1
|
Cheng Y, Zhang T, Yang C, Gebeyew K, Ye C, Zhou X, Zhang T, Feng G, Li R, He Z, Parnas O, Tan Z. Low expression of CCKBR in the acinar cells is associated with insufficient starch hydrolysis in ruminants. Commun Biol 2024; 7:1686. [PMID: 39706905 DOI: 10.1038/s42003-024-07406-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024] Open
Abstract
Unlike monogastric animals, ruminants exhibit significantly lower starch digestibility in the small intestine. A better understanding of the physiological mechanisms that regulate digestion patterns in ruminants could lead to an increased use of starch concentrates. Here we show more robust pancreatic exocrine function in adult goats (AG) than in neonatal goats (NG) by combining scRNA-seq and proteomic analysis. Our findings suggest that inadequate amylase activity could be a limiting factor in starch digestion in ruminants. In addition, we show that insufficient starch hydrolysis in adult goats might be associated with low expression of a CCKBR receptor in the acinar cells. On top of that, the low expression of CCKBR in adult goats, in conjunction with a low distribution of the CCK-I cells in the duodenum, may jointly lead to a slow response of the intestinal-pancreatic reflex and induce an asynchronous process of food entering the small intestine and releasing of digestive enzymes, which ultimately limits the starch digestibility. Overall, the present findings generate a resource that can provide better insight into the mammalian pancreas.
Collapse
Affiliation(s)
- Yan Cheng
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianxi Zhang
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chao Yang
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kefyalew Gebeyew
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chengyu Ye
- The Department of Microbiology and Immunology, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Xinxin Zhou
- LC-Bio Technology (Hanghzhou) co.ltd., Hanghzhou, 310000, China
| | - Tianqi Zhang
- College of Horticulture, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Ganyi Feng
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Li
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhixiong He
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Key Laboratory of Forage Breeding-by-Design and Utilization, Chinese Academy of Science, Beijing, 100093, China.
| | - Oren Parnas
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
| | - Zhiliang Tan
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
2
|
Cui D, Feng X, Lei S, Zhang H, Hu W, Yang S, Yu X, Su Z. Pancreatic β-cell failure, clinical implications, and therapeutic strategies in type 2 diabetes. Chin Med J (Engl) 2024; 137:791-805. [PMID: 38479993 PMCID: PMC10997226 DOI: 10.1097/cm9.0000000000003034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Indexed: 04/06/2024] Open
Abstract
ABSTRACT Pancreatic β-cell failure due to a reduction in function and mass has been defined as a primary contributor to the progression of type 2 diabetes (T2D). Reserving insulin-producing β-cells and hence restoring insulin production are gaining attention in translational diabetes research, and β-cell replenishment has been the main focus for diabetes treatment. Significant findings in β-cell proliferation, transdifferentiation, pluripotent stem cell differentiation, and associated small molecules have served as promising strategies to regenerate β-cells. In this review, we summarize current knowledge on the mechanisms implicated in β-cell dynamic processes under physiological and diabetic conditions, in which genetic factors, age-related alterations, metabolic stresses, and compromised identity are critical factors contributing to β-cell failure in T2D. The article also focuses on recent advances in therapeutic strategies for diabetes treatment by promoting β-cell proliferation, inducing non-β-cell transdifferentiation, and reprograming stem cell differentiation. Although a significant challenge remains for each of these strategies, the recognition of the mechanisms responsible for β-cell development and mature endocrine cell plasticity and remarkable advances in the generation of exogenous β-cells from stem cells and single-cell studies pave the way for developing potential approaches to cure diabetes.
Collapse
Affiliation(s)
- Daxin Cui
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingrong Feng
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siman Lei
- Clinical Translational Innovation Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongmei Zhang
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wanxin Hu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shanshan Yang
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoqian Yu
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhiguang Su
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Clinical Translational Innovation Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
3
|
Wang S, Dong X, Maazi M, Chen N, Mahil A, Kopp JL. GABA treatment does not induce neogenesis of new endocrine cells from pancreatic ductal cells. Islets 2023; 15:2219477. [PMID: 37258189 DOI: 10.1080/19382014.2023.2219477] [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: 02/08/2023] [Revised: 05/25/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023] Open
Abstract
Previous studies indicated that ductal cells can contribute to endocrine neogenesis in adult rodents after alpha cells convert into beta cells. This can occur through Pax4 mis-expression in alpha cells or through long-term administration of gamma-aminobutyric acid (GABA) to healthy mice. GABA has also been reported to increase the number of beta cells through direct effects on their proliferation, but only in specific genetic mouse backgrounds. To test whether GABA induces neogenesis of beta cells from ductal cells or affects pancreatic cell proliferation, we administered GABA or saline over 2 or 6 months to Sox9CreER;R26RYFP mice in which 60-80% of large or small ducts were efficiently lineage labeled. We did not observe any increases in islet neogenesis from ductal cells between 1 and 2 months of age in saline treated mice, nor between 2 and 6 months of saline treatment, supporting previous studies indicating that adult ductal cells do not give rise to new endocrine cells during homeostasis. Unlike previous reports, we did not observe an increase in beta cell neogenesis after 2 or 6 months of GABA administration. Nor did we observe a significant increase in the pancreatic islet area, the number of insulin and glucagon double positive cells, or cell proliferation in the pancreas. This indicates that the effect of long term GABA administration on the pancreas is minimal or highly context dependent.
Collapse
Affiliation(s)
- Shihao Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Xin Dong
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Mahan Maazi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Nan Chen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Amar Mahil
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Janel L Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
An J, Jiang T, Qi L, Xie K. Acinar cells and the development of pancreatic fibrosis. Cytokine Growth Factor Rev 2023; 71-72:40-53. [PMID: 37291030 DOI: 10.1016/j.cytogfr.2023.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/10/2023]
Abstract
Pancreatic fibrosis is caused by excessive deposition of extracellular matrixes of collagen and fibronectin in the pancreatic tissue as a result of repeated injury often seen in patients with chronic pancreatic diseases. The most common causative conditions include inborn errors of metabolism, chemical toxicity and autoimmune disorders. Its pathophysiology is highly complex, including acinar cell injury, acinar stress response, duct dysfunction, pancreatic stellate cell activation, and persistent inflammatory response. However, the specific mechanism remains to be fully clarified. Although the current therapeutic strategies targeting pancreatic stellate cells show good efficacy in cell culture and animal models, they are not satisfactory in the clinic. Without effective intervention, pancreatic fibrosis can promote the transformation from pancreatitis to pancreatic cancer, one of the most lethal malignancies. In the normal pancreas, the acinar component accounts for 82% of the exocrine tissue. Abnormal acinar cells may activate pancreatic stellate cells directly as cellular source of fibrosis or indirectly via releasing various substances and initiate pancreatic fibrosis. A comprehensive understanding of the role of acinar cells in pancreatic fibrosis is critical for designing effective intervention strategies. In this review, we focus on the role of and mechanisms underlying pancreatic acinar injury in pancreatic fibrosis and their potential clinical significance.
Collapse
Affiliation(s)
- Jianhong An
- SCUT-QMPH Joint Laboratory for Pancreatic Cancer Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China; Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Tingting Jiang
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China
| | - Ling Qi
- SCUT-QMPH Joint Laboratory for Pancreatic Cancer Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
| | - Keping Xie
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
5
|
Yu L, Li L, Liu J, Sun H, Li X, Xiao H, Alfred MO, Wang M, Wu X, Gao Y, Luo C. Recombinant Reg3α Prevents Islet β-Cell Apoptosis and Promotes β-Cell Regeneration. Int J Mol Sci 2022; 23:ijms231810584. [PMID: 36142497 PMCID: PMC9504149 DOI: 10.3390/ijms231810584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/05/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Progressive loss and dysfunction of islet β-cells has not yet been solved in the treatment of diabetes. Regenerating protein (Reg) has been identified as a trophic factor which is demonstrated to be associated with pancreatic tissue regeneration. We previously produced recombinant Reg3α protein (rReg3α) and proved that it protects against acute pancreatitis in mice. Whether rReg3α protects islet β-cells in diabetes has been elusive. In the present study, rReg3α stimulated MIN6 cell proliferation and resisted STZ-caused cell death. The protective effect of rReg3α was also found in mouse primary islets. In BALB/c mice, rReg3α administration largely alleviated STZ-induced diabetes by the preservation of β-cell mass. The protective mechanism could be attributed to Akt/Bcl-2/-xL activation and GRP78 upregulation. Scattered insulin-expressing cells and clusters with small size, low insulin density, and exocrine distribution were observed and considered to be neogenic. In isolated acinar cells with wheat germ agglutinin (WGA) labeling, rReg3α treatment generated insulin-producing cells through Stat3/Ngn3 signaling, but these cells were not fully functional in response to glucose stimulation. Our results demonstrated that rReg3α resists STZ-induced β-cell death and promotes β-cell regeneration. rReg3α could serve as a potential drug for β-cell maintenance in anti-diabetic treatment.
Collapse
Affiliation(s)
- Luting Yu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 210037, China
| | - Liang Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Junli Liu
- MeDiC Program, The Research Institute of McGill University Health Centre, Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Hao Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xiang Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Hanyu Xiao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Martin Omondi Alfred
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
- Institute of Primate Research, End of Karen Road, Karen, Nairobi P.O. Box 24481-00502, Kenya
| | - Min Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Nature Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xuri Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Nature Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Gao
- Institute of Suzhou Biobank, Suzhou Center for Disease Prevention and Control, Suzhou 215007, China
- Suzhou Institute of Advanced Study in Public Health, Gusu School, Nanjing Medical University, Suzhou 210029, China
- Correspondence: (Y.G.); (C.L.); Tel.: +86-0512-6826-2385 (Y.G.); +86-138-1388-3828 (C.L.)
| | - Chen Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Nature Medicines, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (Y.G.); (C.L.); Tel.: +86-0512-6826-2385 (Y.G.); +86-138-1388-3828 (C.L.)
| |
Collapse
|
6
|
Novelli M, Masini M, Vecoli C, Moscato S, Funel N, Pippa A, Mattii L, Ippolito C, Campani D, Neglia D, Masiello P. Dysregulated insulin secretion is associated with pancreatic β-cell hyperplasia and direct acinar-β-cell trans-differentiation in partially eNOS-deficient mice. Physiol Rep 2022; 10:e15425. [PMID: 35986504 PMCID: PMC9391603 DOI: 10.14814/phy2.15425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/11/2022] [Accepted: 07/23/2022] [Indexed: 06/15/2023] Open
Abstract
eNOS-deficient mice were previously shown to develop hypertension and metabolic alterations associated with insulin resistance either in standard dietary conditions (eNOS-/- homozygotes) or upon high-fat diet (HFD) (eNOS+/- heterozygotes). In the latter heterozygote model, the present study investigated the pancreatic morphological changes underlying the abnormal glycometabolic phenotype. C57BL6 wild type (WT) and eNOS+/- mice were fed with either chow or HFD for 16 weeks. After being longitudinally monitored for their metabolic state after 8 and 16 weeks of diet, mice were euthanized and fragments of pancreas were processed for histological, immuno-histochemical and ultrastructural analyses. HFD-fed WT and eNOS+/- mice developed progressive glucose intolerance and insulin resistance. Differently from WT animals, eNOS+/- mice showed a blunted insulin response to a glucose load, regardless of the diet regimen. Such dysregulation of insulin secretion was associated with pancreatic β-cell hyperplasia, as shown by larger islet fractional area and β-cell mass, and higher number of extra-islet β-cell clusters than in chow-fed WT animals. In addition, only in the pancreas of HFD-fed eNOS+/- mice, there was ultrastructural evidence of a number of hybrid acinar-β-cells, simultaneously containing zymogen and insulin granules, suggesting the occurrence of a direct exocrine-endocrine transdifferentiation process, plausibly triggered by metabolic stress associated to deficient endothelial NO production. As suggested by confocal immunofluorescence analysis of pancreatic histological sections, inhibition of Notch-1 signaling, likely due to a reduced NO availability, is proposed as a novel mechanism that could favor both β-cell hyperplasia and acinar-β-cell transdifferentiation in eNOS-deficient mice with impaired insulin response to a glucose load.
Collapse
Affiliation(s)
- Michela Novelli
- Department of Translational Research and New Technologies in Medicine and SurgeryUniversity of PisaPisaItaly
| | - Matilde Masini
- Department of Translational Research and New Technologies in Medicine and SurgeryUniversity of PisaPisaItaly
| | - Cecilia Vecoli
- Institute of Clinical PhysiologyNational Research Council (CNR)PisaItaly
| | - Stefania Moscato
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Interdepartmental Research Centre "Nutraceuticals and Food for Health"University of PisaPisaItaly
| | - Niccola Funel
- Department of Translational Research and New Technologies in Medicine and SurgeryUniversity of PisaPisaItaly
| | - Anna Pippa
- Institute of Clinical PhysiologyNational Research Council (CNR)PisaItaly
| | - Letizia Mattii
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Interdepartmental Research Centre "Nutraceuticals and Food for Health"University of PisaPisaItaly
| | - Chiara Ippolito
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Daniela Campani
- Department of Surgical, Medical and Molecular Pathology, and Critical Care MedicineUniversity of PisaPisaItaly
| | - Danilo Neglia
- Cardiovascular DepartmentFondazione Toscana Gabriele Monasterio per la Ricerca Medica e di Sanità PubblicaPisaItaly
| | - Pellegrino Masiello
- Department of Translational Research and New Technologies in Medicine and SurgeryUniversity of PisaPisaItaly
| |
Collapse
|
7
|
Jiang T, Wei F, Xie K. Clinical significance of pancreatic ductal metaplasia. J Pathol 2022; 257:125-139. [PMID: 35170758 DOI: 10.1002/path.5883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 11/08/2022]
Abstract
Pancreatic ductal metaplasia (PDM) is the stepwise replacement of differentiated somatic cells with ductal or ductal-like cells in the pancreas. PDM is usually triggered by cellular and environmental insults. PDM development may involve all cell lineages of the pancreas, and acinar cells with the highest plasticity are the major source of PDM. Pancreatic progenitor cells are also involved as cells of origin or transitional intermediates. PDM is heterogeneous at the histological, cellular, and molecular levels and only certain subsets of PDM develop further into pancreatic intraepithelial neoplasia (PanIN) and then pancreatic ductal adenocarcinoma (PDAC). The formation and evolution of PDM is regulated at the cellular and molecular levels through a complex network of signaling pathways. The key molecular mechanisms that drive PDM formation and its progression into PanIN/PDAC remain unclear, but represent key targets for reversing or inhibiting PDM. Alternatively, PDM could be a source of pancreas regeneration, including both exocrine and endocrine components. Cellular aging and apoptosis are obstacles to PDM-to-PanIN progression or pancreas regeneration. Functional identification of the cellular and molecular events driving senescence and apoptosis in PDM and its progression would help not only to restrict the development of PDM into PanIN/PDAC, but may also facilitate pancreatic regeneration. This review systematically assesses recent advances in the understanding of PDM physiology and pathology, with a focus on its implications for enhancing regeneration and prevention of cancer. © 2022 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Tingting Jiang
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, PR China
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, PR China
| | - Fang Wei
- Institute of Digestive Diseases Research, The South China University of Technology School of Medicine, Guangzhou, PR China
| | - Keping Xie
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, PR China
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, PR China
| |
Collapse
|
8
|
Li S, Xie K. Ductal metaplasia in pancreas. Biochim Biophys Acta Rev Cancer 2022; 1877:188698. [DOI: 10.1016/j.bbcan.2022.188698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023]
|
9
|
Parte S, Nimmakayala RK, Batra SK, Ponnusamy MP. Acinar to ductal cell trans-differentiation: A prelude to dysplasia and pancreatic ductal adenocarcinoma. Biochim Biophys Acta Rev Cancer 2022; 1877:188669. [PMID: 34915061 DOI: 10.1016/j.bbcan.2021.188669] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer (PC) is the deadliest neoplastic epithelial malignancies and is projected to be the second leading cause of cancer-related mortality by 2024. Five years overall survival being ~10%, mortality and incidence rates are disturbing. Acinar to ductal cell metaplasia (ADM) encompasses cellular reprogramming and phenotypic switch-over, making it a cardinal event in tumor initiation. Differential cues and varied regulatory factors drive synchronous functions of metaplastic cell populations leading to multiple cell fates and physiological outcomes. ADM is a precursor for developing early pre-neoplastic lesions further progressing into PC due to oncogenic signaling. Hence delineating molecular events guiding tumor initiation may provide cues for regenerative medicine and precision onco-medicine. Therefore, understanding PC pathogenesis and early diagnosis are crucial. We hereby provide a timely overview of the current progress in this direction and future perspectives we foresee unfolding in the best interest of patient well-being and better clinical management of PC.
Collapse
Affiliation(s)
- Seema Parte
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
10
|
Kryvalap Y, Jiang ML, Kryvalap N, Hendrickson C, Czyzyk J. SerpinB13 antibodies promote β cell development and resistance to type 1 diabetes. Sci Transl Med 2021; 13:13/588/eabf1587. [PMID: 33827974 DOI: 10.1126/scitranslmed.abf1587] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/15/2021] [Indexed: 12/20/2022]
Abstract
Pancreatic endocrine cell development is dependent on the rescue of the neurogenin3 (Ngn3) transcription factor from repression by Notch. The signals that prevent Notch signaling, thereby allowing the formation of pancreatic endocrine cells, remain unclear. We show that inhibiting serpinB13, a cathepsin L (CatL) protease inhibitor expressed in the pancreatic epithelium, caused in vitro and in vivo cleavage of the extracellular domain of Notch1. This was followed by a twofold increase in the Ngn3+ progenitor cell population and enhanced conversion of these cells to express insulin. Conversely, both recombinant serpinB13 protein and CatL deficiency down-regulated pancreatic Ngn3+ cell output. Mouse embryonic exposure to inhibitory anti-serpinB13 antibody resulted in increased islet cell mass and improved outcomes in streptozotocin-induced diabetes at 8 weeks of age. Moreover, anti-serpinB13 autoantibodies stimulated Ngn3+ endocrine progenitor formation in the pancreas and were associated with delayed progression to type 1 diabetes (T1D) in children. These data demonstrate long-term impact of serpinB13 activity on islet biology and suggest that promoting protease activity by blocking this serpin may have prophylactic potential in T1D.
Collapse
Affiliation(s)
- Yury Kryvalap
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Washington Ave. SE, Minneapolis, MN 55455, USA
| | - Matthew L Jiang
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Nadzeya Kryvalap
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Cole Hendrickson
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Washington Ave. SE, Minneapolis, MN 55455, USA
| | - Jan Czyzyk
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Washington Ave. SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
11
|
REST Inhibits Direct Reprogramming of Pancreatic Exocrine to Endocrine Cells by Preventing PDX1-Mediated Activation of Endocrine Genes. Cell Rep 2021; 31:107591. [PMID: 32375045 DOI: 10.1016/j.celrep.2020.107591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/29/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
The emerging appreciation of plasticity among pancreatic lineages has created interest in harnessing cellular reprogramming for β cell replacement therapy of diabetes. Current reprogramming methodologies are inefficient, largely because of a limited understanding of the underlying mechanisms. Using an in vitro reprogramming system, we reveal the transcriptional repressor RE-1 silencing transcription factor (REST) as a barrier for β cell gene expression in the reprogramming of pancreatic exocrine cells. We observe that REST-bound loci lie adjacent to the binding sites of multiple key β cell transcription factors, including PDX1. Accordingly, a loss of REST function combined with PDX1 expression results in the synergistic activation of endocrine genes. This is accompanied by increased histone acetylation and PDX1 binding at endocrine gene loci. Collectively, our data identify a mechanism for REST activity involving the prevention of PDX1-mediated activation of endocrine genes and uncover REST downregulation and the resulting chromatin alterations as key events in β cell reprogramming.
Collapse
|
12
|
Huang D, Wang R. Exploring the mechanism of pancreatic cell fate decisions via cell-cell communication. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:2401-2424. [PMID: 33892552 DOI: 10.3934/mbe.2021122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The endocrine and exocrine cells in pancreas originate initially from a group of apparently identical endoderm cells in the early gut. The endocrine and exocrine tissues are composed of islet/acinar and duct cells respectively. To explore the mechanism of pancreas cell fate decisions, we first construct a minimal mathematical model related to pancreatic regulations. The regulatory mechanism of acinar-to-islet cell conversion is revealed by bifurcation analysis of the model. In addition, Notch signaling is critical in determining the fate of endocrine and exocrine in the developing pancreas and it is a typical mediator of lateral inhibition which instructs adjacent cells to make different fate decisions. Next, we construct a multicellular model of cell-cell communication mediated by Notch signaling with trans-activation and cis-inhibition. The roles of Notch signaling in regulating fate decisions of endocrine and exocrine cells during the differentiation of pancreatic cells are explored. The results indicate that high (or low) level of Notch signaling drive cells to select the fate of exocrine (or endocrine) progenitor cells. The networks and the models presented here might be good candidates for providing qualitative mechanisms of pancreatic cell fate decisions. These results can also provide some insight on choosing perturbation strategies for further experimental analysis.
Collapse
Affiliation(s)
- Dasong Huang
- Department of Mathematics, Shanghai University, Shanghai 200444, China
| | - Ruiqi Wang
- Department of Mathematics, Shanghai University, Shanghai 200444, China
| |
Collapse
|
13
|
Spears E, Serafimidis I, Powers AC, Gavalas A. Debates in Pancreatic Beta Cell Biology: Proliferation Versus Progenitor Differentiation and Transdifferentiation in Restoring β Cell Mass. Front Endocrinol (Lausanne) 2021; 12:722250. [PMID: 34421829 PMCID: PMC8378310 DOI: 10.3389/fendo.2021.722250] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
In all forms of diabetes, β cell mass or function is reduced and therefore the capacity of the pancreatic cells for regeneration or replenishment is a critical need. Diverse lines of research have shown the capacity of endocrine as well as acinar, ductal and centroacinar cells to generate new β cells. Several experimental approaches using injury models, pharmacological or genetic interventions, isolation and in vitro expansion of putative progenitors followed by transplantations or a combination thereof have suggested several pathways for β cell neogenesis or regeneration. The experimental results have also generated controversy related to the limitations and interpretation of the experimental approaches and ultimately their physiological relevance, particularly when considering differences between mouse, the primary animal model, and human. As a result, consensus is lacking regarding the relative importance of islet cell proliferation or progenitor differentiation and transdifferentiation of other pancreatic cell types in generating new β cells. In this review we summarize and evaluate recent experimental approaches and findings related to islet regeneration and address their relevance and potential clinical application in the fight against diabetes.
Collapse
Affiliation(s)
- Erick Spears
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ioannis Serafimidis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Alvin C. Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
- VA Tennessee Valley Healthcare System, Nashville, TN, United States
- *Correspondence: Anthony Gavalas, ; Alvin C. Powers,
| | - Anthony Gavalas
- Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of TU Dresden, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Diabetes Research (DZD), Neuherberg, Germany
- *Correspondence: Anthony Gavalas, ; Alvin C. Powers,
| |
Collapse
|
14
|
Kaya-Dagistanli F, Ozturk M. Transdifferentiation of both intra- and extra-islet cells into beta cells in nicotinamide treated neonatal diabetic rats: An in situ hybridization and double immunohistochemical study. Acta Histochem 2020; 122:151612. [PMID: 33066834 DOI: 10.1016/j.acthis.2020.151612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 12/24/2022]
Abstract
We aimed to study the effect of nicotinamide (NA) on beta (β)-cell regeneration and apoptosis in streptozotocin induced neonatal rats (n-STZ). Three groups were performed: Control group, n2-STZ group (100 mg/kg STZ on the second day-after birth), n2-STZ + NA group (STZ;100 mg/kg + NA;500 mg/kg/day for 5 days). The pancreatic tissue sections were immunostained with insulin, glucagon, somatostatin, Pdx1, Notch1 and active caspase-3 antibodies, and double immunostained with insulin/PCNA, insulin/glucagon and insulin/somatostatin antibodies. In situ hybridization carried out with insulin probe. Apoptotic β-cell were shown by TUNEL assay, followed by immunostaining. The number of insulin/PCNA, insulin/glucagon and insulin/somatostatin double-positive cells significantly increased in n2-STZ + NA group compared with the other groups (p < 0.001). n2- STZ group had lower number of insulin and Pdx1 positive cells in islets, compared to NA treated diabetics. The insulin and Pdx1 immun positive cells were located in the small clusters or scattered through the exocrine tissue and around to ducts in n2-STZ + NA group. Notch1 positive cell numbers were increased, whereas caspase-3 and TUNEL positive β-cell numbers were decreased in n2-STZ + NA group. NA treatment induces the neogenic insulin positive islets orginated from the differentiation of ductal progenitor cells, transdifferentiation of acinar cells into β cells, and transformation of potent precursor cells and centroacinar cells via the activated Notch expression into β-cells in n-STZ rats.
Collapse
|
15
|
Elhanani O, Walker MD. Protocol for Studying Reprogramming of Mouse Pancreatic Acinar Cells to β-like Cells. STAR Protoc 2020; 1:100096. [PMID: 33111125 PMCID: PMC7580220 DOI: 10.1016/j.xpro.2020.100096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The potential of reprogrammed β cells derived from pancreatic exocrine cells to treat diabetes has been demonstrated in animal models. However, the precise mechanisms and regulators involved in this process are not clear. Here, we describe a method that allows mechanistic studies of this process in primary exocrine cultures using adenoviral expression vectors. This rapid 5-day protocol, provides the researcher with a highly controlled experimental system in which the effects of different compounds or genetic manipulations can be studied. For complete details on the use and execution of this protocol, please refer to Elhanani et al. (2020).
Collapse
Affiliation(s)
- Ofer Elhanani
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Michael D Walker
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
16
|
Zhang J, Liu F. The De-, Re-, and trans-differentiation of β-cells: Regulation and function. Semin Cell Dev Biol 2020; 103:68-75. [DOI: 10.1016/j.semcdb.2020.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/09/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
|
17
|
Thakur G, Lee HJ, Jeon RH, Lee SL, Rho GJ. Small Molecule-Induced Pancreatic β-Like Cell Development: Mechanistic Approaches and Available Strategies. Int J Mol Sci 2020; 21:E2388. [PMID: 32235681 PMCID: PMC7178115 DOI: 10.3390/ijms21072388] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a metabolic disease which affects not only glucose metabolism but also lipid and protein metabolism. It encompasses two major types: type 1 and 2 diabetes. Despite the different etiologies of type 1 and 2 diabetes mellitus (T1DM and T2DM, respectively), the defining features of the two forms are insulin deficiency and resistance, respectively. Stem cell therapy is an efficient method for the treatment of diabetes, which can be achieved by differentiating pancreatic β-like cells. The consistent generation of glucose-responsive insulin releasing cells remains challenging. In this review article, we present basic concepts of pancreatic organogenesis, which intermittently provides a basis for engineering differentiation procedures, mainly based on the use of small molecules. Small molecules are more auspicious than any other growth factors, as they have unique, valuable properties like cell-permeability, as well as a nonimmunogenic nature; furthermore, they offer immense benefits in terms of generating efficient functional beta-like cells. We also summarize advances in the generation of stem cell-derived pancreatic cell lineages, especially endocrine β-like cells or islet organoids. The successful induction of stem cells depends on the quantity and quality of available stem cells and the efficient use of small molecules.
Collapse
Affiliation(s)
- Gitika Thakur
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Hyeon-Jeong Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Ryoung-Hoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Sung-Lim Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea; (G.T.); (H.-J.L.); (S.-L.L.)
| |
Collapse
|
18
|
Zhao F, Liu X, Wang Z, Lang H, Zhang T, Wang R, Lin X, He D, Shi P, Pang X. Novel Mouse miRNA Chr13_novelMiR7354-5p Improves Bone-Marrow-Derived Mesenchymal Stem Cell Differentiation into Insulin-Producing Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 19:1110-1122. [PMID: 32059337 PMCID: PMC7016162 DOI: 10.1016/j.omtn.2020.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 12/22/2019] [Accepted: 01/02/2020] [Indexed: 12/18/2022]
Abstract
MicroRNAs (miRNAs) that play key roles in the generation of insulin-producing cells from stem cells provide a cell-based approach for insulin replacement therapy. In this study, we used next-generation sequencing to detect the miRNA expression profile of normal mouse pancreatic β cells, non-β cells, bone marrow mesenchymal stem cells (BM-MSCs), and adipose-derived stem cells (ADSCs) and determined relative miRNA expression levels in mouse pancreatic β cells. After the novel mouse miRNA candidates were identified using miRDeep 2.0, we found that Chr13_novelMiR7354-5p, a novel miRNA candidate, significantly promoted the differentiation of BM-MSCs into insulin-producing cells in vitro. Furthermore, Chr13_novelMiR7354-5p-transfected BM-MSCs reversed hyperglycemia in streptozotocin (STZ)-treated diabetic mice. In addition, bioinformatics analyses, a luciferase reporter assay, and western blotting demonstrated that Chr13_novelMiR7354-5p targeted Notch1 and Rbpj. Our results provide compelling evidence of the existence of 65 novel mouse miRNA candidates and present a new treatment strategy to generate insulin-producing cells from stem cells.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Street, Shenbei New District, Shenyang City 110122, Liaoning Province, China
| | - Xiaoyu Liu
- Department of Obstetrics and Gynecology, Center for Assisted Reproduction, Shengjing Hospital of China Medical University, 39 Huaxiang Street, Tiexi District, Shenyang City 110022, Liaoning Province, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang City 110004, Liaoning Province, China
| | - Hongxin Lang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Street, Shenbei New District, Shenyang City 110122, Liaoning Province, China
| | - Tao Zhang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Street, Shenbei New District, Shenyang City 110122, Liaoning Province, China
| | - Rui Wang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Street, Shenbei New District, Shenyang City 110122, Liaoning Province, China
| | - Xuewen Lin
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Street, Shenbei New District, Shenyang City 110122, Liaoning Province, China
| | - Dan He
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Street, Shenbei New District, Shenyang City 110122, Liaoning Province, China
| | - Ping Shi
- Shenyang Amnion Bioengineering and Technology R&D Center, 155-5 Chuangxin Street, Hunnan District, Shenyang City 110015, Liaoning Province, China
| | - Xining Pang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Street, Shenbei New District, Shenyang City 110122, Liaoning Province, China; Shenyang Amnion Bioengineering and Technology R&D Center, 155-5 Chuangxin Street, Hunnan District, Shenyang City 110015, Liaoning Province, China.
| |
Collapse
|
19
|
DiNicolantonio JJ, McCarty M. Autophagy-induced degradation of Notch1, achieved through intermittent fasting, may promote beta cell neogenesis: implications for reversal of type 2 diabetes. Open Heart 2019; 6:e001028. [PMID: 31218007 PMCID: PMC6546199 DOI: 10.1136/openhrt-2019-001028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2019] [Indexed: 02/06/2023] Open
|
20
|
Adult human pancreatic acinar cells dedifferentiate into an embryonic progenitor-like state in 3D suspension culture. Sci Rep 2019; 9:4040. [PMID: 30858455 PMCID: PMC6411888 DOI: 10.1038/s41598-019-40481-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/15/2019] [Indexed: 01/02/2023] Open
Abstract
Human pancreatic exocrine cells were cultured in 3D suspension and formed pancreatospheres composed of acinar-derived and duct-like cells. We investigated, up to 6 days, the fate of human pancreatic acinar cells using fluorescein-conjugated Ulex Europaeus Agglutinin 1 lectin, a previously published acinar-specific non-genetic lineage tracing strategy. At day 4, fluorescence-activated cell sort for the intracellularly incorporated FITC-conjugated UEA1 lectin and the duct-specific CA19.9 surface marker, distinguished acinar-derived cells (UEA1+CA19.9-) from duct-like cells (UEA1-CA19.9+) and acinar-to-duct-like transdifferentiated cells (UEA1+CA19.9+). mRNA expression analysis of the acinar-derived (UEA1+CA19.9-) and duct-like (UEA1-CA19.9+) cell fractions with concomitant immunocytochemical analysis of the pancreatospheres revealed acquisition of an embryonic signature in the UEA1+CA19.9- acinar-derived cells characterized by de novo expression of SOX9 and CD142, robust expression of PDX1 and surface expression of GP2. The colocalisation of CD142, a multipotent pancreatic progenitor surface marker, PDX1, SOX9 and GP2 is reminiscent of a cellular state present during human embryonic development. Addition of TGF-beta signalling inhibitor Alk5iII, induced a 28-fold increased KI67-labeling in pancreatospheres, more pronounced in the CD142+GP2+ acinar-derived cells. These findings with human cells underscore the remarkable plasticity of pancreatic exocrine acinar cells, previously described in rodents, and could find applications in the field of regenerative medicine.
Collapse
|
21
|
Aguayo-Mazzucato C, Bonner-Weir S. Pancreatic β Cell Regeneration as a Possible Therapy for Diabetes. Cell Metab 2018; 27:57-67. [PMID: 28889951 PMCID: PMC5762410 DOI: 10.1016/j.cmet.2017.08.007] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/05/2017] [Accepted: 08/08/2017] [Indexed: 02/08/2023]
Abstract
Diabetes is the result of having inadequate supply of functional insulin-producing β cells. Two possible approaches for replenishing the β cells are: (1) replacement by transplanting cadaveric islets or β cells derived from human embryonic stem cells/induced pluripotent stem cells and (2) induction of endogenous regeneration. This review focuses on endogenous regeneration, which can follow two pathways: enhanced replication of existing β cells and formation of new β cells from cells not expressing insulin, either by conversion from a differentiated cell type (transdifferentiation) or differentiation from progenitors (neogenesis). Exciting progress on both pathways suggest that regeneration may have therapeutic promise.
Collapse
Affiliation(s)
| | - Susan Bonner-Weir
- Joslin Diabetes Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA.
| |
Collapse
|
22
|
Acinar cells in the neonatal pancreas grow by self-duplication and not by neogenesis from duct cells. Sci Rep 2017; 7:12643. [PMID: 28974717 PMCID: PMC5626771 DOI: 10.1038/s41598-017-12721-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/13/2017] [Indexed: 01/08/2023] Open
Abstract
Pancreatic acinar cells secrete digestive enzymes necessary for nutrient digestion in the intestine. They are considered the initiating cell type of pancreatic cancer and are endowed with differentiation plasticity that has been harnessed to regenerate endocrine beta cells. However, there is still uncertainty about the mechanisms of acinar cell formation during the dynamic period of early postnatal development. To unravel cellular contributions in the exocrine acinar development we studied two reporter mouse strains to trace the fate of acinar and duct cells during the first 4 weeks of life. In the acinar reporter mice, the labelling index of acinar cells remained unchanged during the neonatal pancreas growth period, evidencing that acinar cells are formed by self-duplication. In line with this, duct cell tracing did not show significant increase in acinar cell labelling, excluding duct-to-acinar cell contribution during neonatal development. Immunohistochemical analysis confirms massive levels of acinar cell proliferation in this early period of life. Further, also increase in acinar cell size contributes to the growth of pancreatic mass.We conclude that the growth of acinar cells during physiological neonatal pancreas development is by self-duplication (and hypertrophy) rather than neogenesis from progenitor cells as was suggested before.
Collapse
|
23
|
Siebel C, Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol Rev 2017; 97:1235-1294. [PMID: 28794168 DOI: 10.1152/physrev.00005.2017] [Citation(s) in RCA: 657] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Notch signaling is an evolutionarily highly conserved signaling mechanism, but in contrast to signaling pathways such as Wnt, Sonic Hedgehog, and BMP/TGF-β, Notch signaling occurs via cell-cell communication, where transmembrane ligands on one cell activate transmembrane receptors on a juxtaposed cell. Originally discovered through mutations in Drosophila more than 100 yr ago, and with the first Notch gene cloned more than 30 yr ago, we are still gaining new insights into the broad effects of Notch signaling in organisms across the metazoan spectrum and its requirement for normal development of most organs in the body. In this review, we provide an overview of the Notch signaling mechanism at the molecular level and discuss how the pathway, which is architecturally quite simple, is able to engage in the control of cell fates in a broad variety of cell types. We discuss the current understanding of how Notch signaling can become derailed, either by direct mutations or by aberrant regulation, and the expanding spectrum of diseases and cancers that is a consequence of Notch dysregulation. Finally, we explore the emerging field of Notch in the control of tissue homeostasis, with examples from skin, liver, lung, intestine, and the vasculature.
Collapse
Affiliation(s)
- Chris Siebel
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Urban Lendahl
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
24
|
Tritschler S, Theis FJ, Lickert H, Böttcher A. Systematic single-cell analysis provides new insights into heterogeneity and plasticity of the pancreas. Mol Metab 2017; 6:974-990. [PMID: 28951822 PMCID: PMC5605721 DOI: 10.1016/j.molmet.2017.06.021] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Diabetes mellitus is characterized by loss or dysfunction of insulin-producing β-cells in the pancreas, resulting in failure of blood glucose regulation and devastating secondary complications. Thus, β-cells are currently the prime target for cell-replacement and regenerative therapy. Triggering endogenous repair is a promising strategy to restore β-cell mass and normoglycemia in diabetic patients. Potential strategies include targeting specific β-cell subpopulations to increase proliferation or maturation. Alternatively, transdifferentiation of pancreatic islet cells (e.g. α- or δ-cells), extra-islet cells (acinar and ductal cells), hepatocytes, or intestinal cells into insulin-producing cells might improve glycemic control. To this end, it is crucial to systematically characterize and unravel the transcriptional program of all pancreatic cell types at the molecular level in homeostasis and disease. Furthermore, it is necessary to better determine the underlying mechanisms of β-cell maturation, maintenance, and dysfunction in diabetes, to identify and molecularly profile endocrine subpopulations with regenerative potential, and to translate the findings from mice to man. Recent approaches in single-cell biology started to illuminate heterogeneity and plasticity in the pancreas that might be targeted for β-cell regeneration in diabetic patients. SCOPE OF REVIEW This review discusses recent literature on single-cell analysis including single-cell RNA sequencing, single-cell mass cytometry, and flow cytometry of pancreatic cell types in the context of mechanisms of endogenous β-cell regeneration. We discuss new findings on the regulation of postnatal β-cell proliferation and maturation. We highlight how single-cell analysis recapitulates described principles of functional β-cell heterogeneity in animal models and adds new knowledge on the extent of β-cell heterogeneity in humans as well as its role in homeostasis and disease. Furthermore, we summarize the findings on cell subpopulations with regenerative potential that might enable the formation of new β-cells in diseased state. Finally, we review new data on the transcriptional program and function of rare pancreatic cell types and their implication in diabetes. MAJOR CONCLUSION Novel, single-cell technologies offer high molecular resolution of cellular heterogeneity within the pancreas and provide information on processes and factors that govern β-cell homeostasis, proliferation, and maturation. Eventually, these technologies might lead to the characterization of cells with regenerative potential and unravel disease-associated changes in gene expression to identify cellular and molecular targets for therapy.
Collapse
Affiliation(s)
- Sophie Tritschler
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Am Parkring 11, 85748 Garching-Hochbrück, Germany
- German Center for Diabetes Research, 85764 Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Am Parkring 11, 85748 Garching-Hochbrück, Germany
- German Center for Diabetes Research, 85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Anika Böttcher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Am Parkring 11, 85748 Garching-Hochbrück, Germany
- German Center for Diabetes Research, 85764 Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| |
Collapse
|
25
|
Afelik S, Rovira M. Pancreatic β-cell regeneration: Facultative or dedicated progenitors? Mol Cell Endocrinol 2017; 445:85-94. [PMID: 27838399 DOI: 10.1016/j.mce.2016.11.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 10/21/2016] [Accepted: 11/08/2016] [Indexed: 12/19/2022]
Abstract
The adult pancreas is only capable of limited regeneration. Unlike highly regenerative tissues such as the skin, intestinal crypts and hematopoietic system, no dedicated adult stem cells or stem cell niche have so far been identified within the adult pancreas. New β cells have been shown to form in the adult pancreas, in response to high physiological demand or experimental β-cell ablation, mostly by replication of existing β cells. The possibility that new β cells are formed from other sources is currently a point of major controversy. Under particular injury conditions, fully differentiated pancreatic duct and acinar cells have been shown to dedifferentiate into a progenitor-like state, however the extent, to which ductal, acinar or other endocrine cells contribute to restoring pancreatic β-cell mass remains to be resolved. In this review we focus on regenerative events in the pancreas with emphasis on the restoration of β-cell mass. We present an overview of regenerative responses noted within the different pancreatic lineages, following injury. We also highlight the intrinsic plasticity of the adult pancreas that allows for inter-conversion of fully differentiated pancreatic lineages through manipulation of few genes or growth factors. Taken together, evidence from a number of studies suggest that differentiated pancreatic lineages could act as facultative progenitor cells, but the extent to which these contribute to β-cell regeneration in vivo is still a matter of contention.
Collapse
Affiliation(s)
- Solomon Afelik
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, 840 South Wood Street, CSB 920 (Rm 502), Chicago, IL 60612, USA.
| | - Meritxell Rovira
- Genomic Programming of Beta-Cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
| |
Collapse
|
26
|
Acinar phenotype is preserved in human exocrine pancreas cells cultured at low temperature: implications for lineage-tracing of β-cell neogenesis. Biosci Rep 2016; 36:BSR20150259. [PMID: 26987985 PMCID: PMC4859086 DOI: 10.1042/bsr20150259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/22/2016] [Indexed: 01/22/2023] Open
Abstract
In vitro cultured pancreatic acinar cells rapidly differentiate. Low temperature exposure prevents this process and improves the efficiency of acinar cell labelling with adenovirus vectors. This may help in tracing β-cell neogenesis from human pancreatic acinar cells. The regenerative medicine field is expanding with great successes in laboratory and preclinical settings. Pancreatic acinar cells in diabetic mice were recently converted into β-cells by treatment with ciliary neurotrophic factor (CNTF) and epidermal growth factor (EGF). This suggests that human acinar cells might become a cornerstone for diabetes cell therapy in the future, if they can also be converted into glucose-responsive insulin-producing cells. Presently, studying pancreatic acinar cell biology in vitro is limited by their high plasticity, as they rapidly lose their phenotype and spontaneously transdifferentiate to a duct-like phenotype in culture. We questioned whether human pancreatic acinar cell phenotype could be preserved in vitro by physico-chemical manipulations and whether this could be valuable in the study of β-cell neogenesis. We found that culture at low temperature (4°C) resulted in the maintenance of morphological and molecular acinar cell characteristics. Specifically, chilled acinar cells did not form the spherical clusters observed in controls (culture at 37°C), and they maintained high levels of acinar-specific transcripts and proteins. Five-day chilled acinar cells still transdifferentiated into duct-like cells upon transfer to 37°C. Moreover, adenoviral-mediated gene transfer evidenced an active Amylase promoter in the 7-day chilled acinar cells, and transduction performed in chilled conditions improved acinar cell labelling. Together, our findings indicate the maintenance of human pancreatic acinar cell phenotype at low temperature and the possibility to efficiently label acinar cells, which opens new perspectives for the study of human acinar-to-β-cell transdifferentiation.
Collapse
|
27
|
Wei R, Hong T. Lineage Reprogramming: A Promising Road for Pancreatic β Cell Regeneration. Trends Endocrinol Metab 2016; 27:163-176. [PMID: 26811208 DOI: 10.1016/j.tem.2016.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 12/24/2015] [Accepted: 01/06/2016] [Indexed: 12/18/2022]
Abstract
Cell replacement therapy is a promising method to restore pancreatic β cell function and cure diabetes. Distantly related cells (fibroblasts, keratinocytes, and muscle cells) and developmentally related cells (hepatocytes, gastrointestinal, and pancreatic exocrine cells) have been successfully reprogrammed into β cells in vitro and in vivo. However, while some reprogrammed β cells bear similarities to bona fide β cells, others do not develop into fully functional β cells. Here we review various strategies currently used for β cell reprogramming, including ectopic expression of specific transcription factors associated with islet development, repression of maintenance factors of host cells, regulation of epigenetic modifications, and microenvironmental changes. Development of simple and efficient reprogramming methods is a key priority for developing fully functional β cells suitable for cell replacement therapy.
Collapse
Affiliation(s)
- Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing 100191, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
28
|
Kim HS, Lee MK. β-Cell regeneration through the transdifferentiation of pancreatic cells: Pancreatic progenitor cells in the pancreas. J Diabetes Investig 2016; 7:286-96. [PMID: 27330712 PMCID: PMC4847880 DOI: 10.1111/jdi.12475] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/27/2015] [Accepted: 01/04/2016] [Indexed: 12/17/2022] Open
Abstract
Pancreatic progenitor cell research has been in the spotlight, as these cells have the potential to replace pancreatic β‐cells for the treatment of type 1 and 2 diabetic patients with the absence or reduction of pancreatic β‐cells. During the past few decades, the successful treatment of diabetes through transplantation of the whole pancreas or isolated islets has nearly been achieved. However, novel sources of pancreatic islets or insulin‐producing cells are required to provide sufficient amounts of donor tissues. To overcome this limitation, the use of pancreatic progenitor cells is gaining more attention. In particular, pancreatic exocrine cells, such as duct epithelial cells and acinar cells, are attractive candidates for β‐cell regeneration because of their differentiation potential and pancreatic lineage characteristics. It has been assumed that β‐cell neogenesis from pancreatic progenitor cells could occur in pancreatic ducts in the postnatal stage. Several studies have shown that insulin‐producing cells can arise in the duct tissue of the adult pancreas. Acinar cells also might have the potential to differentiate into insulin‐producing cells. The present review summarizes recent progress in research on the transdifferentiation of pancreatic exocrine cells into insulin‐producing cells, especially duct and acinar cells.
Collapse
Affiliation(s)
- Hyo-Sup Kim
- Division of Endocrinology and Metabolism Department of Medicine Sungkyunkwan University School of Medicine Samsung Biomedical Research Institute Samsung Medical Center Seoul Korea
| | - Moon-Kyu Lee
- Division of Endocrinology and Metabolism Department of Medicine Sungkyunkwan University School of Medicine Samsung Biomedical Research Institute Samsung Medical Center Seoul Korea
| |
Collapse
|
29
|
PNA lectin for purifying mouse acinar cells from the inflamed pancreas. Sci Rep 2016; 6:21127. [PMID: 26884345 PMCID: PMC4756371 DOI: 10.1038/srep21127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/18/2016] [Indexed: 12/14/2022] Open
Abstract
Better methods for purifying human or mouse acinar cells without the need for genetic modification are needed. Such techniques would be advantageous for the specific study of certain mechanisms, such as acinar-to-beta-cell reprogramming and pancreatitis. Ulex Europaeus Agglutinin I (UEA-I) lectin has been used to label and isolate acinar cells from the pancreas. However, the purity of the UEA-I-positive cell fraction has not been fully evaluated. Here, we screened 20 widely used lectins for their binding specificity for major pancreatic cell types, and found that UEA-I and Peanut agglutinin (PNA) have a specific affinity for acinar cells in the mouse pancreas, with minimal affinity for other major pancreatic cell types including endocrine cells, duct cells and endothelial cells. Moreover, PNA-purified acinar cells were less contaminated with mesenchymal and inflammatory cells, compared to UEA-I purified acinar cells. Thus, UEA-I and PNA appear to be excellent lectins for pancreatic acinar cell purification. PNA may be a better choice in situations where mesenchymal cells or inflammatory cells are significantly increased in the pancreas, such as type 1 diabetes, pancreatitis and pancreatic cancer.
Collapse
|
30
|
Li XY, Zhai WJ, Teng CB. Notch Signaling in Pancreatic Development. Int J Mol Sci 2015; 17:ijms17010048. [PMID: 26729103 PMCID: PMC4730293 DOI: 10.3390/ijms17010048] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/22/2015] [Accepted: 12/24/2015] [Indexed: 12/12/2022] Open
Abstract
The Notch signaling pathway plays a significant role in embryonic cell fate determination and adult tissue homeostasis. Various studies have demonstrated the deep involvement of Notch signaling in the development of the pancreas and the lateral inhibition of Notch signaling in pancreatic progenitor differentiation and maintenance. The targeted inactivation of the Notch pathway components promotes premature differentiation of the endocrine pancreas. However, there is still the contrary opinion that Notch signaling specifies the endocrine lineage. Here, we review the current knowledge of the Notch signaling pathway in pancreatic development and its crosstalk with the Wingless and INT-1 (Wnt) and fibroblast growth factor (FGF) pathways.
Collapse
Affiliation(s)
- Xu-Yan Li
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
- College of Life Sciences, Agriculture and Forestry, Qiqihar University, Qiqihar 161006, China.
| | - Wen-Jun Zhai
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| | - Chun-Bo Teng
- College of Life Science, Northeast Forestry University, Harbin 150040, China.
| |
Collapse
|
31
|
Pin CL, Ryan JF, Mehmood R. Acinar cell reprogramming: a clinically important target in pancreatic disease. Epigenomics 2015; 7:267-81. [PMID: 25942535 DOI: 10.2217/epi.14.83] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acinar cells of the pancreas produce the majority of enzymes required for digestion and make up >90% of the cells within the pancreas. Due to a common developmental origin and the plastic nature of the acinar cell phenotype, these cells have been identified as a possible source of β cells as a therapeutic option for Type I diabetes. However, recent evidence indicates that acinar cells are the main source of pancreatic intraepithelial neoplasias (PanINs), the predecessor of pancreatic ductal adenocarcinoma (PDAC). The conversion of acinar cells to either β cells or precursors to PDAC is dependent on reprogramming of the cells to a more primitive, progenitor-like phenotype, which involves changes in transcription factor expression and activity, and changes in their epigenetic program. This review will focus on the mechanisms that promote acinar cell reprogramming, as well as the factors that may affect these mechanisms.
Collapse
Affiliation(s)
- Christopher L Pin
- Department of Paediatrics, Physiology & Pharmacology, & Oncology, University of Western Ontario, London, ON N6C 2V5, Canada
| | | | | |
Collapse
|
32
|
Lemper M, Leuckx G, Heremans Y, German MS, Heimberg H, Bouwens L, Baeyens L. Reprogramming of human pancreatic exocrine cells to β-like cells. Cell Death Differ 2014; 22:1117-30. [PMID: 25476775 PMCID: PMC4572860 DOI: 10.1038/cdd.2014.193] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 09/04/2014] [Accepted: 10/23/2014] [Indexed: 12/31/2022] Open
Abstract
Rodent acinar cells exhibit a remarkable plasticity as they can transdifferentiate to duct-, hepatocyte- and islet β-like cells. We evaluated whether exocrine cells from adult human pancreas can similarly respond to proendocrine stimuli. Exocrine cells from adult human pancreas were transduced directly with lentiviruses expressing activated MAPK (mitogen-activated protein kinase) and STAT3 (signal transducer and activator of transcription 3) and cultured as monolayers or as 3D structures. Expression of STAT3 and MAPK in human exocrine cells activated expression of the proendocrine factor neurogenin 3 in 50% to 80% of transduced exocrine cells. However, the number of insulin-positive cells increased only in the exocrine cells grown initially in suspension before 3D culture. Lineage tracing identified human acinar cells as the source of Ngn3- and insulin-expressing cells. Long-term engraftment into immunocompromised mice increased the efficiency of reprogramming to insulin-positive cells. Our data demonstrate that exocrine cells from human pancreas can be reprogrammed to transplantable insulin-producing cells that acquire functionality. Given the large number of exocrine cells in a donor pancreas, this approach presents a novel strategy to expand cell therapy in type 1 diabetes.
Collapse
Affiliation(s)
- M Lemper
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - G Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Y Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - M S German
- Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143-0669, USA
| | - H Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - L Bouwens
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - L Baeyens
- 1] Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium [2] Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143-0669, USA
| |
Collapse
|
33
|
Puri S, Folias AE, Hebrok M. Plasticity and dedifferentiation within the pancreas: development, homeostasis, and disease. Cell Stem Cell 2014; 16:18-31. [PMID: 25465113 DOI: 10.1016/j.stem.2014.11.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cellular identity is established by genetic, epigenetic, and environmental factors that regulate organogenesis and tissue homeostasis. Although some flexibility in fate potential is beneficial to overall organ health, dramatic changes in cellular identity can have disastrous consequences. Emerging data within the field of pancreas biology are revising current beliefs about how cellular identity is shaped by developmental and environmental cues under homeostasis and stress conditions. Here, we discuss the changes occurring in cellular states upon fate modulation and address how our understanding of the nature of this fluidity is shaping therapeutic approaches to pancreatic disorders such as diabetes and cancer.
Collapse
Affiliation(s)
- Sapna Puri
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexandra E Folias
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
34
|
Marzioni M, Agostinelli L, Candelaresi C, Saccomanno S, De Minicis S, Maroni L, Mingarelli E, Rychlicki C, Trozzi L, Banales JM, Benedetti A, Baroni GS. Activation of the developmental pathway neurogenin-3/microRNA-7a regulates cholangiocyte proliferation in response to injury. Hepatology 2014; 60:1324-35. [PMID: 24925797 DOI: 10.1002/hep.27262] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 06/05/2014] [Indexed: 12/15/2022]
Abstract
UNLABELLED The activation of the biliary stem-cell signaling pathway hairy and enhancer of split 1/pancreatic duodenal homeobox-1 (Hes-1/PDX-1) in mature cholangiocytes determines cell proliferation. Neurogenin-3 (Ngn-3) is required for pancreas development and ductal cell neogenesis. PDX-1-dependent activation of Ngn-3 initiates the differentiation program by inducing microRNA (miR)-7 expression. Here we investigated the role Ngn-3 on cholangiocyte proliferation. Expression levels of Ngn-3 and miR-7 isoforms were tested in cholangiocytes from normal and cholestatic human livers. Ngn-3 was knocked-down in vitro in normal rat cholangiocytes by short interfering RNA (siRNA). In vivo, wild-type and Ngn-3-heterozygous (+/-) mice were subjected to 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) feeding (a model of sclerosing cholangitis) or bile duct ligation (BDL). In the liver, Ngn-3 is expressed specifically in cholangiocytes of primary sclerosing cholangitis (PSC) patients and in mice subjected to DDC or BDL, but not in normal human and mouse livers. Expression of miR-7a-1 and miR-7a-2 isoforms, but not miR-7b, was increased in DDC cholangiocytes compared to normal ones. In normal rat cholangiocytes, siRNA against Ngn-3 blocked the proliferation stimulated by exendin-4. In addition, Ngn-3 knockdown neutralized the overexpression of insulin growth factor-1 (IGF1; promitotic effector) observed after exposure to exendin-4, but not that of PDX-1 or VEGF-A/C. Oligonucleotides anti-miR-7 inhibited the exendin-4-induced proliferation in normal rat cholangiocytes, but did not affect Ngn-3 synthesis. Biliary hyperplasia and collagen deposition induced by DDC or BDL were significantly reduced in Ngn-3(+/-) mice compared to wild-type. CONCLUSION Ngn-3-dependent activation of miR-7a is a determinant of cholangiocyte proliferation. These findings indicate that the reacquisition of a molecular profile typical of organ development is essential for the biological response to injury by mature cholangiocytes.
Collapse
Affiliation(s)
- Marco Marzioni
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
De Waele E, Wauters E, Ling Z, Bouwens L. Conversion of human pancreatic acinar cells toward a ductal-mesenchymal phenotype and the role of transforming growth factor β and activin signaling. Pancreas 2014; 43:1083-92. [PMID: 25003220 DOI: 10.1097/mpa.0000000000000154] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Epithelial-mesenchymal transition may interfere with the differentiation of cultured pancreatic acinar cells toward endocrine cells. Therefore, it will be important to investigate into detail the reprogramming of human pancreatic acinar cells toward a mesenchymal phenotype: the association with acinoductal transdifferentiation, the influence of cell adhesion, and the regulation behind this process. METHODS Human exocrine cells, isolated from donor pancreata, were cultured in suspension or as monolayers. Non-genetic lineage tracing, using labeled ulex europaeus agglutinin 1 lectin, was performed, and the role of the transforming growth factor (TGF-β) superfamily was investigated. RESULTS After 7 days in monolayer culture, the human acinar cells coexpressed the mesenchymal marker vimentin and the ductal marker Sox9. However, when the human exocrine cells were cultured in suspension, epithelial-mesenchymal transition was not observed. The spontaneous transition of the human acinar cells toward a ductal and mesenchymal phenotype was decreased by inhibition of the TGF-β and activin signaling pathways. CONCLUSIONS The human acinar cells spontaneously undergo TGF-β- regulated reprogramming in the monolayer culture. These observations are helpful to develop culture methods for the in vitro reprogramming of pancreatic exocrine to endocrine cells. They are also of potential interest for studies on exocrine acinar cells in the development of pancreatic cancer.
Collapse
Affiliation(s)
- Evelien De Waele
- From the *Cell Differentiation Laboratory, and †Cell Therapy Laboratory, Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | |
Collapse
|
36
|
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
|
37
|
Shimoda M, Chen S, Noguchi H, Takita M, Sugimoto K, Itoh T, Chujo D, Iwahashi S, Naziruddin B, Levy MF, Matsumoto S, Grayburn PA. A new method for generating insulin-secreting cells from human pancreatic epithelial cells after islet isolation transformed by NeuroD1. Hum Gene Ther Methods 2014; 25:206-19. [PMID: 24845703 DOI: 10.1089/hgtb.2013.122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The generation of insulin-secreting cells from nonendocrine pancreatic epithelial cells (NEPEC) has been demonstrated for potential clinical use in the treatment of diabetes. However, previous methods either had limited efficacy or required viral vectors, which hinder clinical application. In this study, we aimed to establish an efficient method of insulin-secreting cell generation from NEPEC without viral vectors. We used nonislet fractions from both research-grade human pancreata from brain-dead donors and clinical pancreata after total pancreatectomy with autologous islet transplantation to treat chronic pancreatitis. It is of note that a few islets could be mingled in the nonislet fractions, but their influence could be limited. The NeuroD1 gene was induced into NEPEC using an effective triple lipofection method without viral vectors to generate insulin-secreting cells. The differentiation was promoted by adding a growth factor cocktail into the culture medium. Using the research-grade human pancreata, the effective method showed high efficacy in the differentiation of NEPEC into insulin-positive cells that secreted insulin in response to a glucose challenge and improved diabetes after being transplanted into diabetic athymic mice. Using the clinical pancreata, similar efficacy was obtained, even though those pancreata suffered chronic pancreatitis. In conclusion, our effective differentiation protocol with triple lipofection method enabled us to achieve very efficient insulin-secreting cell generation from human NEPEC without viral vectors. This method offers the potential for supplemental insulin-secreting cell transplantation for both allogeneic and autologous islet transplantation.
Collapse
|
38
|
Aïello V, Moreno-Asso A, Servitja JM, Martín M. Thyroid hormones promote endocrine differentiation at expenses of exocrine tissue. Exp Cell Res 2014; 322:236-48. [DOI: 10.1016/j.yexcr.2014.01.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 01/17/2014] [Accepted: 01/27/2014] [Indexed: 12/12/2022]
|
39
|
Baeyens L, Lemper M, Leuckx G, De Groef S, Bonfanti P, Stangé G, Shemer R, Nord C, Scheel DW, Pan FC, Ahlgren U, Gu G, Stoffers DA, Dor Y, Ferrer J, Gradwohl G, Wright CVE, Van de Casteele M, German MS, Bouwens L, Heimberg H. Transient cytokine treatment induces acinar cell reprogramming and regenerates functional beta cell mass in diabetic mice. Nat Biotechnol 2014; 32:76-83. [PMID: 24240391 PMCID: PMC4096987 DOI: 10.1038/nbt.2747] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 10/15/2013] [Indexed: 12/20/2022]
Abstract
Reprogramming of pancreatic exocrine cells into cells resembling beta cells may provide a strategy for treating diabetes. Here we show that transient administration of epidermal growth factor and ciliary neurotrophic factor to adult mice with chronic hyperglycemia efficiently stimulates the conversion of terminally differentiated acinar cells to beta-like cells. Newly generated beta-like cells are epigenetically reprogrammed, functional and glucose responsive, and they reinstate normal glycemic control for up to 248 d. The regenerative process depends on Stat3 signaling and requires a threshold number of Neurogenin 3 (Ngn3)-expressing acinar cells. In contrast to previous work demonstrating in vivo conversion of acinar cells to beta-like cells by viral delivery of exogenous transcription factors, our approach achieves acinar-to-beta-cell reprogramming through transient cytokine exposure rather than genetic modification.
Collapse
Affiliation(s)
- Luc Baeyens
- Diabetes Research Center, Vrije Universiteit Brussel, 1090 Brussels, Belgium
- Diabetes Center, California Institute for Regenerative Medicine (CIRM), University of California San Francisco, San Francisco, CA 94143-0669, USA
| | - Marie Lemper
- Diabetes Research Center, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Gunter Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Sofie De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Paola Bonfanti
- Diabetes Research Center, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Geert Stangé
- Diabetes Research Center, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Ruth Shemer
- The Institute for Medical Research Israel-Canada, The Hebrew University, Jerusalem, Israel
| | - Christoffer Nord
- Umeå Center for Molecular Medicine, Umeå University, S-901 87 Umeå, Sweden
| | - David W. Scheel
- Diabetes Center, California Institute for Regenerative Medicine (CIRM), University of California San Francisco, San Francisco, CA 94143-0669, USA
| | - Fong C. Pan
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ulf Ahlgren
- Umeå Center for Molecular Medicine, Umeå University, S-901 87 Umeå, Sweden
| | - Guoqiang Gu
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Doris A. Stoffers
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Yuval Dor
- The Institute for Medical Research Israel-Canada, The Hebrew University, Jerusalem, Israel
| | - Jorge Ferrer
- Institut d’Investigacions Biomediques August Pi i Sunyer, Hospital Clinic de Barcelona, Barcelona, Spain and Imperial College London, London W12 0NN, United Kingdom
| | - Gerard Gradwohl
- Development and Stem Cells Program, Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, 67404, France
| | - Christopher VE Wright
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Michael S. German
- Diabetes Center, California Institute for Regenerative Medicine (CIRM), University of California San Francisco, San Francisco, CA 94143-0669, USA
| | - Luc Bouwens
- Diabetes Research Center, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| |
Collapse
|
40
|
Bar Y, Efrat S. The NOTCH Pathway in β-Cell Growth and Differentiation. THE PANCREATIC BETA CELL 2014; 95:391-405. [DOI: 10.1016/b978-0-12-800174-5.00015-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
41
|
Bouwens L, Houbracken I, Mfopou JK. The use of stem cells for pancreatic regeneration in diabetes mellitus. Nat Rev Endocrinol 2013; 9:598-606. [PMID: 23877422 DOI: 10.1038/nrendo.2013.145] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The endocrine pancreas represents an interesting arena for regenerative medicine and cell therapeutics. One of the major pancreatic diseases, diabetes mellitus is a metabolic disorder caused by having an insufficient number of insulin-producing β cells. Replenishment of β cells by cell transplantation can restore normal metabolic control. The shortage in donor pancreata has meant that the demand for transplantable β cells has outstripped the supply, which could be met by using alternative sources of stem cells. This situation has opened up new areas of research, such as cellular reprogramming and in vivo β-cell regeneration. Pluripotent stem cells seem to be the best option for clinical applications of β-cell regeneration in the near future, as these cells have been demonstrated to represent an unlimited source of functional β cells. Although compelling evidence shows that the adult pancreas retains regenerative capacity, it remains unclear whether this organ contains stem cells. Alternatively, specialized cell types within or outside the pancreas retain plasticity in proliferation and differentiation. Cellular reprogramming or transdifferentiation of exocrine cells or other types of endocrine cells in the pancreas could provide a long-term solution.
Collapse
Affiliation(s)
- Luc Bouwens
- Cell Differentiation Unit, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels B-1090, Belgium
| | | | | |
Collapse
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
Affiliation(s)
- Luc Bouwens
- Cell Differentiation Unit, Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
46
|
Houbracken I, Baeyens L, Ravassard P, Heimberg H, Bouwens L. Gene delivery to pancreatic exocrine cells in vivo and in vitro. BMC Biotechnol 2012; 12:74. [PMID: 23088534 PMCID: PMC3487942 DOI: 10.1186/1472-6750-12-74] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 10/19/2012] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Effective gene transfer to the pancreas or to pancreatic cells has remained elusive although it is essential for studies of genetic lineage tracing and modulation of gene expression. Different transduction methods and viral vectors were tested in vitro and in vivo, in rat and mouse pancreas. RESULTS For in vitro transfection/transduction of rat exocrine cells lipofection reagents, adenoviral vectors, and Mokola- and VSV-G pseudotyped lentiviral vectors were used. For in vivo transduction of mouse and rat pancreas adenoviral vectors and VSV-G lentiviral vectors were injected into the parenchymal tissue. Both lipofection of rat exocrine cell cultures and transduction with Mokola pseudotyped lentiviral vectors were inefficient and resulted in less than 4% EGFP expressing cells. Adenoviral transduction was highly efficient but its usefulness for gene delivery to rat exocrine cells in vitro was hampered by a drastic increase in cell death. In vitro transduction of rat exocrine cells was most optimal with VSV-G pseudotyped lentiviral vectors, with stable transgene expression, no significant effect on cell survival and about 40% transduced cells. In vivo, pancreatic cells could not be transduced by intra-parenchymal administration of lentiviral vectors in mouse and rat pancreas. However, a high efficiency could be obtained by adenoviral vectors, resulting in transient transduction of mainly exocrine acinar cells. Injection in immune-deficient animals diminished leukocyte infiltration and prolonged transgene expression. CONCLUSIONS In summary, our study remarkably demonstrates that transduction of pancreatic exocrine cells requires lentiviral vectors in vitro but adenoviral vectors in vivo.
Collapse
Affiliation(s)
- Isabelle Houbracken
- Cell Differentiation Lab, Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, B-1090, Belgium.
| | | | | | | | | |
Collapse
|
47
|
Guo L, Sans MD, Hou Y, Ernst SA, Williams JA. c-Jun/AP-1 is required for CCK-induced pancreatic acinar cell dedifferentiation and DNA synthesis in vitro. Am J Physiol Gastrointest Liver Physiol 2012; 302:G1381-96. [PMID: 22461029 PMCID: PMC3378092 DOI: 10.1152/ajpgi.00129.2010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endogenous CCK plays an important role in pancreatic regeneration after pancreatitis. We used primary culture of mouse pancreatic acinar cells to evaluate the effect of CCK on acinar cell morphology and gene expression and to determine signaling pathways required for proliferation of acinar cells in vitro. Over 4 days in culture, cells grew out from acini and formed patches of monolayer, which displayed a reduced expression of acinar cell markers including digestive enzymes and Mist1 and an increased expression of ductal and embryonic markers, including cytokeratin 7, β-catenin, E-cadherin, pdx-1, and nestin. There was no appearance of stellate cell markers. CCK enhanced cellular spreading, DNA synthesis, and cyclin D1 expression. When signaling pathways were evaluated, CCK stimulation increased c-Jun expression, JNK and ERK activity, and AP-1 activation. Chemical inhibitors of JNK and ERK pathways, dominant-negative JNK and c-Jun, and c-Jun shRNA significantly inhibited CCK-induced DNA synthesis, CCK-induced AP-1 activation, and cyclin D1 expression. Furthermore, dominant-negative c-Jun reduced the increased expression of β-catenin and the decreased expression of amylase during culture. These results show that MAPK/c-Jun/AP-1 pathway plays an important role in pancreatic acinar cell dedifferentiation and proliferation in culture. Monolayer culture can serve as a model to study acinar cell proliferation similar to regeneration after pancreatitis in vivo.
Collapse
Affiliation(s)
- Lili Guo
- 1Department of Molecular and Integrative Physiology, The University of Michigan Medical School, Ann Arbor, Michigan,
| | - Maria Dolors Sans
- 1Department of Molecular and Integrative Physiology, The University of Michigan Medical School, Ann Arbor, Michigan,
| | - Yanan Hou
- 1Department of Molecular and Integrative Physiology, The University of Michigan Medical School, Ann Arbor, Michigan,
| | - Stephen A. Ernst
- 2Department of Cellular and Developmental Biology, The University of Michigan Medical School, Ann Arbor, Michigan, and
| | - John A. Williams
- 1Department of Molecular and Integrative Physiology, The University of Michigan Medical School, Ann Arbor, Michigan, ,3Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
48
|
Chen S, Shimoda M, Chen J, Matsumoto S, Grayburn PA. Ectopic transgenic expression of NKX2.2 induces differentiation of adult pancreatic progenitors and mediates islet regeneration. Cell Cycle 2012; 11:1544-53. [PMID: 22433950 DOI: 10.4161/cc.19928] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
To pursue islet regeneration in situ in adult pancreas with a diabetic animal model, we used ultrasound targeted microbubble destruction (UTMD) to deliver islet transcription factor genes to the pancreas of STZ-treated rats, specifically using a piggyBac transposon gene delivery system for long-term transgene expression of Nkx2.2 in STZ rat pancreas. Our results show that Nkx2.2 gene induced robust proliferation and differentiation of adult pancreatic progenitors. Our high-resolution confocal images precisely displayed how one single pancreatic progenitor cell differentiated into islets-like clusters and, further, into mature islets with normal morphology in situ in postnatal pancreas. Nkx2.2 targeted to the pancreas by UTMD induces pancreatic progenitor cell proliferation and differentiation with subsequent islet regeneration and cure of STZ-induced diabetes for three months.
Collapse
|
49
|
Mfopou JK, Baeyens L, Bouwens L. Hedgehog signals inhibit postnatal beta cell neogenesis from adult rat exocrine pancreas in vitro. Diabetologia 2012; 55:1024-34. [PMID: 22237687 DOI: 10.1007/s00125-011-2434-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 12/05/2011] [Indexed: 10/14/2022]
Abstract
AIMS/HYPOTHESIS Transdifferentiation of pancreatic exocrine cells into insulin-producing beta cells may represent an important alternative to islets required for diabetes cell therapy. Rat pancreatic acinar cells are known to transdifferentiate into functional beta cells, with recapitulation of several pancreas developmental features. Considering the inhibitory functions of hedgehog signalling in early and mid-stage pancreatic development, we questioned whether it also operates in transdifferentiating acinar cells and whether its modulation would influence postnatal beta cell neogenesis in vitro. METHODS Rat exocrine cells were precultured in suspension for 4 days and then incubated with EGF and leukaemia inhibitory factor (LIF) for 72 h. The hedgehog signalling pathway was modulated during this, and its effects analysed by RT-PCR, immunocytochemistry and western blot. RESULTS Our data indicate induction of Dhh and Ihh, but not Shh, expression during acinar cell culture, resulting in activation of hedgehog targets (Ptc1, Gli1). Exposure of the metaplastic cells to EGF and LIF induced beta cell differentiation without affecting endogenous hedgehog activity. Whereas blocking endogenous hedgehog only slightly increased beta cell neogenesis, exposure to embryoid body-conditioned medium activated hedgehog signalling as well as other pathways such as Notch, resulting in severe blockade of beta cell neogenesis. Interestingly, this effect was partially rescued by treatment with the hedgehog inhibitor, 3-keto-N-(aminoethyl-aminocaproyl-dihydrocinnamoyl)-cyclopamine (KAAD-cyclopamine), alone. CONCLUSIONS/INTERPRETATION We report here Dhh/Ihh-dependent activation of hedgehog targets during pancreatic exocrine cell metaplasia in vitro and a persistent inhibitory function of hedgehog signalling in a model of postnatal beta cell differentiation.
Collapse
Affiliation(s)
- J K Mfopou
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium
| | | | | |
Collapse
|
50
|
Bar Y, Russ HA, Sintov E, Anker-Kitai L, Knoller S, Efrat S. Redifferentiation of expanded human pancreatic β-cell-derived cells by inhibition of the NOTCH pathway. J Biol Chem 2012; 287:17269-17280. [PMID: 22457355 DOI: 10.1074/jbc.m111.319152] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In vitro expansion of β-cells from adult human pancreatic islets would overcome donor β-cell shortage for cell replacement therapy for diabetes. Using a β-cell-specific labeling system we have shown that β-cell expansion is accompanied by dedifferentiation resembling epithelial-mesenchymal transition and loss of insulin expression. Epigenetic analyses indicate that key β-cell genes maintain open chromatin structure in expanded β-cell-derived (BCD) cells, although they are not transcribed. In the developing pancreas important cell-fate decisions are regulated by NOTCH receptors, which signal through the Hairy and Enhancer of Split 1 (HES1) transcription regulator. We have reported that BCD cell dedifferentiation and proliferation in vitro correlate with reactivation of the NOTCH pathway. Inhibition of HES1 expression using shRNA during culture initiation results in reduced β-cell replication and dedifferentiation, suggesting that HES1 inhibition may also affect BCD cell redifferentiation following expansion. Here, we used HES1 shRNA to down-regulate HES1 expression in expanded human BCD cells, showing that HES1 inhibition is sufficient to induce BCD cell redifferentiation, as manifested by a significant increase in insulin expression. Combined treatment with HES1 shRNA, cell aggregation in serum-free medium, and a mixture of soluble factors further stimulated the redifferentiation of BCD cells. In vivo analyses demonstrated the ability of the redifferentiated cells to replace β-cell function in hyperglycemic immunodeficient mice. These findings demonstrate the redifferentiation potential of ex vivo expanded BCD cells and the reproducible differentiating effect of HES1 inhibition in these cells.
Collapse
Affiliation(s)
- Yael Bar
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Holger A Russ
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Elad Sintov
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Leeat Anker-Kitai
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Sarah Knoller
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Shimon Efrat
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel.
| |
Collapse
|