1
|
Sali S, Azzam L, Jaro T, Ali AAG, Mardini A, Al-Dajani O, Khattak S, Butler AE, Azeez JM, Nandakumar M. A perfect islet: reviewing recent protocol developments and proposing strategies for stem cell derived functional pancreatic islets. Stem Cell Res Ther 2025; 16:160. [PMID: 40165291 PMCID: PMC11959787 DOI: 10.1186/s13287-025-04293-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
The search for an effective cell replacement therapy for diabetes has driven the development of "perfect" pancreatic islets from human pluripotent stem cells (hPSCs). These hPSC-derived pancreatic islet-like β cells can overcome the limitations for disease modelling, drug development and transplantation therapies in diabetes. Nevertheless, challenges remain in generating fully functional and mature β cells from hPSCs. This review underscores the significant efforts made by researchers to optimize various differentiation protocols aimed at enhancing the efficiency and quality of hPSC-derived pancreatic islets and proposes methods for their improvement. By emulating the natural developmental processes of pancreatic embryogenesis, specific growth factors, signaling molecules and culture conditions are employed to guide hPSCs towards the formation of mature β cells capable of secreting insulin in response to glucose. However, the efficiency of these protocols varies greatly among different human embryonic stem cell (hESC) and induced pluripotent stem cell (hiPSC) lines. This variability poses a particular challenge for generating patient-specific β cells. Despite recent advancements, the ultimate goal remains to develop a highly efficient directed differentiation protocol that is applicable across all genetic backgrounds of hPSCs. Although progress has been made, further research is required to optimize the protocols and characterization methods that could ensure the safety and efficacy of hPSC-derived pancreatic islets before they can be utilized in clinical settings.
Collapse
Affiliation(s)
- Sujitha Sali
- King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
- Research Department, School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| | - Leen Azzam
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Taraf Jaro
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Ahmed Ali Gebril Ali
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Ali Mardini
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Omar Al-Dajani
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen, 15503, Bahrain
| | - Shahryar Khattak
- King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Alexandra E Butler
- Research Department, School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain.
| | - Juberiya M Azeez
- Research Department, School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| | - Manjula Nandakumar
- Research Department, School of Postgraduate Studies & Research, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| |
Collapse
|
2
|
Hu M, Liu T, Huang H, Ogi D, Tan Y, Ye K, Jin S. Extracellular matrix proteins refine microenvironments for pancreatic organogenesis from induced pluripotent stem cell differentiation. Theranostics 2025; 15:2229-2249. [PMID: 39990212 PMCID: PMC11840725 DOI: 10.7150/thno.104883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/30/2024] [Indexed: 02/25/2025] Open
Abstract
Rationale: The current understanding on manipulating signaling pathways to generate mature human islet organoids with all major hormone-secreting endocrine cell types (i.e., α, β, δ, and γ cells) from induced pluripotent stem cells (iPSCs) is insufficient. However, donor islet shortage necessitates that we produce functional islets in vitro. In this study, we aimed to find decellularized pancreatic extracellular matrix (dpECM) proteins that leverage signaling pathways and promote functional iPSC islet organogenesis. Methods: We performed proteomic analysis to identify key islet promoting factors from porcine and rat dpECM. With this, we identified collagen type II (COL2) as a potential biomaterial cue that endorses islet development from iPSCs. Using global transcriptome profiling, gene set enrichment analysis, immunofluorescence microscopy, flow cytometry, Western blot, and glucose-stimulated hormonal secretion analysis, we examined COL2's role in regulating iPSC pancreatic lineage specification and signaling pathways, critical to islet organogenesis and morphogenesis. Results: We discovered COL2 acts as a functional biomaterial that augments islet development from iPSCs, similar to collagen type V (COL5) as reported in our earlier study. COL2 substantially stimulates the formation of endocrine progenitors and subsequent islet organoids with significantly elevated expressions of pancreatic signature genes and proteins. Furthermore, it enhances islets' glucose sensitivity for hormonal secretion. A cluster of gene expressions associated with various signaling pathways, including but not limited to oxidative phosphorylation, insulin secretion, cell cycle, the canonical WNT, hypoxia, and interferon-γ response, were considerably affected by COL2 and COL5 cues. Conclusion: We demonstrated dpECM's crucial role in refining stem cell differentiation microenvironments for organoid development and maturation. Our findings on biomaterial-stimulated signaling for stem cell specification, organogenesis, and maturation open up a new way to increase the differentiation efficacy of endocrine tissues that can contribute to the production of biologically functional islets.
Collapse
Affiliation(s)
- Ming Hu
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Tianzheng Liu
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Hui Huang
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Derek Ogi
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Yinfei Tan
- Genomics Facility, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Kaiming Ye
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
- Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Sha Jin
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
- Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| |
Collapse
|
3
|
Rotti PG, Yi Y, Gasser G, Yuan F, Sun X, Apak-Evans I, Wu P, Liu G, Choi S, Reeves R, Scioneaux AE, Zhang Y, Winter M, Liang B, Cunicelli N, Uc A, Norris AW, Sussel L, Wells KL, Engelhardt JF. CFTR represses a PDX1 axis to govern pancreatic ductal cell fate. iScience 2024; 27:111393. [PMID: 39687022 PMCID: PMC11647141 DOI: 10.1016/j.isci.2024.111393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/29/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
Inflammation, acinar atrophy, and ductal hyperplasia drive pancreatic remodeling in newborn cystic fibrosis (CF) ferrets lacking a functional cystic fibrosis conductance regulator (CFTR) channel. These changes are associated with a transient phase of glucose intolerance that involves islet destruction and subsequent regeneration near hyperplastic ducts. The phenotypic changes in CF ductal epithelium and their impact on islet function are unknown. Using bulk RNA sequencing (RNA-seq), single-cell RNA sequencing (scRNA-seq), and assay for transposase-accessible chromatin using sequencing (ATAC-seq) on CF ferret models, we demonstrate that ductal CFTR protein constrains PDX1 expression by maintaining PTEN and GSK3β activation. In the absence of CFTR protein, centroacinar cells adopted a bipotent progenitor-like state associated with enhanced WNT/β-Catenin, transforming growth factor β (TGF-β), and AKT signaling. We show that the level of CFTR protein, not its channel function, regulates PDX1 expression. Thus, this study has discovered a cell-autonomous CFTR-dependent mechanism by which CFTR mutations that produced little to no protein could impact pancreatic exocrine/endocrine remodeling in people with CF.
Collapse
Affiliation(s)
| | - Yaling Yi
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Grace Gasser
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Feng Yuan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Xingshen Sun
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Idil Apak-Evans
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Peipei Wu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Guangming Liu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Soon Choi
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Rosie Reeves
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Attilina E. Scioneaux
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yulong Zhang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Michael Winter
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Bo Liang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nathan Cunicelli
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Aliye Uc
- Stead Family Department of Pediatrics, Carver College of Medicine, Iowa City, IA, USA
| | - Andrew W. Norris
- Center for Gene Therapy, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Lori Sussel
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz, Medical Campus, Aurora, CO, USA
| | - Kristen L. Wells
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz, Medical Campus, Aurora, CO, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
4
|
Jarc L, Bandral M, Zanfrini E, Lesche M, Kufrin V, Sendra R, Pezzolla D, Giannios I, Khattak S, Neumann K, Ludwig B, Gavalas A. Regulation of multiple signaling pathways promotes the consistent expansion of human pancreatic progenitors in defined conditions. eLife 2024; 12:RP89962. [PMID: 38180318 PMCID: PMC10945307 DOI: 10.7554/elife.89962] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024] Open
Abstract
The unlimited expansion of human progenitor cells in vitro could unlock many prospects for regenerative medicine. However, it remains an important challenge as it requires the decoupling of the mechanisms supporting progenitor self-renewal and expansion from those mechanisms promoting their differentiation. This study focuses on the expansion of human pluripotent stem (hPS) cell-derived pancreatic progenitors (PP) to advance novel therapies for diabetes. We obtained mechanistic insights into PP expansion requirements and identified conditions for the robust and unlimited expansion of hPS cell-derived PP cells under GMP-compliant conditions through a hypothesis-driven iterative approach. We show that the combined stimulation of specific mitogenic pathways, suppression of retinoic acid signaling, and inhibition of selected branches of the TGFβ and Wnt signaling pathways are necessary for the effective decoupling of PP proliferation from differentiation. This enabled the reproducible, 2000-fold, over 10 passages and 40-45 d, expansion of PDX1+/SOX9+/NKX6-1+ PP cells. Transcriptome analyses confirmed the stabilization of PP identity and the effective suppression of differentiation. Using these conditions, PDX1+/SOX9+/NKX6-1+ PP cells, derived from different, both XY and XX, hPS cell lines, were enriched to nearly 90% homogeneity and expanded with very similar kinetics and efficiency. Furthermore, non-expanded and expanded PP cells, from different hPS cell lines, were differentiated in microwells into homogeneous islet-like clusters (SC-islets) with very similar efficiency. These clusters contained abundant β-cells of comparable functionality as assessed by glucose-stimulated insulin secretion assays. These findings established the signaling requirements to decouple PP proliferation from differentiation and allowed the consistent expansion of hPS cell-derived PP cells. They will enable the establishment of large banks of GMP-produced PP cells derived from diverse hPS cell lines. This approach will streamline SC-islet production for further development of the differentiation process, diabetes research, personalized medicine, and cell therapies.
Collapse
Affiliation(s)
- Luka Jarc
- 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 HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| | - Manuj Bandral
- 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 HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| | - Elisa Zanfrini
- 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 HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| | - Mathias Lesche
- Dresden Concept Genome Centre (DcGC), TU DresdenDresdenGermany
- Center for Molecular and Cellular Bioengineering (CMCB) Technology Platform, TU DresdenDresdenGermany
| | - Vida Kufrin
- 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 HealthNeuherbergGermany
| | - Raquel Sendra
- 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 HealthNeuherbergGermany
| | - Daniela Pezzolla
- German Centre for Diabetes Research (DZD)MunichGermany
- Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, TU DresdenDresdenGermany
| | - Ioannis Giannios
- 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 HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| | - Shahryar Khattak
- Stem Cell Engineering Facility, (SCEF), CRTD, Faculty of Medicine, TU DresdenDresdenGermany
| | - Katrin Neumann
- Stem Cell Engineering Facility, (SCEF), CRTD, Faculty of Medicine, TU DresdenDresdenGermany
| | - Barbara Ludwig
- 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 HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
- Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, TU DresdenDresdenGermany
- Department of Medicine III, University Hospital Carl Gustav Carus and Faculty of Medicine, TU DresdenDresdenGermany
| | - 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 HealthNeuherbergGermany
- German Centre for Diabetes Research (DZD)MunichGermany
| |
Collapse
|
5
|
Ma Z, Zhang X, Zhong W, Yi H, Chen X, Zhao Y, Ma Y, Song E, Xu T. Deciphering early human pancreas development at the single-cell level. Nat Commun 2023; 14:5354. [PMID: 37660175 PMCID: PMC10475098 DOI: 10.1038/s41467-023-40893-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 08/15/2023] [Indexed: 09/04/2023] Open
Abstract
Understanding pancreas development can provide clues for better treatments of pancreatic diseases. However, the molecular heterogeneity and developmental trajectory of the early human pancreas are poorly explored. Here, we performed large-scale single-cell RNA sequencing and single-cell assay for transposase accessible chromatin sequencing of human embryonic pancreas tissue obtained from first-trimester embryos. We unraveled the molecular heterogeneity, developmental trajectories and regulatory networks of the major cell types. The results reveal that dorsal pancreatic multipotent cells in humans exhibit different gene expression patterns than ventral multipotent cells. Pancreato-biliary progenitors that generate ventral multipotent cells in humans were identified. Notch and MAPK signals from mesenchymal cells regulate the differentiation of multipotent cells into trunk and duct cells. Notably, we identified endocrine progenitor subclusters with different differentiation potentials. Although the developmental trajectories are largely conserved between humans and mice, some distinct gene expression patterns have also been identified. Overall, we provide a comprehensive landscape of early human pancreas development to understand its lineage transitions and molecular complexity.
Collapse
Affiliation(s)
- Zhuo Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaofei Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570102, China
| | - Wen Zhong
- Science for Life Laboratory, Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, 581 83, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Hongyan Yi
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570102, China
| | - Xiaowei Chen
- Center for High Throughput Sequencing, Core Facility for Protein Research, Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yinsuo Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570102, China.
| | - Eli Song
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Guangzhou Laboratory, Guangzhou, 510005, China.
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China.
| |
Collapse
|
6
|
Abstract
The islets of Langerhans are highly organized structures that have species-specific, three-dimensional tissue architecture. Islet architecture is critical for proper hormone secretion in response to nutritional stimuli. Islet architecture is disrupted in all types of diabetes mellitus and in cadaveric islets for transplantation during isolation, culture, and perfusion, limiting patient outcomes. Moreover, recapitulating native islet architecture remains a key challenge for in vitro generation of islets from stem cells. In this review, we discuss work that has led to the current understanding of determinants of pancreatic islet architecture, and how this architecture is maintained or disrupted during tissue remodeling in response to normal and pathological metabolic changes. We further discuss both empirical and modeling data that highlight the importance of islet architecture for islet function.
Collapse
Affiliation(s)
- Melissa T. Adams
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barak Blum
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
- CONTACT Barak Blum Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705, USA
| |
Collapse
|
7
|
Basile G, Qadir MMF, Mauvais-Jarvis F, Vetere A, Shoba V, Modell AE, Pastori RL, Russ HA, Wagner BK, Dominguez-Bendala J. Emerging diabetes therapies: Bringing back the β-cells. Mol Metab 2022; 60:101477. [PMID: 35331962 PMCID: PMC8987999 DOI: 10.1016/j.molmet.2022.101477] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Stem cell therapies are finally coming of age as a viable alternative to pancreatic islet transplantation for the treatment of insulin-dependent diabetes. Several clinical trials using human embryonic stem cell (hESC)-derived β-like cells are currently underway, with encouraging preliminary results. Remaining challenges notwithstanding, these strategies are widely expected to reduce our reliance on human isolated islets for transplantation procedures, making cell therapies available to millions of diabetic patients. At the same time, advances in our understanding of pancreatic cell plasticity and the molecular mechanisms behind β-cell replication and regeneration have spawned a multitude of translational efforts aimed at inducing β-cell replenishment in situ through pharmacological means, thus circumventing the need for transplantation. SCOPE OF REVIEW We discuss here the current state of the art in hESC transplantation, as well as the parallel quest to discover agents capable of either preserving the residual mass of β-cells or inducing their proliferation, transdifferentiation or differentiation from progenitor cells. MAJOR CONCLUSIONS Stem cell-based replacement therapies in the mold of islet transplantation are already around the corner, but a permanent cure for type 1 diabetes will likely require the endogenous regeneration of β-cells aided by interventions to restore the immune balance. The promise of current research avenues and a strong pipeline of clinical trials designed to tackle these challenges bode well for the realization of this goal.
Collapse
Affiliation(s)
- G Basile
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - M M F Qadir
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - F Mauvais-Jarvis
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - A Vetere
- Broad Institute, Cambridge, MA, USA
| | - V Shoba
- Broad Institute, Cambridge, MA, USA
| | | | - R L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - H A Russ
- Barbara Davis Center for Diabetes, Colorado University Anschutz Medical Campus, Aurora, CO, USA.
| | | | - J Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
8
|
Wang HL, Wang L, Zhao CY, Lan HY. Role of TGF-Beta Signaling in Beta Cell Proliferation and Function in Diabetes. Biomolecules 2022; 12:373. [PMID: 35327565 PMCID: PMC8945211 DOI: 10.3390/biom12030373] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 12/27/2022] Open
Abstract
Beta (β) cell dysfunction or loss is the common pathological feature in all types of diabetes mellitus (diabetes). Resolving the underlying mechanism may facilitate the treatment of diabetes by preserving the β cell population and function. It is known that TGF-β signaling plays diverse roles in β cell development, function, proliferation, apoptosis, and dedifferentiation. Inhibition of TGF-β signaling expands β cell lineage in the development. However, deletion of Tgfbr1 has no influence on insulin demand-induced but abolishes inflammation-induced β cell proliferation. Among canonical TGF-β signaling, Smad3 but not Smad2 is the predominant repressor of β cell proliferation in response to systemic insulin demand. Deletion of Smad3 simultaneously improves β cell function, apoptosis, and systemic insulin resistance with the consequence of eliminated overt diabetes in diabetic mouse models, revealing Smad3 as a key mediator and ideal therapeutic target for type-2 diabetes. However, Smad7 shows controversial effects on β cell proliferation and glucose homeostasis in animal studies. On the other hand, overexpression of Tgfb1 prevents β cells from autoimmune destruction without influence on β cell function. All these findings reveal the diverse regulatory roles of TGF-β signaling in β cell biology.
Collapse
Affiliation(s)
- Hong-Lian Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (H.-L.W.); (L.W.)
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (H.-L.W.); (L.W.)
| | - Chang-Ying Zhao
- Department of Endocrinology, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China;
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Guangdong Academy of Sciences, Guangdong Provincial People’s Hospital Joint Research Laboratory on Immunological and Genetic Kidney Diseases, The Chinese University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
9
|
Li J, Chen B, Fellows GF, Goodyer CG, Wang R. Activation of Pancreatic Stellate Cells Is Beneficial for Exocrine but Not Endocrine Cell Differentiation in the Developing Human Pancreas. Front Cell Dev Biol 2021; 9:694276. [PMID: 34490247 PMCID: PMC8418189 DOI: 10.3389/fcell.2021.694276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/14/2021] [Indexed: 02/04/2023] Open
Abstract
Pancreatic stellate cells (PaSCs) are non-endocrine, mesenchymal-like cells that reside within the peri-pancreatic tissue of the rodent and human pancreas. PaSCs regulate extracellular matrix (ECM) turnover in maintaining the integrity of pancreatic tissue architecture. Although there is evidence indicating that PaSCs are involved in islet cell survival and function, its role in islet cell differentiation during human pancreatic development remains unclear. The present study examines the expression pattern and functional role of PaSCs in islet cell differentiation of the developing human pancreas from late 1st to 2nd trimester of pregnancy. The presence of PaSCs in human pancreata (8–22 weeks of fetal age) was characterized by ultrastructural, immunohistological, quantitative RT-PCR and western blotting approaches. Using human fetal PaSCs derived from pancreata at 14–16 weeks, freshly isolated human fetal islet-epithelial cell clusters (hIECCs) were co-cultured with active or inactive PaSCs in vitro. Ultrastructural and immunofluorescence analysis demonstrated a population of PaSCs near ducts and newly formed islets that appeared to make complex cell-cell dendritic-like contacts. A small subset of PaSCs co-localized with pancreatic progenitor-associated transcription factors (PDX1, SOX9, and NKX6-1). PaSCs were highly proliferative, with significantly higher mRNA and protein levels of PaSC markers (desmin, αSMA) during the 1st trimester of pregnancy compared to the 2nd trimester. Isolated human fetal PaSCs were identified by expression of stellate cell markers and ECM. Suppression of PaSC activation, using all-trans retinoic acid (ATRA), resulted in reduced PaSC proliferation and ECM proteins. Co-culture of hIECCs, directly on PaSCs or indirectly using Millicell® Inserts or using PaSC-conditioned medium, resulted in a reduction the number of insulin+ cells but a significant increase in the number of amylase+ cells. Suppression of PaSC activation or Notch activity during the co-culture resulted in an increase in beta-cell differentiation. This study determined that PaSCs, abundant during the 1st trimester of pancreatic development but decreased in the 2nd trimester, are located near ductal and islet structures. Direct and indirect co-cultures of hIECCs with PaSCs suggest that activation of PaSCs has opposing effects on beta-cell and exocrine cell differentiation during human fetal pancreas development, and that these effects may be dependent on Notch signaling.
Collapse
Affiliation(s)
- Jinming Li
- Children's Health Research Institute, Western University, London, ON, Canada.,Departments of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Bijun Chen
- Children's Health Research Institute, Western University, London, ON, Canada
| | - George F Fellows
- Department of Obstetrics and Gynecology, Western University, London, ON, Canada
| | | | - Rennian Wang
- Children's Health Research Institute, Western University, London, ON, Canada.,Departments of Physiology and Pharmacology, Western University, London, ON, Canada
| |
Collapse
|
10
|
Abstract
Pancreatic islet beta cells (β-cells) synthesize and secrete insulin in response to rising glucose levels and thus are a prime target in both major forms of diabetes. Type 1 diabetes ensues due to autoimmune destruction of β-cells. On the other hand, the prevailing insulin resistance and hyperglycemia in type 2 diabetes (T2D) elicits a compensatory response from β-cells that involves increases in β-cell mass and function. However, the sustained metabolic stress results in β-cell failure, characterized by severe β-cell dysfunction and loss of β-cell mass. Dynamic changes to β-cell mass also occur during pancreatic development that involves extensive growth and morphogenesis. These orchestrated events are triggered by multiple signaling pathways, including those representing the transforming growth factor β (TGF-β) superfamily. TGF-β pathway ligands play important roles during endocrine pancreas development, β-cell proliferation, differentiation, and apoptosis. Furthermore, new findings are suggestive of TGF-β's role in regulation of adult β-cell mass and function. Collectively, these findings support the therapeutic utility of targeting TGF-β in diabetes. Summarizing the role of the various TGF-β pathway ligands in β-cell development, growth and function in normal physiology, and during diabetes pathogenesis is the topic of this mini-review.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Cell Growth and Metabolism Section, Diabetes, Endocrinology & Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Clinical Research Center, Bethesda, MD, USA
| | - Ji-Hyeon Lee
- Cell Growth and Metabolism Section, Diabetes, Endocrinology & Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Clinical Research Center, Bethesda, MD, USA
| | - Sushil G Rane
- Cell Growth and Metabolism Section, Diabetes, Endocrinology & Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Clinical Research Center, Bethesda, MD, USA
- Correspondence: Sushil G. Rane, PhD, Cell Growth and Metabolism Section, Diabetes, Endocrinology and Obesity Branch, National Institutes of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Clinical Research Center, Building 10, CRC-West 5-5940, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
11
|
Sehrawat A, Shiota C, Mohamed N, DiNicola J, Saleh M, Kalsi R, Zhang T, Wang Y, Prasadan K, Gittes GK. SMAD7 enhances adult β-cell proliferation without significantly affecting β-cell function in mice. J Biol Chem 2020; 295:4858-4869. [PMID: 32122971 DOI: 10.1074/jbc.ra119.011011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 02/18/2020] [Indexed: 12/19/2022] Open
Abstract
The interplay between the transforming growth factor β (TGF-β) signaling proteins, SMAD family member 2 (SMAD2) and 3 (SMAD3), and the TGF-β-inhibiting SMAD, SMAD7, seems to play a vital role in proper pancreatic endocrine development and also in normal β-cell function in adult pancreatic islets. Here, we generated conditional SMAD7 knockout mice by crossing insulin1Cre mice with SMAD7fx/fx mice. We also created a β cell-specific SMAD7-overexpressing mouse line by crossing insulin1Dre mice with HPRT-SMAD7/RosaGFP mice. We analyzed β-cell function in adult islets when SMAD7 was either absent or overexpressed in β cells. Loss of SMAD7 in β cells inhibited proliferation, and SMAD7 overexpression enhanced cell proliferation. However, alterations in basic glucose homeostasis were not detectable following either SMAD7 deletion or overexpression in β cells. Our results show that both the absence and overexpression of SMAD7 affect TGF-β signaling and modulates β-cell proliferation but does not appear to alter β-cell function. Reversible SMAD7 overexpression may represent an attractive therapeutic option to enhance β-cell proliferation without negative effects on β-cell function.
Collapse
Affiliation(s)
- Anuradha Sehrawat
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Chiyo Shiota
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Nada Mohamed
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Julia DiNicola
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Mohamed Saleh
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Ranjeet Kalsi
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Ting Zhang
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Yan Wang
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Krishna Prasadan
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - George K Gittes
- Department of Pediatric Surgery, Children's Hospital of University of Pittsburgh, Pittsburgh, Pennsylvania 15224
| |
Collapse
|
12
|
Zhang F, Ma D, Liu T, Liu YH, Guo J, Song J, Wu Q, Pan Y, Zhang Y, Guo C, Teng C, Jin L. Expansion and Maintenance of CD133-Expressing Pancreatic Ductal Epithelial Cells by Inhibition of TGF-β Signaling. Stem Cells Dev 2019; 28:1236-1252. [PMID: 31311463 DOI: 10.1089/scd.2019.0087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Restoring β-cell mass by the transplantation of pancreatic islets is an effective diabetes treatment, but it is limited by the shortage of donor organs. CD133-expressing pancreatic ductal epithelial cells (PDECs) have the ability to generate insulin-producing cells. The expansion of these cells is dependent on extrinsic niche factors, but few of those signals have been identified. In this study, CD133-expressing PDECs were purified by sorting from adult wild-type C57BL/6 mice and TGFβRIInull/null mice. Furthermore, using immunofluorescence and transplantation assays, we found that the inhibition of the transforming growth factor-β (TGF-β) pathway promoted the expansion of CD133-expressing PDECs for many generations and maintained the ability of CD133-expressing PDECs to generate insulin-producing cells. Moreover, western blot, qRT-PCR, and dual luciferase assays using TGF-β inhibitors were performed to identify the mechanisms by which TGF-β signaling regulates proliferation and differentiation. The results showed that the inhibition of TGF-β signaling enhanced Id2 binding to the promoter region of the cell proliferation repressor p16 and promoted the expansion of CD133-expressing PDECs, and the increased Id2 binding to NeuroD1 decreased the transcription of Pax6 to maintain CD133-expressing PDECs in the Pdx1-expression stage. Taken together, the effect of TGF-β antagonists on CD133-expressing PDECs reveals a novel paradigm of signaling that explains the balance between the expansion and differentiation of pancreatic duct epithelial progenitors.
Collapse
Affiliation(s)
- Fangfang Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Dongshen Ma
- Department of Pathology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tingsheng Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yu Hong Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jiamin Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jing Song
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Qiong Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Changying Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Chunbo Teng
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, School of life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
13
|
Pancreas organogenesis: The interplay between surrounding microenvironment(s) and epithelium-intrinsic factors. Curr Top Dev Biol 2019; 132:221-256. [DOI: 10.1016/bs.ctdb.2018.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
14
|
Wang Z, Ding L, Zhu J, Su Y, Wang L, Liu L, Ma Q, Yao H. Long non-coding RNA MEG3 mediates high glucose-induced endothelial cell dysfunction. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:1088-1100. [PMID: 31938204 PMCID: PMC6958101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/22/2017] [Indexed: 06/10/2023]
Abstract
Long noncoding RNAs (lncRNAs) are implicated in the progression of diabetes mellitus (DM) and diabetes-induced endothelial dysfunction. Maternally expressed gene 3 (MEG3) encodes an lncRNA which is suggested to function as a tumor suppressor. Therefore, the aim of the present study was to investigate whether MEG3 is a potential regulator and molecular biomarker of high glucose-induced endothelial dysfunction. LncRNA Meg3-specific small interfering RNA (siRNA) and scrambled (Scr) siRNA were transfected for MEG3 dysfunction studies. RNA and protein expression were examined by quantitative RT-PCR (qPCR) and Western blot, respectively. The percentage of apoptotic cells was measured by flow cytometry. Cell viability was determined through MTT assay. This study demonstrates involvement of lncRNA MEG3 in high glucose-induced endothelial dysfunction. MEG3 is significantly downregulated in an endothelial cell model of hyperglycemia. In addition, MEG3 knockdown could exacerbate inflammatory damage in endothelial cells. Interestingly, MEG3 knockdown in HUVECs significantly induced proliferation and inhibited apoptosis by upregulating Bcl-2 and downregulating Bax, caspase-3, and P53. It should be noted that MEG3 knockdown could activate the TGF-β signaling pathway via upregulating TGF-β1, SMAD2, and SMAD7 and activate the Wnt/β-catenin signaling pathway via upregulating β-catenin and Cyclin D1 and downregulating TCF7L2. Our results indicate that MEG3 can be regarded as a novel therapeutic target and molecular biomarker for high glucose-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Public Health, Xinjiang Medical UniversityUrumqi, Xinjiang, China
- Xinjiang Key Laboratory of Metabolic Disease Research, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, Xinjiang, China
| | - Lili Ding
- Department of Public Health, Xinjiang Medical UniversityUrumqi, Xinjiang, China
- Department of Infection Control, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, Xinjiang, China
| | - Jun Zhu
- Xinjiang Key Laboratory of Metabolic Disease Research, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, Xinjiang, China
- Department of Endocrinology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, Xinjiang, China
| | - Yinxia Su
- Xinjiang Key Laboratory of Metabolic Disease Research, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, Xinjiang, China
| | - Li Wang
- Xinjiang Key Laboratory of Metabolic Disease Research, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, Xinjiang, China
| | - Lina Liu
- Department of Endocrinology, Branch of The First Affiliated Hospital of Xinjiang Medical UniversityChangji, Xinjiang, China
| | - Qi Ma
- Xinjiang Key Laboratory of Metabolic Disease Research, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, Xinjiang, China
| | - Hua Yao
- Xinjiang Key Laboratory of Metabolic Disease Research, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, Xinjiang, China
| |
Collapse
|
15
|
TGF-β Family Signaling in Ductal Differentiation and Branching Morphogenesis. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031997. [PMID: 28289061 DOI: 10.1101/cshperspect.a031997] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epithelial cells contribute to the development of various vital organs by generating tubular and/or glandular architectures. The fully developed forms of ductal organs depend on processes of branching morphogenesis, whereby frequency, total number, and complexity of the branching tissue define the final architecture in the organ. Some ductal tissues, like the mammary gland during pregnancy and lactation, disintegrate and regenerate through periodic cycles. Differentiation of branched epithelia is driven by antagonistic actions of parallel growth factor systems that mediate epithelial-mesenchymal communication. Transforming growth factor-β (TGF-β) family members and their extracellular antagonists are prominently involved in both normal and disease-associated (e.g., malignant or fibrotic) ductal tissue patterning. Here, we discuss collective knowledge that permeates the roles of TGF-β family members in the control of the ductal tissues in the vertebrate body.
Collapse
|
16
|
Bastidas-Ponce A, Scheibner K, Lickert H, Bakhti M. Cellular and molecular mechanisms coordinating pancreas development. Development 2017; 144:2873-2888. [PMID: 28811309 DOI: 10.1242/dev.140756] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pancreas is an endoderm-derived glandular organ that participates in the regulation of systemic glucose metabolism and food digestion through the function of its endocrine and exocrine compartments, respectively. While intensive research has explored the signaling pathways and transcriptional programs that govern pancreas development, much remains to be discovered regarding the cellular processes that orchestrate pancreas morphogenesis. Here, we discuss the developmental mechanisms and principles that are known to underlie pancreas development, from induction and lineage formation to morphogenesis and organogenesis. Elucidating such principles will help to identify novel candidate disease genes and unravel the pathogenesis of pancreas-related diseases, such as diabetes, pancreatitis and cancer.
Collapse
Affiliation(s)
- Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Katharina Scheibner
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Technical University of Munich, Medical Faculty, 81675 Munich, Germany
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany .,Institute of Stem Cell Research, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
| |
Collapse
|
17
|
Pauerstein PT, Tellez K, Willmarth KB, Park KM, Hsueh B, Efsun Arda H, Gu X, Aghajanian H, Deisseroth K, Epstein JA, Kim SK. A radial axis defined by semaphorin-to-neuropilin signaling controls pancreatic islet morphogenesis. Development 2017; 144:3744-3754. [PMID: 28893946 DOI: 10.1242/dev.148684] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 09/04/2017] [Indexed: 12/24/2022]
Abstract
The islets of Langerhans are endocrine organs characteristically dispersed throughout the pancreas. During development, endocrine progenitors delaminate, migrate radially and cluster to form islets. Despite the distinctive distribution of islets, spatially localized signals that control islet morphogenesis have not been discovered. Here, we identify a radial signaling axis that instructs developing islet cells to disperse throughout the pancreas. A screen of pancreatic extracellular signals identified factors that stimulated islet cell development. These included semaphorin 3a, a guidance cue in neural development without known functions in the pancreas. In the fetal pancreas, peripheral mesenchymal cells expressed Sema3a, while central nascent islet cells produced the semaphorin receptor neuropilin 2 (Nrp2). Nrp2 mutant islet cells developed in proper numbers, but had defects in migration and were unresponsive to purified Sema3a. Mutant Nrp2 islets aggregated centrally and failed to disperse radially. Thus, Sema3a-Nrp2 signaling along an unrecognized pancreatic developmental axis constitutes a chemoattractant system essential for generating the hallmark morphogenetic properties of pancreatic islets. Unexpectedly, Sema3a- and Nrp2-mediated control of islet morphogenesis is strikingly homologous to mechanisms that regulate radial neuronal migration and cortical lamination in the developing mammalian brain.
Collapse
Affiliation(s)
- Philip T Pauerstein
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Krissie Tellez
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kirk B Willmarth
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Keon Min Park
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brian Hsueh
- Departments of Bioengineering and of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - H Efsun Arda
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xueying Gu
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Haig Aghajanian
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karl Deisseroth
- Departments of Bioengineering and of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
18
|
Prasadan K, Shiota C, Xiangwei X, Ricks D, Fusco J, Gittes G. A synopsis of factors regulating beta cell development and beta cell mass. Cell Mol Life Sci 2016; 73:3623-37. [PMID: 27105622 PMCID: PMC5002366 DOI: 10.1007/s00018-016-2231-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/24/2016] [Accepted: 04/14/2016] [Indexed: 12/29/2022]
Abstract
The insulin-secreting beta cells in the endocrine pancreas regulate blood glucose levels, and loss of functional beta cells leads to insulin deficiency, hyperglycemia (high blood glucose) and diabetes mellitus. Current treatment strategies for type-1 (autoimmune) diabetes are islet transplantation, which has significant risks and limitations, or normalization of blood glucose with insulin injections, which is clearly not ideal. The type-1 patients can lack insulin counter-regulatory mechanism; therefore, hypoglycemia is a potential risk. Hence, a cell-based therapy offers a better alternative for the treatment of diabetes. Past research was focused on attempting to generate replacement beta cells from stem cells; however, recently there has been an increasing interest in identifying mechanisms that will lead to the conversion of pre-existing differentiated endocrine cells into beta cells. The goal of this review is to provide an overview of several of the key factors that regulate new beta cell formation (neogenesis) and beta cell proliferation.
Collapse
Affiliation(s)
- Krishna Prasadan
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Chiyo Shiota
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Xiao Xiangwei
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - David Ricks
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Joseph Fusco
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - George Gittes
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
19
|
Dhawan S, Dirice E, Kulkarni RN, Bhushan A. Inhibition of TGF-β Signaling Promotes Human Pancreatic β-Cell Replication. Diabetes 2016; 65:1208-18. [PMID: 26936960 PMCID: PMC4839200 DOI: 10.2337/db15-1331] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/22/2016] [Indexed: 12/19/2022]
Abstract
Diabetes is associated with loss of functional pancreatic β-cells, and restoration of β-cells is a major goal for regenerative therapies. Endogenous regeneration of β-cells via β-cell replication has the potential to restore cellular mass; however, pharmacological agents that promote regeneration or expansion of endogenous β-cells have been elusive. The regenerative capacity of β-cells declines rapidly with age, due to accumulation of p16(INK4a), resulting in limited capacity for adult endocrine pancreas regeneration. Here, we show that transforming growth factor-β (TGF-β) signaling via Smad3 integrates with the trithorax complex to activate and maintain Ink4a expression to prevent β-cell replication. Importantly, inhibition of TGF-β signaling can result in repression of the Ink4a/Arf locus, resulting in increased β-cell replication in adult mice. Furthermore, small molecule inhibitors of the TGF-β pathway promote β-cell replication in human islets transplanted into NOD-scid IL-2Rg(null) mice. These data reveal a novel role for TGF-β signaling in the regulation of the Ink4a/Arf locus and highlight the potential of using small molecule inhibitors of TGF-β signaling to promote human β-cell replication.
Collapse
MESH Headings
- Animals
- Benzamides/pharmacology
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclin-Dependent Kinase Inhibitor p16/agonists
- Cyclin-Dependent Kinase Inhibitor p16/antagonists & inhibitors
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Cyclin-Dependent Kinase Inhibitor p16/metabolism
- Dioxoles/pharmacology
- Female
- Gene Expression Regulation/drug effects
- Humans
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/physiology
- Islets of Langerhans Transplantation/physiology
- Male
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Receptors, Transforming Growth Factor beta/agonists
- Receptors, Transforming Growth Factor beta/antagonists & inhibitors
- Receptors, Transforming Growth Factor beta/metabolism
- Regeneration/drug effects
- Signal Transduction/drug effects
- Smad3 Protein/metabolism
- Tissue Banks
- Transforming Growth Factor beta1/antagonists & inhibitors
- Transforming Growth Factor beta1/metabolism
- Transplantation, Heterologous
- Transplantation, Heterotopic
Collapse
Affiliation(s)
- Sangeeta Dhawan
- Division of Endocrinology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Ercument Dirice
- Islet Cell and Regenerative Biology, Joslin Diabetes Center and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Anil Bhushan
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
20
|
El-Gohary Y, Wiersch J, Tulachan S, Xiao X, Guo P, Rymer C, Fischbach S, Prasadan K, Shiota C, Gaffar I, Song Z, Galambos C, Esni F, Gittes GK. Intraislet Pancreatic Ducts Can Give Rise to Insulin-Positive Cells. Endocrinology 2016; 157:166-75. [PMID: 26505114 PMCID: PMC4701882 DOI: 10.1210/en.2015-1175] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 10/23/2015] [Indexed: 01/31/2023]
Abstract
A key question in diabetes research is whether new β-cells can be derived from endogenous, nonendocrine cells. The potential for pancreatic ductal cells to convert into β-cells is a highly debated issue. To date, it remains unclear what anatomical process would result in duct-derived cells coming to exist within preexisting islets. We used a whole-mount technique to directly visualize the pancreatic ductal network in young wild-type mice, young humans, and wild-type and transgenic mice after partial pancreatectomy. Pancreatic ductal networks, originating from the main ductal tree, were found to reside deep within islets in young mice and humans but not in mature mice or humans. These networks were also not present in normal adult mice after partial pancreatectomy, but TGF-β receptor mutant mice demonstrated formation of these intraislet duct structures after partial pancreatectomy. Genetic and viral lineage tracings were used to determine whether endocrine cells were derived from pancreatic ducts. Lineage tracing confirmed that pancreatic ductal cells can typically convert into new β-cells in normal young developing mice as well as in adult TGF-β signaling mutant mice after partial pancreatectomy. Here the direct visual evidence of ducts growing into islets, along with lineage tracing, not only represents strong evidence for duct cells giving rise to β-cells in the postnatal pancreas but also importantly implicates TGF-β signaling in this process.
Collapse
Affiliation(s)
- Yousef El-Gohary
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - John Wiersch
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Sidhartha Tulachan
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Xiangwei Xiao
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Ping Guo
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Christopher Rymer
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Shane Fischbach
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Krishna Prasadan
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Chiyo Shiota
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Iljana Gaffar
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Zewen Song
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Csaba Galambos
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - Farzad Esni
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| | - George K Gittes
- Departments of Surgery (Y.E.-G., J.W., X.X., P.G., K.P., C.S., I.G., Z.S., F.E., G.K.G.) and Pediatrics (C.R.), Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224; Department of Surgery (Y.E.-G.), Stony Brook University Medical Center, Stony Brook, New York 11794; Department of Surgery (J.W.), University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Section of Gastroenterology/Hepatology (S.T.), Georgia Regents University, Augusta, Georgia 30912; Division of Biology and Medicine (S.F.), Brown University, Providence, Rhode Island 02912; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central South University, Yuelu, Changsha, Hunan 410013, China; and Department of Pathology and Laboratory Medicine (C.G.), Children's Hospital Colorado, Aurora, Colorado 80045
| |
Collapse
|
21
|
Verchere CB, Lynn FC. Reawakening the Duct Cell Progenitor? Endocrinology 2016; 157:52-3. [PMID: 26717475 DOI: 10.1210/en.2015-2008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- C Bruce Verchere
- Departments of Surgery (C.B.V., F.C.L.) and Pathology and Laboratory Medicine (C.B.V.), Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada V5Z4H4
| | - Francis C Lynn
- Departments of Surgery (C.B.V., F.C.L.) and Pathology and Laboratory Medicine (C.B.V.), Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada V5Z4H4
| |
Collapse
|
22
|
Chen L, Chu C, Kong X, Huang G, Huang T, Cai YD. A hybrid computational method for the discovery of novel reproduction-related genes. PLoS One 2015; 10:e0117090. [PMID: 25768094 PMCID: PMC4358884 DOI: 10.1371/journal.pone.0117090] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 12/13/2014] [Indexed: 12/12/2022] Open
Abstract
Uncovering the molecular mechanisms underlying reproduction is of great importance to infertility treatment and to the generation of healthy offspring. In this study, we discovered novel reproduction-related genes with a hybrid computational method, integrating three different types of method, which offered new clues for further reproduction research. This method was first executed on a weighted graph, constructed based on known protein-protein interactions, to search the shortest paths connecting any two known reproduction-related genes. Genes occurring in these paths were deemed to have a special relationship with reproduction. These newly discovered genes were filtered with a randomization test. Then, the remaining genes were further selected according to their associations with known reproduction-related genes measured by protein-protein interaction score and alignment score obtained by BLAST. The in-depth analysis of the high confidence novel reproduction genes revealed hidden mechanisms of reproduction and provided guidelines for further experimental validations.
Collapse
Affiliation(s)
- Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, People’s Republic of China
| | - Chen Chu
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, People’s Republic of China
| | - Xiangyin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, People’s Republic of China
| | - Guohua Huang
- Institute of Systems Biology, Shanghai University, Shanghai, 200444, People’s Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, People’s Republic of China
- * E-mail: (TH); (YDC)
| | - Yu-Dong Cai
- Institute of Systems Biology, Shanghai University, Shanghai, 200444, People’s Republic of China
- * E-mail: (TH); (YDC)
| |
Collapse
|
23
|
Shim JH, Kim J, Han J, An SY, Jang YJ, Son J, Woo DH, Kim SK, Kim JH. Pancreatic Islet-Like Three-Dimensional Aggregates Derived From Human Embryonic Stem Cells Ameliorate Hyperglycemia in Streptozotocin-Induced Diabetic Mice. Cell Transplant 2014; 24:2155-68. [PMID: 25397866 DOI: 10.3727/096368914x685438] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We previously reported the in vitro differentiation of human embryonic stem cells (hESCs) into pancreatic endoderm. Here we demonstrate that islet-like three-dimensional (3D) aggregates can be derived from the pancreatic endoderm by optimizing our previous protocol. Sequential treatment with Wnt3a, activin A, and noggin induced a transient upregulation of T and MixL1, followed by increased expression of endodermal genes, including FOXA2, SOX17, and CXCR4. Subsequent treatment with retinoic acid highly upregulated PDX1 expression. We also show that inhibition of sonic hedgehog signaling by bFGF/activin βB and cotreatment with VEGF and FGF7 produced many 3D cellular clusters that express both SOX17 and PDX1. We found for the first time that proteoglycans and vimentin(+) mesenchymal cells were mainly localized in hESC-derived PDX1(+) clusters. Importantly, treatment with chlorate, an inhibitor of proteoglycan sulfation, together with inhibition of Notch signaling significantly increased the expression of Neurog3 and NeuroD1, promoting a transition from PDX1(+) progenitor cells toward mature pancreatic endocrine cells. Purified dithizone(+) 3D aggregates generated by our refined protocol produced pancreatic hormones and released insulin in response to both glucose and pharmacological drugs in vitro. Furthermore, the islet-like 3D aggregates decreased blood glucose levels and continued to exhibit pancreatic features after transplantation into diabetic mice. Generation of islet-like 3D cell aggregates from human pluripotent stem cells may overcome the shortage of cadaveric donor islets for future cases of clinical islet transplantation.
Collapse
Affiliation(s)
- Joong-Hyun Shim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Schiesser JV, Wells JM. Generation of β cells from human pluripotent stem cells: are we there yet? Ann N Y Acad Sci 2014; 1311:124-37. [PMID: 24611778 DOI: 10.1111/nyas.12369] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In 1998, the landmark paper describing the isolation and culture of human embryonic stem cells (ESCs) was published. Since that time, the main goal of many diabetes researchers has been to derive β cells from ESCs as a renewable cell-based therapy for the treatment of patients with diabetes. In working toward this goal, numerous protocols that attempt to recapitulate normal pancreatic development have been published that result in the formation of pancreatic cell types from human pluripotent cells. This review examines stem cell differentiation methods and places them within the context of pancreatic development. We additionally compare strategies that are currently being used to generate pancreatic cell types and contrast them with approaches that have been used to generate functional cell types in different lineages. In doing this, we aim to identify how new approaches might be used to improve yield and functionality of in vitro-derived pancreatic β cells as an eventual cell-based therapy for type 1 diabetes.
Collapse
Affiliation(s)
- Jacqueline V Schiesser
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | |
Collapse
|
25
|
El-Gohary Y, Tulachan S, Wiersch J, Guo P, Welsh C, Prasadan K, Paredes J, Shiota C, Xiao X, Wada Y, Diaz M, Gittes G. A smad signaling network regulates islet cell proliferation. Diabetes 2014; 63:224-36. [PMID: 24089514 PMCID: PMC3868054 DOI: 10.2337/db13-0432] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Pancreatic β-cell loss and dysfunction are critical components of all types of diabetes. Human and rodent β-cells are able to proliferate, and this proliferation is an important defense against the evolution and progression of diabetes. Transforming growth factor-β (TGF-β) signaling has been shown to affect β-cell development, proliferation, and function, but β-cell proliferation is thought to be the only source of new β-cells in the adult. Recently, β-cell dedifferentiation has been shown to be an important contributory mechanism to β-cell failure. In this study, we tie together these two pathways by showing that a network of intracellular TGF-β regulators, smads 7, 2, and 3, control β-cell proliferation after β-cell loss, and specifically, smad7 is necessary for that β-cell proliferation. Importantly, this smad7-mediated proliferation appears to entail passing through a transient, nonpathologic dedifferentiation of β-cells to a pancreatic polypeptide-fold hormone-positive state. TGF-β receptor II appears to be a receptor important for controlling the status of the smad network in β-cells. These studies should help our understanding of properly regulated β-cell replication.
Collapse
Affiliation(s)
- Yousef El-Gohary
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Sidhartha Tulachan
- Department of Internal Medicine, St. Elizabeth Health Center, Youngstown, OH
| | - John Wiersch
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA
- Department of Surgery, School of Medicine, University of Texas Health Sciences Center at San Antonio, San Antonio, TX
| | - Ping Guo
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Carey Welsh
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Krishna Prasadan
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Jose Paredes
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Chiyo Shiota
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Xiangwei Xiao
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Yoko Wada
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Marilyn Diaz
- Laboratory of Molecular Genetics, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, NC
| | - George Gittes
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA
- Corresponding author: George Gittes,
| |
Collapse
|
26
|
Abstract
Islets form in the pancreas after the first endocrine cells have arisen as either single cells or small cell clusters in the epithelial cords. These cords constitute the developing pancreas in one of its earliest recognizable stages. Islet formation begins at the time the cords transform into a branching ductal system, continues while the ductal system expands, and finally stops before the exocrine tissue of ducts and acini reaches its final expansion. Thus, islets continuously arise from founder cells located in the branching and ramifying ducts. Islets arising from proximal duct cells locate between the exocrine lobules, develop strong autonomic and sensory innervations, and pass their blood to efferent veins (insulo-venous efferent system). Islets arising from cells of more distal ducts locate within the exocrine lobules, respond to nerve impulses ending at neighbouring blood vessels, and pass their blood to the surrounding acini (insulo-acinar portal system). Consequently, the section of the ductal system from which an islet arises determines to a large extent its future neighbouring tissue, architecture, properties, and functions. We note that islets interlobular in position are frequently found in rodents (rats and mice), whereas intralobularly-located, peripheral duct islets prevail in humans and cattle. Also, we expound on bovine foetal Laguesse islets as a prominent foetal type of type 1 interlobular neuro-insular complexes, similar to neuro-insular associations frequently found in rodents. Finally, we consider the probable physiological and pathophysiological implications of the different islet positions within and between species.
Collapse
|
27
|
Guo T, Landsman L, Li N, Hebrok M. Factors expressed by murine embryonic pancreatic mesenchyme enhance generation of insulin-producing cells from hESCs. Diabetes 2013; 62:1581-92. [PMID: 23305648 PMCID: PMC3636645 DOI: 10.2337/db12-0167] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Islet transplantation has proven to be a successful strategy to restore normoglycemia in patients with type 1 diabetes (T1D). However, the dearth of cadaveric islets available for transplantation hampers the widespread application of this treatment option. Although human embryonic stem cells and induced pluripotent stem cells are capable of generating insulin-producing cells in vitro when provided with the appropriate inductive cues, the insulin-expressing cells that develop behave more like immature β-cells with minimal sensitivity to glucose stimulation. Here, we identify a set of signaling factors expressed in mouse embryonic mesenchyme during the time when foregut and pancreatic progenitors are specified and test their activities during in vitro differentiation of human embryonic stem cells. Several of the identified factors work in concert to expand the pancreatic progenitor pool. Interestingly, transforming growth factor (TGF)-β ligands, most potent in inducing pancreatic progenitors, display strong inhibitory effects on subsequent endocrine cell differentiation. Treatment with TGF-β ligands, followed by the addition of a TGF-β receptor antagonist, dramatically increased the number of insulin-producing cells in vitro, demonstrating the need for dynamic temporal regulation of TGF-β signaling during in vitro differentiation. These studies illustrate the need to precisely mimic the in vivo conditions to fully recapitulate pancreatic lineage specification in vitro.
Collapse
|
28
|
Suzuki T, Dai P, Hatakeyama T, Harada Y, Tanaka H, Yoshimura N, Takamatsu T. TGF-β Signaling Regulates Pancreatic β-Cell Proliferation through Control of Cell Cycle Regulator p27 Expression. Acta Histochem Cytochem 2013; 46:51-8. [PMID: 23720603 PMCID: PMC3661777 DOI: 10.1267/ahc.12035] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 01/31/2013] [Indexed: 02/06/2023] Open
Abstract
Proliferation of pancreatic β-cells is an important mechanism underlying β-cell mass adaptation to metabolic demands. Increasing β-cell mass by regeneration may ameliorate or correct both type 1 and type 2 diabetes, which both result from inadequate production of insulin by β-cells of the pancreatic islet. Transforming growth factor β (TGF-β) signaling is essential for fetal development and growth of pancreatic islets. In this study, we exposed HIT-T15, a clonal pancreatic β-cell line, to TGF-β signaling. We found that inhibition of TGF-β signaling promotes proliferation of the cells significantly, while TGF-β signaling stimulation inhibits proliferation of the cells remarkably. We confirmed that this proliferative regulation by TGF-β signaling is due to the changed expression of the cell cycle regulator p27. Furthermore, we demonstrated that there is no observed effect on transcriptional activity of p27 by TGF-β signaling. Our data show that TGF-β signaling mediates the cell-cycle progression of pancreatic β-cells by regulating the nuclear localization of CDK inhibitor, p27. Inhibition of TGF-β signaling reduces the nuclear accumulation of p27, and as a result this inhibition promotes proliferation of β-cells.
Collapse
Affiliation(s)
- Tomoyuki Suzuki
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
- Department of Transplantation and Regenerative Surgery, Kyoto Prefectural University of Medicine
| | - Ping Dai
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
| | - Tomoya Hatakeyama
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
- Division of Digestive Surgery, Department of Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
| | - Hideo Tanaka
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
| | - Norio Yoshimura
- Department of Transplantation and Regenerative Surgery, Kyoto Prefectural University of Medicine
| | - Tetsuro Takamatsu
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine
| |
Collapse
|
29
|
El-Gohary Y, Tulachan S, Guo P, Welsh C, Wiersch J, Prasadan K, Paredes J, Shiota C, Xiao X, Wada Y, Diaz M, Gittes G. Smad signaling pathways regulate pancreatic endocrine development. Dev Biol 2013; 378:83-93. [PMID: 23603491 DOI: 10.1016/j.ydbio.2013.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 03/04/2013] [Accepted: 04/04/2013] [Indexed: 10/26/2022]
Abstract
Expansion of the pancreatic endocrine cell population occurs during both embryonic development and during post-natal pancreatic growth and regeneration. Mechanisms of the expansion of endocrine cells during embryonic development are not completely understood, and no clear mechanistic link has been established between growth of the embryonic endocrine pancreas and the islet cell replication that occurs in an adult animal. We found that transforming growth factor-beta (TGF-β) superfamily signaling, which has been implicated in many developmental processes, plays a key role in regulating pancreatic endocrine maturation and development. Specifically, the intracellular mediators of TGF-β signaling, smad2 and smad3, along with their inhibitor smad7, appear to mediate this process. Smad2, smad3 and smad7 were all broadly expressed throughout the early embryonic pancreatic epithelium. However, during later stages of development, smad2 and smad3 became strongly localized to the nuclei of the endocrine positive cells, whereas the inhibitory smad7 became absent in the endocrine component. Genetic inactivation of smad2 and smad3 led to a significant expansion of the embryonic endocrine compartment, whereas genetic inactivation of smad7 led to a significant decrease in the endocrine compartment. In vitro antisense studies further corroborated these results and supported the possibility that interplay between the inhibitory smad7 and the intracellular mediators smad2/3 is a control point for pancreatic endocrine development. These results should provide a better understanding of the key control mechanisms for β-cell development.
Collapse
Affiliation(s)
- Yousef El-Gohary
- Department of Surgery, Division of Pediatric Surgery, Children's Hospital of Pittsburgh, One Children's Hospital Drive, 4401 Penn Ave., Pittsburgh, PA 15224, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Xiao X, Wiersch J, El-Gohary Y, Guo P, Prasadan K, Paredes J, Welsh C, Shiota C, Gittes GK. TGFβ receptor signaling is essential for inflammation-induced but not β-cell workload-induced β-cell proliferation. Diabetes 2013; 62:1217-26. [PMID: 23248173 PMCID: PMC3609557 DOI: 10.2337/db12-1428] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Protection and restoration of a functional β-cell mass are fundamental strategies for prevention and treatment of diabetes. Consequently, knowledge of signals that determine the functional β-cell mass is of immense clinical relevance. Transforming growth factor β (TGFβ) superfamily signaling pathways play a critical role in development and tissue specification. Nevertheless, the role of these pathways in adult β-cell homeostasis is not well defined. Here, we ablated TGFβ receptor I and II genes in mice undergoing two surgical β-cell replication models (partial pancreatectomy or partial duct ligation), representing two triggers for β-cell proliferation, increased β-cell workload and local inflammation, respectively. Our data suggest that TGFβ receptor signaling is necessary for baseline β-cell proliferation. By either provision of excess glucose or treatment with exogenous insulin, we further demonstrated that inflammation and increased β-cell workload are both stimulants for β-cell proliferation but are TGFβ receptor signaling dependent and independent, respectively. Collectively, by using a pancreas-specific TGFβ receptor-deleted mouse model, we have identified two distinct pathways that regulate adult β-cell proliferation. Our study thus provides important information for understanding β-cell proliferation during normal growth and in pancreatic diseases.
Collapse
Affiliation(s)
- Xiangwei Xiao
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhu Z, Huangfu D. Human pluripotent stem cells: an emerging model in developmental biology. Development 2013; 140:705-17. [PMID: 23362344 DOI: 10.1242/dev.086165] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Developmental biology has long benefited from studies of classic model organisms. Recently, human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells, have emerged as a new model system that offers unique advantages for developmental studies. Here, we discuss how studies of hPSCs can complement classic approaches using model organisms, and how hPSCs can be used to recapitulate aspects of human embryonic development 'in a dish'. We also summarize some of the recently developed genetic tools that greatly facilitate the interrogation of gene function during hPSC differentiation. With the development of high-throughput screening technologies, hPSCs have the potential to revolutionize gene discovery in mammalian development.
Collapse
Affiliation(s)
- Zengrong Zhu
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA.
| | | |
Collapse
|
32
|
Signaling pathways regulating murine pancreatic development. Semin Cell Dev Biol 2012; 23:663-72. [DOI: 10.1016/j.semcdb.2012.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 06/13/2012] [Indexed: 12/24/2022]
|
33
|
Guo P, El-Gohary Y, Prasadan K, Shiota C, Xiao X, Wiersch J, Paredes J, Tulachan S, Gittes GK. Rapid and simplified purification of recombinant adeno-associated virus. J Virol Methods 2012; 183:139-46. [PMID: 22561982 DOI: 10.1016/j.jviromet.2012.04.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 04/09/2012] [Accepted: 04/18/2012] [Indexed: 12/12/2022]
Abstract
Preclinical gene therapy studies both in vitro and in vivo require high purity preparations of adeno-associated virus (AAV). Current methods for purification of AAV entail the use of centrifugation over either a CsCl or iodixanol gradient, or the use of chromatography. These methods can be cumbersome and expensive, necessitating ultrahigh speed gradient centrifugation or, for chromatography the use of other expensive equipment. In addition, these methods are time consuming, and the viral yield is not high. Currently no commercial purification kits are available for other than AAV serotype 2. A simplified method was used for the purification of AAV, with a viral yield that is able to be used effectively in adult and embryo mice. The method does not require ultrahigh speed gradient centrifugation nor chromatography. Instead, polyethylene glycol (PEG)/aqueous two-phase partitioning is used to remove soluble proteins from the PEG8000 precipitated virus-protein mixture. The procedure obtained rapidly up to 95% recovery of high quality purified AAV. The entire purification process, including HEK293 cell transfection, can be completed readily within one week, with purity seemingly higher than that obtained after one round of CsCl gradient purification.
Collapse
Affiliation(s)
- Ping Guo
- Department of Surgery, Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh, PA 15224, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Shin JA, Hong OK, Lee HJ, Jeon SY, Kim JW, Lee SH, Cho JH, Lee JM, Choi YH, Chang SA, Son HY, Kim JH, Yoon KH. Transforming growth factor-β induces epithelial to mesenchymal transition and suppresses the proliferation and transdifferentiation of cultured human pancreatic duct cells. J Cell Biochem 2011; 112:179-88. [PMID: 21069735 DOI: 10.1002/jcb.22929] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Pancreatic duct cells are considered a potential source of β-cell regeneration, and transforming growth factor-β (TGF-β) has been suggested to perform an important role in these processes, but the underlying mechanism of the signal pathways, especially in humans, remains poorly understood. To evaluate the role of TGF-β1, pancreatic duct cells were isolated from three brain-dead organ donors. Pancreatic cell clusters harvested after islet isolation were dispersed to single cells and cultured in monolayers, then treated with TGF-β1. We analyzed the characteristics of the cultured cells, the TGF-β1 intracellular signaling pathway, the proliferation, and transdifferentiation rates of the duct cells. We also evaluated the genes and protein expression patterns after TGF-β1 treatment. After TGF-β1 treatment, typical morphologic changes representative of EMT were observed and Erk1/2, JNK, and AKT phosphorylation, Ras downstream effectors, were increased. β cell-specific transcription factors including PDX-1, Beta2/NeuroD, Ist-1, and NGN3 were markedly suppressed and the rate of transdifferentiation into β cells was also suppressed. Genomic and proteomic analyses suggested that TGF-β1 induces marked changes in a variety of structural genes and proteins associated with EMT. In conclusion, TGF-β1 induces EMT in cultured human pancreatic duct cells, but suppresses its proliferation and transdifferentiation into β cells. Our results are the first report of TGF-β1 effects for EMT and ductal cell transdifferentiation and proliferation at the protein level in human pancreatic duct cells.
Collapse
Affiliation(s)
- Jeong-Ah Shin
- Department of Endocrinology & Metabolism, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Rezania A, Riedel MJ, Wideman RD, Karanu F, Ao Z, Warnock GL, Kieffer TJ. Production of functional glucagon-secreting α-cells from human embryonic stem cells. Diabetes 2011; 60:239-47. [PMID: 20971966 PMCID: PMC3012176 DOI: 10.2337/db10-0573] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Differentiation of human embryonic stem (hES) cells to fully developed cell types holds great therapeutic promise. Despite significant progress, the conversion of hES cells to stable, fully differentiated endocrine cells that exhibit physiologically regulated hormone secretion has not yet been achieved. Here we describe an efficient differentiation protocol for the in vitro conversion of hES cells to functional glucagon-producing α- cells. RESEARCH DESIGN AND METHODS Using a combination of small molecule screening and empirical testing, we developed a six-stage differentiation protocol for creating functional α-cells. An extensive in vitro and in vivo characterization of the differentiated cells was performed. RESULTS A high rate of synaptophysin expression (>75%) and robust expression of glucagon and the α-cell transcription factor ARX was achieved. After a transient polyhormonal state in which cells coexpress glucagon and insulin, maturation in vitro or in vivo resulted in depletion of insulin and other β-cell markers with concomitant enrichment of α-cell markers. After transplantation, these cells secreted fully processed, biologically active glucagon in response to physiologic stimuli including prolonged fasting and amino acid challenge. Moreover, glucagon release from transplanted cells was sufficient to reduce demand for pancreatic glucagon, resulting in a significant decrease in pancreatic α-cell mass. CONCLUSIONS These results indicate that fully differentiated pancreatic endocrine cells can be created via stepwise differentiation of hES cells. These cells may serve as a useful screening tool for the identification of compounds that modulate glucagon secretion as well as those that promote the transdifferentiation of α-cells to β-cells.
Collapse
Affiliation(s)
- Alireza Rezania
- BetaLogics Venture, Centocor Research and Development, Skillman, New Jersey
| | - Michael J. Riedel
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rhonda D. Wideman
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Francis Karanu
- BetaLogics Venture, Centocor Research and Development, Skillman, New Jersey
| | - Ziliang Ao
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Garth L. Warnock
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Timothy J. Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Corresponding author: Timothy J. Kieffer,
| |
Collapse
|
36
|
Limbert C, Ebert R, Schilling T, Path G, Benisch P, Klein-Hitpass L, Seufert J, Jakob F. Functional signature of human islet-derived precursor cells compared to bone marrow-derived mesenchymal stem cells. Stem Cells Dev 2010; 19:679-91. [PMID: 19895235 DOI: 10.1089/scd.2009.0241] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Pancreatic islet beta-cell replenishment can be driven by epithelial cells from exocrine pancreas via epithelial-mesenchymal transition (EMT) and the reverse process MET, while specified pancreatic mesenchymal cells control islet cell development and maintenance. The role of human islet-derived precursor cells (hIPCs) in regeneration and support of endocrine islets is under investigation. Here, we analyzed hIPCs as to their immunophenotype, multilineage differentiation capacity, and gene profiling, in comparison to human bone marrow-derived mesenchymal stem cells (hBM-MSCs). hIPCs and hBM-MSCs display a common mesenchymal character and express lineage-specific marker genes upon induction toward pancreatic endocrine and mesenchymal pathways of differentiation. hIPCs can go further along endocrine pathways while lacking some core mesenchymal differentiation attributes. Significance analysis of microarray (SAM) from 5 hBM-MSC and 3 hIPC donors mirrored such differences. Candidate gene cluster analysis disclosed differential expression of key lineage regulators, indicated a HoxA gene-associated positional memory in hIPCs and hBM-MSCs, and showed as well a clear transition state from mesenchyme to epithelium or vice versa in hIPCs. Our findings raise new research platforms to further clarify the potential of hIPCs to undergo complete MET thus contributing to islet cell replenishment, maintenance, and function.
Collapse
Affiliation(s)
- Catarina Limbert
- Orthopedic Center for Musculoskeletal Research, Stem Cell Division, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Diabetes is characterized by decreased function of insulin-producing beta cells and insufficient insulin output resulting from an absolute (Type 1) or relative (Type 2) inadequate functional beta cell mass. Both forms of the disease would greatly benefit from treatment strategies that could enhance beta cell regeneration and/or function. Successful and reliable methods of generating beta cells or whole islets from progenitor cells in vivo or in vitro could lead to restoration of beta cell mass in individuals with Type 1 diabetes and enhanced beta cell compensation in Type 2 patients. A thorough understanding of the normal developmental processes that occur during pancreatic organogenesis, for example, transcription factors, cell signaling molecules, and cell-cell interactions that regulate endocrine differentiation from the embryonic pancreatic epithelium, is required in order to successfully reach these goals. This review summarizes our current understanding of pancreas development, with particular emphasis on factors intrinsic or extrinsic to the pancreatic epithelium that are involved in regulating the development and differentiation of the various pancreatic cell types. We also discuss the recent progress in generating insulin-producing cells from progenitor sources.
Collapse
Affiliation(s)
- Michelle A Guney
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
38
|
Nichols J, Cooke A. Overcoming self-destruction in the pancreas. Curr Opin Biotechnol 2009; 20:511-5. [PMID: 19857955 DOI: 10.1016/j.copbio.2009.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 09/24/2009] [Accepted: 09/25/2009] [Indexed: 01/12/2023]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease where insulin producing pancreatic beta cells are progressively destroyed. In the absence of a cure, exogenous insulin is given to maintain glucose homeostasis. Tolerogenic strategies to halt destruction and facilitate recovery of beta cells are being explored. This disease is under polygenic control; the identification of specific candidate pathways to target for drug discovery or corrective therapy would enhance therapeutic options. Whilst islet transplantation combined with immune suppression has shown some efficacy, the availability of beta cells restricts its general use. Differentiation protocols directing cultured stem cells into pancreatic tissue are under development. The availability of embryonic stem cells from a mouse model of T1D should facilitate dissection of the causes and progression of this disease.
Collapse
Affiliation(s)
- Jennifer Nichols
- Wellcome Trust Centre for Stem Cell Research, Tennis Court Rd, Cambridge CB2 1QR, United Kingdom.
| | | |
Collapse
|
39
|
Kutlu B, Kayali AG, Jung S, Parnaud G, Baxter D, Glusman G, Goodman N, Behie LA, Hayek A, Hood L. Meta-analysis of gene expression in human pancreatic islets after in vitro expansion. Physiol Genomics 2009; 39:72-81. [PMID: 19622797 DOI: 10.1152/physiolgenomics.00063.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pancreatic islet transplantation as a potential cure for type 1 diabetes (T1D) cannot be scaled up due to a scarcity of human pancreas donors. In vitro expansion of beta-cells from mature human pancreatic islets provides an alternative source of insulin-producing cells. The exact nature of the expanded cells produced by diverse expansion protocols and their potential for differentiation into functional beta-cells remain elusive. We performed a large-scale meta-analysis of gene expression in human pancreatic islet cells, which were processed using three different previously described protocols for expansion and for which redifferentiation was attempted. All three expansion protocols induced dramatic changes in the expression profiles of pancreatic islets; many of these changes are shared among the three protocols. Attempts at redifferentiation of expanded cells induce a limited number of gene expression changes. Nevertheless, these fail to restore a pancreatic islet-like gene expression pattern. Comparison with a collection of public microarray datasets confirmed that expanded cells are highly comparable to mesenchymal stem cells. Genes induced in expanded cells are also enriched for targets of transcription factors important for pluripotency induction. The present data increase our understanding of the active pathways in expanded and redifferentiated islets. Knowledge of the mesenchymal stem cell potential may help development of drug therapeutics to restore beta-cell mass in T1D patients.
Collapse
Affiliation(s)
- B Kutlu
- Institute for Systems Biology, Seattle, Washington, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lin HM, Lee JH, Yadav H, Kamaraju AK, Liu E, Zhigang D, Vieira A, Kim SJ, Collins H, Matschinsky F, Harlan DM, Roberts AB, Rane SG. Transforming growth factor-beta/Smad3 signaling regulates insulin gene transcription and pancreatic islet beta-cell function. J Biol Chem 2009; 284:12246-57. [PMID: 19265200 DOI: 10.1074/jbc.m805379200] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic islet beta-cell dysfunction is a signature feature of Type 2 diabetes pathogenesis. Consequently, knowledge of signals that regulate beta-cell function is of immense clinical relevance. Transforming growth factor (TGF)-beta signaling plays a critical role in pancreatic development although the role of this pathway in the adult pancreas is obscure. Here, we define an important role of the TGF-beta pathway in regulation of insulin gene transcription and beta-cell function. We identify insulin as a TGF-beta target gene and show that the TGF-beta signaling effector Smad3 occupies the insulin gene promoter and represses insulin gene transcription. In contrast, Smad3 small interfering RNAs relieve insulin transcriptional repression and enhance insulin levels. Transduction of adenoviral Smad3 into primary human and non-human primate islets suppresses insulin content, whereas, dominant-negative Smad3 enhances insulin levels. Consistent with this, Smad3-deficient mice exhibit moderate hyperinsulinemia and mild hypoglycemia. Moreover, Smad3 deficiency results in improved glucose tolerance and enhanced glucose-stimulated insulin secretion in vivo. In ex vivo perifusion assays, Smad3-deficient islets exhibit improved glucose-stimulated insulin release. Interestingly, Smad3-deficient islets harbor an activated insulin-receptor signaling pathway and TGF-beta signaling regulates expression of genes involved in beta-cell function. Together, these studies emphasize TGF-beta/Smad3 signaling as an important regulator of insulin gene transcription and beta-cell function and suggest that components of the TGF-beta signaling pathway may be dysregulated in diabetes.
Collapse
Affiliation(s)
- Huei-Min Lin
- Diabetes Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Gittes GK. Developmental biology of the pancreas: a comprehensive review. Dev Biol 2008; 326:4-35. [PMID: 19013144 DOI: 10.1016/j.ydbio.2008.10.024] [Citation(s) in RCA: 315] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2008] [Revised: 10/09/2008] [Accepted: 10/13/2008] [Indexed: 02/06/2023]
Abstract
Pancreatic development represents a fascinating process in which two morphologically distinct tissue types must derive from one simple epithelium. These two tissue types, exocrine (including acinar cells, centro-acinar cells, and ducts) and endocrine cells serve disparate functions, and have entirely different morphology. In addition, the endocrine tissue must become disconnected from the epithelial lining during its development. The pancreatic development field has exploded in recent years, and numerous published reviews have dealt specifically with only recent findings, or specifically with certain aspects of pancreatic development. Here I wish to present a more comprehensive review of all aspects of pancreatic development, though still there is not a room for discussion of stem cell differentiation to pancreas, nor for discussion of post-natal regeneration phenomena, two important fields closely related to pancreatic development.
Collapse
Affiliation(s)
- George K Gittes
- Children's Hospital of Pittsburgh and the University of Pittsburgh School of Medicine, Department of Pediatric Surgery, 3705 Fifth Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|