1
|
Andersson-Rolf A, Groot K, Korving J, Begthel H, Hanegraaf MAJ, VanInsberghe M, Salmén F, van den Brink S, Lopez-Iglesias C, Peters PJ, Krueger D, Beumer J, Geurts MH, Alemany A, Gehart H, Carlotti F, de Koning EJP, Chuva de Sousa Lopes SM, van Oudenaarden A, van Es JH, Clevers H. Long-term in vitro expansion of a human fetal pancreas stem cell that generates all three pancreatic cell lineages. Cell 2024; 187:7394-7413.e22. [PMID: 39626658 DOI: 10.1016/j.cell.2024.10.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/15/2024] [Revised: 09/18/2024] [Accepted: 10/18/2024] [Indexed: 12/29/2024]
Abstract
The mammalian pancreas consists of three epithelial compartments: the acini and ducts of the exocrine pancreas and the endocrine islets of Langerhans. Murine studies indicate that these three compartments derive from a transient, common pancreatic progenitor. Here, we report derivation of 18 human fetal pancreas organoid (hfPO) lines from gestational weeks 8-17 (8-17 GWs) fetal pancreas samples. Four of these lines, derived from 15 to 16 GWs samples, generate acinar-, ductal-, and endocrine-lineage cells while expanding exponentially for >2 years under optimized culture conditions. Single-cell RNA sequencing identifies rare LGR5+ cells in fetal pancreas and in hfPOs as the root of the developmental hierarchy. These LGR5+ cells share multiple markers with adult gastrointestinal tract stem cells. Organoids derived from single LGR5+ organoid-derived cells recapitulate this tripotency in vitro. We describe a human fetal tripotent stem/progenitor cell capable of long-term expansion in vitro and of generating all three pancreatic cell lineages.
Collapse
Affiliation(s)
- Amanda Andersson-Rolf
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands.
| | - Kelvin Groot
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Jeroen Korving
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Harry Begthel
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Maaike A J Hanegraaf
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Michael VanInsberghe
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Fredrik Salmén
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Stieneke van den Brink
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Carmen Lopez-Iglesias
- The Maastricht Multimodal Molecular Imaging Institute, 6229 ER Maastricht, the Netherlands
| | - Peter J Peters
- The Maastricht Multimodal Molecular Imaging Institute, 6229 ER Maastricht, the Netherlands
| | - Daniel Krueger
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Joep Beumer
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche innovation Centre, 4070 Basel, Switzerland
| | - Maarten H Geurts
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Princess Maxima Centre for Pediatric Oncology, 3584 CS Utrecht, the Netherlands
| | - Anna Alemany
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Helmuth Gehart
- ETH Zurich, Institute of Molecular Health Sciences, 8093 Zürich, Schweiz
| | - Françoise Carlotti
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Eelco J P de Koning
- Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | | | - Alexander van Oudenaarden
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Johan H van Es
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Hans Clevers
- Hubrecht Institute, Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), 3584 CT Utrecht, the Netherlands; University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Princess Maxima Centre for Pediatric Oncology, 3584 CS Utrecht, the Netherlands; Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche innovation Centre, 4070 Basel, Switzerland.
| |
Collapse
|
2
|
Wang Y, Liu Z, Li S, Su X, Lai KP, Li R. Biochemical pancreatic β-cell lineage reprogramming: Various cell fate shifts. Curr Res Transl Med 2024; 72:103412. [PMID: 38246021 DOI: 10.1016/j.retram.2023.103412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/22/2022] [Revised: 07/12/2023] [Accepted: 09/19/2023] [Indexed: 01/23/2024]
Abstract
The incidence of pancreatic diseases has been continuously rising in recent years. Thus, research on pancreatic regeneration is becoming more popular. Chronic hyperglycemia is detrimental to pancreatic β-cells, leading to impairment of insulin secretion which is the main hallmark of pancreatic diseases. Obtaining plenty of functional pancreatic β-cells is the most crucial aspect when studying pancreatic biology and treating diabetes. According to the International Diabetes Federation, diabetes has become a global epidemic, with about 3 million people suffering from diabetes worldwide. Hyperglycemia can lead to many dangerous diseases, including amputation, blindness, neuropathy, stroke, and cardiovascular and kidney diseases. Insulin is widely used in the treatment of diabetes; however, innovative approaches are needed in the academic and preclinical stages. A new approach aims at synthesizing patient-specific functional pancreatic β-cells. The present article focuses on how cells from different tissues can be transformed into pancreatic β-cells.
Collapse
Affiliation(s)
- Yuqin Wang
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, 1 Zhiyuan Road, Lingui District, Guilin 541199, China
| | - Zhuoqing Liu
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Shengren Li
- Lingui Clinical College of Guilin Medical University, Guilin, China
| | - Xuejuan Su
- Lingui Clinical College of Guilin Medical University, Guilin, China
| | - Keng Po Lai
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, 1 Zhiyuan Road, Lingui District, Guilin 541199, China
| | - Rong Li
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, 1 Zhiyuan Road, Lingui District, Guilin 541199, China.
| |
Collapse
|
3
|
Yi X, Xie Y, Gerber DA. Pancreas patch grafting to treat type 1 diabetes. Biochem Biophys Res Commun 2023; 686:149200. [PMID: 37926045 DOI: 10.1016/j.bbrc.2023.149200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Stem/progenitor cell therapy is a promising treatment option for patients with type 1 diabetes (T1D) a disease characterized by autoimmune destruction of pancreatic β cells. Actively injecting cells into an organ is one option for cell delivery, but in the pancreas, this contributes to acute inflammation and pancreatitis. We employed a patch grafting approach to transplant biliary tree stem cells/progenitor cells (BTSC) onto the surface of the pancreas in diabetic mice. The cells engraft and differentiate into β-like cells reversing hyperglycemia during a four-month period of observation. In addition, C-peptide and insulin gradually increase in blood circulation without detectable adverse effects during this period. Moreover, the patch graft transplant promoted the proliferation and differentiation of pancreatic β-like cells with co-expression of the β cell biomarker. CONCLUSION: BTSC transplantation can effectively attenuate T1D over a four-month period that is vital important for clinical applications.
Collapse
Affiliation(s)
- Xianwen Yi
- Department of Surgery, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Youmei Xie
- Department of Surgery, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - David A Gerber
- Department of Surgery, University of North Carolina, Chapel Hill, NC, 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
4
|
Joglekar MV, Sahu S, Wong WKM, Satoor SN, Dong CX, Farr RJ, Williams MD, Pandya P, Jhala G, Yang SNY, Chew YV, Hetherington N, Thiruchevlam D, Mitnala S, Rao GV, Reddy DN, Loudovaris T, Hawthorne WJ, Elefanty AG, Joglekar VM, Stanley EG, Martin D, Thomas HE, Tosh D, Dalgaard LT, Hardikar AA. A Pro-Endocrine Pancreatic Islet Transcriptional Program Established During Development Is Retained in Human Gallbladder Epithelial Cells. Cell Mol Gastroenterol Hepatol 2022; 13:1530-1553.e4. [PMID: 35032693 PMCID: PMC9043310 DOI: 10.1016/j.jcmgh.2022.01.008] [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] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/25/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/10/2022]
Abstract
BACKGROUND & AIMS Pancreatic islet β-cells are factories for insulin production; however, ectopic expression of insulin also is well recognized. The gallbladder is a next-door neighbor to the developing pancreas. Here, we wanted to understand if gallbladders contain functional insulin-producing cells. METHODS We compared developing and adult mouse as well as human gallbladder epithelial cells and islets using immunohistochemistry, flow cytometry, enzyme-linked immunosorbent assays, RNA sequencing, real-time polymerase chain reaction, chromatin immunoprecipitation, and functional studies. RESULTS We show that the epithelial lining of developing, as well as adult, mouse and human gallbladders naturally contain interspersed cells that retain the capacity to actively transcribe, translate, package, and release insulin. We show that human gallbladders also contain functional insulin-secreting cells with the potential to naturally respond to glucose in vitro and in situ. Notably, in a non-obese diabetic (NOD) mouse model of type 1 diabetes, we observed that insulin-producing cells in the gallbladder are not targeted by autoimmune cells. Interestingly, in human gallbladders, insulin splice variants are absent, although insulin splice forms are observed in human islets. CONCLUSIONS In summary, our biochemical, transcriptomic, and functional data in mouse and human gallbladder epithelial cells collectively show the evolutionary and developmental similarities between gallbladder and the pancreas that allow gallbladder epithelial cells to continue insulin production in adult life. Understanding the mechanisms regulating insulin transcription and translation in gallbladder epithelial cells would help guide future studies in type 1 diabetes therapy.
Collapse
Affiliation(s)
- Mugdha V Joglekar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Subhshri Sahu
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Wilson K M Wong
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Sarang N Satoor
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Charlotte X Dong
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Ryan J Farr
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Michael D Williams
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Prapti Pandya
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Gaurang Jhala
- Immunology and Diabetes Group, St. Vincent's Institute for Medical Research, Victoria, Australia
| | - Sundy N Y Yang
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Yi Vee Chew
- The Westmead Institute for Medical Research, Westmead Millenium Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Nicola Hetherington
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Dhan Thiruchevlam
- Department of Gastroenterology, St. Vincent's Hospital, Melbourne, Victoria, Australia
| | - Sasikala Mitnala
- Surgical Gastroenterology Research, Asian Institute of Gastroenterology, Hyderabad, India
| | - Guduru V Rao
- Surgical Gastroenterology Research, Asian Institute of Gastroenterology, Hyderabad, India
| | | | - Thomas Loudovaris
- Immunology and Diabetes Group, St. Vincent's Institute for Medical Research, Victoria, Australia
| | - Wayne J Hawthorne
- The Westmead Institute for Medical Research, Westmead Millenium Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Andrew G Elefanty
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | | | - Edouard G Stanley
- Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - David Martin
- Upper Gastrointestinal Surgery, Strathfield Hospital, Strathfield, New South Wales, Australia
| | - Helen E Thomas
- Immunology and Diabetes Group, St. Vincent's Institute for Medical Research, Victoria, Australia
| | - David Tosh
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Louise T Dalgaard
- Section of Eukaryotic Cell Biology, Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia.
| |
Collapse
|
5
|
Wang W, Zhang C. Targeting β-cell dedifferentiation and transdifferentiation: opportunities and challenges. Endocr Connect 2021; 10:R213-R228. [PMID: 34289444 PMCID: PMC8428079 DOI: 10.1530/ec-21-0260] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/21/2021] [Accepted: 07/21/2021] [Indexed: 12/02/2022]
Abstract
The most distinctive pathological characteristics of diabetes mellitus induced by various stressors or immune-mediated injuries are reductions of pancreatic islet β-cell populations and activity. Existing treatment strategies cannot slow disease progression; consequently, research to genetically engineer β-cell mimetics through bi-directional plasticity is ongoing. The current consensus implicates β-cell dedifferentiation as the primary etiology of reduced β-cell mass and activity. This review aims to summarize the etiology and proposed mechanisms of β-cell dedifferentiation and to explore the possibility that there might be a time interval from the onset of β-cell dysfunction caused by dedifferentiation to the development of diabetes, which may offer a therapeutic window to reduce β-cell injury and to stabilize functionality. In addition, to investigate β-cell plasticity, we review strategies for β-cell regeneration utilizing genetic programming, small molecules, cytokines, and bioengineering to transdifferentiate other cell types into β-cells; the development of biomimetic acellular constructs to generate fully functional β-cell-mimetics. However, the maturation of regenerated β-cells is currently limited. Further studies are needed to develop simple and efficient reprogramming methods for assembling perfectly functional β-cells. Future investigations are necessary to transform diabetes into a potentially curable disease.
Collapse
Affiliation(s)
- Wenrui Wang
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Chuan Zhang
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, People’s Republic of China
- Correspondence should be addressed to C Zhang:
| |
Collapse
|
6
|
Abstract
The pancreas is a complex organ with exocrine and endocrine components. Many pathologies impair exocrine function, including chronic pancreatitis, cystic fibrosis and pancreatic ductal adenocarcinoma. Conversely, when the endocrine pancreas fails to secrete sufficient insulin, patients develop diabetes mellitus. Pathology in either the endocrine or exocrine pancreas results in devastating economic and personal consequences. The current standard therapy for treating patients with type 1 diabetes mellitus is daily exogenous insulin injections, but cell sources of insulin provide superior glycaemic regulation and research is now focused on the goal of regenerating or replacing β cells. Stem-cell-based models might be useful to study exocrine pancreatic disorders, and mesenchymal stem cells or secreted factors might delay disease progression. Although the standards that bioengineered cells must meet before being considered as a viable therapy are not yet established, any potential therapy must be acceptably safe and functionally superior to current therapies. Here, we describe progress and challenges in cell-based methods to restore pancreatic function, with a focus on optimizing the site for cell delivery and decreasing requirements for immunosuppression through encapsulation. We also discuss the tools and strategies being used to generate exocrine pancreas and insulin-producing β-cell surrogates in situ and highlight obstacles to clinical application.
Collapse
|
7
|
Galivo F, Benedetti E, Wang Y, Pelz C, Schug J, Kaestner KH, Grompe M. Reprogramming human gallbladder cells into insulin-producing β-like cells. PLoS One 2017; 12:e0181812. [PMID: 28813430 PMCID: PMC5558938 DOI: 10.1371/journal.pone.0181812] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2017] [Accepted: 06/12/2017] [Indexed: 12/20/2022] Open
Abstract
The gallbladder and cystic duct (GBCs) are parts of the extrahepatic biliary tree and share a common developmental origin with the ventral pancreas. Here, we report on the very first genetic reprogramming of patient-derived human GBCs to β-like cells for potential autologous cell replacement therapy for type 1 diabetes. We developed a robust method for large-scale expansion of human GBCs ex vivo. GBCs were reprogrammed into insulin-producing pancreatic β-like cells by a combined adenoviral-mediated expression of hallmark pancreatic endocrine transcription factors PDX1, MAFA, NEUROG3, and PAX6 and differentiation culture in vitro. The reprogrammed GBCs (rGBCs) strongly induced the production of insulin and pancreatic endocrine genes and these responded to glucose stimulation in vitro. rGBCs also expressed an islet-specific surface marker, which was used to enrich for the most highly reprogrammed cells. More importantly, global mRNA and microRNA expression profiles and protein immunostaining indicated that rGBCs adopted an overall β-like state and these rGBCs engrafted in immunodeficient mice. Furthermore, comparative global expression analyses identified putative regulators of human biliary to β cell fate conversion. In summary, we have developed, for the first time, a reliable and robust genetic reprogramming and culture expansion of primary human GBCs—derived from multiple unrelated donors—into pancreatic β-like cells ex vivo, thus showing that human gallbladder is a potentially rich source of reprogrammable cells for autologous cell therapy in diabetes.
Collapse
Affiliation(s)
- Feorillo Galivo
- Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail: (FG); (MG)
| | - Eric Benedetti
- Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Yuhan Wang
- Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Carl Pelz
- Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jonathan Schug
- Department of Genetics, School of Medicine and Institute of Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Klaus H. Kaestner
- Department of Genetics, School of Medicine and Institute of Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Markus Grompe
- Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail: (FG); (MG)
| |
Collapse
|
8
|
Wei R, Hong T. Lineage Reprogramming: A Promising Road for Pancreatic β Cell Regeneration. Trends Endocrinol Metab 2016; 27:163-176. [PMID: 26811208 DOI: 10.1016/j.tem.2016.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/11/2015] [Revised: 12/24/2015] [Accepted: 01/06/2016] [Indexed: 12/18/2022]
Abstract
Cell replacement therapy is a promising method to restore pancreatic β cell function and cure diabetes. Distantly related cells (fibroblasts, keratinocytes, and muscle cells) and developmentally related cells (hepatocytes, gastrointestinal, and pancreatic exocrine cells) have been successfully reprogrammed into β cells in vitro and in vivo. However, while some reprogrammed β cells bear similarities to bona fide β cells, others do not develop into fully functional β cells. Here we review various strategies currently used for β cell reprogramming, including ectopic expression of specific transcription factors associated with islet development, repression of maintenance factors of host cells, regulation of epigenetic modifications, and microenvironmental changes. Development of simple and efficient reprogramming methods is a key priority for developing fully functional β cells suitable for cell replacement therapy.
Collapse
Affiliation(s)
- Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing 100191, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
9
|
Novel surface markers directed against adult human gallbladder. Stem Cell Res 2015; 15:172-81. [PMID: 26079872 DOI: 10.1016/j.scr.2015.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/26/2015] [Revised: 05/19/2015] [Accepted: 06/08/2015] [Indexed: 12/28/2022] Open
Abstract
Novel cell surface-reactive monoclonal antibodies generated against extrahepatic biliary cells were developed for the isolation and characterization of different cell subsets from normal adult human gallbladder. Eleven antigenically distinct gallbladder subpopulations were isolated by fluorescence-activated cell sorting. They were classified into epithelial, mesenchymal, and pancreatobiliary (PDX1(+)SOX9(+)) subsets based on gene expression profiling. These antigenically distinct human gallbladder cell subsets could potentially also reflect different functional properties in regards to bile physiology, cell renewal and plasticity. Three of the novel monoclonal antibodies differentially labeled archival sections of primary carcinoma of human gallbladder relative to normal tissue. The novel monoclonal antibodies described herein enable the identification and characterization of antigenically diverse cell subsets within adult human gallbladder and are putative tumor biomarkers.
Collapse
|
10
|
Hickey RD, Galivo F, Schug J, Brehm MA, Haft A, Wang Y, Benedetti E, Gu G, Magnuson MA, Shultz LD, Lagasse E, Greiner DL, Kaestner KH, Grompe M. Generation of islet-like cells from mouse gall bladder by direct ex vivo reprogramming. Stem Cell Res 2013; 11:503-15. [PMID: 23562832 DOI: 10.1016/j.scr.2013.02.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/27/2012] [Revised: 02/01/2013] [Accepted: 02/09/2013] [Indexed: 01/19/2023] Open
Abstract
Cell replacement is an emerging therapy for type 1 diabetes. Pluripotent stem cells have received a lot of attention as a potential source of transplantable β-cells, but their ability to form teratomas poses significant risks. Here, we evaluated the potential of primary mouse gall bladder epithelial cells (GBCs) as targets for ex vivo genetic reprogramming to the β-cell fate. Conditions for robust expansion and genetic transduction of primary GBCs by adenoviral vectors were developed. Using a GFP reporter for insulin, conditions for reprogramming were then optimized. Global expression analysis by RNA-sequencing was used to quantitatively compare reprogrammed GBCs (rGBCs) to true β-cells, revealing both similarities and differences. Adenoviral-mediated expression of NEUROG3, Pdx1, and MafA in GBCs resulted in robust induction of pancreatic endocrine genes, including Ins1, Ins2, Neurod1, Nkx2-2 and Isl1. Furthermore, expression of GBC-specific genes was repressed, including Sox17 and Hes1. Reprogramming was also enhanced by addition of retinoic acid and inhibition of Notch signaling. Importantly, rGBCs were able to engraft long term in vivo and remained insulin-positive for 15weeks. We conclude that GBCs are a viable source for autologous cell replacement in diabetes, but that complete reprogramming will require further manipulations.
Collapse
Affiliation(s)
- Raymond D Hickey
- Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR 97203, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Icariin potentiates the antitumor activity of gemcitabine in gallbladder cancer by suppressing NF-κB. Acta Pharmacol Sin 2013; 34:301-8. [PMID: 23274410 DOI: 10.1038/aps.2012.162] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/29/2022] Open
Abstract
AIM Gemcitabine has been increasingly prescribed for the treatment of gallbladder cancer. However, the response rate is low. The aim of this study is to determine whether icariin, a flavonoid isolated from Epimedi herba, could potentiate the antitumor activity of gemcitabine in gallbladder cancer. METHODS Human gallbladder carcinoma cell lines GBC-SD and SGC-996 were tested. Cell proliferation and apoptosis were analyzed using MTT assay and flow cytometry, respectively. The expression of apoptosis- and proliferation-related molecules was detected with Western blotting. Caspase-3 activity was analyzed using colorimetric assay, and NF-κB activity was measured with ELISA. A gallbladder cancer xenograft model was established in female BALB/c (nu/nu) mice. The mice were intraperitoneally administered gemcitabine (125 mg/kg) in combination with icariin (40 mg/kg) for 2 weeks. RESULTS Icariin (40-160 μg/mL) dose-dependently suppressed cell proliferation and induced apoptosis in both GBC-SD and SGC-996 cells, with SGC-996 cells being less sensitive to the drug. Icariin (40 μg/mL) significantly enhanced the antitumor activity of gemcitabine (0.5 μmol/L) in both GBC-SD and SGC-996 cells. The mice bearing gallbladder cancer xenograft treated with gemcitabine in combination with icariin exhibited significantly smaller tumor size than the mice treated with either drug alone. In GBC-SD cells, icariin significantly inhibited both the constitutive and gemcitabine-induced NF-κB activity, enhanced caspase-3 activity, induced G(0)-G(1) phase arrest, and suppressed the expression of Bcl-2, Bcl-xL and surviving proteins. CONCLUSION Icariin, by suppressing NF-κB activity, exerts antitumor activity, and potentiates the antitumor activity of gemcitabine in gallbladder cancer. Combined administration of gemcitabine and icariin may offer a better therapeutic option for the patients with gallbladder cancer.
Collapse
|
12
|
Gandou C, Harada K, Sato Y, Igarashi S, Sasaki M, Ikeda H, Nakanuma Y. Hilar cholangiocarcinoma and pancreatic ductal adenocarcinoma share similar histopathologies, immunophenotypes, and development-related molecules. Hum Pathol 2012; 44:811-21. [PMID: 23134772 DOI: 10.1016/j.humpath.2012.08.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/02/2012] [Revised: 08/07/2012] [Accepted: 08/08/2012] [Indexed: 12/15/2022]
Abstract
Embryologically, intrahepatic small bile ducts arise from hepatic progenitor cells via ductal plates, whereas the pancreato-extrahepatic biliary progenitor cells expressing the transcription factors PDX1 and HES1 are reportedly involved in the development of the extrahepatic biliary tract and ventral pancreas. The expression of cellular markers characteristic of the different anatomical levels of the biliary tree and pancreas, as well as PDX1 and HES1, was examined in cholangiocarcinoma components of combined hepatocellular cholangiocarcinoma (12 cases), intrahepatic cholangiocarcinoma (21 cases), hilar cholangiocarcinoma (25 cases), and pancreatic ductal adenocarcinoma (18 cases). Anterior gradient protein-2 and S100P were frequently expressed in hilar cholangiocarcinoma and pancreatic ductal adenocarcinoma, whereas neural cell adhesion molecule and luminal expression of epithelial membrane antigen were common in cholangiocarcinoma components of combined hepatocellular cholangiocarcinoma. PDX1 and HES1 were frequently and markedly expressed in pancreatic ductal adenocarcinoma and, to a lesser degree, in hilar cholangiocarcinoma, although their expression was rare and mild in cholangiocarcinoma components in combined hepatocellular cholangiocarcinoma. The expression patterns of these molecules in intrahepatic cholangiocarcinoma were intermediate between those in hilar cholangiocarcinoma and cholangiocarcinoma components of combined hepatocellular cholangiocarcinoma. Pancreatic ductal adenocarcinoma and hilar cholangiocarcinoma had a similar expression of mucin, immunophenotypes, as well as transcription factors. Pancreatic ductal adenocarcinoma and hilar cholangiocarcinoma showed similar postoperative prognosis. In conclusion, the similar expression of phenotypes related to pancreatobiliary anatomy and embryology may in part explain why these 2 types of carcinoma present similar clinicopathologic features. Further studies on the carcinogenesis of these carcinomas based on their similarities are warranted.
Collapse
Affiliation(s)
- Chihiro Gandou
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa 920-8640, Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Zhao HJ, Wei R, Hong TP. Progress in understanding the relationship of microRNAs with pancreas development and stem cell differentiation. Shijie Huaren Xiaohua Zazhi 2012; 20:1913-1918. [DOI: 10.11569/wcjd.v20.i21.1913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs), a class of non-coding small RNAs involved in post-transcriptional gene regulation, play important roles in embryonic development, cell fate determination, and growth regulation. It has been shown that miRNAs contribute to the embryonic pancreas development by regulating several important transcriptional factors. Stem cells can be induced to differentiate into insulin-producing cells in vitro by mimicking the development of pancreatic β cells in vivo. MiRNAs may have regulatory roles in the maintenance and differentiation of stem cells. Therefore, elucidation of the mechanisms by which miRNAs regulate pancreas development and stem cell differentiation will provide novel insights into the development of therapeutic approach for diabetes.
Collapse
|