1
|
Hu C, Chen Y, Yin X, Xu R, Yin C, Wang C, Zhao Y. Pancreatic endocrine and exocrine signaling and crosstalk in physiological and pathological status. Signal Transduct Target Ther 2025; 10:39. [PMID: 39948335 PMCID: PMC11825823 DOI: 10.1038/s41392-024-02098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/20/2024] [Accepted: 12/03/2024] [Indexed: 02/16/2025] Open
Abstract
The pancreas, an organ with dual functions, regulates blood glucose levels through the endocrine system by secreting hormones such as insulin and glucagon. It also aids digestion through the exocrine system by secreting digestive enzymes. Complex interactions and signaling mechanisms between the endocrine and exocrine functions of the pancreas play a crucial role in maintaining metabolic homeostasis and overall health. Compelling evidence indicates direct and indirect crosstalk between the endocrine and exocrine parts, influencing the development of diseases affecting both. From a developmental perspective, the exocrine and endocrine parts share the same origin-the "tip-trunk" domain. In certain circumstances, pancreatic exocrine cells may transdifferentiate into endocrine-like cells, such as insulin-secreting cells. Additionally, several pancreatic diseases, including pancreatic cancer, pancreatitis, and diabetes, exhibit potential relevance to both endocrine and exocrine functions. Endocrine cells may communicate with exocrine cells directly through cytokines or indirectly by regulating the immune microenvironment. This crosstalk affects the onset and progression of these diseases. This review summarizes the history and milestones of findings related to the exocrine and endocrine pancreas, their embryonic development, phenotypic transformations, signaling roles in health and disease, the endocrine-exocrine crosstalk from the perspective of diseases, and potential therapeutic targets. Elucidating the regulatory mechanisms of pancreatic endocrine and exocrine signaling and provide novel insights for the understanding and treatment of diseases.
Collapse
Grants
- National High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
- cNational High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
Collapse
Affiliation(s)
- Chenglin Hu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chenxue Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chengcheng Wang
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
- Institute of Clinical Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
2
|
Zhao H, Zhou B. Lineage tracing of pancreatic cells for mechanistic and therapeutic insights. Trends Endocrinol Metab 2025:S1043-2760(24)00330-8. [PMID: 39828453 DOI: 10.1016/j.tem.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
Recent advances in lineage-tracing technologies have significantly improved our understanding of pancreatic cell biology, particularly in elucidating the ontogeny and regenerative capacity of pancreatic cells. A deeper appreciation of the mechanisms underlying pancreatic cell identity and plasticity holds the potential to inform the development of new therapeutic modalities for conditions such as diabetes and pancreatitis. With this goal in mind, here we summarize advances, challenges, and future directions in tracing pancreatic cell origins and fates using lineage-tracing technologies. Given their essential role for blood glucose regulation, we pay particular attention on the insights gained from endocrine cells, especially β-cells.
Collapse
Affiliation(s)
- Huan Zhao
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Bin Zhou
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
3
|
Abstract
The pancreas is made from two distinct components: the exocrine pancreas, a reservoir of digestive enzymes, and the endocrine islets, the source of the vital metabolic hormone insulin. Human islets possess limited regenerative ability; loss of islet β-cells in diseases such as type 1 diabetes requires therapeutic intervention. The leading strategy for restoration of β-cell mass is through the generation and transplantation of new β-cells derived from human pluripotent stem cells. Other approaches include stimulating endogenous β-cell proliferation, reprogramming non-β-cells to β-like cells, and harvesting islets from genetically engineered animals. Together these approaches form a rich pipeline of therapeutic development for pancreatic regeneration.
Collapse
|
4
|
Lima MJ, Muir KR, Docherty HM, McGowan NWA, Forbes S, Heremans Y, Heimberg H, Casey J, Docherty K. Generation of Functional Beta-Like Cells from Human Exocrine Pancreas. PLoS One 2016; 11:e0156204. [PMID: 27243814 PMCID: PMC4887015 DOI: 10.1371/journal.pone.0156204] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/10/2016] [Indexed: 12/24/2022] Open
Abstract
Transcription factor mediated lineage reprogramming of human pancreatic exocrine tissue could conceivably provide an unlimited supply of islets for transplantation in the treatment of diabetes. Exocrine tissue can be efficiently reprogrammed to islet-like cells using a cocktail of transcription factors: Pdx1, Ngn3, MafA and Pax4 in combination with growth factors. We show here that overexpression of exogenous Pax4 in combination with suppression of the endogenous transcription factor ARX considerably enhances the production of functional insulin-secreting β-like cells with concomitant suppression of α-cells. The efficiency was further increased by culture on laminin-coated plates in media containing low glucose concentrations. Immunocytochemistry revealed that reprogrammed cultures were composed of ~45% islet-like clusters comprising >80% monohormonal insulin+ cells. The resultant β-like cells expressed insulin protein levels at ~15–30% of that in adult human islets, efficiently processed proinsulin and packaged insulin into secretory granules, exhibited glucose responsive insulin secretion, and had an immediate and prolonged effect in normalising blood glucose levels upon transplantation into diabetic mice. We estimate that approximately 3 billion of these cells would have an immediate therapeutic effect following engraftment in type 1 diabetes patients and that one pancreas would provide sufficient tissue for numerous transplants.
Collapse
Affiliation(s)
- Maria J. Lima
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
- * E-mail:
| | - Kenneth R. Muir
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Hilary M. Docherty
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Neil W. A. McGowan
- Department of Surgery, University of Edinburgh, Edinburgh Royal Infirmary, Edinburgh, EH16 4SU, United Kingdom
| | - Shareen Forbes
- Endocrinology Unit, University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Yves Heremans
- Diabetes Research Centre, Vrije Universiteit Brussel, B1090 Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Centre, Vrije Universiteit Brussel, B1090 Brussels, Belgium
| | - John Casey
- Department of Surgery, University of Edinburgh, Edinburgh Royal Infirmary, Edinburgh, EH16 4SU, United Kingdom
| | - Kevin Docherty
- School of Medical Sciences, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| |
Collapse
|
5
|
Seyedi F, Farsinejad A, Nematollahi-Mahani SA, Eslaminejad T, Nematollahi-Mahani SN. Suspension Culture Alters Insulin Secretion in Induced Human Umbilical Cord Matrix-Derived Mesenchymal Cells. CELL JOURNAL 2016; 18:52-61. [PMID: 27054119 PMCID: PMC4819386 DOI: 10.22074/cellj.2016.3987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 06/29/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Worldwide, diabetes mellitus (DM) is an ever-increasing metabolic disorder. A promising approach to the treatment of DM is the implantation of insulin producing cells (IPC) that have been derived from various stem cells. Culture conditions play a pivotal role in the quality and quantity of the differentiated cells. In this experimental study, we have applied various culture conditions to differentiate human umbilical cord matrix-derived mesenchymal cells (hUCMs) into IPCs and measured insulin production. MATERIALS AND METHODS In this experimental study, we exposed hUCMs cells to pancreatic medium and differentiated them into IPCs in monolayer and suspension cultures. Pancreatic medium consisted of serum-free Dulbecco's modified eagle's medium Nutrient mixture F12 (DMEM/F12) medium with 17.5 mM glucose supplemented by 10 mM nicotinamide, 10 nM exendin-4, 10 nM pentagastrin, 100 pM hepatocyte growth factor, and B-27 serum-free supplement. After differentiation, insulin content was analyzed by gene expression, immunocytochemistry (IHC) and the chemiluminesence immunoassay (CLIA). RESULTS Reverse transcription-polymerase chain reaction (RT-PCR) showed efficient expressions of NKX2.2, PDX1 and INSULIN genes in both groups. IHC analysis showed higher expression of insulin protein in the hanging drop group, and CLIA revealed a significant higher insulin production in hanging drops compared with the monolayer group following the glucose challenge test. CONCLUSION We showed by this novel, simple technique that the suspension culture played an important role in differentiation of hUCMs into IPC. This culture was more efficient than the conventional culture method commonly used in IPC differentiation and cultivation.
Collapse
Affiliation(s)
- Fatemeh Seyedi
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Farsinejad
- Stem Cell Research Lab, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Touba Eslaminejad
- Pharmaceutics Research Center (PRC), Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed Noureddin Nematollahi-Mahani
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Afzal Research Institute, Kerman, Iran
| |
Collapse
|
6
|
Jiang FX, Morahan G. Multipotent pancreas progenitors: Inconclusive but pivotal topic. World J Stem Cells 2015; 7:1251-1261. [PMID: 26730269 PMCID: PMC4691693 DOI: 10.4252/wjsc.v7.i11.1251] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/20/2015] [Accepted: 11/11/2015] [Indexed: 02/07/2023] Open
Abstract
The establishment of multipotent pancreas progenitors (MPP) should have a significant impact not only on the ontology of the pancreas, but also for the translational research of glucose-responding endocrine β-cells. Deficiency of the latter may lead to the pandemic type 1 or type 2 diabetes mellitus, a metabolic disorder. An ideal treatment of which would potentially be the replacement of destroyed or failed β-cells, by restoring function of endogenous pancreatic endocrine cells or by transplantation of donor islets or in vitro generated insulin-secreting cells. Thus, considerable research efforts have been devoted to identify MPP candidates in the pre- and post-natal pancreas for the endogenous neogenesis or regeneration of endocrine insulin-secreting cells. In order to advance this inconclusive but critical field, we here review the emerging concepts, recent literature and newest developments of potential MPP and propose measures that would assist its forward progression.
Collapse
|
7
|
Nukaya D, Minami K, Hoshikawa R, Yokoi N, Seino S. Preferential gene expression and epigenetic memory of induced pluripotent stem cells derived from mouse pancreas. Genes Cells 2015; 20:367-81. [PMID: 25727848 DOI: 10.1111/gtc.12227] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 12/31/2014] [Indexed: 11/27/2022]
Abstract
Induced pluripotent stem cells (iPSCs) have been established from various somatic cell types. Accumulating evidence suggests that iPSCs from different cell sources have distinct molecular and functional properties. Here, we establish iPSC derived from mouse pancreas (Panc-iPSC) and compared their properties with those of iPSC derived from tail-tip fibroblast (TTF-iPSC). The metabolic profile differs between Panc-iPSC and TTF-iPSC, indicating distinct cell properties in these iPSCs. Expression of Pdx1, a marker of pancreas differentiation, is increased through formation of embryoid body (EB) in Panc-iPSC, but the level is similar to that in TTF-iPSC. In contrast, EBs derived from Panc-iPSC express liver-specific albumin (Alb) and alpha-fetoprotein (Afp) genes much more strongly than those from TTF-iPSC. Epigenetic analysis shows a different histone modification pattern between Panc-iPSC and TTF-iPSC. Promoter regions of Alb and Afp genes in Panc-iPSC are suggested to have a more open chromatin structure than those in TTF-iPSC, which also is seen in primary cultured pancreatic cells. Our data suggest that Panc-iPSC possesses distinct differentiation capacity from that of TTF-PSC, which may be influenced by epigenetic memory.
Collapse
Affiliation(s)
- Daiki Nukaya
- Division of Cellular and Molecular Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; Regenerative and Cellular Medicine Office, Sumitomo Dainippon Pharma Co., Ltd, 2-2-2 Minatojimaminami-machi, Chuo-ku, Kobe, 650-0047, Japan
| | | | | | | | | |
Collapse
|
8
|
Abstract
Diabetes mellitus is caused by absolute (type 1) or relative (type 2) deficiency of insulin-secreting islet β cells. An ideal treatment of diabetes would, therefore, be to replace the lost or deficient β cells, by transplantation of donated islets or differentiated endocrine cells or by regeneration of endogenous islet cells. Due to their ability of unlimited proliferation and differentiation into all functional lineages in our body, including β cells, embryonic stem cells and induced pluripotent stem cells are ideally placed as cell sources for a diabetic transplantation therapy. Unfortunately, the inability to generate functional differentiated islet cells from pluripotent stem cells and the poor availability of donor islets have severely restricted the broad clinical use of the replacement therapy. Therefore, endogenous sources that can be directed to becoming insulin-secreting cells are actively sought after. In particular, any cell types in the developing or adult pancreas that may act as pancreatic stem cells (PSC) would provide an alternative renewable source for endogenous regeneration. In this review, we will summarize the latest progress and knowledge of such PSC, and discuss ways that facilitate the future development of this often controversial, but crucial research.
Collapse
Affiliation(s)
- Fang-Xu Jiang
- 1 Islet Cell Development Program, Harry Perkins Institute of Medical Research, and Centre for Medical Research, The University of Western Australia , Perth, Australia
| | | |
Collapse
|
9
|
Minami K, Seino S. Current status of regeneration of pancreatic β-cells. J Diabetes Investig 2014; 4:131-41. [PMID: 24843642 PMCID: PMC4019265 DOI: 10.1111/jdi.12062] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 01/21/2013] [Indexed: 12/13/2022] Open
Abstract
Newly generated insulin‐secreting cells for use in cell therapy for insulin‐deficient diabetes mellitus require properties similar to those of native pancreatic β‐cells. Pancreatic β‐cells are highly specialized cells that produce a large amount of insulin, and secrete insulin in a regulated manner in response to glucose and other stimuli. It is not yet explained how the β‐cells acquire this complex function during normal differentiation. So far, in vitro generation of insulin‐secreting cells from embryonic stem cells, induced‐pluripotent stem cells and adult stem/progenitor‐like cells has been reported. However, most of these cells are functionally immature and show poor glucose‐responsive insulin secretion compared to that of native pancreatic β‐cells (or islets). Strategies to generate functional β‐cells or a whole organ in vivo have also recently been proposed. Establishing a protocol to generate fully functional insulin‐secreting cells that closely resemble native β‐cells is a critical matter in regenerative medicine for diabetes. Understanding the physiological processes of differentiation, proliferation and regeneration of pancreatic β‐cells might open the path to cell therapy to cure patients with absolute insulin deficiency.
Collapse
Affiliation(s)
- Kohtaro Minami
- Division of Cellular and Molecular Medicine Department of Physiology and Cell Biology Kobe University Graduate School of Medicine Kobe Japan
| | - Susumu Seino
- Division of Cellular and Molecular Medicine Department of Physiology and Cell Biology Kobe University Graduate School of Medicine Kobe Japan ; Division of Diabetes and Endocrinology Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan ; Core Research for Evolutional Science and Technology (CREST) Japan Science and Technology Corp. Kawaguchi Saitama Japan
| |
Collapse
|
10
|
Abstract
Aims/Introduction: Studies have suggested that pancreatic β‐cells undergo dedifferentiation during proliferation in vitro. However, due to limitations of the methodologies used, the question remains whether such dedifferentiated cells can redifferentiate into β‐cells. Materials and Methods: We have established a method for cell tracing in combination with fluorescence‐activated cell sorter (FACS). Using this method, mouse pancreatic β‐cells labeled with green fluorescent protein (GFP) under the control of the insulin promoter are collected by FACS. These β‐cells can be traced and characterized throughout the culture process, even when insulin becomes undetectable, because the cells are also marked with monomeric red fluorescent protein (mRFP) driven by the CAG promoter. Results: When cultured with fetal mouse pancreatic cells, FACS sorted β‐cells lost GFP expression, but retained mRFP expression. The cells also lost expressions of genes characteristic of the β‐cell phenotype, such as Pdx1 and glucokinase, indicating dedifferentiation. More than 30% of such dedifferentiated pancreatic β‐cells were detected in S or G2/M phase. Furthermore, these dedifferentiated cells redifferentiated into insulin‐expressing cells on cultivation with a MEK1/2 inhibitor. Conclusions: Our data provide direct evidence that pre‐existing β‐cells can undergo dedifferentiation and redifferentiation in vitro, their phenotype is reversible and that dedifferentiation in β‐cells is associated with progression of the cell cycle. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2010.00051.x, 2010)
Collapse
Affiliation(s)
- Kohtaro Minami
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology ; Laboratory for Somatic Stem Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe
| | - Kazumasa Miyawaki
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology
| | - Manami Hara
- Department of Medicine, University of Chicago, Chicago, Il, USA
| | - Shuichi Yamada
- Animal Research Laboratory, Bioscience Research and Education Center, Akita University, Akita
| | - Susumu Seino
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine ; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Corp., Kawaguchi, Saitama, Japan
| |
Collapse
|
11
|
Iwasaki M, Minami K, Shibasaki T, Miki T, Miyazaki JI, Seino S. Establishment of new clonal pancreatic β-cell lines (MIN6-K) useful for study of incretin/cyclic adenosine monophosphate signaling. J Diabetes Investig 2014; 1:137-42. [PMID: 24843422 PMCID: PMC4008005 DOI: 10.1111/j.2040-1124.2010.00026.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Incretin/cyclic adenosine monophosphate (cAMP) signaling is critical for potentiation of insulin secretion. Although several cell lines of pancreatic β‐cells are currently available, there are no cell lines suitable for investigation of incretin/cAMP signaling. In the present study, we have newly established pancreatic β‐cell lines (named MIN6‐K) from the IT6 mouse, which develops insulinoma. MIN6‐K8 cells respond to both glucose and incretins, such as glucagon‐like peptide‐1 (GLP‐1) and glucose‐dependent insulinotropic polypeptide (GIP), as is the case in pancreatic islets, whereas MIN6‐K20 cells respond to glucose, but not to incretins. Despite the difference in incretin‐potentiated insulin secretion between these two cell lines, the accumulation of cAMP after stimulation of GLP‐1 is comparable in these cells. Interestingly, we also found that incretin responsiveness is drastically induced by the formation of pseudoislets from MIN6‐K20 cells to a level comparable to that of pancreatic islets. Thus, these cell lines are useful for studying incretin/cAMP signaling in β‐cells. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2010.00026.x, 2010)
Collapse
Affiliation(s)
- Masahiro Iwasaki
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe
| | - Kohtaro Minami
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe
| | - Tadao Shibasaki
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe
| | - Takashi Miki
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe ; Department of Autonomic Physiology, Graduate School of Medicine, Chiba University, Chiba
| | - Jun-Ichi Miyazaki
- Department of Nutrition and Physiological Chemistry, Osaka University Graduate School of Medicine, Osaka
| | - Susumu Seino
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe ; Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe ; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
12
|
Minami K, Doi R, Kawaguchi Y, Nukaya D, Hagiwara Y, Noguchi H, Matsumoto S, Seino S. In vitro generation of insulin-secreting cells from human pancreatic exocrine cells. J Diabetes Investig 2014; 2:271-5. [PMID: 24843497 PMCID: PMC4014966 DOI: 10.1111/j.2040-1124.2010.00095.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Transplantation of surrogate β‐cells is a promising option for the treatment of insulin‐deficient diabetes mellitus in the future. Although pancreatic exocrine cells of rodents have been shown to transdifferentiate into insulin‐secreting cells, no studies are reported on human exocrine cells. Here, we report the generation of insulin‐secreting cells from exocrine cells of the human pancreas. When cultured in suspension with epidermal growth factor, human pancreatic exocrine cells readily formed spherical cell clusters. Expression of Pdx1 was induced in all 19 cases in which we successfully isolated exocrine cells, and insulin expression was induced in 11 cases. In addition, insulin secretion was evaluated in four cases, and the newly‐made cells were found to secrete insulin in response to various stimuli. Although further studies are required to improve both the quality and quantity of such insulin‐secreting cells, our data suggest that pancreatic exocrine cells represent a potential source of insulin‐secreting cells for treatment of type 1 diabetes. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2010.00095.x, 2011)
Collapse
Affiliation(s)
- Kohtaro Minami
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology ; Laboratory for Somatic Stem Cell Therapy, Foundation for Biomedical Research and Innovation, Kobe
| | - Ryuichiro Doi
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Kyoto University, Kyoto
| | - Yoshiya Kawaguchi
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Kyoto University, Kyoto
| | - Daiki Nukaya
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology
| | - Yoshiaki Hagiwara
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology
| | - Hirofumi Noguchi
- Baylor All Saints Medical Center, Baylor Research Institute, Fort Worth ; Institute of Biomedical Studies, Baylor University, Waco, TX, USA
| | | | - Susumu Seino
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology ; Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine ; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Corp., Kawaguchi, Saitama, Japan
| |
Collapse
|
13
|
de Back W, Zimm R, Brusch L. Transdifferentiation of pancreatic cells by loss of contact-mediated signaling. BMC SYSTEMS BIOLOGY 2013; 7:77. [PMID: 23938152 PMCID: PMC3751562 DOI: 10.1186/1752-0509-7-77] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 07/04/2013] [Indexed: 12/01/2022]
Abstract
Background Replacement of dysfunctional β-cells in the islets of Langerhans by transdifferentiation of pancreatic acinar cells has been proposed as a regenerative therapy for diabetes. Adult acinar cells spontaneously revert to a multipotent state upon tissue dissociation in vitro and can be stimulated to redifferentiate into β-cells. Despite accumulating evidence that contact-mediated signals are involved, the mechanisms regulating acinar-to-islet cell transdifferentiation remain poorly understood. Results In this study, we propose that the crosstalk between two contact-mediated signaling mechanisms, lateral inhibition and lateral stabilization, controls cell fate stability and transdifferentiation of pancreatic cells. Analysis of a mathematical model combining gene regulation with contact-mediated signaling reveals the multistability of acinar and islet cell fates. Inhibition of one or both modes of signaling results in transdifferentiation from the acinar to the islet cell fate, either by dedifferentiation to a multipotent state or by direct lineage switching. Conclusions This study provides a theoretical framework to understand the role of contact-mediated signaling in pancreatic cell fate control that may help to improve acinar-to-islet cell transdifferentiation strategies for β-cell neogenesis.
Collapse
Affiliation(s)
- Walter de Back
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, 01062, Germany
| | | | | |
Collapse
|
14
|
de Back W, Zhou JX, Brusch L. On the role of lateral stabilization during early patterning in the pancreas. J R Soc Interface 2013. [PMID: 23193107 DOI: 10.1098/rsif.2012.0766] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The cell fate decision of multi-potent pancreatic progenitor cells between the exocrine and endocrine lineages is regulated by Notch signalling, mediated by cell-cell interactions. However, canonical models of Notch-mediated lateral inhibition cannot explain the scattered spatial distribution of endocrine cells and the cell-type ratio in the developing pancreas. Based on evidence from acinar-to-islet cell transdifferentiation in vitro, we propose that lateral stabilization, i.e. positive feedback between adjacent progenitor cells, acts in parallel with lateral inhibition to regulate pattern formation in the pancreas. A simple mathematical model of transcriptional regulation and cell-cell interaction reveals the existence of multi-stability of spatial patterns whose simultaneous occurrence causes scattering of endocrine cells in the presence of noise. The scattering pattern allows for control of the endocrine-to-exocrine cell-type ratio by modulation of lateral stabilization strength. These theoretical results suggest a previously unrecognized role for lateral stabilization in lineage specification, spatial patterning and cell-type ratio control in organ development.
Collapse
Affiliation(s)
- Walter de Back
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
| | | | | |
Collapse
|
15
|
Furuya F, Shimura H, Asami K, Ichijo S, Takahashi K, Kaneshige M, Oikawa Y, Aida K, Endo T, Kobayashi T. Ligand-bound thyroid hormone receptor contributes to reprogramming of pancreatic acinar cells into insulin-producing cells. J Biol Chem 2013; 288:16155-66. [PMID: 23595988 DOI: 10.1074/jbc.m112.438192] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
One goal of diabetic regenerative medicine is to instructively convert mature pancreatic exocrine cells into insulin-producing cells. We recently reported that ligand-bound thyroid hormone receptor α (TRα) plays a critical role in expansion of the β-cell mass during postnatal development. Here, we used an adenovirus vector that expresses TRα driven by the amylase 2 promoter (AdAmy2TRα) to induce the reprogramming of pancreatic acinar cells into insulin-producing cells. Treatment with l-3,5,3-triiodothyronine increases the association of TRα with the p85α subunit of phosphatidylinositol 3-kinase (PI3K), leading to the phosphorylation and activation of Akt and the expression of Pdx1, Ngn3, and MafA in purified acinar cells. Analyses performed with the lectin-associated cell lineage tracing system and the Cre/loxP-based direct cell lineage tracing system indicate that newly synthesized insulin-producing cells originate from elastase-expressing pancreatic acinar cells. Insulin-containing secretory granules were identified in these cells by electron microscopy. The inhibition of p85α expression by siRNA or the inhibition of PI3K by LY294002 prevents the expression of Pdx1, Ngn3, and MafA and the reprogramming to insulin-producing cells. In immunodeficient mice with streptozotocin-induced hyperglycemia, treatment with AdAmy2TRα leads to the reprogramming of pancreatic acinar cells to insulin-producing cells in vivo. Our findings suggest that ligand-bound TRα plays a critical role in β-cell regeneration during postnatal development via activation of PI3K signaling.
Collapse
Affiliation(s)
- Fumihiko Furuya
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo-shi, Yamanashi 409-3898, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Wauters E, Sanchez-Arévalo Lobo VJ, Pinho AV, Mawson A, Herranz D, Wu J, Cowley MJ, Colvin EK, Njicop EN, Sutherland RL, Liu T, Serrano M, Bouwens L, Real FX, Biankin AV, Rooman I. Sirtuin-1 regulates acinar-to-ductal metaplasia and supports cancer cell viability in pancreatic cancer. Cancer Res 2013; 73:2357-67. [PMID: 23370328 DOI: 10.1158/0008-5472.can-12-3359] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The exocrine pancreas can undergo acinar-to-ductal metaplasia (ADM), as in the case of pancreatitis where precursor lesions of pancreatic ductal adenocarcinoma (PDAC) can arise. The NAD(+)-dependent protein deacetylase Sirtuin-1 (Sirt1) has been implicated in carcinogenesis with dual roles depending on its subcellular localization. In this study, we examined the expression and the role of Sirt1 in different stages of pancreatic carcinogenesis, i.e. ADM models and established PDAC. In addition, we analyzed the expression of KIAA1967, a key mediator of Sirt1 function, along with potential Sirt1 downstream targets. Sirt1 was co-expressed with KIAA1967 in the nuclei of normal pancreatic acinar cells. In ADM, Sirt1 underwent a transient nuclear-to-cytoplasmic shuttling. Experiments where during ADM, we enforced repression of Sirt1 shuttling, inhibition of Sirt1 activity or modulation of its expression, all underscore that the temporary decrease of nuclear and increase of cytoplasmic Sirt1 stimulate ADM. Our results further underscore that important transcriptional regulators of acinar differentiation, that is, Pancreatic transcription factor-1a and β-catenin can be deacetylated by Sirt1. Inhibition of Sirt1 is effective in suppression of ADM and in reducing cell viability in established PDAC tumors. KIAA1967 expression is differentially downregulated in PDAC and impacts on the sensitivity of PDAC cells to the Sirt1/2 inhibitor Tenovin-6. In PDAC, acetylation of β-catenin is not affected, unlike p53, a well-characterized Sirt1-regulated protein in tumor cells. Our results reveal that Sirt1 is an important regulator and potential therapeutic target in pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Elke Wauters
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Type 1 and some forms of type 2 diabetes mellitus are caused by deficiency of insulin-secretory islet β cells. An ideal treatment for these diseases would therefore be to replace β cells, either by transplanting donated islets or via endogenous regeneration (and controlling the autoimmunity in type 1 diabetes). Unfortunately, the poor availability of donor islets has severely restricted the broad clinical use of islet transplantation. The ability to differentiate embryonic stem cells into insulin-expressing cells initially showed great promise, but the generation of functional β cells has proven extremely difficult and far slower than originally hoped. Pancreatic stem cells (PSC) or transdifferentiation of other cell types in the pancreas may hence provide an alternative renewable source of surrogate β cells. However, the existence of PSC has been hotly debated for many years. In this review, we will discuss the latest development and future perspectives of PSC research, giving readers an overview of this controversial but important area.
Collapse
Affiliation(s)
- Fang-Xu Jiang
- Centre for Diabetes Research, Western Australian Institute for Medical Research, The University of Western Australia, 50 Murray St (Rear), Perth, WA 6000, Australia.
| | | |
Collapse
|
18
|
Gallego-Perez D, Higuita-Castro N, Reen RK, Palacio-Ochoa M, Sharma S, Lee LJ, Lannutti JJ, Hansford DJ, Gooch KJ. Micro/nanoscale technologies for the development of hormone-expressing islet-like cell clusters. Biomed Microdevices 2012; 14:779-89. [PMID: 22573223 DOI: 10.1007/s10544-012-9657-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin-expressing islet-like cell clusters derived from precursor cells have significant potential in the treatment of type-I diabetes. Given that cluster size and uniformity are known to influence islet cell behavior, the ability to effectively control these parameters could find applications in the development of anti-diabetic therapies. In this work, we combined micro and nanofabrication techniques to build a biodegradable platform capable of supporting the formation of islet-like structures from pancreatic precursors. Soft lithography and electrospinning were used to create arrays of microwells (150-500 μm diameter) structurally interfaced with a porous sheet of micro/nanoscale polyblend fibers (~0.5-10 μm in cross-sectional size), upon which human pancreatic ductal epithelial cells anchored and assembled into insulin-expressing 3D clusters. The microwells effectively regulated the spatial distribution of the cells on the platform, as well as cluster size, shape and homogeneity. Average cluster cross-sectional area (~14000-17500 μm(2)) varied in proportion to the microwell dimensions, and mean circularity values remained above 0.7 for all microwell sizes. In comparison, clustering on control surfaces (fibers without microwells or tissue culture plastic) resulted in irregularly shaped/sized cell aggregates. Immunoreactivity for insulin, C-peptide and glucagon was detected on both the platform and control surfaces; however, intracellular levels of C-peptide/cell were ~60 % higher on the platform.
Collapse
Affiliation(s)
- Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Insulin-producing cells from human pancreatic islet-derived progenitor cells following transplantation in mice. Cell Biol Int 2011; 35:483-90. [PMID: 21080910 DOI: 10.1042/cbi20100152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Stem/progenitor cells hold promise for alleviating/curing type 1 diabetes due to the capacity to differentiate into functional insulin-producing cells. The current study aims to assess the differentiation potential of human pancreatic IPCs (islet-derived progenitor cells). IPCs were derived from four human donors and subjected to more than 2000-fold expansion before turning into ICCs (islet-like cell clusters). The ICCs expressed ISL-1 Glut2, PDX-1, ngn3, insulin, glucagon and somatostatin at the mRNA level and stained positive for insulin and glucagon by immunofluorescence. Following glucose challenge in vitro, C-peptide was detected in the sonicated ICCs, instead of in the conditioned medium. To examine the function of the cells in vivo, IPCs or ICCs were transplanted under the renal capsule of immunodeficient mice. One month later, 19 of 28 mice transplanted with ICCs and 4 of 14 mice with IPCs produced human C-peptide detectable in blood, indicating that the in vivo environment further facilitated the maturation of ICCs. However, among the hormone-positive mice, only 9 of 19 mice with ICCs and two of four mice with IPCs were able to secrete C-peptide in response to glucose.
Collapse
|
20
|
Takahashi R, Sonoda H, Tabata Y, Hisada A. Formation of hepatocyte spheroids with structural polarity and functional bile canaliculi using nanopillar sheets. Tissue Eng Part A 2010; 16:1983-95. [PMID: 20100035 DOI: 10.1089/ten.tea.2009.0662] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We developed a method for controlling the spheroid formation of adult rat primary hepatocytes simply by optimizing the pillar diameters and patterns of nanopillar sheets. To investigate the effects of the pillar parameters on the spheroid formation, rat primary hepatocytes were cultured on nanopillar sheets with pillars that had one of five different diameters and that had been precoated with a solution containing one of two different concentrations of type I collagen. Spheroids with a compact morphology that were adhesive to the substratum and had an optimal size (50 to 100 microm) were obtained using a sheet with a pillar diameter of 2.0 microm that was precoated with 100 ng/mL of type I collagen solution. Immunohistochemistry revealed that the spheroids had a structure similar to that of native liver tissue. We then assessed the effect of overlaying reconstituted spheroids with Matrigel with the aim of achieving a simulated in vivo environment. The mRNA expression levels of MRP2, albumin, and P450-3A3 for spheroids determined by semiquantitative real-time PCR were significantly higher than those for spheroids cultured without the Matrigel overlay or for hepatocytes cultured using a conventional two-dimensional method. The spheroids obtained exhibited higher structural polarity and functional bile canaliculi compared with hepatocytes cultured using a conventional two-dimensional method.
Collapse
Affiliation(s)
- Ryosuke Takahashi
- Advanced Research Laboratory, Hitachi Ltd., Hatoyama, Saitama, Japan
| | | | | | | |
Collapse
|
21
|
Yang S, Wu X, Luo C, Pan C, Pu J. Expression and clinical significance of hepaCAM and VEGF in urothelial carcinoma. World J Urol 2010; 28:473-8. [PMID: 20593288 DOI: 10.1007/s00345-010-0573-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 05/28/2010] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Investigate the expression of hepatocyte cell adhesion molecule (hepaCAM) and vascular endothelial growth factor (VEGF) mRNA in 55 cases of urothelial carcinoma to examine the potential relationship between hepaCAM and VEGF in urothelial carcinoma. METHODS Expression of hepaCAM and VEGF gene was determined by semiquantitative reverse transcriptase polymerase chain reaction (RT-PCR) in 55 paired urothelial carcinoma specimens. T24 cells stably expressing hepaCAM gene were established by Lipofectamine 2000. RT-PCR and western blot analysis were used to detect gene and protein expression of hepaCAM and VEGF before and after transfection. MTT test was used to detect the effect of hepaCAM gene on the cell proliferation. RESULTS RT-PCR showed that hepaCAM expression level was significantly lower, and VEGF was significantly higher in urothelial carcinoma tissues than in adjacent tissues (P < 0.05, P < 0.05). hepaCAM and VEGF were strongly correlated with tumor stage (P < 0.05, P < 0.05). Spearman correlation analysis showed lower hepaCAM level was associated with higher VEGF level (r = -0.277 P = 0.041). Experiments with T24 cells in vitro demonstrated the expression of VEGF mRNA and protein were significantly decreased after transfection of hepaCAM gene (P < 0.05, P < 0.05). Expression of hepaCAM resulted in a significant inhibition of T24 cells proliferation (P < 0.05). CONCLUSION There is a close relationship between hepaCAM and VEGF in urothelial carcinoma. hepaCAM may be defined as a new target for diagnosis and anticancer therapy.
Collapse
Affiliation(s)
- Shuzhe Yang
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | | | | | | | | |
Collapse
|
22
|
Wang C, Roy SK. Expression of E-cadherin and N-cadherin in perinatal hamster ovary: possible involvement in primordial follicle formation and regulation by follicle-stimulating hormone. Endocrinology 2010; 151:2319-30. [PMID: 20219978 PMCID: PMC2869259 DOI: 10.1210/en.2009-1489] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We examined the expression and hormonal regulation of E-cadherin (CDH1) and N-cadherin (CDH2) with respect to primordial follicle formation. Hamster Cdh1 and Cdh2 cDNA and amino acid sequences were more than 90% similar to those of the mouse, rat, and human. Although CDH1 expression remained exclusively in the oocytes during neonatal ovary development, CDH2 expression shifted from the oocytes to granulosa cells of primordial follicles on postnatal day (P)8. Subsequently, strong CDH2 expression was restricted to granulosa cells of growing follicles. Cdh2 mRNA levels in the ovary decreased from embryonic d 13 through P10 with a transient increase on P7, which was the day before the appearance of primordial follicles. Cdh1 mRNA levels decreased from embryonic d 13 through P3 and then showed a transient increase on P8, coinciding with the formation of primordial follicles. CDH1 and CDH2 expression were consistent with that of mRNA. Neutralization of FSH in utero impaired primordial follicle formation with an associated decrease in Cdh2 mRNA and CDH2, but an increase in Cdh1 mRNA and CDH1 expression. The altered expression was reversed by equine chorionic gonadotropin treatment on P1. Whereas a CDH2 antibody significantly reduced the formation of primordial and primary follicles in vitro, a CDH1 antibody had the opposite effect. This is the first evidence to suggest that primordial follicle formation requires a differential spatiotemporal expression and action of CDH1 and CDH2. Further, FSH regulation of primordial follicle formation may involve the action of CDH1 and CDH2.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska 68198-4515, USA
| | | |
Collapse
|
23
|
Seino S, Shibasaki T, Minami K. Pancreatic beta-cell signaling: toward better understanding of diabetes and its treatment. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2010; 86:563-577. [PMID: 20551594 PMCID: PMC3081169 DOI: 10.2183/pjab.86.563] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 04/14/2010] [Indexed: 05/29/2023]
Abstract
Pancreatic beta-cells play a central role in the maintenance glucose homeostasis by secreting insulin, a key hormone that regulates blood glucose levels. Dysfunction of the beta-cells and/or a decrease in the beta-cell mass are associated closely with the pathogenesis and pathophysiology of diabetes mellitus, a major metabolic disease that is rapidly increasing worldwide. Clarification of the mechanisms of insulin secretion and beta-cell fate provides a basis for the understanding of diabetes and its better treatment. In this review, we discuss cell signaling critical for the insulin secretory function based on our recent studies.
Collapse
Affiliation(s)
- Susumu Seino
- Division of Cellular and Molecular Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Hyogo, Japan.
| | | | | |
Collapse
|
24
|
Hickey JG, Myers SM, Tian X, Zhu SJ, V. Shaw JL, Andrew SD, Richardson DS, Brettschneider J, Mulligan LM. RET-mediated gene expression pattern is affected by isoform but not oncogenic mutation. Genes Chromosomes Cancer 2009; 48:429-40. [DOI: 10.1002/gcc.20653] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
25
|
Seeberger KL, Eshpeter A, Rajotte RV, Korbutt GS. Epithelial cells within the human pancreas do not coexpress mesenchymal antigens: epithelial-mesenchymal transition is an artifact of cell culture. J Transl Med 2009; 89:110-21. [PMID: 19079324 DOI: 10.1038/labinvest.2008.122] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pancreatic mesenchymal stem cells (MSCs) may be derived from human beta-cells undergoing reversible epithelial-mesenchymal transition (EMT), suggesting that they could be a potential source of new beta-cells. In this study we sought to determine the origin of pancreatic MSCs in the nonendocrine pancreas. Double immunofluorescent (IF) staining and flow cytometry were used to assess the cell phenotype of nonendocrine pancreas tissue following islet procurement, during in vitro expansion of MSCs, and after differentiation. IF staining of paraffin-embedded pancreatic biopsy sections was used to assess cell phenotype in vivo. In this study we demonstrated that: (1) pancreatic epithelial cells do not express MSC antigens in vivo; (2) following islet isolation EpCAM- and CK19-positive epithelial cells coexpressed the MSC antigens CD44 (32+/-8% and 38+/-10%) and CD29 (85+/-4% and 64+/-4%); (3) during in vitro expansion the number of single-positive epithelial and double-positive epithelial/MSCs decreased whereas the number of single-positive MSCs increased and (4) differentiated MSCs do not revert to a true epithelial cell phenotype in our culture conditions, as epithelial cell surface markers (EpCAM, CK19 and E-Cadherin) are not reexpressed, although the MSC phenotype is altered. This study demonstrates that MSCs may be derived in vitro via a pancreatic epithelial cell undergoing EMT, however it is more likely that a small percentage of MSCs that reside in the adult pancreas are proliferating whereas the epithelial cells are negatively selected by the experimental culture conditions.
Collapse
Affiliation(s)
- Karen L Seeberger
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | | | | | | |
Collapse
|
26
|
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
|
27
|
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
|