1
|
Madkor HR, Abd El-Aziz MK, Abd El-Maksoud MS, Ibrahim IM, Ali FEM. Stem Cells Reprogramming in Diabetes Mellitus and Diabetic Complications: Recent Advances. Curr Diabetes Rev 2025; 21:21-37. [PMID: 38173073 DOI: 10.2174/0115733998275428231210055650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/09/2023] [Accepted: 11/21/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND The incidence of diabetes mellitus (DM) is dramatically increasing worldwide, and it is expected to affect 700 million cases by 2045. Diabetes influences health care economics, human quality of life, morbidity, and mortality, which were primarily seen extensively in developing countries. Uncontrolled DM, which results in consistent hyperglycemia, may lead to severe life-threatening complications such as nephropathy, retinopathy, neuropathy, and cardiovascular complications. METHODOLOGY In addition to traditional therapies with insulin and oral anti-diabetics, researchers have developed new approaches for treatment, including stem cell (SC) therapy, which exhibits promising outcomes. Besides its significant role in treating type one DM (T1DM) and type two DM (T2DM), it can also attenuate diabetic complications. Furthermore, the development of insulin- producing cells can be achieved by using the different types of SCs, such as embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and multiple types of adult stem cells, such as pancreatic, hepatic, and mesenchymal stem cells (MSC). All these types have been extensively studied and proved their ability to develop insulin-producing cells, but every type has limitations. CONCLUSION This review aims to enlighten researchers about recent advances in stem cell research and their potential benefits in DM and diabetic complications.
Collapse
Affiliation(s)
- Hafez R Madkor
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt
| | | | | | - Islam M Ibrahim
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Fares E M Ali
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| |
Collapse
|
2
|
Examining the therapeutic potential of various stem cell sources for differentiation into insulin-producing cells to treat diabetes. ANNALES D'ENDOCRINOLOGIE 2019; 80:47-53. [DOI: 10.1016/j.ando.2018.06.1084] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/24/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022]
|
3
|
Cooper TT, Bell GI, Hess DA. Inhibition of Retinoic Acid Production Expands a Megakaryocyte-Enriched Subpopulation with Islet Regenerative Function. Stem Cells Dev 2018; 27:1449-1461. [PMID: 30039749 DOI: 10.1089/scd.2018.0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Islet regeneration is stimulated after transplantation of human umbilical cord blood (UCB) hematopoietic progenitor cells with high aldehyde dehydrogenase (ALDH)-activity into NOD/SCID mice with streptozotocin (STZ)-induced β cell ablation. ALDHhi progenitor cells represent a rare subset within UCB that will require expansion without the loss of islet regenerative functions for use in cell therapies. ALDHhi cells efficiently expand (>70-fold) under serum-free conditions; however, high ALDH-activity is rapidly diminished during culture coinciding with emergence of a committed megakaryocyte phenotype CD41+/CD42+/CD38+. ALDH-activity is also the rate-limiting step in retinoic acid (RA) production, a potent driver of hematopoietic differentiation. We have previously shown that inhibition of RA production during 9-day cultures, using diethylaminobenzaldehyde (DEAB) treatment, enhanced the expansion of ALDHhi cells (>20-fold) with vascular regenerative paracrine functions. Herein, we sought to determine if DEAB-treatment also expanded ALDHhi cells that retain islet regenerative function following intrapancreatic transplantation into hyperglycemic mice. After DEAB-treatment, expanded ALDHhi cell subset was enriched for CD34+/CD38- expression and demonstrated enhanced myeloid multipotency in vitro compared to the ALDHlo cell subset. Unfortunately, DEAB-treated ALDHhi cells did not support islet regeneration after transplantation. Conversely, expanded ALDHlo cells from DEAB-treated conditions reduced hyperglycemia, and increased islet number and cell proliferation in STZ-induced hyperglycemic NOD/SCID mice. DEAB-treated ALDHlo cells were largely committed to a CD41+/CD42+ megakaryocyte phenotype. Collectively, this study provides preliminary evidence that committed cells of the megakaryocyte-lineage support endogenous islet regeneration and/or function, and the retention of high ALDH-activity did not coincide with islet regenerative function after expansion under serum-free culture conditions.
Collapse
Affiliation(s)
- Tyler Thomas Cooper
- 1 Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University , London, Canada .,2 Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute , London, Canada
| | - Gillian I Bell
- 2 Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute , London, Canada
| | - David A Hess
- 1 Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University , London, Canada .,2 Molecular Medicine Research Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute , London, Canada
| |
Collapse
|
4
|
Aloy-Reverté C, Moreno-Amador JL, Nacher M, Montanya E, Semino CE. Use of RGD-Functionalized Sandwich Cultures to Promote Redifferentiation of Human Pancreatic Beta Cells AfterIn VitroExpansion. Tissue Eng Part A 2018; 24:394-406. [DOI: 10.1089/ten.tea.2016.0493] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Caterina Aloy-Reverté
- Department of Bioengineering, Tissue Engineering Laboratory, IQS School of Engineering, Barcelona, Spain
| | - José L. Moreno-Amador
- Hospital Universitari Bellvitge-Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Diabetes and Metabolic Diseases (CIBERDEM), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Montserrat Nacher
- Hospital Universitari Bellvitge-Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Diabetes and Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Eduard Montanya
- Hospital Universitari Bellvitge-Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Diabetes and Metabolic Diseases (CIBERDEM), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Carlos E. Semino
- Department of Bioengineering, Tissue Engineering Laboratory, IQS School of Engineering, Barcelona, Spain
| |
Collapse
|
5
|
Askenasy N. Less Is More: The Detrimental Consequences of Immunosuppressive Therapy in the Treatment of Type-1 Diabetes. Int Rev Immunol 2015; 34:523-37. [DOI: 10.3109/08830185.2015.1010723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
6
|
Setyowati Karolina D, Sepramaniam S, Tan HZ, Armugam A, Jeyaseelan K. miR-25 and miR-92a regulate insulin I biosynthesis in rats. RNA Biol 2014; 10:1365-78. [PMID: 24084692 DOI: 10.4161/rna.25557] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The 3' UTR of insulin has been identified as a critical region that confers mRNA stability, which is crucial for promoting transcription in response to glucose challenge. miRNAs are endogenously encoded non-coding RNAs that function as regulators of gene expression. This regulatory function is generally mediated by complementary binding to the 3'UTR of its mRNA targets that affects subsequent translational process. Genes involved in the regulation of glucose homeostasis, particularly in insulin production, have been found as targets of several miRNAs. Yet, no direct miRNA-based regulators of insulin biosynthesis have been identified. In this study, identification of possible miRNA-based regulators of insulin production is explored. Members of a miRNA family, miR-25 and miR-92a, are found as direct modulators of insulin expression. Overexpression of miR-25 or miR-92a reduced insulin expression while inhibition of miR-25 and miR-92a expression using corresponding antagomiRs promoted insulin expression and ultimately enhanced glucose-induced insulin secretion. Furthermore, suppression of insulin secretion by pre miR-9 could be attenuated by treatment with anti-miR-25 or miR-92a. Interestingly, we found the binding site of miR-25 and miR-92a to overlap with that of PTBP1, an important RNA binding molecule that stabilizes insulin mRNA for translation. Despite the increase in PTBP1 protein in the pancreas of diabetic rats, we observed insulin expression to be reduced alongside upregulation of miR-25 and miR-92a, suggesting an intricate regulation of insulin (bio)synthesis at its mRNA level.
Collapse
Affiliation(s)
- Dwi Setyowati Karolina
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University Health System; Singapore
| | | | | | | | | |
Collapse
|
7
|
Tang K, Xiao X, Liu D, Shen Y, Chen Y, Wang Y, Li B, Yu F, Ma D, Yan J, Liang H, Yang D, Weng J. Autografting of bone marrow mesenchymal stem cells alleviates streptozotocin‑induced diabetes in miniature pigs: real-time tracing with MRI in vivo. Int J Mol Med 2014; 33:1469-76. [PMID: 24714958 PMCID: PMC4055443 DOI: 10.3892/ijmm.2014.1729] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 03/31/2014] [Indexed: 02/03/2023] Open
Abstract
Cellular replacement therapy for diabetes mellitus (DM) has received much attention. In this study, we investigated the effect of transplantation of autologous bone marrow-derived mesenchymal stem cells (ABMSCs) in streptozotocin (STZ)-induced diabetic miniature pigs. Miniature pig BMSCs were cultured, labeled with superparamagnetic iron oxide (SPIO) and transplanted into the pancreas of diabetic miniature pigs through targeted intervention. Blood glucose levels, intravenous and oral glucose tolerance test (OGTT), serum insulin, C-peptide and islets histology were analyzed. These transplanted cells were then identified by magnetic resonance imaging (MRI). The results showed that transplantation of ABMSCs reversed STZ-induced diabetes in miniature pigs. Blood glucose levels, intravenous, OGTT, serum insulin and C-peptide were significantly recovered in the diabetic minipigs with the autologous BMSC (DMAB) transplantation group. In addition, the number of islets was significantly increased in this group compared to the diabetic minipig control (DMC) group with conventional therapy. These data suggested the implantation of autologous BMSCs for type 1 diabetes treatment can partially restore the function of islet β cells and maintain blood glucose homeostasis. Transplanted autologous BMSCs may improve islet repairing by differentiating for new islets and change pancreatic microcirculation and be identified in a real-time manner using MRI in vivo.
Collapse
Affiliation(s)
- Kuanxiao Tang
- Department of Endocrinology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Xiaoyan Xiao
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Dayue Liu
- Department of Vascular Surgery and Radiology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yunfeng Shen
- Department of Endocrinology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Yingming Chen
- Department of Vascular Surgery and Radiology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yu Wang
- Department of Vascular Surgery and Radiology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Baoying Li
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Fei Yu
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Dedong Ma
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jinhua Yan
- Department of Endocrinology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Hua Liang
- Department of Endocrinology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Daizhi Yang
- Department of Endocrinology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jianping Weng
- Department of Endocrinology, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
8
|
Sarker MMH, Zhou M, Rameshwar P, Hanover JA. Functions and roles of proteins: diabetes as a paradigm. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 114:2-7. [PMID: 24239502 PMCID: PMC10483990 DOI: 10.1016/j.pbiomolbio.2013.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 11/04/2013] [Indexed: 01/01/2023]
Abstract
Molecular and cellular biology has moved towards complete and accurate knowledge of how molecules behave in space and time. Protein is considered as the primary group of molecules responsible for mediating most physiological processes. Changes in the levels of proteins may lead to the altered function and are responsible for many diseases. This review provides a partial molecular explanation of biological force-ratio generation that may act to split protein into branches, and shows molecular functional divergence. Developing a non-reductionist theory of the cellular function in medicine is clearly not sufficient. Finding effective parameters of the models by characterizing molecular interactions becomes necessary. Protein interactivity and stability provides a basis for an integrated understanding of pathologies such diabetes. One example of how a mechanistic analysis of such physiological processes can be of value is the time-delay between mRNA and translation that can act as a fork allowing a slowdown in gene expression.
Collapse
Affiliation(s)
- Md Mosharrof Hossain Sarker
- Dept of Electrical and Computer Engineering, New Jersey Institute of Technology (NJIT), Newark, NJ 07102, USA.
| | - MengChu Zhou
- Dept of Electrical and Computer Engineering, New Jersey Institute of Technology (NJIT), Newark, NJ 07102, USA.
| | - Pranela Rameshwar
- Dept of Medicine-Hematology/Oncology, Graduate School of Biomedical Science, Rutgers-New Jersey Medical School, NJ 07103, USA.
| | - John A Hanover
- Laboratory of Cellular and Molecular Biology, NIDDK, National Institutes of Health (NIH), Bethesda, MD 20892-0851, USA.
| |
Collapse
|
9
|
Verma PR, Itankar PR, Arora SK. Evaluation of antidiabetic antihyperlipidemic and pancreatic regeneration, potential of aerial parts of Clitoria ternatea. REVISTA BRASILEIRA DE FARMACOGNOSIA-BRAZILIAN JOURNAL OF PHARMACOGNOSY 2013. [DOI: 10.1590/s0102-695x2013000500015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
10
|
Shi Q, Schatten G, Hodara V, Simerly C, VandeBerg JL. Endothelial reconstitution by CD34+ progenitors derived from baboon embryonic stem cells. J Cell Mol Med 2013; 17:242-51. [PMID: 23301772 PMCID: PMC3814022 DOI: 10.1111/jcmm.12002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 11/13/2012] [Indexed: 12/17/2022] Open
Abstract
In this study, we used a large non-human primate model, the baboon, to establish a step-wise protocol to generate CD34+ endothelial progenitor cells (EPCs) from embryonic stem cells (ESCs) and to demonstrate their reparative effects. Baboon ESCs were sequentially differentiated from embryoid body cultures for 9 days and then were specified into EPCs by culturing them in monolayer for 12 days. The resulting EPCs expressed CD34, CXCR4 and UEA-1, but neither CD31 nor CD117. The EPCs were able to form intact lumen structures when seeded on Matrigel, took up Dil-LDL, and responded to TNF-α. Angioblasts specified in EGM-2 medium and ECGS medium had 6.41 ± 1.16% (n = 3) and 9.32 ± 3.73% CD34+ cells (n = 3). The efficiency of generating CD34+ EPCs did not differ significantly from ECGS to EGM-2 culture media, however, angioblasts specified in ECGS medium expressed a higher percentage of CD34+/CXCR4+ cells (3.49 ± 1.32%, n = 3) than those specified in EGM-2 medium (0.49 ± 0.52%, n = 3). To observe their reparative capacity, we purified CD34+ progenitors after specification by EGM-2 medium; inoculated fluorescently labelled CD34+ EPCs into an arterial segment denuded of endothelium in an ex vivo system. After 14 days of ex vivo culture, the grafted cells had attached and integrated to the denuded surface; in addition, they had matured further and expressed terminally differentiated endothelial markers including CD31 and CD146. In conclusion, we have proved that specified CD34+ EPCs are promising therapeutic agents for repairing damaged vasculature.
Collapse
Affiliation(s)
- Qiang Shi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245-0549, USA.
| | | | | | | | | |
Collapse
|
11
|
Abouna S, Old RW, Pelengaris S, Epstein D, Ifandi V, Sweeney I, Khan M. Non-β-cell progenitors of β-cells in pregnant mice. Organogenesis 2012; 6:125-33. [PMID: 20885859 DOI: 10.4161/org.6.2.10374] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 10/16/2009] [Accepted: 10/20/2009] [Indexed: 02/06/2023] Open
Abstract
Pregnancy is a normal physiological condition in which the maternal β-cell mass increases rapidly about two-fold to adapt to new metabolic challenges. We have used a lineage tracing of β-cells to analyse the origin of new β-cells during this rapid expansion in pregnancy. Double transgenic mice bearing a tamoxifen-dependent Cre-recombinase construct under the control of a rat insulin promoter, together with a reporter Z/AP gene, were generated. Then, in response to a pulse of tamoxifen before pregnancy, β-cells in these animals were marked irreversibly and heritably with the human placental alkaline phosphatase (HP AP). First, we conclude that the lineage tracing system was highly specific for β-cells. Secondly, we scored the proportion of the β-cells marked with HP AP during a subsequent chase period in pregnant and non-pregnant females. We observed a dilution in this labeling index in pregnant animal pancreata, compared to nonpregnant controls, during a single pregnancy in the chase period. To extend these observations we also analysed the labeling index in pancreata of animals during the second of two pregnancies in the chase period. The combined data revealed statistically-significant dilution during pregnancy, indicating a contribution to new beta cells from a non-β-cell source. Thus for the first time in a normal physiological condition, we have demonstrated not only β-cell duplication, but also the activation of a non-β-cell progenitor population. Further, there was no transdifferentiation of β-cells to other cell types in a two and half month period following labeling, including the period of pregnancy.
Collapse
Affiliation(s)
- Sylvie Abouna
- Department of Biological Sciences, University of Warwick, Coventry, UK
| | | | | | | | | | | | | |
Collapse
|
12
|
Zhu D, Chen L, Hong T. Position Statement of the Chinese Diabetes Society regarding stem cell therapy for diabetes. J Diabetes 2012; 4:18-21. [PMID: 22040058 DOI: 10.1111/j.1753-0407.2011.00166.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Dalong Zhu
- Division of Endocrinology, Affiliated Drum Tower Hospital of Nanjing University, Nanjing, Jiangsu, China
| | | | | |
Collapse
|
13
|
Soejitno A, Prayudi PKA. The prospect of induced pluripotent stem cells for diabetes mellitus treatment. Ther Adv Endocrinol Metab 2011; 2:197-210. [PMID: 23148185 PMCID: PMC3474639 DOI: 10.1177/2042018811420198] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A continuous search for a permanent cure for diabetes mellitus is underway with several remarkable discoveries over the past few decades. One of these is the potential of pancreatic stem/progenitor cells to rejuvenate functional β cells. However, the existence of these cell populations is still obscure and a lack of phenotype characterization hampers their use in clinical settings. Cellular reprogramming through induced pluripotent stem (iPS) cell technology can become an alternative strategy to generate insulin-producing cells in a relatively safe (autologous-derived cells, thus devoid of rejection risk) and efficient way (high cellular proliferation) but retain a precise morphological and genetic composition, similar to that of the native β cells. iPS cell technology is a technique of transducing any cell types with key transcription factors to yield embryonic-like stem cells with high clonogenicity and is able to give rise into all cell lineages from three germ layers (endoderm, ectoderm, and mesoderm). This approach can generate β-like pancreatic cells that are fully functional as proven by either in vitro or in vivo studies. This novel proof-of-concept stem cell technology brings new expectations on applying stem cell therapy for diabetes mellitus in clinical settings.
Collapse
Affiliation(s)
- Andreas Soejitno
- Department of Molecular Medicine and Stem Cell Research, Faculty of Medicine Udayana University, Denpasar, Indonesia
| | - Pande Kadek Aditya Prayudi
- Department of Molecular Medicine and Stem Cell Research, Faculty of Medicine Udayana University, Denpasar, Indonesia
| |
Collapse
|
14
|
Shin JS, Min BH, Lim JY, Kim BK, Han HJ, Yoon KH, Kim SJ, Park CG. Novel culture technique involving an histone deacetylase inhibitor reduces the marginal islet mass to correct streptozotocin-induced diabetes. Cell Transplant 2011; 20:1321-32. [PMID: 21294957 DOI: 10.3727/096368910x557146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Islet transplantation is limited by the difficulties in isolating the pancreatic islets from the cadaveric donor and maintaining them in culture. To increase islet viability and function after isolation, here we present a novel culture technique involving an histone deacetylase inhibitor (HDACi) to rejuvenate the isolated islets. Pancreatic islets were isolated from Sprague-Dawley (SD) rats and one group (FIs; freshly isolated islets) was used after overnight culture and the other group (RIs; rejuvenated islet) was subjected to rejuvenation culture procedure, which is composed of three discrete steps including degranulation, chromatin remodeling, and regranulation. FIs and RIs were compared with regard to intracellular insulin content, glucose-stimulated insulin secretion (GSIS) capacity, gene expression profile, viability and apoptosis rate under oxidative stresses, and the engraftment efficacy in the xenogeneic islet transplantation models. RIs have been shown to have 1.9 ± 0.28- and 1.7 ± 0.31-fold greater intracellular insulin content and GSIS capacity, respectively, than FIs. HDACi increased overall histone acetylation levels, with inducing increased expression of many genes including insulin 1, insulin 2, GLUT2, and Ogg1. This enhanced islet capacity resulted in more resistance against oxidative stresses and increase of the engraftment efficacy shown by reduction of twofold marginal mass of islets in xenogeneic transplantation model. In conclusion, a novel rejuvenating culture technique using HDACi as chromatin remodeling agents improved the function and viability of the freshly isolated islets, contributing to the reduction of islet mass for the control of hyperglycemia in islet transplantation.
Collapse
Affiliation(s)
- Jun-Seop Shin
- Korea Islet Transplantation Institute, Inc., Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Arien-Zakay H, Lazarovici P, Nagler A. Tissue regeneration potential in human umbilical cord blood. Best Pract Res Clin Haematol 2011; 23:291-303. [PMID: 20837341 DOI: 10.1016/j.beha.2010.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerative medicine is the process of creating functional tissue with the aid of stem cells, to repair loss of organ function. Possible targets for regenerative medicine include orthopaedic, cardiac, hepatic, pancreatic and central nervous system (CNS) applications. Umbilical cord blood (CB) has established itself as a legitimate source for haematopoietic stem cell transplantation. It is also considered an accessible and less immunogenic source for mesenchymal, unrestricted somatic and for other stem cells with pluri/multipotent properties. The latter are capable of differentiating into a wide variety of cell types including bone, cartilage, cardiomyocytes and neural. They also possess protective abilities that may contribute to tissue repair even if in vitro differentiation is excluded. In view of the absence of treatment for many devastating diseases, the elucidation of non-haematopoietic applications for CB will facilitate the development of pioneering relevant cell therapy approaches. This review focusses on current studies using human CB-derived cells for regenerative medicine.
Collapse
|
16
|
Wang Y, Rovira M, Yusuff S, Parsons MJ. Genetic inducible fate mapping in larval zebrafish reveals origins of adult insulin-producing β-cells. Development 2011; 138:609-17. [PMID: 21208992 DOI: 10.1242/dev.059097] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The Notch-signaling pathway is known to be fundamental in controlling pancreas differentiation. We now report on using Cre-based fate mapping to indelibly label pancreatic Notch-responsive cells (PNCs) at larval stages and follow their fate in the adult pancreas. We show that the PNCs represent a population of progenitors that can differentiate to multiple lineages, including adult ductal cells, centroacinar cells (CACs) and endocrine cells. These endocrine cells include the insulin-producing β-cells. CACs are a functional component of the exocrine pancreas; however, our fate-mapping results indicate that CACs are more closely related to endocrine cells by lineage as they share a common progenitor. The majority of the exocrine pancreas consists of the secretory acinar cells; however, we only detect a very limited contribution of PNCs to acinar cells. To explain this observation we re-examined early events in pancreas formation. The pancreatic anlage that gives rise to the exocrine pancreas is located in the ventral gut endoderm (called the ventral bud). Ptf1a is a gene required for exocrine pancreas development and is first expressed as the ventral bud forms. We used transgenic marker lines to observe both the domain of cells expressing ptf1a and cells responding to Notch signaling. We do not detect any overlap in expression and demonstrate that the ventral bud consists of two cell populations: a ptf1-expressing domain and a Notch-responsive progenitor core. As pancreas organogenesis continues, the ventral bud derived PNCs align along the duct, remain multipotent and later in development differentiate to form secondary islets, ducts and CACs.
Collapse
Affiliation(s)
- Yiyun Wang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
17
|
Wu HL, Wang Y, Zhang P, Li SF, Chen X, Chen YK, Li JG, Yang SM, Su YP, Wang JP, Chen B. Reversible immortalization of rat pancreatic β cells with a novel immortalizing and tamoxifen-mediated self-recombination tricistronic vector. J Biotechnol 2010; 151:231-41. [PMID: 21167227 DOI: 10.1016/j.jbiotec.2010.12.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2010] [Revised: 12/02/2010] [Accepted: 12/07/2010] [Indexed: 02/07/2023]
Abstract
Although the strategy of "Cre/LoxP-based reversible immortalization" holds great promise to overcome the cellular senescence of primary cell cultures for their further use, a secondary gene transfer for Cre expression is usually utilized to trigger the excision of the immortalizing genes in a large number of cells, thus presenting a formidable hurdle for large-scale application. We modified the strategy by utilizing a tricistronic retroviral vector pLCRSTP, in which Cre-ER, simian virus 40 large T antigen (SV40LTAg) oncogene, and a reporter gene were flanked by the same pair of LoxA sites. Five immortalized rat pancreatic β cell clones transduced with pLCRSTP, and six immortalized rat pancreatic β cell clones co-transduced with pLCRSTP and another vector encoding the human telomerase reverse transcriptase (hTERT) gene, were obtained, respectively. The Cre-ER protein could be induced to translocate from the cytoplasm to the nucleus by 4-hydroxytamoxifen to make SV40LTAg, hTERT and the Cre-ER gene itself excise without a secondary gene transfer. Our studies suggest that this system is useful to expand rat β cells and may allow for large-scale production due to its simpler manipulation.
Collapse
Affiliation(s)
- Hui-Ling Wu
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, No. 29 Gaotanyan Street, Chongqing 400038, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Dinarvand P, Hashemi SM, Soleimani M. Effect of transplantation of mesenchymal stem cells induced into early hepatic cells in streptozotocin-induced diabetic mice. Biol Pharm Bull 2010; 33:1212-7. [PMID: 20606315 DOI: 10.1248/bpb.33.1212] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cellular replacement therapy for diabetes mellitus has received much attention. In this study we investigated the effect of transplantation of bone marrow-derived mesenchymal stem cells (BM-MSCs) induced into endoderm and early hepatic cells in streptozotocin (STZ)-induced diabetic mice. Mouse BM-MSCs were cultured in the presence of hepatocyte growth factor (HGF) and fibroblast growth factor (FGF-4) for 2 weeks and transplanted into diabetic mice. Blood glucose levels, intraperitoneal glucose tolerance test, serum insulin, body weight and islets histology were analyzed. The results demonstrated that transplantation of syngeneic induced MSCs could reverse STZ-induced diabetes in mice. The treatment of mice with hyperglycemia and islet destruction resulted in the repair of pancreatic islets. Blood glucose levels, intraperitoneal glucose tolerance test, and serum insulin were significantly recovered in induced BM-MSCs (iBM-MSCs) group. In addition, in the iBM-MSCs group the body weight and the number of islets were significantly increased compared to other groups. The results demonstrate that BM-MSCs induced into endoderm and early hepatic cells are suitable candidates for cell-based therapy of diabetes mellitus.
Collapse
Affiliation(s)
- Peyman Dinarvand
- Department of Stem Cells and Tissue Engineering, Stem Cell Technology Research Center, Tehran 14155-3117, Iran
| | | | | |
Collapse
|
19
|
Limbert C, Ebert R, Schilling T, Path G, Benisch P, Klein-Hitpass L, Seufert J, Jakob F. Functional signature of human islet-derived precursor cells compared to bone marrow-derived mesenchymal stem cells. Stem Cells Dev 2010; 19:679-91. [PMID: 19895235 DOI: 10.1089/scd.2009.0241] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Pancreatic islet beta-cell replenishment can be driven by epithelial cells from exocrine pancreas via epithelial-mesenchymal transition (EMT) and the reverse process MET, while specified pancreatic mesenchymal cells control islet cell development and maintenance. The role of human islet-derived precursor cells (hIPCs) in regeneration and support of endocrine islets is under investigation. Here, we analyzed hIPCs as to their immunophenotype, multilineage differentiation capacity, and gene profiling, in comparison to human bone marrow-derived mesenchymal stem cells (hBM-MSCs). hIPCs and hBM-MSCs display a common mesenchymal character and express lineage-specific marker genes upon induction toward pancreatic endocrine and mesenchymal pathways of differentiation. hIPCs can go further along endocrine pathways while lacking some core mesenchymal differentiation attributes. Significance analysis of microarray (SAM) from 5 hBM-MSC and 3 hIPC donors mirrored such differences. Candidate gene cluster analysis disclosed differential expression of key lineage regulators, indicated a HoxA gene-associated positional memory in hIPCs and hBM-MSCs, and showed as well a clear transition state from mesenchyme to epithelium or vice versa in hIPCs. Our findings raise new research platforms to further clarify the potential of hIPCs to undergo complete MET thus contributing to islet cell replenishment, maintenance, and function.
Collapse
Affiliation(s)
- Catarina Limbert
- Orthopedic Center for Musculoskeletal Research, Stem Cell Division, Orthopedic Department, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Houbracken I, Bouwens L. The quest for tissue stem cells in the pancreas and other organs, and their application in beta-cell replacement. Rev Diabet Stud 2010; 7:112-23. [PMID: 21060970 PMCID: PMC2989784 DOI: 10.1900/rds.2010.7.112] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Revised: 07/07/2010] [Accepted: 07/10/2010] [Indexed: 02/06/2023] Open
Abstract
Adult stem cell research has drawn a lot of attention by many researchers, due to its medical hope of cell replacement or regenerative therapy for diabetes patients. Despite the many research efforts to date, there is no consensus on the existence of stem cells in adult pancreas. Genetic lineage tracing experiments have put into serious doubt whether β-cell neogenesis from stem/progenitor cells takes place postnatally. Different in vitro experiments have suggested centroacinar, ductal, acinar, stellate, or yet unidentified clonigenic cells as candidate β-cell progenitors. As in the rest of the adult stem cell field, sound and promising observations have been made. However, these observations still need to be replicated. As an alternative to committed stem/progenitor cells in the pancreas, transdifferentiation or lineage reprogramming of exocrine acinar and endocrine α-cells may be used to generate new β-cells. At present, it is unclear which approach is most medically promising. This article highlights the progress being made in knowledge about tissue stem cells, their existence and availability for therapy in diabetes. Particular attention is given to the assessment of methods to verify the existence of tissue stem cells.
Collapse
Affiliation(s)
| | - Luc Bouwens
- Cell Differentiation Lab, Diabetes Research Center, Vrije Universiteit Brussel (Free University of Brussels), Laarbeeklaan 103, 1090 - Brussels, Belgium
| |
Collapse
|
21
|
Haller MJ, Wasserfall CH, McGrail KM, Cintron M, Brusko TM, Wingard JR, Kelly SS, Shuster JJ, Atkinson MA, Schatz DA. Autologous umbilical cord blood transfusion in very young children with type 1 diabetes. Diabetes Care 2009; 32:2041-6. [PMID: 19875605 PMCID: PMC2768209 DOI: 10.2337/dc09-0967] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Interest continues to grow regarding the therapeutic potential for umbilical cord blood therapies to modulate autoimmune disease. We conducted an open-label phase I study using autologous umbilical cord blood infusion to ameliorate type 1 diabetes. RESEARCH DESIGN AND METHODS Fifteen patients diagnosed with type 1 diabetes and for whom autologous umbilical cord blood was stored underwent a single intravenous infusion of autologous cells and completed 1 year of postinfusion follow-up. Intensive insulin regimens were used to optimize glycemic control. Metabolic and immunologic assessments were performed before infusion and at established time periods thereafter. RESULTS Median (interquartile range [IQR]) age at infusion was 5.25 (3.1-7.3) years, with a median postdiagnosis time to infusion of 17.7 (10.9-26.5) weeks. No infusion-related adverse events were observed. Metabolic indexes 1 year postinfusion were peak C-peptide median 0.50 ng/ml (IQR 0.26-1.30), P = 0.002; A1C 7.0% (IQR 6.5-7.7), P = 0.97; and insulin dose 0.67 units * kg(-1) * day(-1) (IQR 0.55-0.77), P = 0.009. One year postinfusion, no changes were observed in autoantibody titers, regulatory T-cell numbers, CD4-to-CD8 ratio, or other T-cell phenotypes. CONCLUSIONS Autologous umbilical cord blood transfusion in children with type 1 diabetes is safe but has yet to demonstrate efficacy in preserving C-peptide. Larger randomized studies as well as 2-year postinfusion follow-up of this cohort are needed to determine whether autologous cord blood-based approaches can be used to slow the decline of endogenous insulin production in children with type 1 diabetes.
Collapse
Affiliation(s)
- Michael J Haller
- Department of Pediatrics, The University of Florida, Gainesville, Florida, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Limbert C, Seufert J. In vitro (re)programming of human bone marrow stromal cells toward insulin-producing phenotypes. Pediatr Diabetes 2009; 10:413-9. [PMID: 19627549 DOI: 10.1111/j.1399-5448.2009.00502.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Catarina Limbert
- Division of Pediatric Endocrinology and Diabetology, Children's University Hospital Dona Estefânia, Lisbon, Portugal
| | | |
Collapse
|
23
|
Bibliography. Current world literature. Curr Opin Endocrinol Diabetes Obes 2009; 16:328-37. [PMID: 19564733 DOI: 10.1097/med.0b013e32832eb365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Jin J, Park J, Kim K, Kang Y, Park SG, Kim JH, Park KS, Jun H, Kim Y. Detection of differential proteomes of human beta-cells during islet-like differentiation using iTRAQ labeling. J Proteome Res 2009; 8:1393-403. [PMID: 19199707 DOI: 10.1021/pr800765t] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A human beta-cell line, RNAKT-15, was recently established from human pancreatic islets, whereby its differentiation into islet-like beta-cells (islet-like RNAKT-15) increased its expression of insulin 2-fold compared with RNAKT-15 cells. To characterize the differentiation of RNAKT-15 cells into islet-like RNAKT-15, microarray and quantitative proteomics were performed. Our analysis of differential proteomic and mRNA expression has resulted in a greater understanding of the molecular functions that are involved in beta-cell differentiation and insulin synthesis and release.
Collapse
Affiliation(s)
- Jonghwa Jin
- Departments of Biomedical Sciences and Internal Medicine, Genome Research Center for Diabetes and Endocrine Disease, Seoul National University College of Medicine, 28 Yongon-Dong, Seoul 110-799, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Moro E, Gnügge L, Braghetta P, Bortolussi M, Argenton F. Analysis of beta cell proliferation dynamics in zebrafish. Dev Biol 2009; 332:299-308. [PMID: 19500567 DOI: 10.1016/j.ydbio.2009.05.576] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 05/28/2009] [Accepted: 05/29/2009] [Indexed: 10/20/2022]
Abstract
Among the different mechanisms invoked to explain the beta cell mass expansion during postnatal stages and adulthood, self-replication is being considered the major cellular event occurring both under physiological conditions and in regenerating pancreas after partial pancreactomy. Neogenesis, i.e. differentiation from pancreatic progenitors, has been demonstrated to act concurrently with beta cell replication during pancreatic regeneration. Both phenomena have been largely elucidated in higher vertebrates (mouse, rat and guinea pig), but an extensive description of beta cell dynamics in other animal models is currently lacking. We, therefore, explored in zebrafish the cellular origins of new beta cells in both adult and larval stages. By integrating the results from in vivo time lapse analysis and immunostaining, we provide a detailed reconstruction of the major processes involved in fish beta cell genesis and maintenance. Moreover, by establishing the selective ablation of proliferating beta cells, through the ganciclovir-HSVTK system, we could show that in larval stages self-replication is the main mechanism of beta cells expansion. Since the same mechanism of proliferation has been observed to occur during early and late life stages, we suggest that zebrafish larvae can be used as an alternative tool for an in vivo exploration and screening of new potential mitogens specifically targeting beta cells.
Collapse
Affiliation(s)
- Enrico Moro
- Department of Biology, University of Padova, Padova, Italy
| | | | | | | | | |
Collapse
|
26
|
von Herrath M. Can we learn from viruses how to prevent type 1 diabetes?: the role of viral infections in the pathogenesis of type 1 diabetes and the development of novel combination therapies. Diabetes 2009; 58:2-11. [PMID: 19114721 PMCID: PMC2606872 DOI: 10.2337/db08-9027] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We will take a journey from basic pathogenetic mechanisms elicited by viral infections that play a role in the development of type 1 diabetes to clinical interventions, where we will discuss novel combination therapies. The role of viral infections in the development of type 1 diabetes is a rather interesting topic because in experimental models viruses appear capable of both accelerating as well as decelerating the immunological processes leading to type 1 diabetes. Consequently, I will discuss some of the underlying mechanisms for each situation and consider methods to investigate the proposed dichotomy for the involvement of viruses in human type 1 diabetes. Prevention of type 1 diabetes by infection supports the so-called "hygiene hypothesis." Interestingly, viruses invoke mechanisms that need to be exploited by novel combinatorial immune-based interventions, the first one being the elimination of autoaggressive T-cells attacking the beta-cells, ultimately leading to their immediate but temporally limited amelioration. The other is the invigoration of regulatory T-cells (Tregs), which can mediate long-term tolerance to beta-cell proteins in the pancreatic islets and draining lymph nodes. In combination, these two immune elements have the potential to permanently stop type 1 diabetes. It is my belief that only combination therapies will enable the permanent prevention and curing of type 1 diabetes.
Collapse
Affiliation(s)
- Matthias von Herrath
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA.
| |
Collapse
|
27
|
Pearl EJ, Horb ME. Promoting ectopic pancreatic fates: pancreas development and future diabetes therapies. Clin Genet 2008; 74:316-24. [PMID: 18783407 DOI: 10.1111/j.1399-0004.2008.01081.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diabetes is a disease that could be treated more effectively with a better understanding of pancreas development. This review examines the role of master regulator genes driving crucial steps in pancreas development, from foregut specification to differentiation of the five endocrine cell types. The roles of Pdx1, Ptf1a, and Ngn3 are particularly examined as they are both necessary and sufficient for promoting pancreatic cell fates (Pdx1, Ptf1a) and endocrine cell development (Ngn3). The roles of Arx and Pax4 are studied as they compose part of the regulatory mechanism balancing development of different types of endocrine cells within the iselts and promote the development of alpha/PP and beta/delta cell progenitors, respectively. The roles of the aforementioned genes, and the consequences of misexpression of them for functionality of the pancreas, are examined through recent studies in model organisms, particularly Xenopus and zebrafish. Recent developments in cell replacement therapy research are also covered, concentrating on stem cell research (coaxing both adult and embryonic stem cells toward a beta cell fate) and transdifferentiation (generating beta cells from other differentiated cell types).
Collapse
Affiliation(s)
- E J Pearl
- Laboratory of Molecular Organogenesis, Institut de Recherches Cliniques de Montréal, Québec, Canada
| | | |
Collapse
|
28
|
Sakurada K, McDonald F, Shimada F. Regenerative Medicine and Stem Cell Based Drug Discovery. Angew Chem Int Ed Engl 2008; 47:5718-38. [DOI: 10.1002/anie.200700724] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
29
|
|
30
|
Autologous umbilical cord blood infusion for type 1 diabetes. Exp Hematol 2008; 36:710-5. [PMID: 18358588 DOI: 10.1016/j.exphem.2008.01.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 10/31/2007] [Accepted: 01/23/2008] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The physical, emotional, and economic costs of type 1 diabetes (T1D) mandate continued efforts to develop effective strategies to prevent or reverse the disease. Herein, we describe the scientific and therapeutic rationale underlying efforts utilizing umbilical cord blood (UCB) as a therapy for ameliorating the progression of this autoimmune disease. MATERIALS AND METHODS We recently embarked on a pilot study to document the safety and potential efficacy of autologous UCB infusion in subjects with T1D. Under this protocol, patients recently diagnosed with the disease and for whom autologous cord blood is stored, undergo infusion. Studies are performed before infusion and every 3 to 6 months postinfusion for immunologic and metabolic assessment. To date, 15 autologous infusions have been performed. RESULTS Preliminary observations suggest that autologous cord blood transfusion is safe and provides some slowing of the loss of endogenous insulin production in children with T1D. Mechanistic studies demonstrate that umbilical cord blood contains highly functional populations of regulatory T cells (Treg) and that increased Treg populations may be found in the peripheral blood of subjects more than 6 months after cord blood infusion. We provide the rationale for cord blood-based therapies, a summary of our initial protocol, and plans for future studies designed to explore the potential of cord blood-derived regulatory T cells to treat T1D. CONCLUSIONS Prolonged follow-up and additional mechanistic efforts are urgently needed to determine if umbilical cord blood-derived stem cells can be used as part of safe and effective therapies for T1D.
Collapse
|