR. At postnatal days 7 and 21, we measured male offspring β-cell fraction, mass, proliferation, aggregate number, and size as well as mRNA level for 13 key genes regulating β-cell development and function in isolated islets. Compared with CC, pre- and postnatal LP (RR) decreased β-cell fraction, mass, proliferation, aggregate size, and number and increased Hnf1a, Hnf4a, Pdx1, Isl1, Rfx6, and Slc2a2 mRNA levels. LP only in pregnancy (RC) also decreased β-cell fraction, mass, proliferation, aggregate size, and number and increased Hnf1a, Hnf4a, Pdx1, Rfx6, and Ins mRNA levels. Postnatal LP offspring (CR) showed decreased β-cell mass but increased β-cell fraction, aggregate number, and Hnf1a, Hnf4a, Rfx6, and Slc2a2 mRNA levels. We conclude that LP in pregnancy sets the trajectory of postnatal β-cell growth and differentiation, whereas LP in lactation has smaller effects. We propose that LP promotes differentiation through upregulation of transcription factors that stimulate differentiation at the expense of proliferation. This results in a decreased β-cell reserve, which can contribute to later-life predisposition to T2D.
Collapse
Affiliation(s)
- Adriana Rodríguez-Trejo
- Universidad Nacional Autónoma de México. Av. Universidad 3000, Facultad de Química, Mexico City, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Benitez CM, Goodyer WR, Kim SK. Deconstructing pancreas developmental biology. Cold Spring Harb Perspect Biol 2012; 4:cshperspect.a012401. [PMID: 22587935 DOI: 10.1101/cshperspect.a012401] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The relentless nature and increasing prevalence of human pancreatic diseases, in particular, diabetes mellitus and adenocarcinoma, has motivated further understanding of pancreas organogenesis. The pancreas is a multifunctional organ whose epithelial cells govern a diversity of physiologically vital endocrine and exocrine functions. The mechanisms governing the birth, differentiation, morphogenesis, growth, maturation, and maintenance of the endocrine and exocrine components in the pancreas have been discovered recently with increasing tempo. This includes recent studies unveiling mechanisms permitting unexpected flexibility in the developmental potential of immature and mature pancreatic cell subsets, including the ability to interconvert fates. In this article, we describe how classical cell biology, genetic analysis, lineage tracing, and embryological investigations are being complemented by powerful modern methods including epigenetic analysis, time-lapse imaging, and flow cytometry-based cell purification to dissect fundamental processes of pancreas development.
Collapse
Affiliation(s)
- Cecil M Benitez
- Department of Developmental Biology, Stanford University School of Medicine, California 94305-5329, USA
| | | | | |
Collapse
|
92
|
Ninov N, Borius M, Stainier DYR. Different levels of Notch signaling regulate quiescence, renewal and differentiation in pancreatic endocrine progenitors. Development 2012; 139:1557-67. [PMID: 22492351 DOI: 10.1242/dev.076000] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Genetic studies have implicated Notch signaling in the maintenance of pancreatic progenitors. However, how Notch signaling regulates the quiescent, proliferative or differentiation behaviors of pancreatic progenitors at the single-cell level remains unclear. Here, using single-cell genetic analyses and a new transgenic system that allows dynamic assessment of Notch signaling, we address how discrete levels of Notch signaling regulate the behavior of endocrine progenitors in the zebrafish intrapancreatic duct. We find that these progenitors experience different levels of Notch signaling, which in turn regulate distinct cellular outcomes. High levels of Notch signaling induce quiescence, whereas lower levels promote progenitor amplification. The sustained downregulation of Notch signaling triggers a multistep process that includes cell cycle entry and progenitor amplification prior to endocrine differentiation. Importantly, progenitor amplification and differentiation can be uncoupled by modulating the duration and/or extent of Notch signaling downregulation, indicating that these processes are triggered by distinct levels of Notch signaling. These data show that different levels of Notch signaling drive distinct behaviors in a progenitor population.
Collapse
Affiliation(s)
- Nikolay Ninov
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics and Human Genetics, Diabetes Center, and Liver Center, University of California, San Francisco, San Francisco, CA 94158, USA.
| | | | | |
Collapse
|
93
|
Metzger DE, Gasperowicz M, Otto F, Cross JC, Gradwohl G, Zaret KS. The transcriptional co-repressor Grg3/Tle3 promotes pancreatic endocrine progenitor delamination and β-cell differentiation. Development 2012; 139:1447-56. [PMID: 22434868 DOI: 10.1242/dev.072892] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Pancreatic β-cells arise from Ngn3(+) endocrine progenitors within the trunk epithelium of the embryonic pancreas. The emergence of endocrine cells requires E-cadherin downregulation, but the crucial steps that elicit such are not clear, yet probably important for ultimately being able to efficiently generate β-cells de novo from stem cells. Grg3 (groucho-related gene 3, also known as Tle3), encodes a member of the Groucho/TLE family of co-repressors and its function in various cell contexts is mediated by recruitment to target genes by different transcription factors. Grg proteins broadly regulate the progression of progenitor cells to differentiated cell types, but specific developmental mechanisms have not been clear. We find that Grg3 is expressed in most β-cells and a subset of other endocrine cell types in the pancreas. Grg3 is highly expressed in Ngn3(+) endocrine progenitor descendants just after transient Ngn3 expression. Grg3-null embryos die at E14.5, which is associated with placental defects, so we explanted E12.5 pancreata to allow endocrine differentiation to occur in culture. Grg3 knockout explants displayed a drastic decrease in the differentiation of all endocrine cell types owing to defects in the delamination of early endocrine progenitors from the trunk epithelium. We find that Grg3 normally suppresses E-cadherin gene expression, thereby allowing delamination of endocrine cells from the trunk epithelium and revealing how this transcriptional co-repressor modulates this crucial step of β-cell development.
Collapse
Affiliation(s)
- David E Metzger
- Institute for Regenerative Medicine, Institute for Diabetes Obesity and Metabolism, Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine,1056 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA.
| | | | | | | | | | | |
Collapse
|
94
|
Oropeza D, Horb M. Transient expression of Ngn3 in Xenopus endoderm promotes early and ectopic development of pancreatic beta and delta cells. Genesis 2012; 50:271-85. [PMID: 22121111 DOI: 10.1002/dvg.20828] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 11/18/2011] [Accepted: 11/21/2011] [Indexed: 01/18/2023]
Abstract
Promoting ectopic development of pancreatic beta cells from other cell types is one of the strategies being pursued for the treatment of diabetes. To achieve this, a detailed outline of the molecular lineage that operates in pancreatic progenitor cells to generate beta cells over other endocrine cell types is necessary. Here, we demonstrate that early transient expression of the endocrine progenitor bHLH protein Neurogenin 3 (Ngn3) favors the promotion of pancreatic beta and delta cell fates over an alpha cell fate, while later transient expression promotes ectopic development of all three endocrine cell fates. We found that short-term activation of Ngn3 in Xenopus laevis endoderm just after gastrulation was sufficient to promote both early and ectopic development of beta and delta cells. By examining gene expression changes 4 h after Ngn3 activation we identified several new downstream targets of Ngn3. We show that several of these are required for the promotion of ectopic beta cells by Ngn3 as well as for normal beta cell development. These results provide new detail regarding the Ngn3 transcriptional network operating in endocrine progenitor cells to specify a beta cell phenotype and should help define new approaches to promote ectopic development of beta cells for diabetes therapy.
Collapse
Affiliation(s)
- Daniel Oropeza
- Laboratory of Molecular Organogenesis, Institut de recherches cliniques de Montréal, Montreal, QC, Canada
| | | |
Collapse
|
95
|
Bramswig NC, Kaestner KH. Organogenesis and functional genomics of the endocrine pancreas. Cell Mol Life Sci 2012; 69:2109-23. [PMID: 22241333 DOI: 10.1007/s00018-011-0915-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 12/04/2011] [Accepted: 12/29/2011] [Indexed: 02/06/2023]
Abstract
Functional genomics, the analysis of the wealth of data produced by genome-wide analyses of gene expression, protein-protein, and protein-DNA interactions, has revolutionized biomedical research. Our ability to determine global gene expression profiles, transcription factor-binding sites, and histone modification maps using microarray-based technologies and next-generation sequencing applications has greatly enhanced our understanding of gene regulatory networks and the molecular wiring diagrams of cells and tissues. The organogenesis of the endocrine pancreas involves numerous signaling events within the endoderm-derived pancreatic epithelium and the surrounding mesenchyme, as well as complex transcription factor networks. Detailed understanding of the differentiation process from foregut endoderm to mature endocrine cells has enabled the rational design of in vitro differentiation protocols that coax embryonic stem cells into β-like cells that might enable cell replacement therapy for diabetes in the future. In this review, we summarize the research studies that have utilized genomic tools to elucidate endocrine pancreatic organogenesis.
Collapse
Affiliation(s)
- Nuria C Bramswig
- Department of Genetics, Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
96
|
Hald J, Galbo T, Rescan C, Radzikowski L, Sprinkel AE, Heimberg H, Ahnfelt-Rønne J, Jensen J, Scharfmann R, Gradwohl G, Kaestner KH, Stoeckert C, Jensen JN, Madsen OD. Pancreatic islet and progenitor cell surface markers with cell sorting potential. Diabetologia 2012; 55:154-65. [PMID: 21947380 DOI: 10.1007/s00125-011-2295-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 07/12/2011] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS The aim of the study was to identify surface bio-markers and corresponding antibody tools that can be used for the imaging and immunoisolation of the pancreatic beta cell and its progenitors. This may prove essential to obtain therapeutic grade human beta cells via stem cell differentiation. METHODS Using bioinformatics-driven data mining, we generated a gene list encoding putative plasma membrane proteins specifically expressed at distinct stages of the developing pancreas and islet beta cells. In situ hybridisation and immunohistochemistry were used to further prioritise and identify candidates. RESULTS In the developing pancreas seizure related 6 homologue like (SEZ6L2), low density lipoprotein receptor-related protein 11 (LRP11), dispatched homologue 2 (Drosophila) (DISP2) and solute carrier family 30 (zinc transporter), member 8 (SLC30A8) were found to be expressed in early islet cells, whereas discoidin domain receptor tyrosine kinase 1 (DDR1) and delta/notch-like EGF repeat containing (DNER) were expressed in early pancreatic progenitors. The expression pattern of DDR1 overlaps with the early pancreatic and duodenal homeobox 1 (PDX1)⁺/NK6 homeobox 1 (NKX6-1)⁺ multipotent progenitor cells from embryonic day 11, whereas DNER expression in part overlaps with neurogenin 3 (NEUROG3)⁺ cells. In the adult pancreas SEZ6L2, LRP11, DISP2 and SLC30A8, but also FXYD domain containing ion transport regulator 2 (FXYD2), tetraspanin 7 (TSPAN7) and transmembrane protein 27 (TMEM27), retain an islet-specific expression, whereas DDR1 is undetectable. In contrast, DNER is expressed at low levels in peripheral mouse and human islet cells. Re-expression of DDR1 and upregulation of DNER is observed in duct-ligated pancreas. Antibodies to DNER and DISP2 have been successfully used in cell sorting. CONCLUSIONS/INTERPRETATION Extracellular epitopes of SEZ6L2, LRP11, DISP2, DDR1 and DNER have been identified as useful tags by applying specific antibodies to visualise pancreatic cell types at specific stages of development. Furthermore, antibodies recognising DISP2 and DNER are suitable for FACS-mediated cell purification.
Collapse
Affiliation(s)
- J Hald
- Department of Beta-Cell Regeneration, Hagedorn Research Institute, Niels Steensens Vej 1, 2820 Gentofte, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Chung MI, Peyrot SM, LeBoeuf S, Park TJ, McGary KL, Marcotte EM, Wallingford JB. RFX2 is broadly required for ciliogenesis during vertebrate development. Dev Biol 2011; 363:155-65. [PMID: 22227339 DOI: 10.1016/j.ydbio.2011.12.029] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Revised: 12/09/2011] [Accepted: 12/19/2011] [Indexed: 10/14/2022]
Abstract
In Caenorhabditis elegans, the RFX (Daf19) transcription factor is a major regulator of ciliogenesis, controlling the expression of the many essential genes required for making cilia. In vertebrates, however, seven RFX genes have been identified. Bioinformatic analysis suggests that Rfx2 is among the closest homologues of Daf19. We therefore hypothesize that Rfx2 broadly controls ciliogenesis during vertebrate development. Indeed, here we show that Rfx2 in Xenopus is expressed preferentially in ciliated tissues, including neural tube, gastrocoel roof plate, epidermal multi-ciliated cells, otic vesicles, and kidneys. Knockdown of Rfx2 results in cilia-defective embryonic phenotypes and fewer or truncated cilia are observed in Rfx2 morphants. These results indicate that Rfx2 is broadly required for ciliogenesis in vertebrates. Furthermore, we show that Rfx2 is essential for expression of several ciliogenic genes, including TTC25, which we show here is required for ciliogenesis, HH signaling, and left-right patterning.
Collapse
Affiliation(s)
- Mei-I Chung
- Section of Molecular Cell and Developmental Biology, University of Texas at Austin, Austin, TX 78712, USA
| | | | | | | | | | | | | |
Collapse
|
98
|
Gupta RK, Rosen ED, Spiegelman BM. Identifying novel transcriptional components controlling energy metabolism. Cell Metab 2011; 14:739-45. [PMID: 22152302 PMCID: PMC3240865 DOI: 10.1016/j.cmet.2011.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 10/06/2011] [Accepted: 11/03/2011] [Indexed: 02/07/2023]
Abstract
The investigation of metabolic regulation at the transcriptional level presents different challenges than those encountered in the study of other important problems like development or cancer. Levels of key components like glucose, insulin, and lipids can be modulated but rarely change in an all-or-none fashion, necessitating quantitative techniques that can be applied to multiple tissues and systems. This review examines recent advances in methods for studying transcriptional regulation, with special emphasis on metabolic science. We compare these methods for investigators trying to decide on the best approach for their particular physiological paradigm or model system.
Collapse
Affiliation(s)
- Rana K. Gupta
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Evan D. Rosen
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Bruce M. Spiegelman
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
99
|
Yang SL, Aw SS, Chang C, Korzh S, Korzh V, Peng J. Depletion of Bhmt elevates sonic hedgehog transcript level and increases β-cell number in zebrafish. Endocrinology 2011; 152:4706-17. [PMID: 21952238 DOI: 10.1210/en.2011-1306] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Betaine homocysteine S-methyltransferase (BHMT, EC 2.1.1.5) is a key enzyme in the methionine cycle and is highly expressed in the liver. Despite its important biochemical function, it is not known whether BHMT plays a role during organ development. In this report, we showed that early in development of zebrafish before endoderm organogenesis, bhmt is first expressed in the yolk syncytial layer and then after liver formation becomes a liver-enriched gene. By using the anti-bhmt morpholinos that deplete the Bhmt, we found that in morphant embryos, several endoderm-derived organs, including liver, exocrine pancreas, and intestine are hypoplastic. Strikingly, the number of β-cells in the pancreatic islet was increased rather than reduced in the morphant. Additional studies showed that Bhmt depletion elevates the sonic hedgehog (shh) transcript level in the morphant, whereas Bhmt-depletion in the Shh-deficient mutant syu failed to rescue the isletless phenotype. These molecular and genetic data strongly suggest that Shh functions downstream of Bhmt to promote β-cell development. Therefore, although there are still many intriguing questions to be answered, our finding may identify a novel function for Bhmt involving modulation of Shh signaling to control β-cell development.
Collapse
Affiliation(s)
- Shu-Lan Yang
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Proteos, Singapore
| | | | | | | | | | | |
Collapse
|
100
|
Delhanty PJD, van der Lely AJ. Ghrelin and glucose homeostasis. Peptides 2011; 32:2309-18. [PMID: 21396419 DOI: 10.1016/j.peptides.2011.03.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/01/2011] [Accepted: 03/01/2011] [Indexed: 12/25/2022]
Abstract
Ghrelin plays an important physiological role in modulating GH secretion, insulin secretion and glucose metabolism. Ghrelin has direct effects on pancreatic islet function. Also, ghrelin is part of a mechanism that integrates the physiological response to fasting. However, pharmacologic studies indicate the important obesogenic/diabetogenic properties of ghrelin. This is very likely of physiological relevance, deriving from a requirement to protect against seasonal periods of food scarcity by building energy reserves, predominantly in the form of fat. Available data indicate the potential of ghrelin blockade as a means to prevent its diabetogenic effects. Several studies indicate a negative correlation between ghrelin levels and the incidence of type 2 diabetes and insulin resistance. However, it is unclear if low ghrelin levels are a risk factor or a compensatory response. Direct antagonism of the receptor does not always have the desired effects, however, since it can cause increased body weight gain. Pharmacological suppression of the ghrelin/des-acyl ghrelin ratio by treatment with des-acyl ghrelin may also be a viable alternative approach which appears to improve insulin sensitivity. A promising recently developed approach appears to be through the blockade of GOAT activity, although the longer term effects of this treatment remain to be investigated.
Collapse
Affiliation(s)
- P J D Delhanty
- Department of Internal Medicine, Erasmus MC, 3000 CA Rotterdam, The Netherlands.
| | | |
Collapse
|