1
|
Palihaderu PADS, Mendis BILM, Premarathne JMKJK, Dias WKRR, Yeap SK, Ho WY, Dissanayake AS, Rajapakse IH, Karunanayake P, Senarath U, Satharasinghe DA. Therapeutic Potential of miRNAs for Type 2 Diabetes Mellitus: An Overview. Epigenet Insights 2022; 15:25168657221130041. [PMID: 36262691 PMCID: PMC9575458 DOI: 10.1177/25168657221130041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/14/2022] [Indexed: 11/05/2022] Open
Abstract
MicroRNA(miRNA)s have been identified as an emerging class for therapeutic
interventions mainly due to their extracellularly stable presence in humans and
animals and their potential for horizontal transmission and action. However,
treating Type 2 diabetes mellitus using this technology has yet been in a
nascent state. MiRNAs play a significant role in the pathogenesis of Type 2
diabetes mellitus establishing the potential for utilizing miRNA-based
therapeutic interventions to treat the disease. Recently, the administration of
miRNA mimics or antimiRs in-vivo has resulted in positive modulation of glucose
and lipid metabolism. Further, several cell culture-based interventions have
suggested beta cell regeneration potential in miRNAs. Nevertheless, few such
miRNA-based therapeutic approaches have reached the clinical phase. Therefore,
future research contributions would identify the possibility of miRNA
therapeutics for tackling T2DM. This article briefly reported recent
developments on miRNA-based therapeutics for treating Type 2 Diabetes mellitus,
associated implications, gaps, and recommendations for future studies.
Collapse
Affiliation(s)
- PADS Palihaderu
- Department of Basic Veterinary
Sciences, Faculty of Veterinary Medicine and Animal Science, University of
Peradeniya, Peradeniya, Sri Lanka
| | - BILM Mendis
- Department of Basic Veterinary
Sciences, Faculty of Veterinary Medicine and Animal Science, University of
Peradeniya, Peradeniya, Sri Lanka
| | - JMKJK Premarathne
- Department of Livestock and Avian
Sciences, Faculty of Livestock, Fisheries, and Nutrition, Wayamba University of Sri
Lanka, Makandura, Gonawila (NWP), Sri Lanka
| | - WKRR Dias
- Department of North Indian Music,
Faculty of Music, University of the Visual and Performing Arts, Colombo, Sri
Lanka
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences,
Xiamen University Malaysia Campus, Jalan Sunsuria, Bandar Sunsuria, Sepang,
Selangor, Malaysia
| | - Wan Yong Ho
- Division of Biomedical Sciences,
Faculty of Medicine and Health Sciences, University of Nottingham (Malaysia Campus),
Semenyih, Malaysia
| | - AS Dissanayake
- Department of Clinical Medicine,
Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | - IH Rajapakse
- Department of Psychiatry, Faculty of
Medicine, University of Ruhuna, Galle, Sri Lanka
| | - P Karunanayake
- Department of Clinical Medicine,
Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - U Senarath
- Department of Community Medicine,
Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - DA Satharasinghe
- Department of Basic Veterinary
Sciences, Faculty of Veterinary Medicine and Animal Science, University of
Peradeniya, Peradeniya, Sri Lanka,DA Satharasinghe, Department of Basic
Veterinary Sciences, Faculty of Veterinary Medicine and Animal Science,
University of Peradeniya, Peradeniya, 20400, Sri Lanka.
| |
Collapse
|
2
|
Differentiation of multipotent stem cells to insulin-producing cells for treatment of diabetes mellitus: bone marrow- and adipose tissue-derived cells comparison. Mol Biol Rep 2022; 49:3539-3548. [PMID: 35107740 DOI: 10.1007/s11033-022-07194-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 01/25/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) from human adipose tissue and bone marrow have a great potential for use in cell therapy due to their ease of isolation, expansion, and differentiation. Our intention was to isolate and promote in vitro expansion and differentiation of MSCs from human adipose and bone marrow tissue into cells with a pancreatic endocrine phenotype and to compare the potency of these cells together. METHODS AND RESULTS MSCs were pre-induced with nicotinamide, mercaptoethanol, B-27 and b-FGF in L-DMEM for 2 days and re-induced again in supplemented H-DMEM for another 3 days. Expression of five genes in differentiated beta cells was evaluated by Real-time PCR and western blotting and the potency of insulin release in response to glucose stimulation was evaluated by insulin and C-peptide ELISA kit. The differentiated cells were evaluated by immunocytochemistry staining for Insulin and PDX-1. Quantitative RT-PCR results showed up-regulation of four genes in differentiated beta-islet cells (Insulin, Ngn-3, Pax-4 and Pdx-1) compared with the control. Western blot analysis showed that MSCs cells mainly produced proinsulin and insulin after differentiation but nestin was more expressed in pre-differentiated stem cells. Glucose and insulin secretion assay showed that insulin levels and C-peptide secretion were significantly increased in response to 10 mM glucose. CONCLUSIONS Our study showed that both adipose and bone marrow stem cells could differentiate into functional beta-islet cells but it seems that adipose stem cells could be a better choice for treatment of diabetes mellitus according to their higher potency.
Collapse
|
3
|
Cai Z, Liu F, Yang Y, Li D, Hu S, Song L, Yu S, Li T, Liu B, Luo H, Zhang W, Zhou Z, Zhang J. GRB10 regulates β cell mass by inhibiting β cell proliferation and stimulating β cell dedifferentiation. J Genet Genomics 2021; 49:208-216. [PMID: 34861413 DOI: 10.1016/j.jgg.2021.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/07/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022]
Abstract
Decreased functional β-cell mass is the hallmark of diabetes, but the cause of this metabolic defect remains elusive. Here, we show that the expression levels of the growth factor receptor-bound protein 10 (GRB10), a negative regulator of insulin and mTORC1 signaling, are markedly induced in islets of diabetic mice and high glucose-treated insulinoma cell line INS-1cells. β-cell-specific knockout of Grb10 in mice increased β-cell mass and improved β-cell function. Grb10-deficient β-cells exhibit enhanced mTORC1 signaling and reduced β-cell dedifferentiation, which could be blocked by rapamycin. On the contrary, Grb10 overexpression induced β-cell dedifferentiation in MIN6 cells. Our study identifies GRB10 as a critical regulator of β-cell dedifferentiation and β-cell mass, which exerts its effect by inhibiting mTORC1 signaling.
Collapse
Affiliation(s)
- Zixin Cai
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Fen Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yan Yang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Dandan Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Shanbiao Hu
- Department of Urological Organ Transplantation, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Lei Song
- Department of Urological Organ Transplantation, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Shaojie Yu
- Department of Urological Organ Transplantation, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ting Li
- Department of Liver Organ Transplantation, the Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Bilian Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Hairong Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Weiping Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai 200433, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
4
|
Bai C, Gao Y, Zhang X, Yang W, Guan W. Melatonin promotes self-renewal of nestin-positive pancreatic stem cells through activation of the MT2/ERK/SMAD/nestin axis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:62-74. [PMID: 29037070 DOI: 10.1080/21691401.2017.1389747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Although melatonin has been shown to exhibit a wide variety of biological functions, its effects on promotion of self-renewal in pancreatic stem cells remain unknown. In this study, we incubated murine pancreatic stem cells (PSCs) with various concentrations of melatonin (0.01, 0.1, 1, 10 or 100 μM) to screen for the optimum culture medium for increasing cell proliferation. We found that 10 μM melatonin can significantly increase proliferation and enhance expression of a stem cell marker, nestin, in PSCs via melatonin receptor 2 (MT2). Thus, we used 10 μM melatonin to study the melatonin-mediated molecular mechanisms of cell proliferation in PSCs. We applied extracellular signal-regulated kinase (ERK) pathway inhibitor SCH772984 and transforming growth factor beta (TGF-β) pathway inhibitor SB431542, along with interfering RNAs siERK1, siERK2, siSmad2, siSmad3, siSmad4 and siNestin, to melatonin-treated PSCs to research the roles of these genes in self-renewal. The results revealed a novel molecular mechanism by which melatonin promotes self-renewal of PSCs: a chain reaction in the MT2/ERK/SMAD/nestin axis promoted the aforementioned self-renewal as well as inhibited differentiation. In addition, upregulation of nestin created a positive feedback loop in the regulation of the transforming growth factor beta 1 (TGF-β1)/SMADs pathway by promoting expression of Smad4. Conversely, knockdown of nestin significantly suppressed the proliferative effect in melatonin-treated PSCs. These are all novel mechanisms through which the ERK pathway cooperatively crosstalks with the SMAD pathway to regulate nestin expression, thereby enhancing self-renewal in PSCs.
Collapse
Affiliation(s)
- Chunyu Bai
- a Key Laboratory of Precision Oncology of Shandong Higher Education , Institute of precision medicine , Jining , Shandong Province , P. R. China.,b Institute of Animal Sciences , Chinese Academy of Agricultural Sciences , Beijing , P. R. China
| | - Yuhua Gao
- b Institute of Animal Sciences , Chinese Academy of Agricultural Sciences , Beijing , P. R. China.,c College of Basic Medicine , Jining Medical University , Jining , Shandong Province , P. R. China
| | - Xiangyang Zhang
- c College of Basic Medicine , Jining Medical University , Jining , Shandong Province , P. R. China
| | - Wancai Yang
- a Key Laboratory of Precision Oncology of Shandong Higher Education , Institute of precision medicine , Jining , Shandong Province , P. R. China.,d Department of Pathology , University of Illinois at Chicago , Chicago , IL , USA
| | - Weijun Guan
- b Institute of Animal Sciences , Chinese Academy of Agricultural Sciences , Beijing , P. R. China
| |
Collapse
|
5
|
Bai C, Gao Y, Zhang X, Yang W, Guan W. MicroRNA-34c acts as a bidirectional switch in the maturation of insulin-producing cells derived from mesenchymal stem cells. Oncotarget 2017; 8:106844-106857. [PMID: 29290993 PMCID: PMC5739778 DOI: 10.18632/oncotarget.21883] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/21/2017] [Indexed: 01/08/2023] Open
Abstract
miRNAs regulate insulin secretion, pancreatic development, and beta-cell differentiation. However, their function in the differentiation of IPCs from MSCs is poorly understood. In this study, to screen for miRNAs and their targets that function during the formation of IPCs from MSCs, we examined the miRNA expression profiles of MSCs and IPCs using RNA-seq and qPCR to confirm the above results. We found that miR-34c exhibited transient upregulation at an early stage of the formation of IPCs derived from MSCs. Next, we analyzed the biological function of miR-34c by predicting its targets using bioinformatic tools. Combining our data with those from previous reports, we found that miR-34c and its targets play an important role in the formation of IPCs. Therefore, we overexpressed miR-34c and expressed small interfering RNAs of its targets in MSCs to investigate their functions in IPC formation. We found that miR-34c acts as a bidirectional switch in the formation of IPCs derived from MSCs by regulating the expression of targets to affect insulin synthesis and secretion. miR-34c was shown to downregulate its targets, including PDE7B, PDGFRA, and MAP2K1, to increase proinsulin synthesis, but when miR-34c continually dysregulated such expression, it suppressed the expression of other targets, namely ACSL4 and SAR1A, weakening insulin secretion in IPCs. These results suggest that endogenous miRNAs involved in the formation of IPCs from stem cells should be considered in the development of effective cell transplant therapy for diabetes.
Collapse
Affiliation(s)
- Chunyu Bai
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, 272067, PR China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Yuhua Gao
- College of Basic Medicine, Jining Medical University, Jining, 272067, PR China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Xiangyang Zhang
- College of Basic Medicine, Jining Medical University, Jining, 272067, PR China
| | - Wancai Yang
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, 272067, PR China.,Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Weijun Guan
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| |
Collapse
|
6
|
Bai C, Gao Y, Li X, Wang K, Xiong H, Shan Z, Zhang P, Wang W, Guan W, Ma Y. MicroRNAs can effectively induce formation of insulin-producing cells from mesenchymal stem cells. J Tissue Eng Regen Med 2017; 11:3457-3468. [DOI: 10.1002/term.2259] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 03/28/2016] [Accepted: 07/03/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Chunyu Bai
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Yuhua Gao
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Xiangchen Li
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Kunfu Wang
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Hui Xiong
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Zhiqiang Shan
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Ping Zhang
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Wenjie Wang
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Weijun Guan
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| | - Yuehui Ma
- Institute of Animal Science; Chinese Academy of Agricultural Sciences; Beijing 100193 China
| |
Collapse
|
7
|
Role of microRNA-21 in the formation of insulin-producing cells from pancreatic progenitor cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:280-93. [DOI: 10.1016/j.bbagrm.2015.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/17/2015] [Accepted: 12/02/2015] [Indexed: 12/20/2022]
|
8
|
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
|
9
|
Thakkar UG, Trivedi HL, Vanikar AV, Dave SD. Insulin-secreting adipose-derived mesenchymal stromal cells with bone marrow-derived hematopoietic stem cells from autologous and allogenic sources for type 1 diabetes mellitus. Cytotherapy 2015; 17:940-7. [PMID: 25869301 DOI: 10.1016/j.jcyt.2015.03.608] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/24/2015] [Indexed: 01/24/2023]
Abstract
BACKGROUND AIMS Stem cell therapy (SCT) is now the up-coming therapeutic modality for treatment of type 1 diabetes mellitus (T1DM). METHODS Our study was a prospective, open-labeled, two-armed trial for 10 T1DM patients in each arm of allogenic and autologous adipose-derived insulin-secreting mesenchymal stromal cells (IS-AD-MSC)+bone marrow-derived hematopoietic stem cell (BM-HSC) infusion. Group 1 received autologous SCT: nine male patients and one female patient; mean age, 20.2 years, disease duration 8.1 years; group 2 received allogenic SCT: six male patients and four female patients, mean age, 19.7 years and disease duration, 7.9 years. Glycosylated hemoglobin (HbA1c) was 10.99%; serum (S.) C-peptide, 0.22 ng/mL and insulin requirement, 63.9 IU/day in group 1; HbA1c was 11.93%, S.C-peptide, 0.028 ng/mL and insulin requirement, 57.55 IU/day in group 2. SCs were infused into the portal+thymic circulation and subcutaneous tissue under non-myelo-ablative conditioning. Patients were monitored for blood sugar, S.C-peptide, glutamic acid decarboxylase antibodies and HbA1c at 3-month intervals. RESULTS Group 1 received mean SCs 103.14 mL with 2.65 ± 0.8 × 10(4) ISCs/kg body wt, CD34+ 0.81% and CD45-/90+/73+, 81.55%. Group 2 received mean SCs 95.33 mL with 2.07 ± 0.67 × 10(4) ISCs/kg body wt, CD34+ 0.32% and CD45-/90+/73+ 54.04%. No untoward effect was observed with sustained improvement in HbA1c and S.C-peptide in both groups with a decrease in glutamic acid decarboxylase antibodies and reduction in mean insulin requirement. CONCLUSIONS SCT is a safe and viable treatment option for T1DM. Autologous IS-AD-MSC+ BM-HSC co-infusion offers better long-term control of hyperglycemia as compared with allogenic SCT.
Collapse
Affiliation(s)
- 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.
| | - 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
| | - 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
| | - Shruti D Dave
- 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
| |
Collapse
|
10
|
Talebi S, Aleyasin A, Soleimani M, Massumi M. Derivation of islet-like cells from mesenchymal stem cells using PDX1-transducing lentiviruses. Biotechnol Appl Biochem 2012; 59:205-12. [DOI: 10.1002/bab.1013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Accepted: 02/17/2012] [Indexed: 12/18/2022]
|
11
|
Ma F, Chen F, Chi Y, Yang S, Lu S, Han Z. Isolation of pancreatic progenitor cells with the surface marker of hematopoietic stem cells. Int J Endocrinol 2012; 2012:948683. [PMID: 23316230 PMCID: PMC3536067 DOI: 10.1155/2012/948683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 11/27/2012] [Accepted: 11/29/2012] [Indexed: 11/17/2022] Open
Abstract
To isolate pancreatic progenitor cells with the surface markers of hematopoietic stem cells, the expression of stem cell antigen (Sca-1) and c-Kit and the coexpression of them with pancreatic duodenal homeobox-1 (PDX-1), neurogenin 3 (Ngn3), and insulin were examined in murine embryonic pancreas. Then different pancreatic cell subpopulations were isolated by magnet-activated cell sorting. Isolated cells were cultured overnight in hanging drops. When cells formed spheres, they were laid on floating filters at the air/medium interface. With this new culture system, pancreatic progenitor cells were induced to differentiate to endocrine and exocrine cells. It was shown that c-Kit and Sca-1 were expressed differently in embryonic pancreas at 12.5, 15.5, and 17.5 days of gestation. The expression of c-Kit and Sca-1 was the highest at 15.5 days of gestation. c-Kit rather than Sca-1 coexpressed with PDX-1, Ngn3, and insulin. Cells differentiated from c-Kit-positive cells contained more insulin-producing cells and secreted more insulin in response to glucose stimulation than that from c-Kit-negative cells. These results suggested that c-Kit could be used to isolate pancreatic progenitor cells and our new culture system permitted pancreatic progenitor cells to differentiate to mature endocrine cells.
Collapse
|
12
|
Jones MB, Chu CH, Pendleton JC, Betenbaugh MJ, Shiloach J, Baljinnyam B, Rubin JS, Shamblott MJ. Proliferation and pluripotency of human embryonic stem cells maintained on type I collagen. Stem Cells Dev 2010; 19:1923-35. [PMID: 20367282 DOI: 10.1089/scd.2009.0326] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Human embryonic stem cells (hESC) require a balance of growth factors and signaling molecules to proliferate and retain pluripotency. Conditioned medium (CM) from a human embryonic germ-cell-derived cell culture, SDEC, was observed to support the growth of hESC on type I collagen (COL I) and on Matrigel (MAT) biomatricies. After 1 month, the population doubling of hESC grown in SDEC CM on COL I was equivalent to that of hESC grown in mouse embryonic fibroblast (MEF) CM on MAT. hESC grown in SDEC CM on COL I expressed OCT4, NANOG, SSEA-4, alkaline phosphatase (AP), and TRA-1-60; retained a normal karyotype; and were capable of forming teratomas. DNA microarray analysis was used to compare the transcriptional profiles of SDEC and the less supportive WI38 and Detroit 551 human cell lines. The mRNA level of secreted frizzled-related protein (sFRP-1), a known antagonist of the WNT/β-catenin signaling pathway, was significantly reduced in SDEC as compared with the other 2 cell lines, whereas the mRNA levels of prostaglandin-endoperoxide synthase 2 (PTGS2 or COX-2) and prostaglandin I₂ synthase (PGIS), two prostaglandin biosynthesis genes, were significantly increased in SDEC. The level of sFRP-1 protein was significantly reduced, and levels of 2 prostaglandins that are downstream products of PTGS2 and PGIS, prostaglandin E₂ and 6-keto-prostaglandin F(1α), were significantly elevated in SDEC CM compared with WI38, Detroit 551, and MEF CM. Further, addition of purified sFRP-1 to SDEC CM reduced the proliferation of hESC grown on COL I as well as MAT in a dose-dependent manner.
Collapse
Affiliation(s)
- Meredith B Jones
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Mesenchymal stem cells (MSCs) can be derived from adult bone marrow, fat and several foetal tissues. In vitro, MSCs have the capacity to differentiate into multiple mesodermal and non-mesodermal cell lineages. Besides, MSCs possess immunosuppressive effects by modulating the immune function of the major cell populations involved in alloantigen recognition and elimination. The intriguing biology of MSCs makes them strong candidates for cell-based therapy against various human diseases. Type 1 diabetes is caused by a cell-mediated autoimmune destruction of pancreatic β-cells. While insulin replacement remains the cornerstone treatment for type 1 diabetes, the transplantation of pancreatic islets of Langerhans provides a cure for this disorder. And yet, islet transplantation is limited by the lack of donor pancreas. Generation of insulin-producing cells (IPCs) from MSCs represents an attractive alternative. On the one hand, MSCs from pancreas, bone marrow, adipose tissue, umbilical cord blood and cord tissue have the potential to differentiate into IPCs by genetic modification and/or defined culture conditions In vitro. On the other hand, MSCs are able to serve as a cellular vehicle for the expression of human insulin gene. Moreover, protein transduction technology could offer a novel approach for generating IPCs from stem cells including MSCs. In this review, we first summarize the current knowledge on the biological characterization of MSCs. Next, we consider MSCs as surrogate β-cell source for islet transplantation, and present some basic requirements for these replacement cells. Finally, MSCs-mediated therapeutic neovascularization in type 1 diabetes is discussed.
Collapse
Affiliation(s)
- Meng Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, PR China
| | | |
Collapse
|
14
|
Morton RA, Geras-Raaka E, Wilson LM, Raaka BM, Gershengorn MC. Endocrine precursor cells from mouse islets are not generated by epithelial-to-mesenchymal transition of mature beta cells. Mol Cell Endocrinol 2007; 270:87-93. [PMID: 17363142 PMCID: PMC1987709 DOI: 10.1016/j.mce.2007.02.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 01/30/2007] [Accepted: 02/02/2007] [Indexed: 02/03/2023]
Abstract
We previously presented evidence that proliferative human islet precursor cells may be derived in vitro from adult islets by epithelial-to-mesenchymal transition (EMT) and show here that similar fibroblast-like cells can be derived from mouse islets. These mouse cell populations exhibited changes in gene expression consistent with EMT. Both C-peptide and insulin mRNAs were undetectable in expanded cultures of mouse islet-derived precursor cells (mIPCs). After expansion, mIPCs could be induced to migrate into clusters and differentiate into hormone-expressing islet-like aggregates. Although early morphological changes suggesting EMT were observed by time-lapse microscopy when green fluorescent protein-labeled beta cells were placed in culture, the expanded precursor cell population was not fluorescent. Using two mouse models in which beta cells were permanently made either to express alkaline phosphatase or to have a deleted M(3) muscarinic receptor, we provide evidence that mIPCs in long term culture are not derived from beta cells.
Collapse
Affiliation(s)
- Russell A Morton
- Clinical Endocrinology Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20892-8029, USA
| | | | | | | | | |
Collapse
|
15
|
Dowling P, O'Driscoll L, O'Sullivan F, Dowd A, Henry M, Jeppesen PB, Meleady P, Clynes M. Proteomic screening of glucose-responsive and glucose non-responsive MIN-6 beta cells reveals differential expression of proteins involved in protein folding, secretion and oxidative stress. Proteomics 2007; 6:6578-87. [PMID: 17163442 DOI: 10.1002/pmic.200600298] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The glucose-sensitive insulin-secretion (GSIS) phenotype is relatively unstable in long-term culture of beta cells. The purpose of this study was to investigate relative changes in the proteome between glucose-responsive (low passage) and glucose non-responsive (high passage) murine MIN-6 pancreatic beta cells. The 2D-DIGE and subsequent DeCyder analysis detected 3351 protein spots in the pH range of 4-7. Comparing MIN-6(H) to MIN-6(L) and using a threshold of 1.2-fold, the number of proteins with a decrease in expression level was 152 (4.5%), similar was 3140 (93.7%) and increased 59 (1.8%). From the differentially expressed proteins identified in this study, groups of proteins associated with the endoplasmic reticulum (ER) and proteins involved in oxidative stress were found to be significantly decreased in the high-passage (H passage) cells. These proteins included endoplasmic reticulum protein 29 (ERp29); 78-kDa glucose-related protein, (GRP78); 94-kDa glucose-related protein (GRP94); protein disulphide isomerase; carbonyl reductase 3; peroxidoxin 4 and superoxide dismutase 1. These results suggest that non-GSIS MIN-6 cells do not have the same ability/capacity of glucose-responsive MIN-6 cells to successfully fold, modify or secrete proteins and counteract the problems associated with oxidative stress.
Collapse
Affiliation(s)
- Paul Dowling
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Narang AS, Mahato RI. Biological and biomaterial approaches for improved islet transplantation. Pharmacol Rev 2006; 58:194-243. [PMID: 16714486 DOI: 10.1124/pr.58.2.6] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Islet transplantation may be used to treat type I diabetes. Despite tremendous progress in islet isolation, culture, and preservation, the clinical use of this modality of treatment is limited due to post-transplantation challenges to the islets such as the failure to revascularize and immune destruction of the islet graft. In addition, the need for lifelong strong immunosuppressing agents restricts the use of this option to a limited subset of patients, which is further restricted by the unmet need for large numbers of islets. Inadequate islet supply issues are being addressed by regeneration therapy and xenotransplantation. Various strategies are being tried to prevent beta-cell death, including immunoisolation using semipermeable biocompatible polymeric capsules and induction of immune tolerance. Genetic modification of islets promises to complement all these strategies toward the success of islet transplantation. Furthermore, synergistic application of more than one strategy is required for improving the success of islet transplantation. This review will critically address various insights developed in each individual strategy and for multipronged approaches, which will be helpful in achieving better outcomes.
Collapse
Affiliation(s)
- Ajit S Narang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 26 S. Dunlap St., Feurt Building, Room 413, Memphis, TN 38163, USA
| | | |
Collapse
|
17
|
Hendee WR, Gazelle GS. Biomedical Imaging Research Opportunities Workshop III: A White Paper. Ann Biomed Eng 2006; 34:188-98. [PMID: 16482416 DOI: 10.1007/s10439-005-9040-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 09/07/2005] [Indexed: 10/25/2022]
Abstract
The third Biomedical Imaging Research Opportunities Workshop (BIROW III) was held on March 11-12, 2005, in Bethesda, MD. The workshop addressed four areas of imaging that present opportunities for research and development: Multimodality Image-Guided Therapy, Imaging Informatics, Imaging Cell Trafficking, and Technology Improvement and Commercialization. The first three areas were individually addressed in their own plenary sessions, followed by audience discussions that explored research opportunities and challenges. This paper synthesizes these discussions into a strategy for future research directions in biomedical imaging.
Collapse
Affiliation(s)
- William R Hendee
- Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| | | |
Collapse
|
18
|
Abstract
The implementation and integration of systems biology approaches with the emerging nanosciences and microchip technology will revolutionize profoundly molecular imaging and fuel the drive toward a more predictive and individualized health care. In combination with informatics platforms, key gene and protein targets will be identified, and serve as more effective targets for diagnostic and therapeutic interventions. Drug development also will be expedited by the judicious selection of more appropriate molecular biomarkers that will serve as objective end points of treatment efficacy, in addition to facilitating the development of new target-specific therapeutics. Finally, with the more widespread proliferation of high-field magnets and advancements in imaging hardware; acquisition methods; and novel,"smart" MR agents, the ability to achieve higher resolution analyses of tumor biology, cell track-ing, and gene expression will be realized more fully. Although radiologists will continue to serve as diagnostic consultants and assist in management decisions, the contributions from new developments in the biologic and molecular sciences will significantly alter the scope of our profession. Radiologists will be required to participate more actively in the individualized care of the patient and cultivate a deeper understanding of the underlying molecular basis of disease and molecular pharmacology for facilitating selection of the most appropriate combination of imaging studies that address biologically relevant questions. These radical changes in our profession will necessitate the re-education and emergence of a small cadre of professionals that is educated broadly in multiple scientific disciplines, and demonstrate expertise in clinical care and the basic sciences. The optimistic view is that this already is happening.
Collapse
Affiliation(s)
- Michelle Bradbury
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| | | |
Collapse
|
19
|
Shi CM, Cheng TM. Differentiation of dermis-derived multipotent cells into insulin-producing pancreatic cells in vitro. World J Gastroenterol 2004; 10:2550-2. [PMID: 15300903 PMCID: PMC4572160 DOI: 10.3748/wjg.v10.i17.2550] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To observe the plasticity of whether dermis-derived multipotent cells to differentiate into insulin-producing pancreatic cells in vitro.
METHODS: A clonal population of dermis-derived multipotent stem cells (DMCs) from newborn rat with the capacity to produce osteocytes, chondrocytes, adipocytes and neurons was used. The gene expression of cultured DMCs was assessed by DNA microarray using rat RGU34A gene expression probe arrays. DMCs were further cultured in the presence of insulin complex components (Insulin-transferrin-selenium, ITS) to observe whether DMCs could be induced into insulin-producing pancreatic cells in vitro.
RESULTS: DNA microarray analysis showed that cultured DMCs simultaneously expressed several genes associated with pancreatic cell, neural cell, epithelial cell and hepatocyte, widening its transcriptomic repertoire. When cultured in the specific induction medium containing ITS for pancreatic cells, DMCs differentiated into epithelioid cells that were positive for insulin detected by immunohistochemistry.
CONCLUSION: Our data indicate that dermal multipotent cells may serve as a source of stem/progenitor cells for insulin-producing pancreatic cells.
Collapse
Affiliation(s)
- Chun-Meng Shi
- Institute of Combined Injury, Third Military Medical University, Gaotanyan 400038, Chongqing City, China.
| | | |
Collapse
|
20
|
Liu XX, Miao B, Li F, Ma XF, Shi Q, Shen BJ. Insulin production by insulin-producing cells induced from embryonic stem cells. Shijie Huaren Xiaohua Zazhi 2004; 12:1857-1860. [DOI: 10.11569/wcjd.v12.i8.1857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the insulin secretion of insulin-producing cells (IPCs) induced from embryonic stem cells (ESCs).
METHODS: ESCs were allowed to grow on mouse fetal fibroblast feeder layer to keep undifferentiated state, and then transferred into serum-free DMEM supplemented with bFGF to form outgrowths in the culture. At day 21 after induction, the outgrowths were incubated in DTZ solution (final concentration, 100 mg/L) for 15 minutes before being observed microscopically. In addition, insulin production was examined immunohistochemically, and its secretion was determined using ELISA. The gene expression of endocrine pancreatic markers, including PDX-1, insulin1, insulin2 and Glut2, was also analyzed by RT-PCR, and the activity of secreted insulin was determined by glucose-reducing experiment on mice.
RESULTS: ESCs grew and formed embryoid bodies at day 4, and the addition of bFGF promoted the differentiation of ESCs into IPCs. The induced IPCs self-assembled to form three-dimentional clusters, and were stained crimson red by DTZ at day 21 after differentiation. They were found to be immunoreactive to insulin, express pancreatic-duodenal homeobox 1 (PDX1) and insulin2 mRNA. They were also able to secrete detectable amounts of active insulin, which could reduce mouse blood glucose significantly.
CONCLUSION: ES cell-induced IPCs can synthesize and secrete active insulin that is able to reduce blood glucose significantly.
Collapse
|
21
|
Liu XX, Miao B, Li F, Ma XF, Shi Q, Shen BJ. Therapeutic effect of insulin-producing cells induced from embryonic stem cells on diabetic mice. Shijie Huaren Xiaohua Zazhi 2004; 12:1853-1856. [DOI: 10.11569/wcjd.v12.i8.1853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the therapeutic effect on diabetic mice of insulin-producing cells induced from embryonic stem cells.
METHODS: Firstly, ESCs were induced to differentiate in serum-free DMEM supplemented with bFGF for more than 3 weeks, and DTZ staining was used to identify the induced IPCs; Secondly, experimental diabetes was induced in 6- to 8-week-old male Balb/c mice by a single intraperitoneal injection (200 mg/kg) of streptozotocin freshly dissolved in 0.1 moL/L of citrate buffer, pH 4.5; Finally, the induced IPCs were harvested at day 21 after differentiation, and grafted subcutaneously in the shoulder of streptozotocin-diabetic mice to observe their glucose-reducing effect.
RESULTS: ESCs could be induced to differentiate into IPCs in serum-free DMEM supplemented with bFGF. The induced IPCs were stained crimson red by DTZ, and their transplantation could reduce blood glucose of diabetic mouse significantly.
CONCLUSION: ESCs can be induced to differentiate into IPCs in serum-free DMEM supplemented with bFGF, and the induced IPCs transplantation has a certain therapeutic effect on diabetic mice.
Collapse
|
22
|
Abstract
This article focuses on molecular imaging of novel cell-based therapies, particularly stem cell therapies and the adoptive transfer of immunocytes. The animal models,potential clinical applications, and likely future prospects of these therapies are discussed in the context of imaging.
Collapse
Affiliation(s)
- Dawid Schellingerhout
- Department of Radiology, Center for Molecular Imaging Research, Massachusetts General Hospital, Room 5403, Building 149, 13th Street, Charlestown, MA 02129, USA.
| | | |
Collapse
|