251
|
Soyer J, Flasse L, Raffelsberger W, Beucher A, Orvain C, Peers B, Ravassard P, Vermot J, Voz ML, Mellitzer G, Gradwohl G. Rfx6 is an Ngn3-dependent winged helix transcription factor required for pancreatic islet cell development. Development 2010; 137:203-12. [PMID: 20040487 DOI: 10.1242/dev.041673] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The transcription factor neurogenin 3 (Neurog3 or Ngn3) controls islet cell fate specification in multipotent pancreatic progenitor cells in the mouse embryo. However, our knowledge of the genetic programs implemented by Ngn3, which control generic and islet subtype-specific properties, is still fragmentary. Gene expression profiling in isolated Ngn3-positive progenitor cells resulted in the identification of the uncharacterized winged helix transcription factor Rfx6. Rfx6 is initially expressed broadly in the gut endoderm, notably in Pdx1-positive cells in the developing pancreatic buds, and then becomes progressively restricted to the endocrine lineage, suggesting a dual function in both endoderm development and islet cell differentiation. Rfx6 is found in postmitotic islet progenitor cells in the embryo and is maintained in all developing and adult islet cell types. Rfx6 is dependent on Ngn3 and acts upstream of or in parallel with NeuroD, Pax4 and Arx transcription factors during islet cell differentiation. In zebrafish, the Rfx6 ortholog is similarly found in progenitors and hormone expressing cells of the islet lineage. Loss-of-function studies in zebrafish revealed that rfx6 is required for the differentiation of glucagon-, ghrelin- and somatostatin-expressing cells, which, in the absence of rfx6, are blocked at the progenitor stage. By contrast, beta cells, whose number is only slightly reduced, were no longer clustered in a compact islet. These data unveil Rfx6 as a novel regulator of islet cell development.
Collapse
Affiliation(s)
- Josselin Soyer
- Institute of Genetics and Molecular and Cell Biology (IGBMC), Inserm U-964, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
252
|
Abstract
Over the last years, there has been great success in driving stem cells toward insulin-expressing cells. However, the protocols developed to date have some limitations, such as low reliability and low insulin production. The most successful protocols used for generation of insulin-producing cells from stem cells mimic in vitro pancreatic organogenesis by directing the stem cells through stages that resemble several pancreatic developmental stages. Islet cell fate is coordinated by a complex network of inductive signals and regulatory transcription factors that, in a combinatorial way, determine pancreatic organ specification, differentiation, growth, and lineage. Together, these signals and factors direct the progression from multipotent progenitor cells to mature pancreatic cells. Later in development and adult life, several of these factors also contribute to maintain the differentiated phenotype of islet cells. A detailed understanding of the processes that operate in the pancreas during embryogenesis will help us to develop a suitable source of cells for diabetes therapy. In this chapter, we will discuss the main transcription factors involved in pancreas specification and beta-cell formation.
Collapse
|
253
|
Vetere A, Li WC, Paroni F, Juhl K, Guo L, Nishimura W, Dai X, Bonner-Weir S, Sharma A. OVO homologue-like 1 (Ovol1) transcription factor: a novel target of neurogenin-3 in rodent pancreas. Diabetologia 2010; 53:115-22. [PMID: 19882138 PMCID: PMC3066144 DOI: 10.1007/s00125-009-1567-5] [Citation(s) in RCA: 4] [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] [Received: 04/30/2009] [Accepted: 09/14/2009] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS The basic helix-loop-helix transcription factor neurogenin-3 (NGN3) commits the fates of pancreatic progenitors to endocrine cell types, but knowledge of the mechanisms regulating the choice between proliferation and differentiation of these progenitors is limited. METHODS Using a chromatin immunoprecipitation cloning approach, we searched for direct targets of NGN3 and identified a zinc-finger transcription factor, OVO homologue-like 1 (OVOL1). Transactivation experiments were carried out to elucidate the functional role of NGN3 in Ovol1 gene expression. Embryonic and adult rodents pancreases were immunostained for OVOL1, Ki67 and NGN3. RESULTS We showed that NGN3 negatively regulates transcription of Ovol1 in an E-box-dependent fashion. The presence of either NGN3 or NEUROD1, but not MYOD, reduced endogenous Ovol1 mRNA. OVOL1 was detected in pancreatic tissue around embryonic day 15.5, after which OVOL1 levels dramatically increased. In embryonic pancreas, OVOL1 protein levels were low in NGN3(+) or Ki67(+) cells, but high in quiescent differentiated cells. OVOL1 presence was maintained in adult pancreas, where it was detected in islets, pancreatic ducts and some acinar cells. Additionally OVOL1 presence was lacking in proliferating ductules in regenerating pancreas and induced in cells as they began to acquire their differentiated phenotype. CONCLUSIONS/INTERPRETATION The timing of OVOL1 appearance in pancreas and its increased levels in differentiated cells suggest that OVOL1 promotes the transition of cells from a proliferating, less-differentiated state to a quiescent more-differentiated state. We conclude that OVOL1, a downstream target of NGN3, may play an important role in regulating the balance between proliferation and differentiation of pancreatic cells.
Collapse
Affiliation(s)
- A Vetere
- Section of Islet Transplantation and Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
254
|
Insulin-producing cells derived from stem/progenitor cells: therapeutic implications for diabetes mellitus. Med Mol Morphol 2009; 42:195-200. [DOI: 10.1007/s00795-009-0471-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 09/14/2009] [Indexed: 10/20/2022]
|
255
|
Notch signaling in pancreatic endocrine cell and diabetes. Biochem Biophys Res Commun 2009; 392:247-51. [PMID: 20035712 DOI: 10.1016/j.bbrc.2009.12.115] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 12/21/2009] [Indexed: 12/21/2022]
Abstract
Recent studies have improved our understanding of the physiological function of Notch signaling pathway and now there is compelling evidence demonstrating that Notch is a key regulator of embryonic development and tissue homeostasis. Although further extensive studies are necessary to illustrate the molecular mechanisms, new insights into the role of Notch signaling in pancreas development and diabetes have been achieved. Importantly, the ability to regulate Notch signaling intensity both positively and negatively may have therapeutic relevance for diabetes. Thus, this paper reviews the current knowledge of the roles of Notch signaling in the pancreatic endocrine cell system.
Collapse
|
256
|
|
257
|
Wang S, Yan J, Anderson DA, Xu Y, Kanal MC, Cao Z, Wright CVE, Gu G. Neurog3 gene dosage regulates allocation of endocrine and exocrine cell fates in the developing mouse pancreas. Dev Biol 2009; 339:26-37. [PMID: 20025861 DOI: 10.1016/j.ydbio.2009.12.009] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 12/05/2009] [Accepted: 12/07/2009] [Indexed: 10/20/2022]
Abstract
The basic helix-loop-helix transcription factor Neurog3 (Neurogenin3 or Ngn3) actively drives endodermal progenitor cells towards endocrine islet cell differentiation during embryogenesis. Here, we manipulate Neurog3 expression levels in endocrine progenitor cells without altering its expression pattern using heterozygosity and a hypomorph. Lowered Neurog3 gene dosage in the developing pancreatic epithelium reduces the overall production of endocrine islet cells without significantly affecting the proportions of various islet cell types that do form. A reduced Neurog3 production level in the endocrine-directed pancreatic progenitor population activates the expression of Neurog3 in an increased number of epithelial progenitors. Yet a significant number of these Neurog3+ cells detected in heterozygous and hypomorphic pancreata, possibly those that express low levels of Neurog3, move on to adopt pancreatic ductal or acinar fates. These data directly demonstrate that achieving high levels of Neurog3 expression is a critical step for endocrine commitment from multipotent pancreatic progenitors. These findings also suggest that a high level of Neurog3 expression could mediate lateral inhibition or other unknown feedback mechanisms to regulate the number of cells that initiate Neurog3 transcription and protein production. The control of Neurog3+ cell number and the Neurog3 threshold-dependent endocrine differentiation mechanism combine to select a specific proportion of pancreatic progenitor cells to adopt the islet cell fate.
Collapse
Affiliation(s)
- Sui Wang
- Program in Developmental Biology and Department of Cell and Developmental Biology, Center for Stem Cell Biology, Vanderbilt University Medical Center, 465 21st Avenue South, Rm 4128, Vanderbilt Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
258
|
Anderson KR, White P, Kaestner KH, Sussel L. Identification of known and novel pancreas genes expressed downstream of Nkx2.2 during development. BMC DEVELOPMENTAL BIOLOGY 2009; 9:65. [PMID: 20003319 PMCID: PMC2799404 DOI: 10.1186/1471-213x-9-65] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Accepted: 12/10/2009] [Indexed: 11/10/2022]
Abstract
BACKGROUND The homeodomain containing transcription factor Nkx2.2 is essential for the differentiation of pancreatic endocrine cells. Deletion of Nkx2.2 in mice leads to misspecification of islet cell types; insulin-expressing beta cells and glucagon-expressing alpha cells are replaced by ghrelin-expressing cells. Additional studies have suggested that Nkx2.2 functions both as a transcriptional repressor and activator to regulate islet cell formation and function. To identify genes that are potentially regulated by Nkx2.2 during the major wave of endocrine and exocrine cell differentiation, we assessed gene expression changes that occur in the absence of Nkx2.2 at the onset of the secondary transition in the developing pancreas. RESULTS Microarray analysis identified 80 genes that were differentially expressed in e12.5 and/or e13.5 Nkx2.2-/- embryos. Some of these genes encode transcription factors that have been previously identified in the pancreas, clarifying the position of Nkx2.2 within the islet transcriptional regulatory pathway. We also identified signaling factors and transmembrane proteins that function downstream of Nkx2.2, including several that have not previously been described in the pancreas. Interestingly, a number of known exocrine genes are also misexpressed in the Nkx2.2-/- pancreas. CONCLUSIONS Expression profiling of Nkx2.2-/- mice during embryogenesis has allowed us to identify known and novel pancreatic genes that function downstream of Nkx2.2 to regulate pancreas development. Several of the newly identified signaling factors and transmembrane proteins may function to influence islet cell fate decisions. These studies have also revealed a novel function for Nkx2.2 in maintaining appropriate exocrine gene expression. Most importantly, Nkx2.2 appears to function within a complex regulatory loop with Ngn3 at a key endocrine differentiation step.
Collapse
Affiliation(s)
- Keith R Anderson
- Department of Biochemistry and Program in Molecular Biology, University of Colorado Health Science Center, Denver, CO 80045, USA
| | | | | | | |
Collapse
|
259
|
Abstract
Diabetes is characterized by decreased function of insulin-producing beta cells and insufficient insulin output resulting from an absolute (Type 1) or relative (Type 2) inadequate functional beta cell mass. Both forms of the disease would greatly benefit from treatment strategies that could enhance beta cell regeneration and/or function. Successful and reliable methods of generating beta cells or whole islets from progenitor cells in vivo or in vitro could lead to restoration of beta cell mass in individuals with Type 1 diabetes and enhanced beta cell compensation in Type 2 patients. A thorough understanding of the normal developmental processes that occur during pancreatic organogenesis, for example, transcription factors, cell signaling molecules, and cell-cell interactions that regulate endocrine differentiation from the embryonic pancreatic epithelium, is required in order to successfully reach these goals. This review summarizes our current understanding of pancreas development, with particular emphasis on factors intrinsic or extrinsic to the pancreatic epithelium that are involved in regulating the development and differentiation of the various pancreatic cell types. We also discuss the recent progress in generating insulin-producing cells from progenitor sources.
Collapse
Affiliation(s)
- Michelle A Guney
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
260
|
Lefebvre B, Belaich S, Longue J, Vandewalle B, Oberholzer J, Gmyr V, Pattou F, Kerr-Conte J. 5'-AZA induces Ngn3 expression and endocrine differentiation in the PANC-1 human ductal cell line. Biochem Biophys Res Commun 2009; 391:305-9. [PMID: 19913512 DOI: 10.1016/j.bbrc.2009.11.054] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Accepted: 11/07/2009] [Indexed: 12/24/2022]
Abstract
Neurogenin 3 is necessary for endocrine cell development in the embryonic pancreas and has been shown to induce transdifferentiation duct cells from adult pancreas toward a neuro-endocrine phenotype. Here we discovered that the demethylating agent 5'-Azadeoxycytidine (AZA) induced Ngn3 expression and endocrine differentiation from the PANC-1 human ductal cell line. The expression of markers specific to mature islet cells, i.e., glucagon and somatostatin, was also observed. In addition, we demonstrated that growth factors (betacellulin and soluble factors released during pancreas embryogenesis) increased the level of maturation. Our studies revealed that the PANC-1 model system may provide a basis for elucidating the ductal/endocrine differentiation.
Collapse
|
261
|
Abstract
Beta-cell regeneration represents a major goal of therapy for diabetes. Unravelling the origin of beta cells during pancreatic regeneration could help restore a functional beta-cell mass in diabetes patients. This scientific question has represented a longstanding interest still intensively investigated today. This review focuses on pioneering observations and subsequent theories made 100 years ago and describes how technical innovation helped resolve some, but not all, of the controversies generated by these early investigators. At the end of the 19th century, complete pancreatectomy demonstrated the crucial physiological role of the pancreas and its link with diabetes. Pancreatic injury models, including pancreatectomy and ductal ligation, allowed investigators to describe islet function and to assess the regenerative capacity of the pancreas. Three main theories were proposed to explain the origins of newly formed islets: (i) transdifferentiation of acinar cells into islets, (ii) islet neogenesis, a process reminiscent of islet formation during embryonic development, and (iii) replication of preexisting islet cells. Despite considerable technical innovation in the last 50 years, the origin of new adult beta cells remains highly controversial and the same three theories are still debated today.
Collapse
Affiliation(s)
- A Granger
- Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, USA
| | | |
Collapse
|
262
|
Multiple, temporal-specific roles for HNF6 in pancreatic endocrine and ductal differentiation. Mech Dev 2009; 126:958-73. [PMID: 19766716 DOI: 10.1016/j.mod.2009.09.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Revised: 08/20/2009] [Accepted: 09/11/2009] [Indexed: 01/02/2023]
Abstract
Within the developing pancreas Hepatic Nuclear Factor 6 (HNF6) directly activates the pro-endocrine transcription factor, Ngn3. HNF6 and Ngn3 are each essential for endocrine differentiation and HNF6 is also required for embryonic duct development. Most HNF6(-/-) animals die as neonates, making it difficult to study later aspects of HNF6 function. Here, we describe, using conditional gene inactivation, that HNF6 has specific functions at different developmental stages in different pancreatic lineages. Loss of HNF6 from Ngn3-expressing cells (HNF6(Delta endo)) resulted in fewer multipotent progenitor cells entering the endocrine lineage, but had no effect on beta cell terminal differentiation. Early, pancreas-wide HNF6 inactivation (HNF6(Delta panc)) resulted in endocrine and ductal defects similar to those described for HNF6 global inactivation. However, all HNF6(Delta panc) animals survived to adulthood. HNF6(Delta panc) pancreata displayed increased ductal cell proliferation and metaplasia, as well as characteristics of pancreatitis, including up-regulation of CTGF, MMP7, and p8/Nupr1. Pancreatitis was most likely caused by defects in ductal primary cilia. In addition, expression of Prox1, a known regulator of pancreas development, was decreased in HNF6(Delta panc) pancreata. These data confirm that HNF6 has both early and late functions in the developing pancreas and is essential for maintenance of Ngn3 expression and proper pancreatic duct morphology.
Collapse
|
263
|
Derivation of insulin-producing cells from human embryonic stem cells. Stem Cell Res 2009; 3:73-87. [DOI: 10.1016/j.scr.2009.08.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Revised: 08/01/2009] [Accepted: 08/18/2009] [Indexed: 12/21/2022] Open
|
264
|
Abstract
OBJECTIVE Neurogenin 3 plays a pivotal role in pancreatic endocrine differentiation. Whereas mouse models expressing reporters such as eGFP or LacZ under the control of the Neurog3 gene enable us to label cells in the pancreatic endocrine lineage, the long half-life of most reporter proteins makes it difficult to distinguish cells actively expressing neurogenin 3 from differentiated cells that have stopped transcribing the gene. RESEARCH DESIGN AND METHODS In order to separate the transient neurogenin 3 -expressing endocrine progenitor cells from the differentiating endocrine cells, we developed a mouse model (Ngn3-Timer) in which DsRed-E5, a fluorescent protein that shifts its emission spectrum from green to red over time, was expressed transgenically from the NEUROG3 locus. RESULTS In the Ngn3-Timer embryos, green-dominant cells could be readily detected by microscopy or flow cytometry and distinguished from green/red double-positive cells. When fluorescent cells were sorted into three different populations by a fluorescence-activated cell sorter, placed in culture, and then reanalyzed by flow cytometry, green-dominant cells converted to green/red double-positive cells within 6 h. The sorted cell populations were then used to determine the temporal patterns of expression for 145 transcriptional regulators in the developing pancreas. CONCLUSIONS The precise temporal resolution of this model defines the narrow window of neurogenin 3 expression in islet progenitor cells and permits sequential analyses of sorted cells as well as the testing of gene regulatory models for the differentiation of pancreatic islet cells.
Collapse
Affiliation(s)
- Takeshi Miyatsuka
- From the Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Zhongmei Li
- From the Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Michael S. German
- From the Diabetes Center and Department of Medicine, University of California, San Francisco, San Francisco, California
- Corresponding author: Michael S. German,
| |
Collapse
|
265
|
Oliver-Krasinski JM, Kasner MT, Yang J, Crutchlow MF, Rustgi AK, Kaestner KH, Stoffers DA. The diabetes gene Pdx1 regulates the transcriptional network of pancreatic endocrine progenitor cells in mice. J Clin Invest 2009; 119:1888-98. [PMID: 19487809 DOI: 10.1172/jci37028] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Accepted: 04/02/2009] [Indexed: 12/12/2022] Open
Abstract
Heterozygous mutations in the gene encoding the pancreatic homeodomain transcription factor pancreatic duodenal homeobox 1 (PDX1) are associated with maturity onset diabetes of the young, type 4 (MODY4) and type 2 diabetes. Pdx1 governs the early embryonic development of the pancreas and the later differentiation of the insulin-producing islet beta cells of the endocrine compartment. We derived a Pdx1 hypomorphic allele that reveals a role for Pdx1 in the specification of endocrine progenitors. Mice homozygous for this allele displayed a selective reduction in endocrine lineages associated with decreased numbers of endocrine progenitors and a marked reduction in levels of mRNA encoding the proendocrine transcription factor neurogenin 3 (Ngn3). During development, Pdx1 occupies an evolutionarily conserved enhancer region of Ngn3 and interacts with the transcription factor one cut homeobox 1 (Hnf6) to activate this enhancer. Furthermore, mRNA levels of all 4 members of the transcription factor network that regulates Ngn3 expression, SRY-box containing gene 9 (Sox9), Hnf6, Hnf1b, and forkhead box A2 (Foxa2), were decreased in homozygous mice. Pdx1 also occupied regulatory sequences in Foxa2 and Hnf1b. Thus, Pdx1 contributes to specification of endocrine progenitors both by regulating expression of Ngn3 directly and by participating in a cross-regulatory transcription factor network during early pancreas development. These results provide insights that may be applicable to beta cell replacement strategies involving the guided differentiation of ES cells or other progenitor cell types into the beta cell lineage, and they suggest a molecular mechanism whereby human PDX1 mutations cause diabetes.
Collapse
Affiliation(s)
- Jennifer M Oliver-Krasinski
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
266
|
Díaz Pérez JA. [Neuroendocrine system of the pancreas and gastrointestinal tract: origin and development]. ACTA ACUST UNITED AC 2009; 56 Suppl 2:2-9. [PMID: 19627763 DOI: 10.1016/s1575-0922(09)70858-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Gastroenteropancreatic neuroendocrine tumours (GEP NETs) originate from the neuroendocrine cells through the gastrointestinal tract and endocrine pancreas. The embryologic development of the pancreas is a complex process that begins with the "stem cell" that come from the endodermus. These cells go through two phases: in the first transition the "stem cell" differentiates in exocrine and endocrine cells. This process is regulated by transcription factors such as Pdx1 ("insulin promoter factor 1"), Hlxb6 and SOX9. In the second transition the neuroendocrine cell differentiates in the 5 cell types (alpha, beta, delta, PP y epsilon.). This process is regulated through the balance between factors favoring differentiation (mainly neurogenin 3) and inhibitor factors which depend on Notch signals. The existence of a third transition in postnatal pancreas is hypothesized. The "stem cell" from pancreatic ducts would become adult beta cells, through autoduplication and neogenesis. In the small gut of the adult the stem cell are placed in the intestinal crypts and develop to villi in secretor lines (enterocytes, globet and Paneths cells) or neuroendocrine cells from which at least 10 cell types depend. This process is regulated by transcription factors: Math1, neurogenina 3 and NeuroD.
Collapse
|
267
|
Kaneto H, Matsuoka TA, Kawashima S, Yamamoto K, Kato K, Miyatsuka T, Katakami N, Matsuhisa M. Role of MafA in pancreatic beta-cells. Adv Drug Deliv Rev 2009; 61:489-96. [PMID: 19393272 DOI: 10.1016/j.addr.2008.12.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 12/15/2008] [Indexed: 01/01/2023]
Abstract
Pancreatic beta-cell-specific insulin gene expression is regulated by a variety of pancreatic transcription factors and the conserved A3, C1 and E1 elements in the insulin gene enhancer region are very important for activation of insulin gene. Indeed, PDX-1 binding to the A3 element and NeuroD binding to the E1 element are crucial for insulin gene transcription. Recently, C1 element-binding transcription factor was identified as MafA, which is a basic-leucine zipper transcription factor and functions as a potent transactivator for the insulin gene. Under diabetic conditions, chronic hyperglycemia gradually deteriorates pancreatic beta-cell function, which is accompanied by decreased expression and/or DNA binding activities of MafA and PDX-1. Furthermore, MafA overexpression, together with PDX-1 and NeuroD, markedly induces insulin biosynthesis in various non-beta-cells and thereby is a useful tool to efficiently induce insulin-producing surrogate beta-cells. These results suggest that MafA plays a crucial role in pancreatic beta-cells and could be a novel therapeutic target for diabetes.
Collapse
|
268
|
Identification of a pancreatic stellate cell population with properties of progenitor cells: new role for stellate cells in the pancreas. Biochem J 2009; 421:181-91. [PMID: 19379129 DOI: 10.1042/bj20081466] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Numerous studies conducted in a diversity of adult tissues have shown that certain stem cells are characterized by the expression of a protein known as the ABCG2 transporter (where ABC is ATP- binding cassette). In the adult pancreas, although various multipotent progenitors have been proposed, the ABCG2 marker has only been detected in the so-called 'side population' (a primitive haematopoietic cell population with a multipotential capacity). In the present study we sought to identify new ABCG2+ pancreatic cell populations and to explore whether they exhibit the properties of progenitor cells. We isolated and expanded mitoxantrone-resistant cells from pancreata of lactating rats by drug selection. These cells were characterized and maintained in different stages of differentiation using several media 'cocktails' plus Matrigel (BD Biosciences). Differentiation was assessed by RT-PCR (reverse transcription-PCR), immunocytochemistry, electron microscopy and ELISA. The expanded cell population demonstrated a phenotype of PaSCs (pancreatic stellate cells). Spontaneous cell clusters occurred during cell expansion and they showed weak expression of the transcription factor Pdx1 (pancreatic and duodenal homeobox 1). Moreover, the presence of inductive factors in the Matrigel plus exendin-4 led to an increase in Pdx1 and endocrine genes, such as insulin, islet amyloid polypeptide, glucagon, the glucose transporter GLUT2, chromogranin A and the convertases PC1/3 and PC2 were also detected. Immunocytochemical analysis showed co-localization of insulin and C-peptide, whereas ultrastructural studies revealed the presence of granules. Insulin secretion from cell clusters was detected in the cell culture medium. We identified a population of PaSCs that express the ABCG2+ transporter and have the capacity to transdifferentiate into insulin-producing cells. Although the potential therapeutic application remains to be tested, PaSCs could represent a future option for insulin replacement in diabetes research.
Collapse
|
269
|
Sustained Neurog3 expression in hormone-expressing islet cells is required for endocrine maturation and function. Proc Natl Acad Sci U S A 2009; 106:9715-20. [PMID: 19487660 DOI: 10.1073/pnas.0904247106] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neurog3 (Neurogenin 3 or Ngn3) is both necessary and sufficient to induce endocrine islet cell differentiation from embryonic pancreatic progenitors. Since robust Neurog3 expression has not been detected in hormone-expressing cells, Neurog3 is used as an endocrine progenitor marker and regarded as dispensable for the function of differentiated islet cells. Here we used 3 independent lines of Neurog3 knock-in reporter mice and mRNA/protein-based assays to examine Neurog3 expression in hormone-expressing islet cells. Neurog3 mRNA and protein are detected in hormone-producing cells at both embryonic and adult stages. Significantly, inactivating Neurog3 in insulin-expressing beta cells at embryonic stages or in Pdx1-expressing islet cells in adults impairs endocrine function, a phenotype that is accompanied by reduced expression of several Neurog3 target genes that are essential for islet cell differentiation, maturation, and function. These findings demonstrate that Neurog3 is required not only for initiating endocrine cell differentiation, but also for promoting islet cell maturation and maintaining islet function.
Collapse
|
270
|
Abstract
Understanding how the pancreas develops is vital to finding new treatments for a range of pancreatic diseases, including diabetes and pancreatic cancer. Xenopus is a relatively new model organism for the elucidation of pancreas development, and has already made contributions to the field. Recent studies have shown benefits of using Xenopus for understanding both early patterning and lineage specification aspects of pancreas organogenesis. This review focuses specifically on Xenopus pancreas development, and covers events from the end of gastrulation, when regional specification of the endoderm is occurring, right through metamorphosis, when the mature pancreas is fully formed. We have attempted to cover pancreas development in Xenopus comprehensively enough to assist newcomers to the field and also to enable those studying pancreas development in other model organisms to better place the results from Xenopus research into the context of the field in general and their studies specifically. Developmental Dynamics 238:1271-1286, 2009. (c) 2009 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Esther J. Pearl
- Laboratory of Molecular Organogenesis, Institut de Recherches Cliniques de Montréal, Montréal, QC Canada
- Département de Médecine, Université de Montréal, Montréal, Canada
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, Canada
| | - Cassandra K. Bilogan
- Laboratory of Molecular Organogenesis, Institut de Recherches Cliniques de Montréal, Montréal, QC Canada
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, Canada
- Division of Experimental Medicine and Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Sandeep Mukhi
- Department of Embryology, Carnegie Institution, Baltimore, Maryland
| | - Donald D. Brown
- Department of Embryology, Carnegie Institution, Baltimore, Maryland
| | - Marko E. Horb
- Laboratory of Molecular Organogenesis, Institut de Recherches Cliniques de Montréal, Montréal, QC Canada
- Département de Médecine, Université de Montréal, Montréal, Canada
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, Canada
- Division of Experimental Medicine and Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| |
Collapse
|
271
|
Vincent RK, Odorico JS. Reduced serum concentration is permissive for increased in vitro endocrine differentiation from murine embryonic stem cells. Differentiation 2009; 78:24-34. [PMID: 19446949 DOI: 10.1016/j.diff.2009.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Accepted: 03/15/2009] [Indexed: 10/20/2022]
Abstract
Embryonic stem cells (ESCs) have been shown to be capable of differentiating into pancreatic progenitors and insulin-producing cells in vitro. However, before ESC derivatives can be used in clinical settings, efficient selective differentiation needs to be achieved. Essential to improving ESC differentiation to islet endocrine cells is an understanding of the influences of extrinsic signals and transcription factors on cell specification. Herein, we investigate the influence of serum-supplemented growth conditions on the differentiation of murine ESCs to endocrine lineages in the context of over-expression of two pancreatic transcription factors, Pdx1 and Ngn3. To study the effect of different serum formulations and concentrations on the ability of murine ESCs to differentiate into endocrine cells in vitro, cells were grown into embryoid bodies and then differentiated in various serum replacement (SR), fetal calf serum (FCS) and serum-free conditions. Using immunohistochemistry and quantitative real-time RT-PCR (QPCR), we found that, of the conditions tested, 1% SR differentiation medium resulted in the highest levels of insulin-1 mRNA and significantly increased the total number of insulin-expressing cells. Applying this knowledge to cell lines in which Pdx1 or Ngn3 transgene expression could be induced by exposure to doxycycline we differentiated TetPDX1 and TetNgn3 ESCs under conditions of either 10% FCS or 1% SR medium. In the presence of 10% serum, induced expression of either Pdx1 or Ngn3 in differentiating ESCs resulted in modest increases in hormone transcripts and cell counts. However, changing the serum formulation from 10% FCS to 1% SR significantly enhanced the number of insulin+/C-peptide+ cells in parallel with increased insulin-1 transcript levels in both inducible cell lines. In summary, these data demonstrate that induced expression of key pancreatic transcription factors in combination with low serum/SR concentrations increases endocrine cell differentiation from murine ESCs.
Collapse
Affiliation(s)
- Robert K Vincent
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | | |
Collapse
|
272
|
Baeyens L, Bonné S, Bos T, Rooman I, Peleman C, Lahoutte T, German M, Heimberg H, Bouwens L. Notch signaling as gatekeeper of rat acinar-to-beta-cell conversion in vitro. Gastroenterology 2009; 136:1750-60.e13. [PMID: 19208356 DOI: 10.1053/j.gastro.2009.01.047] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 12/08/2008] [Accepted: 01/16/2009] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Exocrine acinar cells in the pancreas are highly differentiated cells that retain a remarkable degree of plasticity. After isolation and an initial phase of dedifferentiation in vitro, rodent acinar cells can convert to endocrine beta-cells when cultured in the presence of appropriate factors. The mechanisms regulating this phenotypic conversion are largely unknown. METHODS Using rat acinar cell cultures, we studied the role of Notch signaling in a model of acinar-to-beta-cell conversion. RESULTS We report a novel lectin-based cell labeling method to demonstrate the acinar origin of newly formed insulin-expressing beta-cells. This method allows for specific tracing of the acinar cells. We demonstrate that growth factor-induced conversion of adult acinar cells to beta-cells is negatively regulated by Notch1 signaling. Activated Notch1 signaling prevents the reexpression of the proendocrine transcription factor Neurogenin-3, the key regulator of endocrine development in the embryonic pancreas. Interfering with Notch1 signaling allows modulating the acinar cell susceptibility to the differentiation-inducing factors. Its inhibition significantly improves beta-cell neoformation with approximately 30% of acinar cells that convert to beta-cells. The newly formed beta-cells mature when transplanted ectopically and are capable of restoring normal blood glycemia in diabetic recipients. CONCLUSIONS We report for the first time an efficient way to reprogram one third of the acinar cells to beta-cells by adult cell type conversion. This could find application in cell replacement therapy of type 1 diabetes, provided that it can be translated from rodent to human models.
Collapse
Affiliation(s)
- Luc Baeyens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
273
|
Abstract
The number of patients worldwide suffering from the chronic disease diabetes mellitus is growing at an alarming rate. Insulin-secreting beta-cells in the islet of Langerhans are damaged to different extents in diabetic patients, either through an autoimmune reaction present in type 1 diabetic patients or through inherent changes within beta-cells that affect their function in patients suffering from type 2 diabetes. Cell replacement strategies via islet transplantation offer potential therapeutic options for diabetic patients. However, the discrepancy between the limited number of donor islets and the high number of patients who could benefit from such a treatment reflects the dire need for renewable sources of high-quality beta-cells. Human embryonic stem cells (hESCs) are capable of self-renewal and can differentiate into components of all three germ layers, including all pancreatic lineages. The ability to differentiate hESCs into beta-cells highlights a promising strategy to meet the shortage of beta-cells. Here, we review the different approaches that have been used to direct differentiation of hESCs into pancreatic and beta-cells. We will focus on recent progress in the understanding of signaling pathways and transcription factors during embryonic pancreas development and how this knowledge has helped to improve the methodology for high-efficiency beta-cell differentiation in vitro.
Collapse
Affiliation(s)
- Tingxia Guo
- Department of Medicine, Diabetes Center, University of California, San Francisco, San Francisco, California 94143, USA
| | | |
Collapse
|
274
|
Neurogenin 3 and neurogenic differentiation 1 are retained in the cytoplasm of multiple endocrine neoplasia type 1 islet and pancreatic endocrine tumor cells. Pancreas 2009; 38:259-66. [PMID: 19307926 DOI: 10.1097/mpa.0b013e3181930818] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES To investigate if transcription factors involved in pancreatic differentiation and regeneration are present in pancreatic endocrine tumors and if they are differentially expressed in normal pancreas compared with multiple endocrine neoplasia type 1 (MEN1) nontumorous pancreas. METHODS The expression of neurogenin 3 (NEUROG3), neurogenic differentiation 1 (NEUROD1), POU class 3 homeobox 4 (POU3F4), pancreatic duodenal homeobox factor 1 (PDX1), ribosomal protein L10 (RPL10), delta-like 1 homolog (Drosophila; DLK1), and menin was analyzed by immunohistochemistry in normal pancreas and pancreatic endocrine tumors from 6 patients with MEN1 and 16 patients with sporadic tumors, as well as pancreatic specimens from Men1 heterozygous and wild type mice. Quantitative polymerase chain reaction was performed in a subset of human tumors. RESULTS Tumors and MEN1 nontumorous endocrine cells showed a prominent cytoplasmatic NEUROG3 and NEUROD1 expression. These factors were significantly more expressed in the cytoplasm of Men1 heterozygous mouse islet cells compared with wild type islets; the latter showed an exclusively nuclear reactivity. The degree of Pou3f4, Rpl10, and Dlk1 immunoreactivities differed significantly between islets of heterozygous and wild type mice. The expressions of RPL10 and NEUROD1 were prominent in the MEN1 human and heterozygous mouse exocrine pancreas. Insulinomas had significantly higher PDX1 and DLK1 messenger RNA levels compared with other tumor types. CONCLUSIONS Transcription factors involved in pancreatic development show altered expression and subcellular localization in MEN1 nontumorous pancreas and pancreatic endocrine tumors.
Collapse
|
275
|
Activin A-Induced Differentiation of Embryonic Stem Cells into Endoderm and Pancreatic Progenitors—The Influence of Differentiation Factors and Culture Conditions. Stem Cell Rev Rep 2009; 5:159-73. [DOI: 10.1007/s12015-009-9061-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 02/19/2009] [Indexed: 02/07/2023]
|
276
|
Yechoor V, Liu V, Espiritu C, Paul A, Oka K, Kojima H, Chan L. Neurogenin3 is sufficient for transdetermination of hepatic progenitor cells into neo-islets in vivo but not transdifferentiation of hepatocytes. Dev Cell 2009; 16:358-73. [PMID: 19289082 PMCID: PMC2676438 DOI: 10.1016/j.devcel.2009.01.012] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 11/06/2008] [Accepted: 01/23/2009] [Indexed: 12/30/2022]
Abstract
The transcription factor Neurogenin3 (Ngn3) is required for islet-cell type specification. Here, we show that hepatic gene transfer of Ngn3 transiently induces insulin in terminally differentiated hepatocytes but fails to transdifferentiate them, i.e., switch their lineage into islet cells. However, Ngn3 leads to long-term diabetes reversal in mice due to the emergence of periportal islet-like cell clusters. These neo-islets display glycemia-regulated insulin, beta-cell-specific transcripts, and an islet-specific transcription cascade, and they produce all four major islet hormones. They appear to arise from hepatic progenitor cells, most likely endoderm-derived oval cells. Thus, transfer of a single lineage-defining transcription factor, Ngn3, is sufficient to induce cell-lineage switching from a hepatic to an islet lineage in these progenitor cells, a process consistent with transdetermination, i.e, lineage switching in lineage-determined, but not terminally differentiated, cells. This paradigm of induced transdetermination of receptive progenitor cells in vivo may be generally applicable to therapeutic organogenesis for multiple diseases, including diabetes.
Collapse
MESH Headings
- Animals
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/physiology
- Betacellulin
- Cell Differentiation/physiology
- Cell Transdifferentiation/physiology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/therapy
- Gene Transfer Techniques
- Hepatocytes/cytology
- Hepatocytes/physiology
- Insulin/biosynthesis
- Insulin/genetics
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/physiology
- Islets of Langerhans/cytology
- Islets of Langerhans/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Models, Biological
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Stem Cells/cytology
- Stem Cells/physiology
- Transcription, Genetic
Collapse
Affiliation(s)
- Vijay Yechoor
- Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Victoria Liu
- Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Christie Espiritu
- Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Antoni Paul
- Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Kazuhiro Oka
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Hideto Kojima
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular Genetics in Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Lawrence Chan
- Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
277
|
Wang H, Wang S, Hu J, Kong Y, Chen S, Li L, Li L. Oct4 is expressed in Nestin-positive cells as a marker for pancreatic endocrine progenitor. Histochem Cell Biol 2009; 131:553-63. [PMID: 19224238 DOI: 10.1007/s00418-009-0560-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2009] [Indexed: 01/18/2023]
Abstract
There are abundant progenitor cells in the developing pancreas, but molecular markers for these cells are lacking. Octamer-binding transcription factor-4 (Oct4) is an important transcription factor for keeping the features of self-renewal and pluripotency of embryonic stem cells. It's well known that Oct4, as a totipotent stem cells marker, just is expressed in totipotent stem cells. In the present study, we collected ten human fetal pancreases, and found that Oct4 mRNA and protein were expressed in human fetal pancreas samples by RT-PCR, western blot and immunohistochemistry assays. Using double-staining, we demonstrated that Oct4 was not co-expressed with Chromogranin A (a peptide expressed in endocrine cells), but partially co-expressed with Ngn3 (a transcription factor expressed in pancreatic endocrine precursor cells) and Nestin (a intermediate filament, Nestin-positive cells isolated from islets can be induced to express insulin) in human fetal pancreases. Indeed, we prepared Nestin-positive cells from human fetal pancreas by cell selection, and found that these cells expressed Oct4 and Ngn3. The Nestin-positive cells displayed a rapid duplication and could differentiate into osteoblasts, fat and endocrine cells in vitro. These results indicated that the Nestin-positive cells in the fetal age should be pancreatic progenitor cells. Overall, our study suggested that Oct4 was a marker for pancreatic endocrine progenitor.
Collapse
Affiliation(s)
- Hong Wang
- Stem Cell Research Center, Health Science Center, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing, China.
| | | | | | | | | | | | | |
Collapse
|
278
|
Gao N, LeLay J, Vatamaniuk MZ, Rieck S, Friedman JR, Kaestner KH. Dynamic regulation of Pdx1 enhancers by Foxa1 and Foxa2 is essential for pancreas development. Genes Dev 2009; 22:3435-48. [PMID: 19141476 DOI: 10.1101/gad.1752608] [Citation(s) in RCA: 242] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The onset of pancreas development in the foregut endoderm is marked by activation of the homeobox gene Pdx1 (IPF1). Pdx1 is essential for the expansion of the pancreatic primordium and the development of endocrine islets. The control of Pdx1 expression has been only partially elucidated. We demonstrate here that the winged-helix transcription factors Foxa1 and Foxa2 co-occupy multiple regulatory domains in the Pdx1 gene. Compound conditional ablation of both Foxa1 and Foxa2 in the pancreatic primordium results in complete loss of Pdx1 expression and severe pancreatic hypoplasia. Mutant mice exhibit hyperglycemia with severely disrupted acinar and islet development, and die shortly after birth. Assessment of developmental markers in the mutant pancreas revealed a failure in the expansion of the pancreatic anlage, a blockage of exocrine and endocrine cell differentiation, and an arrest at the primitive duct stage. Comparing their relative developmental activity, we find that Foxa2 is the major regulator in promoting pancreas development and cell differentiation. Using chromatin immunoprecipitations (ChIP) and ChIP sequencing (ChIPSeq) of fetal pancreas and islet chromatin, we demonstrate that Foxa1 and Foxa2 predominantly occupy a distal enhancer at -6.4 kb relative to the transcriptional start site in the Pdx1 gene. In addition, occupancy of the well-characterized proximal Pdx1 enhancer by Foxa1 and Foxa2 is developmental stage-dependent. Thus, the regulation of Pdx1 expression by Foxa1 and Foxa2 is a key early event controlling the expansion and differentiation of the pancreatic primordia.
Collapse
Affiliation(s)
- Nan Gao
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
279
|
Bernardo AS, Cho CHH, Mason S, Docherty HM, Pedersen RA, Vallier L, Docherty K. Biphasic induction of Pdx1 in mouse and human embryonic stem cells can mimic development of pancreatic beta-cells. Stem Cells 2009; 27:341-51. [PMID: 19056911 DOI: 10.1634/stemcells.2008-0310] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Embryonic stem (ES) cells represent a possible source of islet tissue for the treatment of diabetes. Achieving this goal will require a detailed understanding of how the transcription factor cascade initiated by the homeodomain transcription factor Pdx1 culminates in pancreatic beta-cell development. Here we describe a genetic approach that enables fine control of Pdx1 transcriptional activity during endoderm differentiation of mouse and human ES cell. By activating an exogenous Pdx1VP16 protein in populations of cells enriched in definitive endoderm we show a distinct lineage-dependent requirement for this transcription factor's activity. Mimicking the natural biphasic pattern of Pdx1 expression was necessary to induce an endocrine pancreas-like cell phenotype, in which 30% of the cells were beta-cell-like. Cell markers consistent with the different beta-cell differentiation stages appeared in a sequential order following the natural pattern of pancreatic development. Furthermore, in mouse ES-derived cultures the differentiated beta-like cells secreted C-peptide (insulin) in response to KCl and 3-isobutyl-1-methylxanthine, suggesting that following a natural path of development in vitro represents the best approach to generate functional pancreatic cells. Together these results reveal for the first time a significant effect of the timed expression of Pdx1 on the non-beta-cells in the developing endocrine pancreas. Collectively, we show that this method of in vitro differentiation provides a template for inducing and studying ES cell differentiation into insulin-secreting cells.
Collapse
Affiliation(s)
- Andreia S Bernardo
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
280
|
Zhang T, Liu WD, Saunee NA, Breslin MB, Lan MS. Zinc finger transcription factor INSM1 interrupts cyclin D1 and CDK4 binding and induces cell cycle arrest. J Biol Chem 2009; 284:5574-81. [PMID: 19124461 DOI: 10.1074/jbc.m808843200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
INSM1 is a zinc finger transcription factor that plays an important role in pancreatic beta-cell development. To further evaluate its role in cell fate determination, we investigated INSM1 effects on cell cycle function. The cyclin box of cyclin D1 is essential for INSM1 binding. Competitive pull-down and co-immunoprecipitation revealed that INSM1 binding to cyclin D1 interrupts its association with CDK4 and induces hypophosphorylation of the retinoblastoma protein. An inducible Tet-on system was established in Cos-7 and Panc-1 cells. Using serum starvation, we synchronized the cell cycle and subsequently induced cell cycle progression by serum stimulation. Comparison of the INSM1 induction group with the noninduced control group, INSM1 ectopic expression causes cell cycle arrest, whereas the INSM1-mediated cell cycle arrest could be reversed by cyclin D1 and CDK4 overexpression. The proline-rich N-terminal portion of INSM1 is required for cyclin D1 binding. Mutation of proline residues abolished cyclin D1 binding and also diminished its ability to induce cell cycle arrest. Cellular proliferation of Panc-1 cells was inhibited by INSM1 overexpression demonstrated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, soft agar colony formation, as well as tumor growth in a nude mouse model. Taken together, we provide evidence to support that INSM1 binds to cyclin D1, interrupts cell cycle signaling, and inhibits cellular proliferation.
Collapse
Affiliation(s)
- Tao Zhang
- Research Institute for Children, Children's Hospital, New Orleans, Louisiana 70118, USA
| | | | | | | | | |
Collapse
|
281
|
Miyatsuka T, Matsuoka TA, Kaneto H. Transcription factors as therapeutic targets for diabetes. Expert Opin Ther Targets 2009; 12:1431-42. [PMID: 18851698 DOI: 10.1517/14728222.12.11.1431] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Islet cell implantation and pancreas transplantation have been used as treatments for diabetes but are limited by the shortage of donors and the requirement for lifelong immunosuppression. As an alternative, the generation of surrogate insulin-producing cells has been an area of interest for many researchers. Understanding how pancreatic beta-cells are generated during pancreas development will provide information that can be applied to generating surrogate beta-cells. OBJECTIVE To outline the current knowledge of pancreas development and differentiation, with a focus on the regulatory network of pancreas-enriched transcription factors and their targets. METHODS A review of relevant literature. CONCLUSIONS Pancreatic and duodenal homeobox 1 (Pdx1), Neurogenin 3 (Ngn3), and musculoaponeurotic fibrosarcoma oncogene homolog A (MafA) have been shown to play essential roles in pancreas development and beta-cell differentiation, and gain-of-function approaches indicate the potency of these factors for inducing differentiation of non-beta-cells into insulin-producing cells, which could lead to a novel therapy to cure diabetes.
Collapse
Affiliation(s)
- Takeshi Miyatsuka
- Osaka University Graduate School of Medicine, Department of Internal Medicine and Therapeutics, 2-2 Yamadaoka, Suita 565-0871, Osaka, Japan
| | | | | |
Collapse
|
282
|
Bernardo AS, Hay CW, Docherty K. Pancreatic transcription factors and their role in the birth, life and survival of the pancreatic beta cell. Mol Cell Endocrinol 2008; 294:1-9. [PMID: 18687378 DOI: 10.1016/j.mce.2008.07.006] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 05/15/2008] [Accepted: 07/04/2008] [Indexed: 12/26/2022]
Abstract
In recent years major progress has been made in understanding the role of transcription factors in the development of the endocrine pancreas in the mouse. Here we describe how a number of these transcription factors play a role in maintaining the differentiated phenotype of the beta cell, and in the mechanisms that allow the beta cell to adapt to changing metabolic demands that occur throughout life. Amongst these factors, Pdx1 plays a critical role in defining the region of the primitive gut that will form the pancreas, Ngn3 expression drives cells towards an endocrine lineage, and a number of additional proteins including Pdx1, in a second wave of expression, Pax4, NeuroD1/beta2, and MafA act as beta cell differentiation factors. In the mature beta cell Pdx1, MafA, beta2, and Nkx2.2 play important roles in regulating expression of insulin and to some extent other genes responsible for maintaining beta cell function. We emphasise here that data from gene expression studies in rodents seldom map on to the known structure of the corresponding human promoters. In the adult the beta cell is particularly susceptible to autoimmune-mediated attack and to the toxic metabolic milieu associated with over-eating, and utilises a number of these transcription factors in its defence. Pdx1 has anti-apoptotic and proliferative activities that help facilitate the maintenance of beta cell mass, while Ngn3 may be involved in the recruitment of progenitor cells, and Pax4 (and possibly HNF1alpha and Hnf4alpha) in the proliferation of beta cells in the adult pancreas. Other transcription factors with a more widespread pattern of expression that play a role in beta cell survival or proliferation include Foxo1, CREB family members, NFAT, FoxM1, Snail and Asc-2.
Collapse
Affiliation(s)
- Andreia S Bernardo
- University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | | | | |
Collapse
|
283
|
Abstract
Here we review progress on isolation and characterization of progenitor cells in the pancreas. We discuss advantages and current limitations of experiments with purified pancreatic cells, and areas where future growth in our understanding is needed to advance experiments in pancreas biology based on cell purification.
Collapse
Affiliation(s)
- T Sugiyama
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305-5329, USA.
| | | |
Collapse
|
284
|
Seymour PA, Freude KK, Dubois CL, Shih HP, Patel NA, Sander M. A dosage-dependent requirement for Sox9 in pancreatic endocrine cell formation. Dev Biol 2008; 323:19-30. [PMID: 18723011 PMCID: PMC2879081 DOI: 10.1016/j.ydbio.2008.07.034] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Revised: 06/28/2008] [Accepted: 07/16/2008] [Indexed: 11/21/2022]
Abstract
We have previously shown the transcription factor SOX9 to be required for the maintenance of multipotential pancreatic progenitor cells in the early embryonic pancreas. However, the association of pancreatic endocrine defects with the Sox9-haploinsufficiency syndrome campomelic dysplasia (CD) implies additional later roles for Sox9 in endocrine development. Using short-term lineage tracing in mice, we demonstrate here that SOX9 marks a pool of multipotential pancreatic progenitors throughout the window of major cell differentiation. During mid-pancreogenesis, both endocrine and exocrine cells simultaneously arise from the SOX9(+) epithelial cords. Our analysis of mice with 50%-reduced Sox9 gene dosage in pancreatic progenitors reveals endocrine-specific defects phenocopying CD. By birth, these mice display a specific reduction in endocrine cell mass, while their exocrine compartment and total organ size is normal. The decrease in endocrine cells is caused by reduced generation of endocrine progenitors from the SOX9(+) epithelium. Conversely, formation of exocrine progenitors is insensitive to reduced Sox9 gene dosage, thus explaining the normal organ size at birth. Our results show that not only is SOX9 required for the maintenance of early pancreatic progenitors, but also governs their adoption of an endocrine fate. Our findings therefore suggest that defective endocrine specification might underlie the pancreatic phenotype of individuals with CD.
Collapse
Affiliation(s)
- Philip A. Seymour
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| | - Kristine K. Freude
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| | - Claire L. Dubois
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| | - Hung-Ping Shih
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| | - Nisha A. Patel
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| | - Maike Sander
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| |
Collapse
|
285
|
Abstract
Ngn3 is a bHLH transcription factor critical for the specification of endocrine cells in the pancreatic Islets of Langerhans. Previous studies in mouse embryos have reported transient expression of Ngn3 in scattered cells within the developing pancreatic epithelium during midgestation (Schwitzgebel et al. [2000] Development 127:3533-3542). Specifically, these Ngn3-expressing cells have been shown to be progenitor cells fated to give rise to islet endocrine cells (Gradwohl et al. [2000] Proc Natl Acad Sci USA 97:1607-1611). Here, we characterize the expression of Ngn3 transcripts and protein throughout pancreatic development. Interestingly, we identify and define a dramatic and previously unnoticed gap in developmental Ngn3 expression. We show that both Ngn3 transcript and protein expression occur in two distinct temporal waves, the first occurring early from approximately E8.5 to E11.0, and the second initiating at approximately E12.0. Strikingly, this observed biphasic expression correlates with the "first" and "second" transitions, which encompass two distinct waves of embryonic endocrine differentiation. In addition, our studies demonstrate that Ngn3 transcripts are markedly more widespread in the pancreatic epithelium than NGN3 protein, indicating that post-transcriptional regulation is likely to play a critical role during endocrine differentiation.
Collapse
Affiliation(s)
- Alethia Villasenor
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9148, USA
| | | | | |
Collapse
|
286
|
Jia D, Sun Y, Konieczny SF. Mist1 regulates pancreatic acinar cell proliferation through p21 CIP1/WAF1. Gastroenterology 2008; 135:1687-97. [PMID: 18762186 PMCID: PMC2853247 DOI: 10.1053/j.gastro.2008.07.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 07/07/2008] [Accepted: 07/17/2008] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Mist1 is a basic helix-loop-helix (bHLH) transcription factor that is important to the proper development of the exocrine pancreas. The aim of this study was to investigate the role of Mist1 in modulating acinar cell proliferation. METHODS Ductal and acinar pancreatic cell lines were engineered to express an inducible Mist1 complementary DNA or to express a short hairpin RNA that targeted endogenous Mist1. Alterations in RNA and protein levels were detected by real-time reverse-transcription polymerase chain reaction and immunoblots. Chromatin immunoprecipitation and reporter gene assays were performed to map Mist1-responsive elements on target genes; the overall proliferation index of acinar cells from Mist1 null pancreata was evaluated by immunohistochemistry. RESULTS Expression of Mist1 resulted in a significant decrease in the proliferative potential of cells that was associated with induced expression of p21(CIP1/WAF1). Short hairpin RNA-directed knockdown of p21(CIP1/WAF1) generated cells that were refractory to Mist1 expression, whereas knockdown of Mist1 transcripts or deletion of Mist1 from the mouse genome led to increased cell proliferation and a concomitant decrease in p21(CIP1/WAF1) protein levels. Surprisingly, Mist1-dependent activation of the p21(CIP1/WAF1) promoter was independent of classic basic helix-loop-helix protein binding sites. Instead, Sp1 binding sites were essential for Mist1-dependent transcription, suggesting that Mist1 activates p21(CIP1/WAF1) expression through a unique Sp1 pathway. Indeed, coimmunoprecipitation studies demonstrated that Mist1 and Sp1 were found within the same transcription complex. CONCLUSIONS Our results show that Mist1 has a dual role in the development of the exocrine pancreas: controlling cell proliferation and promoting terminal differentiation.
Collapse
Affiliation(s)
- Di Jia
- Department of Biological Sciences and the Purdue Cancer Center, Purdue University, West Lafayette, IN
| | - Yan Sun
- Department of Biological Sciences and the Purdue Cancer Center, Purdue University, West Lafayette, IN
| | - Stephen F. Konieczny
- Department of Biological Sciences and the Purdue Cancer Center, Purdue University, West Lafayette, IN
| |
Collapse
|
287
|
Gittes GK. Developmental biology of the pancreas: a comprehensive review. Dev Biol 2008; 326:4-35. [PMID: 19013144 DOI: 10.1016/j.ydbio.2008.10.024] [Citation(s) in RCA: 317] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2008] [Revised: 10/09/2008] [Accepted: 10/13/2008] [Indexed: 02/06/2023]
Abstract
Pancreatic development represents a fascinating process in which two morphologically distinct tissue types must derive from one simple epithelium. These two tissue types, exocrine (including acinar cells, centro-acinar cells, and ducts) and endocrine cells serve disparate functions, and have entirely different morphology. In addition, the endocrine tissue must become disconnected from the epithelial lining during its development. The pancreatic development field has exploded in recent years, and numerous published reviews have dealt specifically with only recent findings, or specifically with certain aspects of pancreatic development. Here I wish to present a more comprehensive review of all aspects of pancreatic development, though still there is not a room for discussion of stem cell differentiation to pancreas, nor for discussion of post-natal regeneration phenomena, two important fields closely related to pancreatic development.
Collapse
Affiliation(s)
- George K Gittes
- Children's Hospital of Pittsburgh and the University of Pittsburgh School of Medicine, Department of Pediatric Surgery, 3705 Fifth Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
288
|
Abstract
Diabetes is a disease that could be treated more effectively with a better understanding of pancreas development. This review examines the role of master regulator genes driving crucial steps in pancreas development, from foregut specification to differentiation of the five endocrine cell types. The roles of Pdx1, Ptf1a, and Ngn3 are particularly examined as they are both necessary and sufficient for promoting pancreatic cell fates (Pdx1, Ptf1a) and endocrine cell development (Ngn3). The roles of Arx and Pax4 are studied as they compose part of the regulatory mechanism balancing development of different types of endocrine cells within the iselts and promote the development of alpha/PP and beta/delta cell progenitors, respectively. The roles of the aforementioned genes, and the consequences of misexpression of them for functionality of the pancreas, are examined through recent studies in model organisms, particularly Xenopus and zebrafish. Recent developments in cell replacement therapy research are also covered, concentrating on stem cell research (coaxing both adult and embryonic stem cells toward a beta cell fate) and transdifferentiation (generating beta cells from other differentiated cell types).
Collapse
Affiliation(s)
- Esther J. Pearl
- Laboratory of Molecular Organogenesis, Institut de Recherches Cliniques de Montréal, Montreal, QC Canada
| | - Marko E. Horb
- Laboratory of Molecular Organogenesis, Institut de Recherches Cliniques de Montréal, Montreal, QC Canada
- Division of Experimental Medicine and Department of Anatomy and Cell Biology, McGill University
- Département de Médecine, Université de Montréal
- Montreal Diabetes Research Center, Centre de recherche du Centre Hospitalier de l'Université de Montréal
| |
Collapse
|
289
|
Nakayama S, Arakawa M, Uchida T, Ogihara T, Kanno R, Ikeda F, Azuma K, Hirose T, Kawamori R, Fujitani Y, Watada H. Dose-dependent requirement of patched homologue 1 in mouse pancreatic beta cell mass. Diabetologia 2008; 51:1883-92. [PMID: 18654758 DOI: 10.1007/s00125-008-1080-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Accepted: 05/23/2008] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS Ectopic activation of hedgehog (HH) signalling in pancreas induces various abnormal morphogenetic events in the pancreas. This study analysed the dose-dependent requirement of patched homologue 1 (PTCH1), a negative regulator of HH signalling on pancreatic development. METHODS We used a recessive spontaneous mutant mouse denoted as mes which carries a mutated Ptch1 resulting in deletion of the most carboxy-terminal cytoplasmic domain of the PTCH1 protein. In this study, we analysed pancreatic morphology in Ptch1 ( +/+ ), Ptch1 ( +/mes ), Ptch1 (+/-), Ptch1 ( mes/me ) (s) and Ptch1 (-/mes ) mouse embryos, as well as the islet mass in adult Ptch1 (+/+), Ptch1 (+/mes ) and Ptch1 (+/-) mice. RESULTS Until embryonic day (E) 12.5, no obvious abnormality of pancreas was observed in any of the Ptch1 mutants. The levels of PDX1 and glucagon were also not evidently different among the mice genotypes studied. Thereafter, morphological abnormalities appeared in the Ptch1 mutant mice. The beta, alpha and exocrine cell masses decreased at E18.5 in parallel with increased HH signalling, with beta cell mass showing the highest sensitivity to HH signalling with a significant decrease even in Ptch1 (+/mes ) mice. Adult Ptch1 (+/-) mice also showed a significant decrease in beta cell mass compared with wild-type mice. CONCLUSIONS/INTERPRETATION Our findings indicate that the carboxy-terminal domain of Ptch1 is essential for pancreatic development. In addition, the loss of Ptch1 function decreases both the endocrine and exocrine cell mass in a dose-dependent manner, with beta cells particularly sensitive to changes in HH signalling.
Collapse
Affiliation(s)
- S Nakayama
- Department of Medicine, Metabolism and Endocrinology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
290
|
Abstract
The major forms of diabetes are characterized by pancreatic islet beta-cell dysfunction and decreased beta-cell numbers, raising hope for cell replacement therapy. Although human islet transplantation is a cell-based therapy under clinical investigation for the treatment of type 1 diabetes, the limited availability of human cadaveric islets for transplantation will preclude its widespread therapeutic application. The result has been an intense focus on the development of alternate sources of beta cells, such as through the guided differentiation of stem or precursor cell populations or the transdifferentiation of more plentiful mature cell populations. Realizing the potential for cell-based therapies, however, requires a thorough understanding of pancreas development and beta-cell formation. Pancreas development is coordinated by a complex interplay of signaling pathways and transcription factors that determine early pancreatic specification as well as the later differentiation of exocrine and endocrine lineages. This review describes the current knowledge of these factors as they relate specifically to the emergence of endocrine beta cells from pancreatic endoderm. Current therapeutic efforts to generate insulin-producing beta-like cells from embryonic stem cells have already capitalized on recent advances in our understanding of the embryonic signals and transcription factors that dictate lineage specification and will most certainly be further enhanced by a continuing emphasis on the identification of novel factors and regulatory relationships.
Collapse
Affiliation(s)
- Jennifer M. Oliver-Krasinski
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Doris A. Stoffers
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
291
|
Kawasaki H, Mizuguchi T, Oshima H, Nobuoka T, Shibata T, Kaji S, Kokai Y, Katsuramaki T, Mitaka T, Hirata K. Efficient transformation of small hepatocytes into insulin-expressing cells by forced expression of Pdx1. JOURNAL OF HEPATO-BILIARY-PANCREATIC SURGERY 2008; 15:403-409. [PMID: 18670842 DOI: 10.1007/s00534-007-1318-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Accepted: 09/21/2007] [Indexed: 01/30/2023]
Abstract
BACKGROUND/PURPOSE The expression of ectopic pancreatic and duodenal homeobox factor 1 (Pdx1) can transform hepatocytes into pancreatic endocrine cells. Small hepatocytes (SHs) have a high possibility to be a cellular source for islet cell transplantation. However, the efficacy of the transformation of SHs into pancreatic endocrine cells is not fully understood. The focus of our study was to compare the efficacy of the transformation into pancreatic endocrine cells of SHs and mature hepatocytes (MHs). METHODS MHs and SHs were cultured for 3 and 10 days, respectively, before Adeno-Pdx1 gene transduction. Western blot analysis was performed for pancreatic transcription factors, and reverse-transcription polymerase chain reaction (RT-PCR) was performed for the gene expression of pancreatic hormones. Confocal laser microscanning analysis was used to observe insulin and glucagon expression. RESULTS Although the pancreatic transcription factors Pdx1, Ngn3, NeuroD, and Pax6 were induced in both SHs and MHs after Adeno-Pdx1 gene expression, the pancreatic transcription factors Nkx2.2 and Nkx6.1 were induced in SHs more than in MHs. Glucagon mRNA expression was seen in both SHs and MHs, whereas insulin mRNA expression was higher in SHs than in MHs. Confocal laser microscanning analysis showed that SHs expressed both insulin and glucagon, whereas MHs predominantly expressed glucagon. CONCLUSIONS SHs were transformed into both insulin-and glucagon-expressing cells, and the efficacy of the transformation into insulin-expressing cells of SHs was higher than that for MHs. Thus, SHs could be a more suitable source of future cell therapy than MHs.
Collapse
Affiliation(s)
- Hiroyuki Kawasaki
- Department of Surgery I, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-Ku, Sapporo, Hokkaido 060-8543, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
292
|
Ogihara T, Fujitani Y, Uchida T, Kanno R, Choi JB, Hirose T, Kawamori R, Watada H. Combined expression of transcription factors induces AR42J-B13 cells to differentiate into insulin-producing cells. Endocr J 2008; 55:691-8. [PMID: 18506085 DOI: 10.1507/endocrj.k07e-169] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Neurogenin 3 (Ngn3) is a transcription factor that regulates an initial step of differentiation from uncommitted pancreatic progenitors into endocrine cells. Additional transcription factors are required for complete differentiation into mature pancreatic beta cells. In this study, we established an in vitro model system of beta-cell differentiation by adenovirus-mediated expression of several transcription factors in AR42J-B13 cells, a pancreatic progenitor-like cell line derived from exocrine pancreas. Exogenous expression of Ngn3 in AR42J-B13 cells induced expression of Nkx2.2, Pax4, and Pax6, which are all essential for beta-cell differentiation in mouse embryos. However, Ngn3 did not induce more downstream regulators of beta-cell differentiation, Nkx6.1 and Maf A. Coexpression of Nkx6.1 and Ngn3 induced endogenous expression of the insulin 2 gene, while coexpression of Maf A and Ngn3 induced both insulin 1 and 2 genes in AR42J-B13 cells. Our data demonstrated that Ngn3 expressed together with Nkx6.1 or MafA induces AR42J-B13 cells to differentiate into insulin-producing cells, supporting the use of these cells as a model system for studying beta-cell differentiation in vitro.
Collapse
Affiliation(s)
- Takeshi Ogihara
- Department of Medicine, Metabolism and Endocrinology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
293
|
Öström M, Loffler KA, Edfalk S, Selander L, Dahl U, Ricordi C, Jeon J, Correa-Medina M, Diez J, Edlund H. Retinoic acid promotes the generation of pancreatic endocrine progenitor cells and their further differentiation into beta-cells. PLoS One 2008; 3:e2841. [PMID: 18665267 PMCID: PMC2475501 DOI: 10.1371/journal.pone.0002841] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Accepted: 07/07/2008] [Indexed: 02/07/2023] Open
Abstract
The identification of secreted factors that can selectively stimulate the generation of insulin producing beta-cells from stem and/or progenitor cells represent a significant step in the development of stem cell-based beta-cell replacement therapy. By elucidating the molecular mechanisms that regulate the generation of beta-cells during normal pancreatic development such putative factors may be identified. In the mouse, beta-cells increase markedly in numbers from embryonic day (e) 14.5 and onwards, but the extra-cellular signal(s) that promotes the selective generation of beta-cells at these stages remains to be identified. Here we show that the retinoic acid (RA) synthesizing enzyme Raldh1 is expressed in developing mouse and human pancreas at stages when beta-cells are generated. We also provide evidence that RA induces the generation of Ngn3(+) endocrine progenitor cells and stimulates their further differentiation into beta-cells by activating a program of cell differentiation that recapitulates the normal temporal program of beta-cell differentiation.
Collapse
Affiliation(s)
- Maria Öström
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Kelly A. Loffler
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Sara Edfalk
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Lars Selander
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Ulf Dahl
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Jongmin Jeon
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Mayrin Correa-Medina
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Juan Diez
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
| | - Helena Edlund
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
294
|
Mesenchymal stem cells derived from human exocrine pancreas express transcription factors implicated in beta-cell development. Pancreas 2008; 37:75-84. [PMID: 18580448 DOI: 10.1097/mpa.0b013e31815fcb1e] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Transplantation of in vitro generated islets or insulin-producing cells represents an attractive option to overcome organ shortage. The aim of this study was to isolate, expand, and characterize cells from human exocrine pancreas and analyze their potential to differentiate into beta cells. METHODS Fibroblast-like cells growing out of human exocrine tissue were characterized by flow cytometry and by their capacity to differentiate into mesenchymal cell lineages. During cell expansion and after differentiation toward beta cells, expression of transcription factors of endocrine pancreatic progenitors was analyzed by reverse transcription polymerase chain reaction. RESULTS Cells emerged from 14/18 human pancreatic exocrine fractions and were expanded up to 40 population doublings. These cells displayed surface antigens similar to mesenchymal stem cells from bone marrow. A culture of these cells in adipogenic and chondrogenic differentiation media allowed differentiation into adipocyte- and chondrocyte-like cells. During expansion, cells expressed transcription factors implicated in islet development such as Isl1, Nkx2.2, Nkx6.1, nestin, Ngn3, Pdx1, and NeuroD. Activin A and hepatocyte growth factor induced an expression of insulin, glucagon, and glucokinase. CONCLUSIONS Proliferating cells with characteristics of mesenchymal stem cells and endocrine progenitors were isolated from exocrine tissue. Under specific conditions, these cells expressed little insulin. Human pancreatic exocrine tissue might thus be a source of endocrine cell progenitors.
Collapse
|
295
|
Abstract
The origin of insulin-expressing beta-cells in the adult mammalian pancreas is controversial. During normal tissue turnover and following injury, beta-cells may be replaced by duplication of existing beta-cells.1 However, an alternative source of beta-cells has recently been proposed based on neogenesis from a Ngn3-positive population present in regenerating pancreatic ducts.2 The appearance of beta-cells from Ngn3-positive progenitors is reminiscent of normal pancreas development, and Ngn3-expressing cells isolated from regenerating pancreas can generate the full repertoire of endocrine phenotypes. The isolation and characterisation of the equivalent human progenitors may represent a significant step forward in the hunt for a cure for diabetes.
Collapse
Affiliation(s)
- Kathy E O'Neill
- Centre for Regenerative Medicine, Department of Biology and Biochemistry, University of Bath, UK
| | | | | |
Collapse
|
296
|
Best M, Carroll M, Hanley NA, Piper Hanley K. Embryonic stem cells to beta-cells by understanding pancreas development. Mol Cell Endocrinol 2008; 288:86-94. [PMID: 18487011 DOI: 10.1016/j.mce.2008.03.008] [Citation(s) in RCA: 24] [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: 12/08/2007] [Revised: 03/18/2008] [Accepted: 03/18/2008] [Indexed: 12/25/2022]
Abstract
Insulin injections treat but do not cure Type 1 diabetes (T1DM). The success of islet transplantation suggests cell replacement therapies may offer a curative strategy. However, cadaver islets are of insufficient number for this to become a widespread treatment. To address this deficiency, the production of beta-cells from pluripotent stem cells offers an ambitious far-sighted opportunity. Recent progress in generating insulin-producing cells from embryonic stem cells has shown promise, highlighting the potential of trying to mimic normal developmental pathways. Here, we provide an overview of the current methodology that has been used to differentiate stem cells toward a beta-cell fate. Parallels are drawn with what is known about normal development, especially regarding the human pancreas.
Collapse
Affiliation(s)
- Marie Best
- Centre for Human Development, Stem Cells & Regeneration, UK
| | | | | | | |
Collapse
|
297
|
Hanley NA, Hanley KP, Miettinen PJ, Otonkoski T. Weighing up beta-cell mass in mice and humans: self-renewal, progenitors or stem cells? Mol Cell Endocrinol 2008; 288:79-85. [PMID: 18450368 DOI: 10.1016/j.mce.2008.03.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 03/07/2008] [Accepted: 03/07/2008] [Indexed: 01/09/2023]
Abstract
Understanding how beta-cells maintain themselves in the adult pancreas is important for prioritizing strategies aimed at ameliorating or ideally curing different forms of diabetes. There has been much debate over whether beta-cell proliferation, as a means of self-renewal, predominates over the existence and differentiation of a pancreatic stem cell or progenitor cell population. This article describes the two opposing positions based largely on research in laboratory rodents and its extrapolation to humans.
Collapse
Affiliation(s)
- Neil A Hanley
- Centre for Human Development, Stem Cells & Regeneration, University of Southampton, Southampton, UK.
| | | | | | | |
Collapse
|
298
|
Kokubu H, Ohtsuka T, Kageyama R. Mash1 is required for neuroendocrine cell development in the glandular stomach. Genes Cells 2008; 13:41-51. [PMID: 18173746 DOI: 10.1111/j.1365-2443.2007.01146.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In the epithelium of the developing glandular stomach, neuroendocrine cells differentiate from common progenitors, but the mechanism of how these cells are specified remains to be determined. Here, we show that the basic helix-loop-helix (bHLH) gene, mammalian achaete-scute homologue 1 (Mash1), is highly expressed in the glandular stomach epithelium. In Mash1-null mice, almost all gastric neuroendocrine cells are missing, whereas development of non-neuroendocrine cells is not significantly affected. The bHLH gene Neurogenin3 (Ngn3), which is known to regulate formation of subsets of gastric neuroendocrine cells (gastrin-, glucagon- and somatostatin-producing cells), is expressed normally in the Mash1-null stomach. Thus, Ngn3 alone is not sufficient but Mash1 is additionally required for the differentiation of these neuroendocrine cells. Taken together, these results indicate that formation of gastrin-, glucagon- and somatostatin-producing cells depends on both Mash1 and Ngn3, while that of other neuroendocrine cells depends on Mash1 alone, suggesting that combinations of bHLH genes may contribute to cell type diversity.
Collapse
Affiliation(s)
- Hiroshi Kokubu
- Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | | |
Collapse
|
299
|
Lynn FC, Sanchez L, Gomis R, German MS, Gasa R. Identification of the bHLH factor Math6 as a novel component of the embryonic pancreas transcriptional network. PLoS One 2008; 3:e2430. [PMID: 18560595 PMCID: PMC2413403 DOI: 10.1371/journal.pone.0002430] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Accepted: 05/07/2008] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Basic helix-loop-helix (bHLH) transcription factors play important roles in differentiation processes during embryonic development of vertebrates. In the pancreas, the atonal-related bHLH gene Neurogenin3 (Neurog3) controls endocrine cell fate specification in uncommitted progenitor cells. Therefore, it is likely that Neurog3-regulated factors will have important functions during pancreatic endocrine cell differentiation. The gene for the atonal-related bHLH factor Math6 was recognized as a potential target of Neurog3 in a genomic scale profiling during endocrine differentiation. Herein we have explored the role of Math6 during endocrine pancreas development. RESULTS We demonstrate that the Math6 gene is a direct target of Neurog3 in vitro and that, during mouse development, Math6 is expressed in both endocrine and exocrine pancreatic precursor cells. We have investigated the role of Math6 in endocrine differentiation by over-expressing this factor in pancreatic duct cells. Math6 possesses intrinsic transcriptional repressor activity and, in contrast to Neurog3 it does not induce the endocrine differentiation program; however, it can modulate some of the pro-endocrine functions of Neurog3 in this system. In addition, we show that Math6 is broadly expressed in mouse embryonic tissues and its expression is induced by tissue-specific bHLH genes other than Neurog3. Furthermore, inactivation of the Math6 gene in the mouse results in early embryonic lethality demonstrating an essential role of this factor in organismal development. CONCLUSIONS These data demonstrate that Math6 is a novel component of the pancreatic transcriptional network during embryonic development and suggest a potential role for Math6 as a modulator of the differentiation program initiated by the pro-endocrine factor Neurog3. Furthermore, our results demonstrate that Math6 is indispensable for early embryonic development and indicate a more widespread function for this factor in tissue-specific differentiation processes that are dependent on class II bHLH genes.
Collapse
Affiliation(s)
- Francis C. Lynn
- Diabetes Center, Hormone Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Lidia Sanchez
- Diabetes and Obesity Laboratory-Endocrinology and Nutrition Unit, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Ramon Gomis
- Diabetes and Obesity Laboratory-Endocrinology and Nutrition Unit, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Michael S. German
- Diabetes Center, Hormone Research Institute, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Rosa Gasa
- Diabetes and Obesity Laboratory-Endocrinology and Nutrition Unit, Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- * E-mail:
| |
Collapse
|
300
|
Gao X, Song L, Shen K, Wang H, Niu W, Qin X. Transplantation of bone marrow derived cells promotes pancreatic islet repair in diabetic mice. Biochem Biophys Res Commun 2008; 371:132-7. [DOI: 10.1016/j.bbrc.2008.04.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Accepted: 04/03/2008] [Indexed: 01/30/2023]
|