101
|
Chitosan-assisted differentiation of porcine adipose tissue-derived stem cells into glucose-responsive insulin-secreting clusters. PLoS One 2017; 12:e0172922. [PMID: 28253305 PMCID: PMC5333835 DOI: 10.1371/journal.pone.0172922] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 02/11/2017] [Indexed: 12/25/2022] Open
Abstract
The unique advantage of easy access and abundance make the adipose-derived stem cells (ADSCs) a promising system of multipotent cells for transplantation and regenerative medicine. Among the available sources, porcine ADSCs (pADSCs) deserve especial attention due to the close resemblance of human and porcine physiology, as well as for the upcoming availability of humanized porcine models. Here, we report on the isolation and conversion of pADSCs into glucose-responsive insulin-secreting cells. We used the stromal-vascular fraction of the dorsal subcutaneous adipose from 9-day-old male piglets to isolate pADSCs, and subjected the cells to an induction scheme for differentiation on chitosan-coated plates. This one-step procedure promoted differentiation of pADSCs into pancreatic islet-like clusters (PILC) that are characterized by the expression of a repertoire of pancreatic proteins, including pancreatic and duodenal homeobox (Pdx-1), insulin gene enhancer protein (ISL-1) and insulin. Upon glucose challenge, these PILC secreted high amounts of insulin in a dose-dependent manner. Our data also suggest that chitosan plays roles not only to enhance the differentiation potential of pADSCs, but also to increase the glucose responsiveness of PILCs. Our novel approach is, therefore, of great potential for transplantation-based amelioration of type 1 diabetes.
Collapse
|
102
|
N. Randolph L, Jiang Y, Lian X. Stem Cell Engineering and Differentiation for Disease Modeling and Cell-based Therapies. ACTA ACUST UNITED AC 2017. [DOI: 10.3934/celltissue.2017.2.140] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
103
|
Penaforte-Saboia JG, Montenegro RM, Couri CE, Batista LA, Montenegro APDR, Fernandes VO, Akhtar H, Negrato CA, Malmegrim KCR, Moraes DA, Dias JBE, Simões BP, Gomes MB, Oliveira MC. Microvascular Complications in Type 1 Diabetes: A Comparative Analysis of Patients Treated with Autologous Nonmyeloablative Hematopoietic Stem-Cell Transplantation and Conventional Medical Therapy. Front Endocrinol (Lausanne) 2017; 8:331. [PMID: 29218029 PMCID: PMC5703738 DOI: 10.3389/fendo.2017.00331] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/08/2017] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To explore the impact on microvascular complications, long-term preservation of residual B-cell function and glycemic control of patients with type 1 diabetes treated with autologous nonmyeloablative hematopoietic stem-cell transplantation (AHST) compared with conventional medical therapy (CT). RESEARCH DESIGN AND METHODS Cross-sectional data of patients treated with AHST were compared with patients who received conventional therapy from the Brazilian Type 1 Diabetes Study Group, the largest multicenter observational study in type 1 diabetes mellitus in Brazil. Both groups of patients had diabetes for 8 years on average. An assessment comparison was made on the presence of microvascular complications, residual function of B cell, A1c, and insulin dose of the patients. RESULTS After a median of 8 years of diagnosis, none of the AHST-treated patients (n = 24) developed microvascular complications, while 21.5% (31/144) had at least one (p < 0.005) complication in the CT group (n = 144). Furthermore, no case of nephropathy was reported in the AHST group, while 13.8% of CT group (p < 0.005) developed nephropathy during the same period. With regard of residual B-cell function, the percentage of individuals with predicted higher C-peptide levels (IDAA1C ≤ 9) was about 10-fold higher in the AHST group compared with CT (75 vs. 8.3%) (p < 0.001) group. Among AHST patients, 54.1% (13/24) had the HbA1c < 7.0 compared with 13.1% in the CT (p < 0.001) group. CONCLUSION Patients with newly diagnosed type 1 diabetes treated with AHST presented lower prevalence of microvascular complications, higher residual B-cell function, and better glycemic control compared with the CT group.
Collapse
Affiliation(s)
| | - Renan M. Montenegro
- Post Graduate Program in Medical Sciences, Federal University of Ceará, Ceará, Brazil
- Federal University of Ceará, Ceará, Brazil
- *Correspondence: Renan M. Montenegro Jr.,
| | - Carlos E. Couri
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Livia A. Batista
- Post Graduate Program in Medical Sciences, Federal University of Ceará, Ceará, Brazil
| | | | | | - Hussain Akhtar
- Federal University of Ceará, Ceará, Brazil
- University of Oslo, UIO, Oslo, Noruega
| | | | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela Aparecida Moraes
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Juliana B. E. Dias
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Belinda P. Simões
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Maria Carolina Oliveira
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
104
|
Jeong J, Kim KN, Chung MS, Kim HJ. Functional comparison of human embryonic stem cells and induced pluripotent stem cells as sources of hepatocyte-like cells. Tissue Eng Regen Med 2016; 13:740-749. [PMID: 30603455 DOI: 10.1007/s13770-016-0094-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/20/2016] [Accepted: 07/25/2016] [Indexed: 12/17/2022] Open
Abstract
Pluripotent stem cells can differentiate into many cell types including mature hepatocytes, and can be used in the development of new drugs, treatment of diseases, and in basic research. In this study, we established a protocol leading to efficient hepatic differentiation, and compared the capacity to differentiate into the hepatocyte lineage of human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Optimal combinations of cytokines and growth factors were added to embryoid bodies produced by both types of cell. Differentiation of the cells was assessed with optical and electron microscopes, and hepatic-specific transcripts and proteins were detected by quantitative reverse transcription polymerase chain reaction and immunocytochemistry, respectively. Both types of embryoid body produced polygonal hepatocyte-like cells accompanied by time-dependent up regulation of genes for α-fetoprotein, albumin (ALB), asialoglycoprotein1, CK8, CK18, CK19, CYP1A2, and CYP3A4, which are expressed in fetal and adult hepatocytes. Both types of cell displayed functions characteristic of mature hepatocytes such as accumulation of glycogen, secretion of ALB, and uptake of indocyanine green. And these cells are transplanted into mouse model. Our findings indicate that hESCs and hiPSCs have similar abilities to differentiate into hepatocyte in vitro using the protocol developed here, and these cells are transplantable into damaged liver. Electronic Supplementary Material Supplementary material is available for this article at 10.1007/s13770-016-0094-y and is accessible for authorized users.
Collapse
Affiliation(s)
- Jaemin Jeong
- 1Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Kyu Nam Kim
- 2Department of Anesthesiology and Pain Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Min Sung Chung
- 1Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- 3Department of Surgery, Hanyang University College of Medicine, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763 Korea
| | - Han Joon Kim
- 1Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- 3Department of Surgery, Hanyang University College of Medicine, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763 Korea
| |
Collapse
|
105
|
Johnson JD. The quest to make fully functional human pancreatic beta cells from embryonic stem cells: climbing a mountain in the clouds. Diabetologia 2016; 59:2047-57. [PMID: 27473069 DOI: 10.1007/s00125-016-4059-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/23/2016] [Indexed: 01/10/2023]
Abstract
The production of fully functional insulin-secreting cells to treat diabetes is a major goal of regenerative medicine. In this article, I review progress towards this goal over the last 15 years from the perspective of a beta cell biologist. I describe the current state-of-the-art, and speculate on the general approaches that will be required to identify and achieve our ultimate goal of producing functional beta cells. The need for deeper phenotyping of heterogeneous cultures of stem cell derived islet-like cells in parallel with a better understanding of the heterogeneity of the target cell type(s) is emphasised. This deep phenotyping should include high-throughput single-cell analysis, as well as comprehensive 'omics technologies to provide unbiased characterisation of cell products and human beta cells. There are justified calls for more detailed and well-powered studies of primary human pancreatic beta cell physiology, and I propose online databases of standardised human beta cell responses to physiological stimuli, including both functional and metabolomic/proteomic/transcriptomic profiles. With a concerted, community-wide effort, including both basic and applied scientists, beta cell replacement will become a clinical reality for patients with diabetes.
Collapse
Affiliation(s)
- James D Johnson
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, 5358-2350 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3.
| |
Collapse
|
106
|
Yu L, Wang H, Guo Z, Li F, Cui H. Role of nucleolar protein NOM1 in pancreatic islet β cell apoptosis in diabetes. Exp Ther Med 2016; 12:2275-2280. [PMID: 27698723 DOI: 10.3892/etm.2016.3576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/05/2016] [Indexed: 12/15/2022] Open
Abstract
Diabetes is a metabolic disease that results from impairment in insulin secretion. The present study aimed to investigate the potential role of NOM1 in the function of pancreatic islet β cells and insulin secretion. MIN6 cells isolated from mice were transfected with siRNA-NOM1 to assess the influence of NOM1 on the expression of the cell apoptosis-associated proteins, such as caspase-3. In addition, MIN6 cells were cultured in medium containing different glucose concentrations in order to assess the sensitivity of MIN6 cells to glucose. The effect of NOM1 expression and glucose on MIN6 cell proliferation was also analyzed using an MTT assay. Furthermore, the mRNA expression levels of insulin 1 and 2 in MIN6 cells were detected using reverse transcription-quantitative polymerase chain reaction, while the expression levels of various cell apoptosis-associated proteins, Bcl-2 and Bax, were analyzed using western blot analysis. Compared with the control group, downregulation NOM1 and high glucose concentration of 25 mM significantly increased the cleaved caspase-3 level in MIN6 cells (P<0.05). In addition, downregulation of NOM1 significantly inhibited the MIN6 cell proliferation ability and reduced the insulin 2 mRNA expression (P<0.05). NOM1 knockdown also resulted in significantly increased Bax2 level and decreased Bcl-2 level in MIN6 cells (P<0.05). However no significant difference in insulin mRNA expression was observed between the control and siRNA-NOM1-transfected group (P>0.05). In conclusion, the present study suggested that NOM1 expression may be affected by blood glucose, and that NOM1 may be associated with pancreatic islet β cell apoptosis. In addition, NOM1 may serve a pivotal role in diabetes by affecting insulin synthesis and secretion in pancreatic islet β cells.
Collapse
Affiliation(s)
- Leilei Yu
- Department of Endocrinology, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Huifeng Wang
- Department of Reproduction and Genetics, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Zhongxiu Guo
- Department of Cardiology, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Fenghua Li
- Department of Endocrinology, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Hong Cui
- Department of Reproduction and Genetics, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
107
|
Xun Q, Wang H, He F. Bone marrow mesenchymal stem cells for treatment of pancreatic diseases: Research status and prospects. Shijie Huaren Xiaohua Zazhi 2016; 24:3232-3237. [DOI: 10.11569/wcjd.v24.i21.3232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The incidence of pancreatic diseases is increasing year by year. Current treatments for pancreatic diseases are mainly symptomatic, and the research on the repair and restoration of function of the pancreatic cells progresses slowly. Stem cells have been widely used in the treatment of diseases in recent years because of their ability of multi-directional differentiation and repair of cell damage caused by disease and injury. Numerous studies confirm that pancreatic stem cells after transplantation can differentiate into pancreatic cells and play an important role in the recovery of external secretory function and repair of the damaged pancreatic cells. Particularly, both in vivo and in vitro studies show that bone marrow mesenchymal stem cells have achieved remarkable results in the treatment of pancreatic diseases, laying a theoretical and practical basis for clinical treatment of pancreatic diseases with stem cells. This article outlines the progress in treatment of acute pancreatitis, chronic pancreatitis and pancreatic cancer with bone marrow mesenchymal stem cells, demonstrating that stem cells are expected to become one of new methods for the treatment of pancreatic diseases.
Collapse
|
108
|
Vanikar AV, Trivedi HL, Thakkar UG. Stem cell therapy emerging as the key player in treating type 1 diabetes mellitus. Cytotherapy 2016; 18:1077-86. [PMID: 27424148 DOI: 10.1016/j.jcyt.2016.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/24/2016] [Accepted: 06/07/2016] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease causing progressive destruction of pancreatic β cells, ultimately resulting in loss of insulin secretion producing hyperglycemia usually affecting children. Replacement of damaged β cells by cell therapy can treat it. Currently available strategies are insulin replacement and islet/pancreas transplantation. Unfortunately these offer rescue for variable duration due to development of autoantibodies. For pancreas/islet transplantation a deceased donor is required and various shortfalls of treatment include quantum, cumbersome technique, immune rejection and limited availability of donors. Stem cell therapy with assistance of cellular reprogramming and β-cell regeneration can open up new therapeutic modalities. The present review describes the history and current knowledge of T1DM, evolution of cell therapies and different cellular therapies to cure this condition.
Collapse
Affiliation(s)
- Aruna V Vanikar
- Department of Regenerative Medicine and Stem Cell Therapy, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India; Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India.
| | - Hargovind L Trivedi
- Department of Regenerative Medicine and Stem Cell Therapy, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India; Department of Nephrology and Transplantation Medicine, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India
| | - Umang G Thakkar
- Department of Regenerative Medicine and Stem Cell Therapy, G.R. Doshi and K.M. Mehta Institute of Kidney Diseases & Research Centre, Dr. H.L. Trivedi Institute of Transplantation Sciences, Gujarat, India
| |
Collapse
|
109
|
Okere B, Lucaccioni L, Dominici M, Iughetti L. Cell therapies for pancreatic beta-cell replenishment. Ital J Pediatr 2016; 42:62. [PMID: 27400873 PMCID: PMC4940879 DOI: 10.1186/s13052-016-0273-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 06/21/2016] [Indexed: 12/19/2022] Open
Abstract
The current treatment approach for type 1 diabetes is based on daily insulin injections, combined with blood glucose monitoring. However, administration of exogenous insulin fails to mimic the physiological activity of the islet, therefore diabetes often progresses with the development of serious complications such as kidney failure, retinopathy and vascular disease. Whole pancreas transplantation is associated with risks of major invasive surgery along with side effects of immunosuppressive therapy to avoid organ rejection. Replacement of pancreatic beta-cells would represent an ideal treatment that could overcome the above mentioned therapeutic hurdles. In this context, transplantation of islets of Langerhans is considered a less invasive procedure although long-term outcomes showed that only 10 % of the patients remained insulin independent five years after the transplant. Moreover, due to shortage of organs and the inability of islet to be expanded ex vivo, this therapy can be offered to a very limited number of patients. Over the past decade, cellular therapies have emerged as the new frontier of treatment of several diseases. Furthermore the advent of stem cells as renewable source of cell-substitutes to replenish the beta cell population, has blurred the hype on islet transplantation. Breakthrough cellular approaches aim to generate stem-cell-derived insulin producing cells, which could make diabetes cellular therapy available to millions. However, to date, stem cell therapy for diabetes is still in its early experimental stages. This review describes the most reliable sources of stem cells that have been developed to produce insulin and their most relevant experimental applications for the cure of diabetes.
Collapse
Affiliation(s)
- Bernard Okere
- Division of Pediatric Oncology, Hematology and Marrow Transplantation, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena Policlinic, Modena, 41100, Italy
| | - Laura Lucaccioni
- Division of Pediatric Oncology, Hematology and Marrow Transplantation, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena Policlinic, Modena, 41100, Italy.,Child Health, School of Medicine, Dentistry & Nursing, University of Glasgow, Glasgow, UK
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena Policlinic, Modena, 41100, Italy
| | - Lorenzo Iughetti
- Division of Pediatric Oncology, Hematology and Marrow Transplantation, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena Policlinic, Modena, 41100, Italy.
| |
Collapse
|
110
|
Jiang FX, Morahan G. Insulin-secreting β cells require a post-genomic concept. World J Diabetes 2016; 7:198-208. [PMID: 27226815 PMCID: PMC4873311 DOI: 10.4239/wjd.v7.i10.198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 03/18/2016] [Indexed: 02/05/2023] Open
Abstract
Pancreatic insulin-secreting β cells are essential in maintaining normal glucose homeostasis accomplished by highly specialized transcription of insulin gene, of which occupies up to 40% their transcriptome. Deficiency of these cells causes diabetes mellitus, a global public health problem. Although tremendous endeavors have been made to generate insulin-secreting cells from human pluripotent stem cells (i.e., primitive cells capable of giving rise to all cell types in the body), a regenerative therapy to diabetes has not yet been established. Furthermore, the nomenclature of β cells has become inconsistent, confusing and controversial due to the lack of standardized positive controls of developmental stage-matched in vivo cells. In order to minimize this negative impact and facilitate critical research in this field, a post-genomic concept of pancreatic β cells might be helpful. In this review article, we will briefly describe how β cells were discovered and islet lineage is developed that may help understand the cause of nomenclatural controversy, suggest a post-genomic definition and finally provide a conclusive remark on future research of this pivotal cell.
Collapse
|
111
|
Cui JP, Zhang MJ, Liu BL. Research progress of mesenchymal stem cells combined with islet transplantation in treatment of type I diabetes mellitus. Shijie Huaren Xiaohua Zazhi 2016; 24:2213-2218. [DOI: 10.11569/wcjd.v24.i14.2213] [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] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The most significant feature of type I diabetes is β-cell loss, which results in a series of complications. While β-cell loss occurs, β-cells are ultimately damaged by macrophages and T cells in the presence of inflammatory mediators. Because of this characteristic, five kinds of antibodies are commonly used in clinical practice to diagnose and evaluate β-cell loss, including islet cell antibody, insulin antibody, GAD65, IA-2 and IA-2b. In addition to the HLA gene related factors, environmental factors, such as infection, diet and physiological and psychological factors, are suspected to be causes of this disease. At present, there are many treatments for type I diabetes, and the clinical goal is to control blood glucose, prevent further damage of βcells and control patients' own immune response. In 1992, the discovery of insulin, which converts the fatal diabetes into a chronic disease, to some extent, delayed the progression of microvascular complications; however, it is not able to delay the progression of the disease. β-cell transplantation is currently the only minimally invasive means for reasonable control of blood glucose control disease related complications. Although whole pancreas transplantation can achieve a promising effect to some extent, it is accompanied by high incidence and mortality, as well as lifelong mandatory immune suppression. Bone marrow mesenchymal stem cells transplantation, lipopolysaccharideon (LPS) bone marrow mesenchymal stem cell pretreatment and islet cell exendin-4 liquid preservation reduce warm ischemia time damage and provide new avenues for islet cell transplantation.
Collapse
|
112
|
Abstract
Since insulin discovery, islet transplantation was the first protocol to show the possibility to cure patients with type 1 diabetes using low-risk procedures. The scarcity of pancreas donors triggered a burst of studies focused on the production of new β cells in vitro. These were rapidly dominated by pluripotent stem cells (PSCs) demonstrating diabetes-reversal potential in diabetic mice. Subsequent enthusiasm fostered a clinical trial with immunoisolated embryonic-derived pancreatic progenitors. Yet safety is the Achilles' heel of PSCs, and a whole branch of β cell engineering medicine focuses on transdifferentiation of adult pancreatic cells. New data showed the possibility to chemically stimulate acinar or α cells to undergo β cell neogenesis and provide opportunities to intervene in situ without the need for a transplant, at least after weighing benefits against systemic adverse effects. The current studies suggested the pancreas as a reservoir of facultative progenitors (e.g., in the duct lining) could be exploited ex vivo for expansion and β cell differentiation in timely fashion and without the hurdles of PSC use. Diabetes cell therapy is thus a growing field not only with great potential but also with many pitfalls to overcome for becoming fully envisioned as a competitor to the current treatment standards.
Collapse
Affiliation(s)
- Philippe A Lysy
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium.
- Pediatric Endocrinology Unit, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium.
| | - Elisa Corritore
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| | - Etienne M Sokal
- Institut de Recherche Expérimentale et Clinique, Pediatric Research Laboratory, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
113
|
Zhang S, Bai C, Ma Y, Li X, Gao Y, Fan Y, Guan W, Zheng D. The characterisation and functional β-cell differentiation of duck pancreas-derived mesenchymal cells. Br Poult Sci 2016; 57:201-10. [DOI: 10.1080/00071668.2015.1135505] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
114
|
Generating Mini-Organs in Culture. CURRENT PATHOBIOLOGY REPORTS 2016. [DOI: 10.1007/s40139-016-0101-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
115
|
Mehrfarjam Z, Esmaeili F, Shabani L, Ebrahimie E. Induction of pancreatic β cell gene expression in mesenchymal stem cells. Cell Biol Int 2016; 40:486-500. [DOI: 10.1002/cbin.10567] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Zahra Mehrfarjam
- Razi Herbal Medicines Research Center; Lorestan University of Medical Sciences; P.O. Box 681499468 Khorramabad Iran
| | - Fariba Esmaeili
- Faculty of Basic Sciences; Department of Biology; University of Isfahan; P.O. Box 8174673441 Isfahan Iran
- Research Institute of Biotechnology; Shahrekord University; P.O. Box 115 Shahrekord Iran
| | - Leila Shabani
- Research Institute of Biotechnology; Shahrekord University; P.O. Box 115 Shahrekord Iran
| | - Esmaeil Ebrahimie
- Institute of Biotechnology; Shiraz University; Shiraz Iran
- Division of Information Technology, Engineering & Environment; School of Information Technology and Mathematical Sciences; University of South Australia; Adelaide Australia
- Department of Genetics and Evolution; The University of Adelaide; Adelaide Australia
- Faculty of Science and Engineering; School of Biological Sciences; Flinders University; Adelaide Australia
| |
Collapse
|
116
|
Microtissues in Cardiovascular Medicine: Regenerative Potential Based on a 3D Microenvironment. Stem Cells Int 2016; 2016:9098523. [PMID: 27073399 PMCID: PMC4814701 DOI: 10.1155/2016/9098523] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/01/2016] [Accepted: 02/21/2016] [Indexed: 02/06/2023] Open
Abstract
More people die annually from cardiovascular diseases than from any other cause. In particular, patients who suffer from myocardial infarction may be affected by ongoing adverse remodeling processes of the heart that may ultimately lead to heart failure. The introduction of stem and progenitor cell-based applications has raised substantial hope for reversing these processes and inducing cardiac regeneration. However, current stem cell therapies using single-cell suspensions have failed to demonstrate long-lasting efficacy due to the overall low retention rate after cell delivery to the myocardium. To overcome this obstacle, the concept of 3D cell culture techniques has been proposed to enhance therapeutic efficacy and cell engraftment based on the simulation of an in vivo-like microenvironment. Of great interest is the use of so-called microtissues or spheroids, which have evolved from their traditional role as in vitro models to their novel role as therapeutic agents. This review will provide an overview of the therapeutic potential of microtissues by addressing primarily cardiovascular regeneration. It will accentuate their advantages compared to other regenerative approaches and summarize the methods for generating clinically applicable microtissues. In addition, this review will illustrate the unique properties of the microenvironment within microtissues that makes them a promising next-generation therapeutic approach.
Collapse
|
117
|
Takahashi Y, Takebe T, Taniguchi H. Engineering pancreatic tissues from stem cells towards therapy. Regen Ther 2016; 3:15-23. [PMID: 31245468 PMCID: PMC6581807 DOI: 10.1016/j.reth.2016.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/17/2015] [Accepted: 01/20/2016] [Indexed: 12/20/2022] Open
Abstract
Pancreatic islet transplantation is performed as a potential treatment for type 1 diabetes mellitus. However, this approach is significantly limited due to the critical shortage of islet sources. Recently, a number of publications have developed protocols for directed β-cell differentiation of pluripotent cells, such as embryonic stem (ES) or induced pluripotent stem (iPS) cells. Decades of studies have led to the development of modified protocols that recapitulate molecular developmental cues by combining various growth factors and small molecules with improved efficiency. However, the later step of pancreatic differentiation into functional β-cells has yet to be satisfactory in vitro, highlighting alternative approach by recapitulating spatiotemporal multicellular interaction in three-dimensional (3D) culture. Here, we summarize recent progress in the directed differentiation into pancreatic β-cells with a focus on both two-dimensional (2D) and 3D differentiation settings. We also discuss the potential transplantation strategies in combination with current bioengineering approaches towards diabetes therapy. Transplantation of stem cell derived pancreatic progenitors is a possible approach for generating mature β-cell in vivo. Promise of 3-D (or 4-D) culture has started to be explored by reconstituting pancreatic tissue structures. Self-condensation culture is a basic technique of integrating multiple heterotypic lineages including vasculatures. Bioengineering approach has been combined for developing effective transplant strategies.
Collapse
Key Words
- 2D, two-dimensional
- 3D, three-dimensional
- BMP, bone morphogenic protein
- Diabetes
- ES, embryonic stem
- FGF, fibroblast growth factors
- Heterotypic cellular interaction
- IBMIR, instant blood-mediated reaction
- ILV, indolactam V
- Ngn3, neurogenin 3
- PEG, polyethylene glycol
- PI3K, phosphatidylinositol-3 kinase
- PIPAAm, poly-N-isopropylacrylamide
- PVA, polyvinyl alcohol
- Pancreas
- Pdx1, pancreatic and duodenal homeobox 1
- Ptf1a, pancreatic transcription factor 1a
- Regenerative medicine
- VEGF, vascular endothelial growth factor
- Vascularization
- iPS, induced pluripotent stem
- iPS/ES cell
Collapse
Affiliation(s)
- Yoshinobu Takahashi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan
| | - Takanori Takebe
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan.,Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan.,PRESTO, Japan Science and Technology Agency, 4-1-8, Honcho, Kawaguchi, Saitama, 332-0012, Japan.,Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH, 45229- 3039, USA
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan.,Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan
| |
Collapse
|
118
|
Zhao Q, Yang Y, Hu J, Shan Z, Wu Y, Lei L. Exendin-4 enhances expression of Neurod1 and Glut2 in insulin-producing cells derived from mouse embryonic stem cells. Arch Med Sci 2016; 12:199-207. [PMID: 26925137 PMCID: PMC4754381 DOI: 10.5114/aoms.2016.57596] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/25/2014] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Stem cells involved cell replacement therapies for type 1 diabetes mellitus is promising, yet time-consuming and inefficient. Exendin-4 is a glucagon-like peptide-1 (GLP-1) receptor agonist which has been reported to possess anti-apoptotic effects, thereby increasing β-cell mass and improving β-cell function. The present study aimed to investigate whether exendin-4 would enhance the differentiation of embryonic stem cells into insulin-secreting cells and improve the pancreatic differentiation strategy. MATERIAL AND METHODS R1 embryonic stem cells were treated with different concentrations of exendin-4 and divided into three groups. In the high dosage group (group H), exendin-4 was added at the dosage of 10 nmol/l. In the low dosage group (group L), exendin-4 was added at the dosage of 0.1 nmol/l. Group C was a control. Expression of genes related to the β-cell phenotype and immunofluorescence staining of insulin and C-peptide were detected. RESULTS Compared with groups L and C, group H had the highest mRNA expression levels of Isl1, Pdx1, Ngn3, and Insulin1 (p < 0.05). Neurod1 and Glut2 only emerged at the final stage of differentiation in group H. Immunofluorescence analysis revealed that exendin-4 upregulated the protein expression of insulin and C-peptide. CONCLUSIONS Exendin-4 remarkably facilitated Neurod1 and Glut2 gene transcription, and was able to induce differentiation of embryonic stem cells into endocrine and insulin-producing cells.
Collapse
Affiliation(s)
- Qiaoshi Zhao
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Yuzhi Yang
- Division of Endocrinology, Heilongjiang Provincial Hospital, Harbin, China
| | - Jing Hu
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Zhiyan Shan
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Yanshuang Wu
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Lei Lei
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| |
Collapse
|
119
|
Wan Y, Garner J, Wu N, Phillip L, Han Y, McDaniel K, Annable T, Zhou T, Francis H, Glaser S, Huang Q, Alpini G, Meng F. Role of stem cells during diabetic liver injury. J Cell Mol Med 2016; 20:195-203. [PMID: 26645107 PMCID: PMC4727564 DOI: 10.1111/jcmm.12723] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/24/2015] [Indexed: 12/18/2022] Open
Abstract
Diabetes mellitus is one of the most severe endocrine metabolic disorders in the world that has serious medical consequences with substantial impacts on the quality of life. Type 2 diabetes is one of the main causes of diabetic liver diseases with the most common being non-alcoholic fatty liver disease. Several factors that may explain the mechanisms related to pathological and functional changes of diabetic liver injury include: insulin resistance, oxidative stress and endoplasmic reticulum stress. The realization that these factors are important in hepatocyte damage and lack of donor livers has led to studies concentrating on the role of stem cells (SCs) in the prevention and treatment of liver injury. Possible avenues that the application of SCs may improve liver injury include but are not limited to: the ability to differentiate into pancreatic β-cells (insulin producing cells), the contribution for hepatocyte regeneration, regulation of lipogenesis, glucogenesis and anti-inflammatory actions. Once further studies are performed to explore the underlying protective mechanisms of SCs and the advantages and disadvantages of its application, there will be a greater understand of the mechanism and therapeutic potential. In this review, we summarize the findings regarding the role of SCs in diabetic liver diseases.
Collapse
Affiliation(s)
- Ying Wan
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research, Southern Medical University, Guangzhou, China
| | - Jessica Garner
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Nan Wu
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
| | - Levine Phillip
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Yuyan Han
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
| | - Kelly McDaniel
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Tami Annable
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Tianhao Zhou
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Heather Francis
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
| | - Qiaobing Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research, Southern Medical University, Guangzhou, China
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Temple, TX, USA
- Department of Internal Medicine, Scott & White Digestive Disease Research Center, Texas A&M University Health Science Center and Baylor Scott & White Healthcare, Temple, TX, USA
- Academic Operations, Baylor Scott & White Healthcare, Temple, TX, USA
| |
Collapse
|
120
|
Khorsandi L, Saremy S, Khodadadi A, Dehbashi F. Effects of Exendine-4 on The Differentiation of Insulin Producing Cells from Rat Adipose-Derived Mesenchymal Stem Cells. CELL JOURNAL 2016; 17:720-9. [PMID: 26862531 PMCID: PMC4746422 DOI: 10.22074/cellj.2016.3844] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 01/07/2015] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To evaluate the effect of Exendine-4 (EX-4), a Glucagon-like peptide 1 (GLP-1) receptor agonist, on the differentiation of insulin-secreting cells (IPCs) from rat adipose-derived mesenchymal stem cells(ADMSCs). MATERIALS AND METHODS In this experimental study, ADMSCs were isolated from rat adi- pose tissue and exposed to induction media with or without EX-4. After induction, the existence of IPCs was confirmed by morphology analysis, expression pattern analysis of islet-specific genes (Pdx-1, Glut-2 and Insulin) and insulin synthesis and secretion. RESULTS IPCs induced in presence of EX-4 were morphologically similar to pancre- atic islet-like cells. Expression of Pdx-1, Glut-2 and Insulin genes in EX-4 treated cells was significantly higher than the cells exposed to differentiation media without EX-4. Compared to EX-4 untreated ADMSCs, insulin release from EX-4 treated ADMSCs showed a nearly 2.5 fold (P<0.05) increase when exposed to a high glucose (25 mM) medium. The percentage of insulin positive cells in the EX-4 treated group was ap- proximately 4-fold higher than in the EX-4 untreated ADMSCs. CONCLUSION The present study has demonstrated that EX-4 enhances the differen- tiation of ADMSCs into IPCs. Improvement of this method may help the formation of an unlimited source of cells for transplantation.
Collapse
Affiliation(s)
- Layasadat Khorsandi
- Cell and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Anatomical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sadegh Saremy
- Cell and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Khodadadi
- Department of Anatomical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fereshteh Dehbashi
- Cell and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
121
|
Abouzaripour M, Pasbakhsh P, Atlasi N, Shahverdi AH, Mahmoudi R, Kashani IR. In Vitro Differentiation of Insulin Secreting Cells from Mouse Bone Marrow Derived Stage-Specific Embryonic Antigen 1 Positive Stem Cells. CELL JOURNAL 2016; 17:701-10. [PMID: 26862529 PMCID: PMC4746420 DOI: 10.22074/cellj.2016.3842] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 02/02/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Bone marrow has recently been recognized as a novel source of stem cells for the treatment of wide range of diseases. A number of studies on murine bone mar- row have shown a homogenous population of rare stage-specific embryonic antigen 1 (SSEA-1) positive cells that express markers of pluripotent stem cells. This study focuses on SSEA-1 positive cells isolated from murine bone marrow in an attempt to differentiate them into insulin-secreting cells (ISCs) in order to investigate their differentiation potential for future use in cell therapy. MATERIALS AND METHODS This study is an experimental research. Mouse SSEA-1 positive cells were isolated by Magnetic-activated cell sorting (MACS) followed by characteriza- tion with flow cytometry. Induced SSEA-1 positive cells were differentiated into ISCs with specific differentiation media. In order to evaluate differentiation quality and analysis, dithizone (DTZ) staining was use, followed by reverse transcription polymerase chain reaction (RT-PCR), immunocytochemistry and insulin secretion assay. Statistical results were analyzed by one-way ANOVA. RESULTS The results achieved in this study reveal that mouse bone marrow contains a population of SSEA-1 positive cells that expresses pluripotent stem cells markers such as SSEA-1, octamer-binding transcription factor 4 (OCT-4) detected by immunocytochem- istry and C-X-C chemokine receptor type 4 (CXCR4) and stem cell antigen-1 (SCA-1) detected by flow cytometric analysis. SSEA-1 positive cells can differentiate into ISCs cell clusters as evidenced by their DTZ positive staining and expression of genes such as Pdx1 (pancreatic transcription factors), Ngn3 (endocrine progenitor marker), Insulin1 and Insulin2 (pancreaticβ-cell markers). Additionally, our results demonstrate expression of Pdx1 and Glut2 protein and insulin secretion in response to a glucose challenge in the differentiated cells. CONCLUSION Our study clearly demonstrates the potential of SSEA-1 positive cells to differentiate into insulin secreting cells in defined culture conditions for clinical ap- plications.
Collapse
Affiliation(s)
- Morteza Abouzaripour
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nader Atlasi
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdol Hossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Medicine, ACECR, Tehran, Iran
| | - Reza Mahmoudi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
122
|
El-Badawy A, El-Badri N. The cell cycle as a brake for β-cell regeneration from embryonic stem cells. Stem Cell Res Ther 2016; 7:9. [PMID: 26759123 PMCID: PMC4711007 DOI: 10.1186/s13287-015-0274-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The generation of insulin-producing β cells from stem cells in vitro provides a promising source of cells for cell transplantation therapy in diabetes. However, insulin-producing cells generated from human stem cells show deficiency in many functional characteristics compared with pancreatic β cells. Recent reports have shown molecular ties between the cell cycle and the differentiation mechanism of embryonic stem (ES) cells, assuming that cell fate decisions are controlled by the cell cycle machinery. Both β cells and ES cells possess unique cell cycle machinery yet with significant contrasts. In this review, we compare the cell cycle control mechanisms in both ES cells and β cells, and highlight the fundamental differences between pluripotent cells of embryonic origin and differentiated β cells. Through critical analysis of the differences of the cell cycle between these two cell types, we propose that the cell cycle of ES cells may act as a brake for β-cell regeneration. Based on these differences, we discuss the potential of modulating the cell cycle of ES cells for the large-scale generation of functionally mature β cells in vitro. Further understanding of the factors that modulate the ES cell cycle will lead to new approaches to enhance the production of functional mature insulin-producing cells, and yield a reliable system to generate bona fide β cells in vitro.
Collapse
Affiliation(s)
- Ahmed El-Badawy
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Sheikh Zayed District, 12588, 6th of October City, Giza, Egypt.
| |
Collapse
|
123
|
Abstract
Regenerative medicine with stem cells holds great hope for the treatment of degenerative disease. The medical potential of embryonic stem cells remains relatively untapped at this point, and significant scientific hurdles remain to be overcome before these cells might be considered safe and effective for uses in patients. Meanwhile, adult stem cells have begun to show significant capabilities of their own in repair of damaged tissues, in both animal models and early patient trials.
Collapse
|
124
|
Abdelalim EM, Emara MM. Pluripotent Stem Cell-Derived Pancreatic β Cells: From In Vitro Maturation to Clinical Application. RECENT ADVANCES IN STEM CELLS 2016. [DOI: 10.1007/978-3-319-33270-3_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
125
|
Abstract
In this chapter the state of the art of live cell microarrays for high-throughput biological assays are reviewed. The fabrication of novel microarrays with respect to material science and cell patterning methods is included. A main focus of the chapter is on various aspects of the application of cell microarrays by providing selected examples in research fields such as biomaterials, stem cell biology and neuroscience. Additionally, the importance of microfluidic technologies for high-throughput on-chip live-cell microarrays is highlighted for single-cell and multi-cell assays as well as for 3D tissue constructs.
Collapse
|
126
|
Liu Y, Tang SCW. Recent Progress in Stem Cell Therapy for Diabetic Nephropathy. KIDNEY DISEASES 2015; 2:20-7. [PMID: 27536688 DOI: 10.1159/000441913] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 10/18/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Diabetic nephropathy (DN) represents the leading cause of end-stage renal disease. Current therapeutic strategies for DN are very limited, and none of them can stop end-stage renal disease progression. Stem cell-based therapy showed encouraging outcomes in kidney disease, including experimental DN. SUMMARY Both podocytes and proximal tubular epithelial cells play key roles in the pathogenesis of DN and, accordingly, could be regarded as treatment targets. Multiple kinds of stem cells contribute to the regeneration of the injured kidney, including embryonic stem cells (ESCs), mesenchymal stem cells, and induced pluripotent stem cells (iPSCs). Stem cells exert reparatory effects mainly by homing to injured sites, directing differentiation, paracrine action, and immunoregulation. However, poor survival after transplantation under diabetic conditions and unsatisfactory animal models of advanced DN are major obstacles for achieving an efficacious therapeutic effect from stem cell transplantation. Recently, remarkable progress has been made both in the direct differentiation of human ESCs and iPSCs into renal cells and in the generation of tissue- and patient-specific iPSCs, offering a powerful tool to investigate DN mechanisms and to identify the ideal candidate cell for future clinical application. KEY MESSAGE This review provides updated information on recent progress and limitations of stem cell-based therapy for DN.
Collapse
Affiliation(s)
- Yang Liu
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR, China
| | - Sydney C W Tang
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR, China
| |
Collapse
|
127
|
Quiskamp N, Bruin JE, Kieffer TJ. Differentiation of human pluripotent stem cells into β-cells: Potential and challenges. Best Pract Res Clin Endocrinol Metab 2015; 29:833-47. [PMID: 26696513 DOI: 10.1016/j.beem.2015.10.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) hold great potential as the basis for cell-based therapies of degenerative diseases, including diabetes. Current insulin-based therapies for diabetes do not prevent hyperglycaemia or the associated long-term organ damage. While transplantation of pancreatic islets can achieve insulin independence and improved glycemic control, it is limited by donor tissue scarcity, challenges of purifying islets from the pancreas, and the need for immunosuppression to prevent rejection of transplants. Large-scale production of β-cells from stem cells is a promising alternative. Recent years have seen considerable progress in the optimization of in vitro differentiation protocols to direct hESCs/iPSCs into mature insulin-secreting β-cells and clinical trials are now under way to test the safety and efficiency of hESC-derived pancreatic progenitor cells in patients with type 1 diabetes. Here, we discuss key milestones leading up to these trials in addition to recent developments and challenges for hESC/iPSC-based diabetes therapies and disease modeling.
Collapse
Affiliation(s)
- Nina Quiskamp
- Laboratory of Molecular and Cellular Medicine, Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| | - Jennifer E Bruin
- Laboratory of Molecular and Cellular Medicine, Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine, Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
128
|
Abstract
Regenerative medicine using patient's own stem cells (SCs) to repair dysfunctional tissues is an attractive approach to complement surgical and pharmacological treatments for aging and degenerative disorders. Recently, dental SCs have drawn much attention owing to their accessibility, plasticity and applicability for regenerative use not only for dental, but also other body tissues. In ophthalmology, there has been increasing interest to differentiate dental pulp SC and periodontal ligament SC (PDLSC) towards ocular lineage. Both can commit to retinal fate expressing eye field transcription factors and generate rhodopsin-positive photoreceptor-like cells. This proposes a novel therapeutic alternative for retinal degeneration diseases. Moreover, as PDLSC shares similar cranial neural crest origin and proteoglycan secretion with corneal stromal keratoctyes and corneal endothelial cells, this offers the possibility of differentiating PDLSC to these corneal cell types. The advance could lead to a shift in the medical management of corneal opacities and endothelial disorders from highly invasive corneal transplantation using limited donor tissue to cell therapy utilizing autologous cells. This article provides an overview of dental SC research and the perspective of utilizing dental SCs for ocular regenerative medicine.
Collapse
|
129
|
El-Demerdash RF, Hammad LN, Kamal MM, El Mesallamy HO. A comparison of Wharton's jelly and cord blood as a source of mesenchymal stem cells for diabetes cell therapy. Regen Med 2015; 10:841-855. [PMID: 26541176 DOI: 10.2217/rme.15.49] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM In this study, we investigated the differences between mesenchymal stem cells (MSCs), isolated from umbilical cord blood (UCB-MSCs) and Wharton's jelly (WJ-MSCs) as sources of diabetes mellitus cell therapy. METHODS After isolation, both cell types were induced to differentiate into insulin producing cells, then the differentiated cells were assessed genetically and functionally. UCB-MSCs and WJ-MSCs were transplanted in the tail veins of streptozotocin-induced diabetic rats. Blood glucose levels were monitored post-transplantation. RESULTS & CONCLUSION Wharton's jelly was more homogeneous, can better differentiate into insulin producing cells in vitro and better control hyperglycemia in diabetic rats in vivo, as compared with UCB. These results indicate that WJ-MSCs represent a potential source of cells in the field of diabetes mellitus cell therapy.
Collapse
Affiliation(s)
- Rasha F El-Demerdash
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt, 44971
| | - Lamiaa N Hammad
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt, 44971
| | - Mohamed M Kamal
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt, 11566
| | - Hala O El Mesallamy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt, 11566
| |
Collapse
|
130
|
Recent advances and future applications of microfluidic live-cell microarrays. Biotechnol Adv 2015; 33:948-61. [DOI: 10.1016/j.biotechadv.2015.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/16/2015] [Accepted: 06/19/2015] [Indexed: 12/31/2022]
|
131
|
Tan Y, AlKhamees B, Jia D, Li L, Couture JF, Figeys D, Jinushi M, Wang L. MFG-E8 Is Critical for Embryonic Stem Cell-Mediated T Cell Immunomodulation. Stem Cell Reports 2015; 5:741-752. [PMID: 26455415 PMCID: PMC4649138 DOI: 10.1016/j.stemcr.2015.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 02/03/2023] Open
Abstract
The molecules and mechanisms pertinent to the low immunogenicity of undifferentiated embryonic stem cells (ESCs) remain poorly understood. Here, we provide evidence that milk fat globule epidermal growth factor 8 (MFG-E8) is a vital mediator in this phenomenon and directly suppresses T cell immune responses. MFG-E8 is enriched in undifferentiated ESCs but diminished in differentiated ESCs. Upregulation of MFG-E8 in ESCs increases the successful engraftment of both undifferentiated and differentiated ESCs across major histocompatibility complex barriers. MFG-E8 suppresses T cell activation/proliferation and inhibits Th1, Th2, and Th17 subpopulations while increasing regulatory T cell subsets. Neutralizing MFG-E8 substantially abrogates these effects, whereas addition of recombinant MFG-E8 to differentiated ESCs restores immunosuppression. Furthermore, we provide the evidence that MFG-E8 suppresses T cell activation and regulates T cell polarization by inhibiting PKCθ phosphorylation through the α3/5βV integrin receptor. Our findings offer an approach to facilitate transplantation acceptance. MFG-E8 is enriched in undifferentiated but diminished in differentiated ESCs MFG-E8 promotes allogeneic engraftment of ESC-derived tissues across the MHC barrier ESC-produced MFG-E8 inhibits Th1/Th2/Th17 while promoting regulatory T cells MFG-E8 modulates T cell polarization via inhibiting PKCθ phosphorylation
Collapse
Affiliation(s)
- Yuan Tan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Bodour AlKhamees
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Deyong Jia
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Li Li
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Jean-François Couture
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daniel Figeys
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Masahisa Jinushi
- Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjiku-ku, Tokyo 160-8582, Japan.
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|
132
|
Wang Y, Hai T, Liu L, Liu Z, Zhou Q. Cell therapy in diabetes: current progress and future prospects. Sci Bull (Beijing) 2015. [DOI: 10.1007/s11434-015-0844-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
133
|
Ali S, Wall IB, Mason C, Pelling AE, Veraitch FS. The effect of Young's modulus on the neuronal differentiation of mouse embryonic stem cells. Acta Biomater 2015; 25:253-267. [PMID: 26159105 DOI: 10.1016/j.actbio.2015.07.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 05/27/2015] [Accepted: 07/05/2015] [Indexed: 12/15/2022]
Abstract
There is substantial evidence that cells produce a diverse response to changes in ECM stiffness depending on their identity. Our aim was to understand how stiffness impacts neuronal differentiation of embryonic stem cells (ESC's), and how this varies at three specific stages of the differentiation process. In this investigation, three effects of stiffness on cells were considered; attachment, expansion and phenotypic changes during differentiation. Stiffness was varied from 2 kPa to 18 kPa to finally 35 kPa. Attachment was found to decrease with increasing stiffness for both ESC's (with a 95% decrease on 35 kPa compared to 2 kPa) and neural precursors (with a 83% decrease on 35 kPa). The attachment of immature neurons was unaffected by stiffness. Expansion was independent of stiffness for all cell types, implying that the proliferation of cells during this differentiation process was independent of Young's modulus. Stiffness had no effect upon phenotypic changes during differentiation for mESC's and neural precursors. 2 kPa increased the proportion of cells that differentiated from immature into mature neurons. Taken together our findings imply that the impact of Young's modulus on attachment diminishes as neuronal cells become more mature. Conversely, the impact of Young's modulus on changes in phenotype increased as cells became more mature.
Collapse
|
134
|
Yu T, Qing Q, Deng N, Min XH, Zhao LN, Li JY, Xia ZS, Chen QK. CXCR4 positive cell-derived Pdx1-high/Shh-low cells originated from embryonic stem cells improve the repair of pancreatic injury in mice. Cell Biol Int 2015; 39:995-1006. [PMID: 25820869 DOI: 10.1002/cbin.10470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 03/14/2015] [Accepted: 03/18/2015] [Indexed: 01/05/2023]
Abstract
Treatments for pancreatic injuries have been significantly improved recently, but full recovery of pancreatic function remains difficult. Embryonic stem cells have great potentialities for self-renewal and multiple differentiations. In this study, we explored an approach to induce the differentiation of pancreatic progenitor cells from embryonic stem cells in vitro. Male mouse embryonic stem cells were cultured by the hanging-drop method to form embryoid bodies. The definitive endoderm marked by CXCR4 in embryoid bodies was sorted by magnetic activated cell sorting and subsequently administrated with b-FGF, exendin-4, and cyclopamine to induce the differentiation of putative pancreatic progenitor cells, which was monitored by Pdx1, and Shh expressions. The putative pancreatic progenitor cells were transplanted into female BALB/c mice with pancreatitis induced by L-Arginine. Male donor cells were located by detecting sex-determining region of Y-chromosome DNA. Definitive endoderm cells (CXCR4(+) cells) were sorted from 5-day embryoid bodies. After 3-day administration with b-FGF, exendin-4, and cyclopamine, Pdx1-high/Shh-low cells were differentiated from CXCR4(+) cells. These cells developed into more amylase-secreted cells in vitro and could specifically reside in the damaged pancreas acinar area in mice with acute pancreatitis to enhance the regeneration. The putative pancreatic progenitor cells (Pdx1-high/Shh-low cells) derived from mouse embryonic stem cells through the administration of b-FGF, exendin-4, and cyclopamine on the CXCR4(+) cells in vitro could improve the regeneration of injured pancreatic acini in vivo.
Collapse
Affiliation(s)
- Tao Yu
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Qing Qing
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Na Deng
- Department of Gastroenterology, Yuebei People's Hospital, Shaoguan, Guangdong, People's Republic of China
| | - Xiao-Hui Min
- Department of Infectious Disease, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Li-Na Zhao
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jie-Yao Li
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Zhong-Sheng Xia
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Qi-Kui Chen
- Departmentof Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
135
|
Sahraneshin Samani F, Ebrahimi M, Zandieh T, Khoshchehreh R, Baghaban Eslaminejad M, Aghdami N, Baharvand H. In Vitro Differentiation of Human Umbilical Cord Blood CD133(+)Cells into Insulin Producing Cells in Co-Culture with Rat Pancreatic Mesenchymal Stem Cells. CELL JOURNAL 2015. [PMID: 26199900 PMCID: PMC4503835 DOI: 10.22074/cellj.2016.3717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Objective Pancreatic stroma plays an important role in the induction of pancreatic cells
by the use of close range signaling. In this respect, we presume that pancreatic mesenchymal cells (PMCs) as a fundamental factor of the stromal niche may have an effective
role in differentiation of umbilical cord blood cluster of differentiation 133+ (UCB-CD133+)
cells into newly-formed β-cells in vitro.
Materials and Methods This study is an experimental research. The UCB-CD133+cells
were purified by magnetic activated cell sorting (MACS) and differentiated into insulin
producing cells (IPCs) in co-culture, both directly and indirectly with rat PMCs. Immunocytochemistry and enzyme linked immune sorbent assay (ELISA) were used to determine
expression and production of insulin and C-peptide at the protein level.
Results Our results demonstrated that UCB-CD133+differentiated into IPCs. Cells in
islet-like clusters with (out) co-cultured with rat pancreatic stromal cells produced insulin
and C-peptide and released them into the culture medium at the end of the induction protocol. However they did not respond well to glucose challenges.
Conclusion Rat PMCs possibly affect differentiation of UCB-CD133+cells into IPCs by
increasing the number of immature β-cells.
Collapse
Affiliation(s)
- Fazel Sahraneshin Samani
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran ; Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran ; Department of Regenerative Biomedicine at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tahereh Zandieh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reyhaneh Khoshchehreh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran ; Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Biomedicine at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
136
|
Thakkar UG, Trivedi HL, Vanikar AV, Dave SD. Co-infusion of insulin-secreting adipose tissue-derived mesenchymal stem cells and hematopoietic stem cells: novel approach to management of type 1 diabetes mellitus. Int J Diabetes Dev Ctries 2015. [DOI: 10.1007/s13410-015-0409-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
137
|
Márquez-Aguirre AL, Canales-Aguirre AA, Padilla-Camberos E, Esquivel-Solis H, Díaz-Martínez NE. Development of the endocrine pancreas and novel strategies for β-cell mass restoration and diabetes therapy. ACTA ACUST UNITED AC 2015; 48:765-76. [PMID: 26176316 PMCID: PMC4568803 DOI: 10.1590/1414-431x20154363] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 03/22/2015] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus represents a serious public health problem owing to its global
prevalence in the last decade. The causes of this metabolic disease include
dysfunction and/or insufficient number of β cells. Existing diabetes mellitus
treatments do not reverse or control the disease. Therefore, β-cell mass restoration
might be a promising treatment. Several restoration approaches have been developed:
inducing the proliferation of remaining insulin-producing cells, de
novo islet formation from pancreatic progenitor cells (neogenesis), and
converting non-β cells within the pancreas to β cells (transdifferentiation) are the
most direct, simple, and least invasive ways to increase β-cell mass. However, their
clinical significance is yet to be determined. Hypothetically, β cells or islet
transplantation methods might be curative strategies for diabetes mellitus; however,
the scarcity of donors limits the clinical application of these approaches. Thus,
alternative cell sources for β-cell replacement could include embryonic stem cells,
induced pluripotent stem cells, and mesenchymal stem cells. However, most
differentiated cells obtained using these techniques are functionally immature and
show poor glucose-stimulated insulin secretion compared with native β cells.
Currently, their clinical use is still hampered by ethical issues and the risk of
tumor development post transplantation. In this review, we briefly summarize the
current knowledge of mouse pancreas organogenesis, morphogenesis, and maturation,
including the molecular mechanisms involved. We then discuss two possible approaches
of β-cell mass restoration for diabetes mellitus therapy: β-cell regeneration and
β-cell replacement. We critically analyze each strategy with respect to the
accessibility of the cells, potential risk to patients, and possible clinical
outcomes.
Collapse
Affiliation(s)
- A L Márquez-Aguirre
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| | - A A Canales-Aguirre
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| | - E Padilla-Camberos
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| | - H Esquivel-Solis
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| | - N E Díaz-Martínez
- Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C., Guadalajara, Jalisco, MX
| |
Collapse
|
138
|
Soria B, Gauthier BR, Martín F, Tejedo JR, Bedoya FJ, Rojas A, Hmadcha A. Using stem cells to produce insulin. Expert Opin Biol Ther 2015; 15:1469-89. [PMID: 26156425 DOI: 10.1517/14712598.2015.1066330] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Tremendous progress has been made in generating insulin-producing cells from pluripotent stem cells. The best outcome of the refined protocols became apparent in the first clinical trial announced by ViaCyte, based on the implantation of pancreatic progenitors that would further mature into functional insulin-producing cells inside the patient's body. AREAS COVERED Several groups, including ours, have contributed to improve strategies to generate insulin-producing cells. Of note, the latest results have gained a substantial amount of interest as a method to create a potentially functional and limitless supply of β-cell to revert diabetes mellitus. This review analyzes the accomplishments that have taken place over the last few decades, summarizes the state-of-art methods for β-cell replacement therapies based on the differentiation of embryonic stem cells into glucose-responsive and insulin-producing cells in a dish and discusses alternative approaches to obtain new sources of insulin-producing cells. EXPERT OPINION Undoubtedly, recent events preface the beginning of a new era in diabetes therapy. However, in our opinion, a number of significant hurdles still stand in the way of clinical application. We believe that the combination of the private and public sectors will accelerate the process of obtaining the desired safe and functional β-cell surrogates.
Collapse
Affiliation(s)
- Bernat Soria
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Benoit R Gauthier
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ;
| | - Franz Martín
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Juan R Tejedo
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Francisco J Bedoya
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Anabel Rojas
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| | - Abdelkrim Hmadcha
- a 1 CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine , Avda. Americo Vespucio s/n, 41092 Seville, Spain ; .,b 2 CIBERDEM, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders , 08036 Barcelona, Spain
| |
Collapse
|
139
|
Dadheech N, Srivastava A, Paranjape N, Gupta S, Dave A, Shah GM, Bhonde RR, Gupta S. Swertisin an Anti-Diabetic Compound Facilitate Islet Neogenesis from Pancreatic Stem/Progenitor Cells via p-38 MAP Kinase-SMAD Pathway: An In-Vitro and In-Vivo Study. PLoS One 2015; 10:e0128244. [PMID: 26047129 PMCID: PMC4457488 DOI: 10.1371/journal.pone.0128244] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/23/2015] [Indexed: 12/12/2022] Open
Abstract
Transplanting islets serves best option for restoring lost beta cell mass and function. Small bio-chemical agents do have the potential to generate new islets mass, however lack of understanding about mechanistic action of these small molecules eventually restricts their use in cell-based therapies for diabetes. We recently reported "Swertisin" as a novel islet differentiation inducer, generating new beta cells mass more effectively. Henceforth, in the present study we attempted to investigate the molecular signals that Swertisin generate for promoting differentiation of pancreatic progenitors into islet cells. To begin with, both human pancreatic progenitors (PANC-1 cells) and primary cultured mouse intra-islet progenitor cells (mIPC) were used and tested for Swertisin induced islet neogenesis mechanism, by monitoring immunoblot profile of key transcription factors in time dependent manner. We observed Swertisin follow Activin-A mediated MEPK-TKK pathway involving role of p38 MAPK via activating Neurogenin-3 (Ngn-3) and Smad Proteins cascade. This MAP Kinase intervention in differentiation of cells was confirmed using strong pharmacological inhibitor of p38 MAPK (SB203580), which effectively abrogated this process. We further confirmed this mechanism in-vivo in partial pancreatectomised (PPx) mice model, where we could show Swertisin exerted potential increase in insulin transcript levels with persistent down-regulation of progenitor markers like Nestin, Ngn-3 and Pancreatic Duodenal Homeobox Gene-1 (PDX-1) expression, within three days post PPx. With detailed molecular investigations here in, we first time report the molecular mode of action of Swertisin for islet neogenesis mediated through MAP Kinase (MEPK-TKK) pathway involving Ngn-3 and Smad transcriptional regulation. These findings held importance for developing Swertisin as potent pharmacological drug candidate for effective and endogenous differentiation of islets in cell based therapy for diabetes.
Collapse
Affiliation(s)
- Nidheesh Dadheech
- Molecular Endocrinology and Stem Cell Research Lab, Department of Biochemistry, Faculty of Science, The M S University of Baroda, Vadodara, Gujarat, India
| | - Abhay Srivastava
- Molecular Endocrinology and Stem Cell Research Lab, Department of Biochemistry, Faculty of Science, The M S University of Baroda, Vadodara, Gujarat, India
| | - Neha Paranjape
- Hislope College of Biotechnology, Nagpur, Maharashtra, India
| | - Shivika Gupta
- Hislope College of Biotechnology, Nagpur, Maharashtra, India
| | - Arpita Dave
- Molecular Endocrinology and Stem Cell Research Lab, Department of Biochemistry, Faculty of Science, The M S University of Baroda, Vadodara, Gujarat, India
| | - Girish M. Shah
- Skin Cancer Research Laboratory, Centre de Recherche du CHUL, CHUQ, Univerisity Laval, Quebec City, Quebec, Canada
| | - Ramesh R. Bhonde
- Manipal Hospital and Regenerative Medicine Centre, Manipal Hospital, Manipal, Karnataka, India
| | - Sarita Gupta
- Molecular Endocrinology and Stem Cell Research Lab, Department of Biochemistry, Faculty of Science, The M S University of Baroda, Vadodara, Gujarat, India
| |
Collapse
|
140
|
Moshrefi M, Yari N, Nabipour F, Bazrafshani MR, Nematollahi-mahani SN. Transplantation of differentiated umbilical cord mesenchymal cells under kidney capsule for control of type I diabetes in rat. Tissue Cell 2015; 47:395-405. [PMID: 26025422 DOI: 10.1016/j.tice.2015.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 04/24/2015] [Accepted: 04/24/2015] [Indexed: 12/29/2022]
Abstract
Nowadays, stem cells have been introduced as an appropriate source of regenerative medicine for treatment of type I diabetes. Human umbilical cord matrix-derived mesenchymal cells (hUCMC) have successfully been differentiated into insulin producing cells. The isolated hUCM cells were characterized by the expression of stem cell surface markers and by differentiation into adipocytes and osteocytes. The hUCMCs were cultured with different concentrations of neural conditional medium (NCM) and were induced to differentiate into insulin producing cells (IPCs). As 60% NCM concentration resulted in higher nestin and PDX1 expression, the cells were first exposed to 60% NCM and were then induced for IPCs differentiation. PDX1 and insulin gene expression was evaluated in the treated cells. Also, the secretion capacity of the IPCs was assessed by glucose challenge test. IPCs were transferred under the rat kidney capsule. Blood glucose level, weight gain and immunohistochemistry assessments were done in the treated animals. hUCMC expressed mesenchymal cell surface markers and successfully differentiated into adipocytes and osteocytes. Higher NCM concentration resulted in higher PDX1 and nestin expression. The IPCs expressed insulin and PDX1. IPCs were detectable under the kidney capsule 2 months after injection. IPCs transplantation resulted in a sharp decline of blood sugar level and less weight loss. Differentiated hUCM cells could alleviate the insulin deprivation in the rat model of type I diabetes. In addition, higher NCM concentration leads to more differentiation into IPCs and more nestin and PDX1 expression. Kidney capsule can serve as a suitable nominee for IPCs transplantation.
Collapse
Affiliation(s)
- Mojgan Moshrefi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Afzal Research Institute (NGO), Kerman, Iran
| | - Nahid Yari
- Department of Reproductive Biology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Nabipour
- Department of Pathology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Reza Bazrafshani
- Department of Medical Genetics, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed Noureddin Nematollahi-mahani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Afzal Research Institute (NGO), Kerman, Iran.
| |
Collapse
|
141
|
Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells into Insulin-Producing Cells: Evidence for Further Maturation In Vivo. BIOMED RESEARCH INTERNATIONAL 2015; 2015:575837. [PMID: 26064925 PMCID: PMC4443784 DOI: 10.1155/2015/575837] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/11/2015] [Indexed: 12/24/2022]
Abstract
The aim of this study was to provide evidence for further in vivo maturation of insulin-producing cells (IPCs) derived from human bone marrow-derived mesenchymal stem cells (HBM-MSCs). HBM-MSCs were obtained from three insulin-dependent type 2 diabetic volunteers. Following expansion, cells were differentiated according to a trichostatin-A/GLP protocol. One million cells were transplanted under the renal capsule of 29 diabetic nude mice. Blood glucose, serum human insulin and c-peptide levels, and glucose tolerance curves were determined. Mice were euthanized 1, 2, 4, or 12 weeks after transplantation. IPC-bearing kidneys were immunolabeled, number of IPCs was counted, and expression of relevant genes was determined. At the end of in vitro differentiation, all pancreatic endocrine genes were expressed, albeit at very low values. The percentage of IPCs among transplanted cells was small (≤3%). Diabetic animals became euglycemic 8 ± 3 days after transplantation. Thereafter, the percentage of IPCs reached a mean of ~18% at 4 weeks. Relative gene expression of insulin, glucagon, and somatostatin showed a parallel increase. The ability of the transplanted cells to induce euglycemia was due to their further maturation in the favorable in vivo microenvironment. Elucidation of the exact mechanism(s) involved requires further investigation.
Collapse
|
142
|
Duan FF, Liu JH, March JC. Engineered commensal bacteria reprogram intestinal cells into glucose-responsive insulin-secreting cells for the treatment of diabetes. Diabetes 2015; 64:1794-803. [PMID: 25626737 PMCID: PMC4407861 DOI: 10.2337/db14-0635] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 12/10/2014] [Indexed: 12/25/2022]
Abstract
The inactive full-length form of GLP-1(1-37) stimulates conversion of both rat and human intestinal epithelial cells into insulin-secreting cells. We investigated whether oral administration of human commensal bacteria engineered to secrete GLP-1(1-37) could ameliorate hyperglycemia in a rat model of diabetes by reprogramming intestinal cells into glucose-responsive insulin-secreting cells. Diabetic rats were fed daily with human lactobacilli engineered to secrete GLP-1(1-37). Diabetic rats fed GLP-1-secreting bacteria showed significant increases in insulin levels and, additionally, were significantly more glucose tolerant than those fed the parent bacterial strain. These rats developed insulin-producing cells within the upper intestine in numbers sufficient to replace ∼25-33% of the insulin capacity of nondiabetic healthy rats. Intestinal tissues in rats with reprogrammed cells expressed MafA, PDX-1, and FoxA2. HNF-6 expression was observed only in crypt epithelia expressing insulin and not in epithelia located higher on the villous axis. Staining for other cell markers in rats treated with GLP-1(1-37)-secreting bacteria suggested that normal function was not inhibited by the close physical proximity of reprogrammed cells. These results provide evidence of the potential for a safe and effective nonabsorbed oral treatment for diabetes and support the concept of engineered commensal bacterial signaling to mediate enteric cell function in vivo.
Collapse
Affiliation(s)
- Franklin F Duan
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY
| | - Joy H Liu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY
| | - John C March
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY
| |
Collapse
|
143
|
Li L, Li T, Zhang Y, Pan Z, Wu B, Huang X, Zhang Y, Mei Y, Ge L, Shen G, Ge RS, Zhu D, Lou Y. Peroxisome proliferator-activated receptorβ/δ activation is essential for modulating p-Foxo1/Foxo1 status in functional insulin-positive cell differentiation. Cell Death Dis 2015; 6:e1715. [PMID: 25855963 PMCID: PMC4650555 DOI: 10.1038/cddis.2015.88] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/26/2015] [Accepted: 03/02/2015] [Indexed: 11/25/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) participate in energy homeostasis and play essential roles in diabetes therapy through their effects on non-pancreas tissues. Pathological microenvironment may influence the metabolic requirements for the maintenance of stem cell differentiation. Accordingly, understanding the mechanisms of PPARs on pancreatic β-cell differentiation may be helpful to find the underlying targets of disrupted energy homeostasis under the pancreatic disease condition. PPARs are involved in stem cell differentiation via mitochondrial oxidative phosphorylation, but the subtype member activation and the downstream regulation in functional insulin-positive (INS+) cell differentiation remain unclear. Here, we show a novel role of PPARβ/δ activation in determining INS+ cell differentiation and functional maturation. We found PPARβ/δ expression selectively upregulated in mouse embryonic pancreases or stem cells-derived INS+ cells at the pancreatic mature stage in vivo and in vitro. Strikingly, given the inefficiency of generating INS+ cells in vitro, PPARβ/δ activation displayed increasing mouse and human ES cell-derived INS+ cell numbers and insulin secretion. This phenomenon was closely associated with the forkhead box protein O1 (Foxo1) nuclear shuttling, which was dependent on PPARβ/δ downstream PI3K/Akt signaling transduction. The present study reveals the essential role of PPARβ/δ activation on p-Foxo1/Foxo1 status, and in turn, determining INS+ cell generation and insulin secretion via affecting pancreatic and duodenal homeobox-1 expression. The results demonstrate the underlying mechanism by which PPARβ/δ activation promotes functional INS+ cell differentiation. It also provides potential targets for anti-diabetes drug discovery and hopeful clinical applications in human cell therapy.
Collapse
Affiliation(s)
- L Li
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - T Li
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Y Zhang
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Z Pan
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - B Wu
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - X Huang
- Cardiovascular Key Laboratory of Zhejiang Province, The 2nd Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Y Zhang
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Y Mei
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - L Ge
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - G Shen
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - R-s Ge
- 1] The Population Council at the Rockefeller University, New York, NY 10021, USA [2] Institute of Reproductive Biomedicine, the 2nd Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - D Zhu
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Y Lou
- Institute of Pharmacology, Toxicology and Biochemical Pharmaceutics, Key Innovation Team for Stem Cell Translational Medicine of Cardiovascular Disease of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
144
|
Yu F, Li Y, Liu CS, Chen Q, Wang GH, Guo W, Wu XE, Li DH, Wu WD, Chen XD. Enteric-coated capsules filled with mono-disperse micro-particles containing PLGA-lipid-PEG nanoparticles for oral delivery of insulin. Int J Pharm 2015; 484:181-91. [DOI: 10.1016/j.ijpharm.2015.02.055] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/16/2015] [Accepted: 02/23/2015] [Indexed: 01/09/2023]
|
145
|
Khorsandi L, Khodadadi A, Nejad-Dehbashi F, Saremy S. Three-dimensional differentiation of adipose-derived mesenchymal stem cells into insulin-producing cells. Cell Tissue Res 2015; 361:745-53. [PMID: 25795142 DOI: 10.1007/s00441-015-2140-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 01/28/2015] [Indexed: 01/17/2023]
Abstract
The aim of this study is to evaluate the collagen/hyaluronic acid (Col/HA) scaffold effect on the differentiation of insulin-producing cells (IPCs) from adipose-derived mesenchymal stem cells (ASCs). In this experimental study, ASCs were cultured and seeded in a Col/HA scaffold (3D culture) and then treated with induction media. After induction, the presence of IPCs was evaluated using gene expression (PDX-1, GLUT-2 and insulin) analysis and immunocytochemistry, while functional maturity was determined by measuring insulin release in response to low- and high-glucose media. The induced IPCs were morphologically similar to pancreatic islet-like cells. Expression of the islet-associated genes PDX-1, GLUT-2 and insulin genes in 3D-cultured cells was markedly higher than the 2D-cultured cells exposure differentiation media. Compared to the 2D culture of ASCs-derived IPCs, the insulin release from 3D ASCs-derived IPCs showed a nearly 4-fold (p < 0.05) increase when exposed to a high glucose (25 mmol) medium. The percentage of insulin-positive cells in the 3D experimental group showed an approximately 4-fold increase compared to the 2D experimental culture cells. The results of this study demonstrated that the COL/HA scaffold can enhance the differentiation of IPCs from rat ASCs.
Collapse
Affiliation(s)
- Layasadat Khorsandi
- Cell & Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran, P.O. Box: 61335,
| | | | | | | |
Collapse
|
146
|
Sinagoga KL, Wells JM. Generating human intestinal tissues from pluripotent stem cells to study development and disease. EMBO J 2015; 34:1149-63. [PMID: 25792515 DOI: 10.15252/embj.201490686] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/09/2015] [Indexed: 01/05/2023] Open
Abstract
As one of the largest and most functionally complex organs of the human body, the intestines are primarily responsible for the breakdown and uptake of macromolecules from the lumen and the subsequent excretion of waste from the body. However, the intestine is also an endocrine organ, regulating digestion, metabolism, and feeding behavior. Intricate neuronal, lymphatic, immune, and vascular systems are integrated into the intestine and are required for its digestive and endocrine functions. In addition, the gut houses an extensive population of microbes that play roles in digestion, global metabolism, barrier function, and host-parasite interactions. With such an extensive array of cell types working and performing in one essential organ, derivation of functional intestinal tissues from human pluripotent stem cells (PSCs) represents a significant challenge. Here we will discuss the intricate developmental processes and cell types that are required for assembly of this highly complex organ and how embryonic processes, particularly morphogenesis, have been harnessed to direct differentiation of PSCs into 3-dimensional human intestinal organoids (HIOs) in vitro. We will further describe current uses of HIOs in development and disease research and how additional tissue complexity might be engineered into HIOs for better functionality and disease modeling.
Collapse
Affiliation(s)
- Katie L Sinagoga
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
147
|
Saito M, Kaneda A, Shigeto H, Hanata N, Otokuni K, Matsuoka H. Development of an optimized 5-stage protocol for the in vitro preparation of insulin-secreting cells from mouse ES cells. Cytotechnology 2015; 68:987-98. [PMID: 25749915 DOI: 10.1007/s10616-015-9853-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/04/2015] [Indexed: 12/12/2022] Open
Abstract
In order to produce insulin-secreting cells with a high value of glucose-stimulated insulin secretion (GSIS) from mouse embryonic stem cells, we have developed an optimized 5-stage protocol by referring to culture conditions so far reported elsewhere. This protocol is characterized by 4 points: (1) use of an activin-free medium in the first stage, (2) use of gelatin/fibronectin coated culture dishes in 1-4 stages throughout, (3) removal of undifferentiated cells by cell sorter at the end of 4th stage, and (4) sedimental culture in the 5th stage. GSIS value of the produced cells reached 2.4, that was at a higher rank of those so far reported. The produced cells were transplanted in diabetes model mice but no remedy effect was observed. Then transplantation was conducted in pre-diabetes model mice, in which GSIS was impaired without affecting insulin producing function. The transplantation of 5 × 10(6) cells resulted in a marked improvement of glucose tolerance within 20 days. This effect decreased but was still observed at 120 days post-transplantation. This demonstrates the feasibility of the novel optimized protocol.
Collapse
Affiliation(s)
- Mikako Saito
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| | - Asako Kaneda
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Hajime Shigeto
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Nobuaki Hanata
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Keiko Otokuni
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Hideaki Matsuoka
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan
| |
Collapse
|
148
|
Tsuchiya M, Misaka R, Nitta K, Tsuchiya K. Transcriptional factors, Mafs and their biological roles. World J Diabetes 2015; 6:175-183. [PMID: 25685288 PMCID: PMC4317310 DOI: 10.4239/wjd.v6.i1.175] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/17/2014] [Accepted: 12/10/2014] [Indexed: 02/05/2023] Open
Abstract
The Maf family of transcription factors is characterized by a typical bZip structure; these transcription factors act as important regulators of the development and differentiation of many organs and tissues, including the kidney. The Maf family consists of two subgroups that are characterized according to their structure: large Maf transcription factors and small Maf transcription factors. The large Maf subgroup consists of four proteins, designated as MAFA, MAFB, c-MAF and neural retina-specific leucine zipper. In particular, MAFA is a distinct molecule that has been attracting the attention of researchers because it acts as a strong transactivator of insulin, suggesting that Maf transcription factors are likely to be involved in systemic energy homeostasis. In this review, we focused on the regulation of glucose/energy balance by Maf transcription factors in various organs.
Collapse
|
149
|
Differentiation of human skin-derived precursor cells into functional islet-like insulin-producing cell clusters. In Vitro Cell Dev Biol Anim 2015; 51:595-603. [DOI: 10.1007/s11626-015-9866-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/01/2015] [Indexed: 01/09/2023]
|
150
|
Abdelalim EM, Emara MM. Advances and challenges in the differentiation of pluripotent stem cells into pancreatic β cells. World J Stem Cells 2015; 7:174-181. [PMID: 25621117 PMCID: PMC4300928 DOI: 10.4252/wjsc.v7.i1.174] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/16/2014] [Accepted: 09/19/2014] [Indexed: 02/06/2023] Open
Abstract
Pluripotent stem cells (PSCs) are able to differentiate into several cell types, including pancreatic β cells. Differentiation of pancreatic β cells depends on certain transcription factors, which function in a coordinated way during pancreas development. The existing protocols for in vitro differentiation produce pancreatic β cells, which are not highly responsive to glucose stimulation except after their transplantation into immune-compromised mice and allowing several weeks for further differentiation to ensure the maturation of these cells in vivo. Thus, although the substantial improvement that has been made for the differentiation of induced PSCs and embryonic stem cells toward pancreatic β cells, several challenges still hindering their full generation. Here, we summarize recent advances in the differentiation of PSCs into pancreatic β cells and discuss the challenges facing their differentiation as well as the different applications of these potential PSC-derived β cells.
Collapse
|