1
|
Nemati M, Alizadeh AA, Dastghaib S, Saki F. Vitamin D supplementation affects bone marrow-derived mesenchymal stem cells differentiation into insulin-producing cells. Mol Biol Rep 2024; 51:748. [PMID: 38874843 DOI: 10.1007/s11033-024-09681-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Background this study was conducted to assess the effects of vitamin D on differentiation of bone marrow- derived mesenchymal stem cells (BM-MSCs) into insulin producing cells (IPCs). Method BM-MSCs were isolated from femur and tibia of rats and incubated in low (LG) or high glucose (HG) (5mM or 25mM), or high glucose DMEM media supplemented with vitamin D (0.2nM) (HGD) for 14 days. Cells viability was analysis by MTT assay. Differentiation of SCs was confirmed using measuring genes expression level of pdx1 and insulin, and insulin secretion, glucose stimulated insulin secretion, and insulin content by ELISA method. Results Cell viability was significantly higher in HGD than LG (p < 0.05) in day 3, also, in HG and HGD than LG (p < 0.001), and HGD vs. HG (p < 0.001) in day 7. Pdx1 and insulin level was markedly higher in HGD than LG (p < 0.05 and p < 0.01). pdx1 expression was markedly higher in HGD (p < 0.05) than LG, also insulin expression the HG (p < 0.05), and HGD (p < 0.01) groups compared to the LG group. Insulin release at 5mM glucose was notably higher in the HGD group compared to LG (p < 0.05), and at 25mM glucose, both HG and HGD showed significant increases vs. LG (p < 0.05 and p < 0.01, respectively). Insulin content was significantly higher in both 5mM and 25mM glucose for HG and HGD vs. LG (p < 0.01 and p < 0.001, respectively). In conclusion, treatment BM-MSCs with vitamin D could increase their differentiation into IPCs and it can be considered as a potential supplementary agent in enhancing differentiation SCs into insulin generating cells.
Collapse
Affiliation(s)
- Marzieh Nemati
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Ali Akbar Alizadeh
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Science, Shiraz, Iran
- Department of Tissue Engineering, School of Advanced Technology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Dastghaib
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Science, Shiraz, Iran
- Authophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Forough Saki
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Science, Shiraz, Iran.
| |
Collapse
|
2
|
Ghoneim MA, Gabr MM, El-Halawani SM, Refaie AF. Current status of stem cell therapy for type 1 diabetes: a critique and a prospective consideration. Stem Cell Res Ther 2024; 15:23. [PMID: 38281991 PMCID: PMC10823744 DOI: 10.1186/s13287-024-03636-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024] Open
Abstract
Over the past decade, there had been progress in the development of cell therapy for insulin-dependent diabetes. Nevertheless, important hurdles that need to be overcome still remain. Protocols for the differentiation of pluripotent stem cells into pancreatic progenitors or fully differentiated β-cells have been developed. The resulting insulin-producing cells can control chemically induced diabetes in rodents and were the subject of several clinical trials. However, these cells are immunogenic and possibly teratogenic for their transplantation, and an immunoisolation device and/or immunosuppression is needed. A growing number of studies have utilized genetic manipulations to produce immune evasive cells. Evidence must be provided that in addition to the expected benefit, gene manipulations should not lead to any unforeseen complications. Mesenchymal stem/stromal cells (MSCs) can provide a viable alternative. MSCs are widely available from many tissues. They can form insulin-producing cells by directed differentiation. Experimentally, evidence has shown that the transplantation of allogenic insulin-producing cells derived from MSCs is associated with a muted allogeneic response that does not interfere with their functionality. This can be explained by the immunomodulatory functions of the MSC subpopulation that did not differentiate into insulin-producing cells. Recently, exosomes derived from naive MSCs have been used in the experimental domain to treat diabetes in rodents with varying degrees of success. Several mechanisms for their beneficial functions were proposed including a reduction in insulin resistance, the promotion of autophagy, and an increase in the T regulatory population. However, euglycemia was not achieved in any of these experiments. We suggest that exosomes derived from β-cells or insulin-producing cells (educated) can provide a better therapeutic effect than those derived from undifferentiated cells.
Collapse
|
3
|
Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines 2023; 11:2558. [PMID: 37761001 PMCID: PMC10527322 DOI: 10.3390/biomedicines11092558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by a gradual destruction of insulin-producing β-cells in the endocrine pancreas due to innate and specific immune responses, leading to impaired glucose homeostasis. T1D patients usually require regular insulin injections after meals to maintain normal serum glucose levels. In severe cases, pancreas or Langerhans islet transplantation can assist in reaching a sufficient β-mass to normalize glucose homeostasis. The latter procedure is limited because of low donor availability, high islet loss, and immune rejection. There is still a need to develop new technologies to improve islet survival and implantation and to keep the islets functional. Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells with high plasticity that can support human pancreatic islet function both in vitro and in vivo and islet co-transplantation with MSCs is more effective than islet transplantation alone in attenuating diabetes progression. The beneficial effect of MSCs on islet function is due to a combined effect on angiogenesis, suppression of immune responses, and secretion of growth factors essential for islet survival and function. In this review, various aspects of MSCs related to islet function and diabetes are described.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ronit Ahdut-HaCohen
- Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel;
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| |
Collapse
|
4
|
Ghoneim MA, Gabr MM, Refaie AF, El-Halawani SM, Al-Issawi MM, Elbassiouny BL, Kader MAAE, Ismail AM, Zidan MF, Karras MS, Magar RW, Khater SM, Ashamallah SA, Zakaria MM, Kloc M. Transplantation of insulin-producing cells derived from human mesenchymal stromal/stem cells into diabetic humanized mice. Stem Cell Res Ther 2022; 13:350. [PMID: 35883190 PMCID: PMC9327173 DOI: 10.1186/s13287-022-03048-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background The purpose of this study was to investigate allogenic immune responses following the transplantation of insulin-producing cells (IPCs) differentiated from human adipose tissue-derived stem cells (hAT-MSCs) into humanized mice. Methods hAT-MSCs were isolated from liposuction aspirates obtained from HLA-A2-negative healthy donors. These cells were expanded and differentiated into IPCs. HLA-A2-positive humanized mice (NOG-EXL) were divided into 4 groups: diabetic mice transplanted with IPCs, diabetic but nontransplanted mice, nondiabetic mice transplanted with IPCs and normal untreated mice. Three million differentiated cells were transplanted under the renal capsule. Animals were followed-up to determine their weight, glucose levels (2-h postprandial), and human and mouse insulin levels. The mice were euthanized 6–8 weeks posttransplant. The kidneys were explanted for immunohistochemical studies. Blood, spleen and bone marrow samples were obtained to determine the proportion of immune cell subsets (CD4+, CD8+, CD16+, CD19+ and CD69+), and the expression levels of HLA-ABC and HLA-DR. Results Following STZ induction, blood glucose levels increased sharply and were then normalized within 2 weeks after cell transplantation. In these animals, human insulin levels were measurable while mouse insulin levels were negligible throughout the observation period. Immunostaining of cell-bearing kidneys revealed sparse CD45+ cells. Immunolabeling and flow cytometry of blood, bone marrow and splenic samples obtained from the 3 groups of animals did not reveal a significant difference in the proportions of immune cell subsets or in the expression levels of HLA-ABC and HLA-DR. Conclusion Transplantation of IPCs derived from allogenic hAT-MSCs into humanized mice was followed by a muted allogenic immune response that did not interfere with the functionality of the engrafted cells. Our findings suggest that such allogenic cells could offer an opportunity for cell therapy for insulin-dependent diabetes without immunosuppression, encapsulation or gene manipulations. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03048-y.
Collapse
Affiliation(s)
| | - Mahmoud M Gabr
- Biotechnology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Ayman F Refaie
- Nephrology Department, Urology and Nephrology Center, Mansoura, Egypt
| | | | - Mohga M Al-Issawi
- Biotechnology Department, Urology and Nephrology Center, Mansoura, Egypt
| | | | - Mai A Abd El Kader
- Biotechnology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Amani M Ismail
- Immunology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Mona F Zidan
- Microbiology and Immunology Research Program, Children's Hospital 57357, Cairo, Egypt
| | - Mary S Karras
- Immunology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Raghda W Magar
- Immunology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Sherry M Khater
- Pathology Department, Urology and Nephrology Center, Mansoura, Egypt
| | | | - Mahmoud M Zakaria
- Biotechnology Department, Urology and Nephrology Center, Mansoura, Egypt
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA.,The Houston Methodist Hospital, Houston, TX, USA.,The University of Texas, M.D. Anderson Cancer Center, Houston TX, USA
| |
Collapse
|
5
|
Hashemi J, Barati G, Bibak B. Decellularized Matrix Bioscaffolds: Implementation of Native Microenvironment in Pancreatic Tissue Engineering. Pancreas 2021; 50:942-951. [PMID: 34643609 DOI: 10.1097/mpa.0000000000001868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
ABSTRACT Type 1 diabetes is an autoimmune disease, and its incidence is usually estimated in the range of 5% to 10%. Currently, the administration of exogenous insulin is the standard of care therapy. However, this therapy is not effective in some patients who may develop some chronic complications. Islet transplantation into the liver is another therapy with promising outcomes; however, the long-term efficacy of this therapeutic option is limited to a small number of patients. Because native extracellular matrix (ECM) components provide a suitable microenvironment for islet functions, engineering a 3-dimensional construct that recapitulates the native pancreatic environment could address these obstacles. Many attempts have been conducted to mimic an in vivo microenvironment to increase the survival of islets or islet-like clusters. With the advent of decellularization technology, it is possible to use a native ECM in organ engineering. Pancreatic decellularized bioscaffold provides proper cell-cell and cell-ECM interactions and retains growth factors that are critical in the determination of cell fate within a native organ. This review summarizes the current knowledge of decellularized matrix technology and addresses its possible limitations before use in the clinic.
Collapse
Affiliation(s)
- Javad Hashemi
- From the Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd
| | | | | |
Collapse
|
6
|
Generation of high yield insulin-producing cells (IPCs) from various sources of stem cells. VITAMINS AND HORMONES 2021; 116:235-268. [PMID: 33752820 DOI: 10.1016/bs.vh.2021.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Type 1 diabetes mellitus occurs when beta cell mass is reduced to less than 20% of the normal level due to immune system destruction of beta cell resulting in an inability to secrete enough insulin. The prevalence of diabetes is expanding according to the American Diabetes Association and the World Health Organization (WHO), foretold to exceed 350 million by 2030. The current treatment does not cure many of the serious complications associated with the disease such as neuropathy, nephropathy, dyslipidemia, retinopathy and cardiovascular disease. Whole pancreas or isolated pancreatic islet transplantation as an alternative therapy can prevent or reduce some of the complications of diabetes. However, the shortage of matched organ or islets cells donor and alloimmune responses limit this therapeutic strategy. Recently, several reports have raised extremely promising results to use different sources of stem cells to differentiate insulin-producing cells and focus on the expansion of these alternative sources. Stem cells, due to their potential for multiple differentiation and self-renewal can differentiate into all cell types, including insulin-producing cells (IPCs). Generation of new beta cells can be achieved from various stem cell sources, including embryonic stem cells (ESCs), adult stem cells, such as mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs). Thus, this chapter discusses on the assistance of cellular reprogramming of various stem cells as candidates for the generation of IPCs using transcription factors/miRNA, cytokines/small molecules and tissue engineering.
Collapse
|
7
|
Efficiency of Stem Cell (SC) Differentiation into Insulin-Producing Cells for Treating Diabetes: a Systematic Review. Stem Cells Int 2021; 2021:6652915. [PMID: 33727934 PMCID: PMC7935591 DOI: 10.1155/2021/6652915] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 01/04/2023] Open
Abstract
Over the recent years, the use of stem cells has provided a new opportunity to treat various disorders including diabetes. Stem cells are unspecialized cells with a capacity for self-renewal and differentiation into more specialized cell types. Many factors contribute to the differentiation of SCs and thus play an important role in regulating the fate of stem cells. Accordingly, a wide range of protocols has been used to differentiate SCs to insulin-producing cells but the effectiveness of SC differentiation varies. The aim of this systematic review was to evaluate the results obtained from different studies on SC differentiation for higher efficacy to treat diabetes. This search was done in PubMed, Web of Science (WOS), and Scopus using keywords “insulin-producing cell (IPC),” “pancreatic B cell,” “insulin-secreting cell,” “stem cell,” “progenitor cells,” “mother cell,” and “colony-forming unit.” Among more than 3646 papers, 32 studies were considered eligible for more evaluations. The obtained results indicated that most of the studies were performed on the mesenchymal stem cells (MSCs) derived from different tissues as compared with other types of SCs. Different evaluations of in vitro studies as well as animal models supported their role in the recovery of diabetes. In the present review, we summarize and discuss recent advances in increasing the efficiency of SC differentiation using different materials, but despite the promising results of this systematic review, further studies are needed to assess the efficiency and safety of transplantation of these cells in diabetes recovery.
Collapse
|
8
|
Ghoneim MA, Refaie AF, Elbassiouny BL, Gabr MM, Zakaria MM. From Mesenchymal Stromal/Stem Cells to Insulin-Producing Cells: Progress and Challenges. Stem Cell Rev Rep 2020; 16:1156-1172. [PMID: 32880857 PMCID: PMC7667138 DOI: 10.1007/s12015-020-10036-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) are an attractive option for cell therapy for type 1 diabetes mellitus (DM). These cells can be obtained from many sources, but bone marrow and adipose tissue are the most studied. MSCs have distinct advantages since they are nonteratogenic, nonimmunogenic and have immunomodulatory functions. Insulin-producing cells (IPCs) can be generated from MSCs by gene transfection, gene editing or directed differentiation. For directed differentiation, MSCs are usually cultured in a glucose-rich medium with various growth and activation factors. The resulting IPCs can control chemically-induced diabetes in immune-deficient mice. These findings are comparable to those obtained from pluripotent cells. PD-L1 and PD-L2 expression by MSCs is upregulated under inflammatory conditions. Immunomodulation occurs due to the interaction between these ligands and PD-1 receptors on T lymphocytes. If this function is maintained after differentiation, life-long immunosuppression or encapsulation could be avoided. In the clinical setting, two sites can be used for transplantation of IPCs: the subcutaneous tissue and the omentum. A 2-stage procedure is required for the former and a laparoscopic procedure for the latter. For either site, cells should be transplanted within a scaffold, preferably one from fibrin. Several questions remain unanswered. Will the transplanted cells be affected by the antibodies involved in the pathogenesis of type 1 DM? What is the functional longevity of these cells following their transplantation? These issues have to be addressed before clinical translation is attempted. Graphical Abstract Bone marrow MSCs are isolated from the long bone of SD rats. Then they are expanded and through directed differentiation insulin-producing cells are formed. The differentiated cells are loaded onto a collagen scaffold. If one-stage transplantation is planned, a drug delivery system must be incorporated to ensure immediate oxygenation, promote vascularization and provide some growth factors. Some mechanisms involved in the immunomodulatory function of MSCs. These are implemented either by cell to cell contact or by the release of soluble factors. Collectively, these pathways results in an increase in T-regulatory cells.
Collapse
|
9
|
Afflerbach AK, Kiri MD, Detinis T, Maoz BM. Mesenchymal Stem Cells as a Promising Cell Source for Integration in Novel In Vitro Models. Biomolecules 2020; 10:E1306. [PMID: 32927777 PMCID: PMC7565384 DOI: 10.3390/biom10091306] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023] Open
Abstract
The human-relevance of an in vitro model is dependent on two main factors-(i) an appropriate human cell source and (ii) a modeling platform that recapitulates human in vivo conditions. Recent years have brought substantial advancements in both these aspects. In particular, mesenchymal stem cells (MSCs) have emerged as a promising cell source, as these cells can differentiate into multiple cell types, yet do not raise the ethical and practical concerns associated with other types of stem cells. In turn, advanced bioengineered in vitro models such as microfluidics, Organs-on-a-Chip, scaffolds, bioprinting and organoids are bringing researchers ever closer to mimicking complex in vivo environments, thereby overcoming some of the limitations of traditional 2D cell cultures. This review covers each of these advancements separately and discusses how the integration of MSCs into novel in vitro platforms may contribute enormously to clinical and fundamental research.
Collapse
Affiliation(s)
- Ann-Kristin Afflerbach
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
- Faculty of Biosciences, Universität Heidelberg, 69120 Heidelberg, Germany
| | - Mark D. Kiri
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
| | - Tahir Detinis
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
| | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel; (A.-K.A.); (M.D.K.); (T.D.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
10
|
Ebrahiminia M, Esmaeili F, Shabani L. In vitro differentiation induction of embryonal carcinoma stem cells into insulin-producing cells by Cichorium intybus L. leaf extract. JOURNAL OF ETHNOPHARMACOLOGY 2020; 246:112214. [PMID: 31491437 DOI: 10.1016/j.jep.2019.112214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/26/2019] [Accepted: 09/02/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Medicinal herb Cichorium intybus L. (chicory) has been used traditionally for the treatment of various diseases, including diabetes. One of the promising therapeutic options to treat diabetes is replacing the degenerative pancreatic β cells by stem cell-derived IPCs (insulin-producing cells). AIM OF THE STUDY By the combination of cell therapy as a modern approach and traditional medicine, the current study was designed to evaluate the effects of chicory leaf extract (LE) on the differentiation potential of P19 EC cells (an embryonal carcinoma stem cell line) into IPCs. MATERIALS AND METHODS The plant (voucher no. 4567) were collected and deposited in the herbarium of Shahrekord University. In vitro experiments were designed to compare the effects of various concentrations of LE on the differentiation potential of P19 EC cells. RESULTS The differentiated cells showed morphological characteristics of pancreatic β cells. They could also synthesized and secreted insulin when exposed to glucose. Moreover, the cells expressed specific proteins and genes of mature pancreatic β cells. CONCLUSIONS In conclusion, LE as a natural herbal extract was efficiently able to induce the differentiation of P19 EC cells into the clusters similar to pancreatic islets with the molecular, cellular and functional characteristics of mature β cells.
Collapse
Affiliation(s)
- M Ebrahiminia
- Research Institute of Biotechnology, Shahrekord University, Shahrekord, 115, Iran
| | - F Esmaeili
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezarjerib Avenue, Isfahan, 8174673441, Iran.
| | - L Shabani
- Research Institute of Biotechnology, Shahrekord University, Shahrekord, 115, Iran; Department of Biology, Faculty of Sciences, Shahrekord University, Shahrekord, Iran.
| |
Collapse
|
11
|
Abazari MF, Nasiri N, Nejati F, Zare Karizi S, Amini Faskhodi M, Saburi E, Aghapur N, Mahdavi MR, Ardeshirylajimi A, Enderami SE, Soleimanifar F. Comparison of human‐induced pluripotent stem cells and mesenchymal stem cell differentiation potential to insulin producing cells in 2D and 3D culture systems in vitro. J Cell Physiol 2019; 235:4239-4246. [DOI: 10.1002/jcp.29298] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/30/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Mohammad Foad Abazari
- Research Center for Clinical Virology Tehran University of Medical Sciences Tehran Iran
| | - Navid Nasiri
- Department of Biology, Central Tehran Branch Islamic Azad University Tehran Iran
| | - Fatemeh Nejati
- Department of Biology, Central Tehran Branch Islamic Azad University Tehran Iran
| | - Shohreh Zare Karizi
- Department of biology, Varamin Pishva branch Islamic Azad University Pishva Varamin Iran
| | | | - Ehsan Saburi
- Medical Genetics and Molecular Medicine Department, School of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Nasrin Aghapur
- Department of Biotechnology, Faculty of Science University of Tehran Tehran Iran
| | | | | | - Seyed Ehsan Enderami
- Diabetes Research Center Mazandaran University of Medical Sciences Sari Iran
- Immunogenetics Research Center, Department of Medical Biotechnology Mazandaran University of Medical Sciences Sari Iran
| | - Fatemeh Soleimanifar
- Dietary Supplements and Probiotic Research Center Alborz University of Medical Sciences Karaj Iran
- Department of Medical Biotechnology, School of Medicine Alborz University of Medical Sciences Karaj Iran
| |
Collapse
|
12
|
Chehelcheraghi F, Chien S, Bayat M. Mesenchymal stem cells improve survival in ischemic diabetic random skin flap via increased angiogenesis and VEGF expression. J Cell Biochem 2019; 120:17491-17499. [PMID: 31127644 DOI: 10.1002/jcb.29013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 01/13/2023]
Abstract
Random skin flaps (RSFs) are cutaneous flaps. Despite the negative impact of diabetes mellitus (DM) on RSF viability, they are commonly used in diabetic patients. In this study, we have assessed bone marrow mesenchymal stem cell (BMMSC) treatment on RSF survival, tensiometrical parameters, angiogenesis, and mast cells (MCs) count in an ischemic RSF model in rats with type 1 DM (T1DM). We induced T1DM in 30 Wistar adult male rats. The animals were assigned to three groups of 10 rats per group as follows: group 1 (control); group 2 (placebo), and group 3 (BMMSCs). A 30 × 80 mm RSF was created in each rat. On day 7, we measured the viable portion of each RSF. A sample was taken for histological and immunohistochemistry studies, fibroblasts, MCs, angiogenesis, collagen bundle density, and the presence of vascular endothelial growth factor (VEGF)+ cells. An additional sample was taken to evaluate the flap's incision strength. Treatment with BMMSCs (17.8 ± 0.37) significantly increased RSF survival compared with the control (13.3 ± 0.35) and placebo (16.1 ± 0.27) groups (one-way analysis of variance, P = .000; least significant difference, P = .000, P = .002). There was a significant improvement in angiogenesis, as confirmed by stereologic examination. Assessment of VEGF+ cells showed prominent neovascularization in BMMSC-treated RSFs compared with the control and placebo groups. Subdermal injection of BMMSC significantly increased ischemic RSF survival as a result of stimulated neovascularization in T1DM rats. Treatment of diabetic RSF with BMMSCs showed no beneficial effects in the fibroblast number and biomechanical parameters for the repair of ischemic wounds in the rat model. Treatment with BMMSCs significantly increased collagen bundle density.
Collapse
Affiliation(s)
- Farzaneh Chehelcheraghi
- Department of Anatomical Sciences, School of Medicine, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky
| | - Mohammad Bayat
- Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Noveratech LLC of Louisville, Louisville, Kentucky
| |
Collapse
|
13
|
Cañibano-Hernández A, Saenz del Burgo L, Espona-Noguera A, Orive G, Hernández RM, Ciriza J, Pedraz JL. Hyaluronic Acid Promotes Differentiation of Mesenchymal Stem Cells from Different Sources toward Pancreatic Progenitors within Three-Dimensional Alginate Matrixes. Mol Pharm 2019; 16:834-845. [DOI: 10.1021/acs.molpharmaceut.8b01126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Alberto Cañibano-Hernández
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine, CIBER-BBN, Vitoria-Gasteiz 01006, Spain
| | - Laura Saenz del Burgo
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine, CIBER-BBN, Vitoria-Gasteiz 01006, Spain
| | - Albert Espona-Noguera
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine, CIBER-BBN, Vitoria-Gasteiz 01006, Spain
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine, CIBER-BBN, Vitoria-Gasteiz 01006, Spain
| | - Rosa M. Hernández
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine, CIBER-BBN, Vitoria-Gasteiz 01006, Spain
| | - Jesús Ciriza
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine, CIBER-BBN, Vitoria-Gasteiz 01006, Spain
| | - José Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine, CIBER-BBN, Vitoria-Gasteiz 01006, Spain
| |
Collapse
|
14
|
Salg GA, Giese NA, Schenk M, Hüttner FJ, Felix K, Probst P, Diener MK, Hackert T, Kenngott HG. The emerging field of pancreatic tissue engineering: A systematic review and evidence map of scaffold materials and scaffolding techniques for insulin-secreting cells. J Tissue Eng 2019; 10:2041731419884708. [PMID: 31700597 PMCID: PMC6823987 DOI: 10.1177/2041731419884708] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/04/2019] [Indexed: 12/18/2022] Open
Abstract
A bioartificial endocrine pancreas is proposed as a future alternative to current treatment options. Patients with insulin-secretion deficiency might benefit. This is the first systematic review that provides an overview of scaffold materials and techniques for insulin-secreting cells or cells to be differentiated into insulin-secreting cells. An electronic literature survey was conducted in PubMed/MEDLINE and Web of Science, limited to the past 10 years. A total of 197 articles investigating 60 different materials met the inclusion criteria. The extracted data on materials, cell types, study design, and transplantation sites were plotted into two evidence gap maps. Integral parts of the tissue engineering network such as fabrication technique, extracellular matrix, vascularization, immunoprotection, suitable transplantation sites, and the use of stem cells are highlighted. This systematic review provides an evidence-based structure for future studies. Accumulating evidence shows that scaffold-based tissue engineering can enhance the viability and function or differentiation of insulin-secreting cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Gabriel Alexander Salg
- Department of General, Abdominal and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Nathalia A Giese
- Department of General, Abdominal and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Miriam Schenk
- Department of General, Abdominal and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix J Hüttner
- Department of General, Abdominal and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus Felix
- Department of General, Abdominal and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Pascal Probst
- Department of General, Abdominal and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus K Diener
- Department of General, Abdominal and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Abdominal and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Hannes Götz Kenngott
- Department of General, Abdominal and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
15
|
In vitro differentiation of human multilineage differentiating stress-enduring (Muse) cells into insulin producing cells. J Genet Eng Biotechnol 2018; 16:433-440. [PMID: 30733757 PMCID: PMC6354004 DOI: 10.1016/j.jgeb.2018.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/09/2018] [Indexed: 12/30/2022]
Abstract
Mesenchymal stem cells (MSCs) is a heterogeneous population. Muse cells is a rare pluripotent subpopulation within MSCs. This study aims to evaluate the pulirpotency and the ability of Muse cells to generate insulin producing cells (IPCs) after in vitro differentiation protocol compared to the non-Muse cells. Muse cells were isolated by FACSAria III cell sorter from adipose-derived MSCs and were evaluated for its pluripotency. Following in vitro differentiation, IPCs derived from Muse and non-Muse cells were evaluated for insulin production. Muse cells comprised 3.2 ± 0.7% of MSCs, approximately 82% of Muse cells were positive for anti stage-specific embryonic antigen-3 (SSEA-3). Pluripotent markers were highly expressed in Muse versus non-Muse cells. The percentage of generated IPCs by flow cytometric analysis was higher in Muse cells. Under confocal microscopy, Muse cells expressed insulin and c-peptide while it was undetected in non-Muse cells. Our results introduced Muse cells as a new adult pluripotent subpopulation, which is capable to produce higher number of functional IPCs.
Collapse
|
16
|
Gharravi AM, Jafar A, Ebrahimi M, Mahmodi A, Pourhashemi E, Haseli N, Talaie N, Hajiasgarli P. Current status of stem cell therapy, scaffolds for the treatment of diabetes mellitus. Diabetes Metab Syndr 2018; 12:1133-1139. [PMID: 30168429 DOI: 10.1016/j.dsx.2018.06.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 06/25/2018] [Indexed: 12/24/2022]
Abstract
Diabetes mellitus (DM) remains the 7th leading cause of death in the world. Daily insulin injection is one component of a treatment plan for people with Diabetes mellitus type 1 (T1DM) that restores normal or near-normal blood sugar levels. However, Insulin treatment depends upon a variety of individual factors and leads to poor and drastic glycemic control. The need for an effective cell replacement strategy will be the aim of future clinical trials. Therefore, the aim of this systematic review is to outline the latest advances in scaffolding and stem cell therapy as a non-pharmacologic treatment for T1DM. It also emphasizes on some pancreas differentiation protocols and the clinical trials associated with stem cell therapy regarding T1DM in vitro and in vivo.
Collapse
Affiliation(s)
- Anneh Mohammad Gharravi
- Stem Cells and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Alireza Jafar
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mehrdad Ebrahimi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ahmad Mahmodi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Erfan Pourhashemi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Nasrin Haseli
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Niloofar Talaie
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Parinaz Hajiasgarli
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
17
|
Abazari MF, Soleimanifar F, Nouri Aleagha M, Torabinejad S, Nasiri N, Khamisipour G, Amini Mahabadi J, Mahboudi H, Enderami SE, Saburi E, Hashemi J, Kehtari M. PCL/PVA nanofibrous scaffold improve insulin-producing cells generation from human induced pluripotent stem cells. Gene 2018; 671:50-57. [DOI: 10.1016/j.gene.2018.05.115] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 05/28/2018] [Accepted: 05/30/2018] [Indexed: 02/08/2023]
|
18
|
Karimi S, Ai J, Khorsandi L, Bijan Nejad D, Saki G. Vildagliptin Enhances Differentiation of Insulin Producing Cells from Adipose-Derived Mesenchymal Stem Cells. CELL JOURNAL 2018; 20:477-482. [PMID: 30123993 PMCID: PMC6099143 DOI: 10.22074/cellj.2019.5542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/20/2017] [Indexed: 12/11/2022]
Abstract
Objective Type 1 diabetes is caused by destruction of beta cells of pancreas. Vildagliptin (VG), a dipeptidyl peptidase IV
(DPP IV) inhibitor, is an anti-diabetic drug, which increases beta cell mass. In the present study, the effects of VG on generation
of insulin-producing cells (IPCs) from adipose-derived mesenchymal stem cells (ASCs) is investigated.
Materials and Methods In this experimental study, ASCs were isolated and after characterization were exposed to
differentiation media with or without VG. The presence of IPCs was confirmed by morphological analysis and gene expression
(Pdx-1, Glut-2 and Insulin). Newport Green staining was used to determine insulin-positive cells. Insulin secretion under
different concentrations of glucose was measured using radioimmunoassay method.
Results In the presence of VG the morphology of differentiated cells was similar to the pancreatic islet cells. Expression
of Pdx-1, Glut-2 and Insulin genes in VG-treated cells was significantly higher than the cells exposed to induction media
only. Insulin release from VG-treated ASCs showed a nearly 3.6 fold (P<0.05) increase when exposed to a high-
glucose medium in comparison to untreated ASCs. The percentage of insulin-positive cells in the VG-treated cells was
approximately 2.9-fold higher than the untreated ASCs.
Conclusion The present study has demonstrated that VG elevates differentiation of ASCs into IPCs. Improvement of this
protocol may be used in cell therapy in diabetic patients.
Collapse
Affiliation(s)
- Samaneh Karimi
- Cell and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Jafar Ai
- Tissue Engineering and Applied Cell Sciences, Department-School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Layasadat Khorsandi
- Cell and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. Electronic Address:
| | - Darioush Bijan Nejad
- Cell and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ghasem Saki
- Cell and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
19
|
Human urine-derived stem cells play a novel role in the treatment of STZ-induced diabetic mice. J Mol Histol 2018; 49:419-428. [PMID: 29675567 DOI: 10.1007/s10735-018-9772-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Human urine-derived stem cells (hUSCs) are a potential stem cell source for cell therapy. However, the effect of hUSCs on glucose metabolism regulation in type 1 diabetes was not clear. Therefore, the aim of the study was to evaluate whether hUSCs have protective effect on streptozotocin (STZ)-induced diabetes. hUSCs were extracted and cultivated with a special culture medium. Flow cytometry analysis was applied to detect cell surface markers. BALB/c male nude mice were either injected with high-dose STZ (HD-STZ) or multiple low-dose STZ (MLD-STZ). Serum and pancreatic insulin were measured, islet morphology and its vascularization were investigated. hUSCs highly expressed CD29, CD73, CD90 and CD146, and could differentiate into, at least, bone and fat in vitro. Transplantation of hUSCs into HD-STZ treated mice prolonged the median survival time and improved their blood glucose, and into those with MLD-STZ improved the glucose tolerance, islet morphology and increased the serum and pancreas insulin content. Furthermore, CD31 expression increased significantly in islets of BALB/c nude mice treated with hUSCs compared to those of un-transplanted MLD-STZ mice. hUSCs could improve the median survival time and glucose homeostasis in STZ-treated mice through promoting islet vascular regeneration and pancreatic beta-cell survival.
Collapse
|
20
|
Kumar N, Joisher H, Ganguly A. Polymeric Scaffolds for Pancreatic Tissue Engineering: A Review. Rev Diabet Stud 2018; 14:334-353. [PMID: 29590227 PMCID: PMC6230446 DOI: 10.1900/rds.2017.14.334] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/24/2018] [Accepted: 02/05/2018] [Indexed: 12/17/2022] Open
Abstract
In recent years, there has been an alarming increase in the incidence of diabetes, with one in every eleven individuals worldwide suffering from this debilitating disease. As the available treatment options fail to reduce disease progression, novel avenues such as the bioartificial pancreas are being given serious consideration. In the past decade, the research focus has shifted towards the field of tissue engineering, which helps to design biological substitutes for repair and replacement of non-functional or damaged organs. Scaffolds constitute an integral part of tissue engineering; they have been shown to mimic the native extracellular matrix, thereby supporting cell viability and proliferation. This review offers a novel compilation of the recent advances in polymeric scaffolds, which are used for pancreatic tissue engineering. Furthermore, in this article, the design strategies for bioartificial pancreatic constructs and their future applications in cell-based therapy are discussed.
Collapse
Affiliation(s)
| | | | - Anasuya Ganguly
- Department of Biological Sciences, BITS-Pilani, K.K Birla Goa Campus, Goa, India 403726
| |
Collapse
|
21
|
Chehelcheraghi F, Abbaszadeh A, Tavafi M. Skin Mast Cell Promotion in Random Skin Flaps in Rats using Bone Marrow Mesenchymal Stem Cells and Amniotic Membrane. IRANIAN BIOMEDICAL JOURNAL 2018; 22:322-30. [PMID: 29506347 PMCID: PMC6058190 DOI: 10.29252/ibj.22.5.322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background Skin flap procedures are employed in plastic surgery, but failure can lead to necrosis of the flap. Studies have used bone marrow mesenchymal stem cells (BM-MSCs) to improve flap viability. BM-MSCs and acellular amniotic membrane (AAM) have been introduced as alternatives. The objective of this study was to evaluate the effect of BM-MSCs and AAM on mast cells of random skin flaps (RSF) in rats. Methods RSFs (80 × 30 mm) were created on 40 rats that were randomly assigned to one of four groups, including (I) AAM, (II) BM-MSCs, (III) BM-MSCs/AAM, and (IV) saline (control). Transplantation was carried out during the procedure (zero day). Flap necrosis was observed on day 7, and skin samples were collected from the transition line of the flap to evaluate the total number and types of mast cells. The development and the total number of mast cells were related to the development of capillaries. Results The results of one-way ANOVA indicated that there was no statistically significant difference between the mean numbers of mast cell types for different study groups. However, the difference between the total number of mast cells in the study groups was statistically significant (p = 0.001). Conclusion The present study suggests that the use of AAM/BM-MSCs can improve the total number of mast cells and accelerate the growth of capillaries at the transient site in RSFs in rats.
Collapse
Affiliation(s)
- Farzaneh Chehelcheraghi
- Department of Anatomical Sciences, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Abolfazl Abbaszadeh
- Department of Surgery, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Magid Tavafi
- Department of Surgery, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
22
|
Enderami SE, Kehtari M, Abazari MF, Ghoraeian P, Nouri Aleagha M, Soleimanifar F, Soleimani M, Mortazavi Y, Nadri S, Mostafavi H, Askari H. Generation of insulin-producing cells from human induced pluripotent stem cells on PLLA/PVA nanofiber scaffold. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:1062-1069. [DOI: 10.1080/21691401.2018.1443466] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Seyed Ehsan Enderami
- Stem Cell Technology Research Center, Tehran, Iran
- Department of Medical Biotechnology and Nanotechnology, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mousa Kehtari
- School of Biology, College of Sciences, University of Tehran, Tehran, Iran
| | - Mohammad Foad Abazari
- Department of Genetics, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Pegah Ghoraeian
- Department of Genetics, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Nouri Aleagha
- Department of Genetics, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Soleimanifar
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Masoud Soleimani
- Department of Hematology, Tarbiat Modares University, Tehran, Iran
| | - Yousef Mortazavi
- Department of Medical Biotechnology and Nanotechnology, Zanjan University of Medical Sciences, Zanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Samad Nadri
- Department of Medical Biotechnology and Nanotechnology, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Mostafavi
- Department of Physiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hassan Askari
- Department of Physiology, School of Medicine, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Im GB, Bhang SH. Recent research trend in cell and drug delivery system for type 1 diabetes treatment. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2018. [DOI: 10.1007/s40005-017-0380-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
24
|
Wang H, Strange C, Nietert PJ, Wang J, Turnbull TL, Cloud C, Owczarski S, Shuford B, Duke T, Gilkeson G, Luttrell L, Hermayer K, Fernandes J, Adams DB, Morgan KA. Autologous Mesenchymal Stem Cell and Islet Cotransplantation: Safety and Efficacy. Stem Cells Transl Med 2018; 7:11-19. [PMID: 29159905 PMCID: PMC5746145 DOI: 10.1002/sctm.17-0139] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/28/2017] [Indexed: 01/01/2023] Open
Abstract
Islet engraftment after transplantation is impaired by high rates of islet/β cell death caused by cellular stressors and poor graft vascularization. We studied whether cotransplantation of ex vivo expanded autologous bone marrow-derived mesenchymal stem cells (MSCs) with islets is safe and beneficial in chronic pancreatitis patients undergoing total pancreatectomy with islet autotransplantation. MSCs were harvested from the bone marrow of three islet autotransplantation patients and expanded at our current Good Manufacturing Practices (cGMP) facility. On the day of islet transplantation, an average dose of 20.0 ± 2.6 ×106 MSCs was infused with islets via the portal vein. Adverse events and glycemic control at baseline, 6, and 12 months after transplantation were compared with data from 101 historical control patients. No adverse events directly related to the MSC infusions were observed. MSC patients required lower amounts of insulin during the peritransplantation period (p = .02 vs. controls) and had lower 12-month fasting blood glucose levels (p = .02 vs. controls), smaller C-peptide declines over 6 months (p = .01 vs. controls), and better quality of life compared with controls. In conclusion, our pilot study demonstrates that autologous MSC and islet cotransplantation may be a safe and potential strategy to improve islet engraftment after transplantation. (Clinicaltrials.gov registration number: NCT02384018). Stem Cells Translational Medicine 2018;7:11-19.
Collapse
Affiliation(s)
- Hongjun Wang
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Charlie Strange
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Paul J. Nietert
- Department of Public Health SciencesMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Jingjing Wang
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Taylor L. Turnbull
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Colleen Cloud
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Stefanie Owczarski
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Betsy Shuford
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Tara Duke
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Gary Gilkeson
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Louis Luttrell
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Kathie Hermayer
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Jyotika Fernandes
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - David B. Adams
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Katherine A. Morgan
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
25
|
Sharma A, Rani R. Do we really need to differentiate mesenchymal stem cells into insulin-producing cells for attenuation of the autoimmune responses in type 1 diabetes: immunoprophylactic effects of precursors to insulin-producing cells. Stem Cell Res Ther 2017; 8:167. [PMID: 28701182 PMCID: PMC5508489 DOI: 10.1186/s13287-017-0615-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 05/16/2017] [Accepted: 06/20/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a multifactorial autoimmune disorder where pancreatic beta cells are lost before the clinical manifestations of the disease. Administration of mesenchymal stem cells (MSCs) or MSCs differentiated into insulin-producing cells (IPCs) have yielded limited success when used therapeutically. We have evaluated the immunoprophylactic potentials of precursors to insulin-producing cells (pIPCs) and IPCs in nonobese diabetic (NOD) mice to ask a basic question: do we need to differentiate MSCs into IPCs or will pIPCs suffice to attenuate autoimmune responses in T1D? METHODS Bone marrow-derived MSCs from Balb/c mice were characterized following the International Society for Cellular Therapy (ISCT) guidelines. MSCs cultured in high-glucose media for 11 to 13 passages were characterized for the expression of pancreatic lineage genes using real-time polymerase chain reaction. Expression of the PDX1 gene in pIPCs was assessed using Western blot and fluorescence-activated cell sorting (FACS). Triple-positive MSCs were differentiated into IPCs using a three-step protocol after sorting them for cell surface markers, i.e. CD29, CD44, and SCA-1. Nonobese diabetic mice were administered pIPCs, IPCs, or phosphate-buffered saline (PBS) into the tail vein at weeks 9 or 10 and followed-up for 29-30 weeks for fasting blood glucose levels. Two consecutive blood sugar levels of more than 250 mg/dl were considered diabetic. RESULTS MSCs grown in high-glucose media for 11 to 13 passages expressed genes of the pancreatic lineage such as PDX1, beta2, neurogenin, PAX4, Insulin, and glucagon. Furthermore, Western blot and FACS analysis for PDX-1, a transcription factor necessary for beta cell maturation, confirmed that these cells were precursors of insulin-producing cells (pIPCs). NOD mice administered with pIPCs were better protected from developing diabetes with a protective efficacy of 78.4% (p < 0.009); however, administration of IPCs gave protective efficacy of 55% at the end of 28-30 weeks. CONCLUSIONS Precursors to insulin-producing cells seem to have better potential to arrest autoimmune response in type 1 diabetes when administered before the onset of the disease in NOD mice. When translated to humans, autologous mesenchymal stem cells grown in high-glucose media for 10 to 13 passages may have beneficial effects in individuals at high risk of developing type 1 diabetes.
Collapse
Affiliation(s)
- Anshu Sharma
- Molecular Immunogenetics Group, National Institute of Immunology, New Delhi, 110067, India
| | - Rajni Rani
- Molecular Immunogenetics Group, National Institute of Immunology, New Delhi, 110067, India. .,Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110025, India.
| |
Collapse
|
26
|
Vieira A, Druelle N, Avolio F, Napolitano T, Navarro-Sanz S, Silvano S, Collombat P. β-Cell Replacement Strategies: The Increasing Need for a "β-Cell Dogma". Front Genet 2017. [PMID: 28634486 PMCID: PMC5459879 DOI: 10.3389/fgene.2017.00075] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes is an auto-immune disease resulting in the loss of pancreatic β-cells and, consequently, in chronic hyperglycemia. Insulin supplementation allows diabetic patients to control their glycaemia quite efficiently, but treated patients still display an overall shortened life expectancy and an altered quality of life as compared to their healthy counterparts. In this context and due to the ever increasing number of diabetics, establishing alternative therapies has become a crucial research goal. Most current efforts therefore aim at generating fully functional insulin-secreting β-like cells using multiple approaches. In this review, we screened the literature published since 2011 and inventoried the selected markers used to characterize insulin-secreting cells generated by in vitro differentiation of stem/precursor cells or by means of in vivo transdifferentiation. By listing these features, we noted important discrepancies when comparing the different approaches for the initial characterization of insulin-producing cells as true β-cells. Considering the recent advances achieved in this field of research, the necessity to establish strict guidelines has become a subject of crucial importance, especially should one contemplate the next step, which is the transplantation of in vitro or ex vivo generated insulin-secreting cells in type 1 diabetic patients.
Collapse
Affiliation(s)
- Andhira Vieira
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Noémie Druelle
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Fabio Avolio
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Tiziana Napolitano
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Sergi Navarro-Sanz
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Serena Silvano
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| | - Patrick Collombat
- Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, iBV, Université Côte d'AzurNice, France
| |
Collapse
|
27
|
Enderami SE, Mortazavi Y, Soleimani M, Nadri S, Biglari A, Mansour RN. Generation of Insulin-Producing Cells From Human-Induced Pluripotent Stem Cells Using a Stepwise Differentiation Protocol Optimized With Platelet-Rich Plasma. J Cell Physiol 2017; 232:2878-2886. [DOI: 10.1002/jcp.25721] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 11/30/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Seyed Ehsan Enderami
- Department of Medical Biotechnology Nanotechnology; Faculty of Medicine; Zanjan University of Medical Sciences; Zanjan Iran
| | - Yousef Mortazavi
- Department of Medical Biotechnology Nanotechnology; Faculty of Medicine; Zanjan University of Medical Sciences; Zanjan Iran
- Cancer Gene Therapy Research Center; Zanjan University of Medical Sciences; Zanjan Iran
| | - Masoud Soleimani
- Department of Hematology; Faculty of Medical Sciences; Tarbiat Modares University; Tehran Iran
| | - Samad Nadri
- Department of Medical Biotechnology Nanotechnology; Faculty of Medicine; Zanjan University of Medical Sciences; Zanjan Iran
| | - Alireza Biglari
- Department of Molecular Medicine and Genetics; Faculty of Medicine; Zanjan University of Medical Sciences; Zanjan Iran
| | - Reyhaneh Nassiri Mansour
- Department of Clinical Biochemistry; Faculty of Medicine; Zanjan University of Medical Sciences; Zanjan Iran
| |
Collapse
|
28
|
Seyedi F, Farsinejad A, Nematollahi-Mahani SN. Fibrin scaffold enhances function of insulin producing cells differentiated from human umbilical cord matrix-derived stem cells. Tissue Cell 2017; 49:227-232. [DOI: 10.1016/j.tice.2017.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/05/2017] [Indexed: 11/29/2022]
|
29
|
Therapeutic efficacy of differentiated versus undifferentiated mesenchymal stem cells in experimental type I diabetes in rat. Biochem Biophys Rep 2016; 5:468-475. [PMID: 28955854 PMCID: PMC5600460 DOI: 10.1016/j.bbrep.2016.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 01/25/2016] [Accepted: 02/01/2016] [Indexed: 12/11/2022] Open
Abstract
Selective MSCs differentiation protocol into pancreatic beta cells was conducted in the present study using exendin-4 and TGF-beta. Differentiated and undifferentiated MSCs were assessed in experimental type I diabetes in rats. Ninety female white albino rats were included in the study and divided equally (n=15/group) into 6 groups: healthy control, healthy control rats received acellular tissue culture medium, diabetic rats, diabetic rats received acellular tissue culture medium, diabetic rats received undifferentiated MSCs and diabetic rats received differentiated MSCs. Therapeutic efficacy of undifferentiated versus differentiated MSCs was evaluated via assessment of quantitative gene expressions of insulin1, insulin 2, Smad-2, Smad-3, PDX-1, PAX-4, neuroD. Blood glucose and insulin hormone levels were also assessed. Results showed that quantitative gene expressions of all studied genes showed significant decrease in diabetic rat groups. Use of undifferentiated and differentiated MSCs led to a significant elevation of expression levels of all genes with more superior effect with differentiated MSCs except smad-2 gene. As regards insulin hormone levels, use of either undifferentiated or differentiated MSCs led to a significant elevation of its levels with more therapeutic effect with differentiated MSCs. Blood glucose levels were significantly decreased with both undifferentiated and differentiated MSCs in comparison to diabetic groups but its levels were normalized 2 months after injection of differentiated MSCs. In conclusion, use of undifferentiated or differentiated MSCs exhibited significant therapeutic potentials in experimental type I diabetes in rats with more significant therapeutic effect with the use of differentiated MSCs. Differentiated MSCs exhibited significant therapeutic potentials in type I diabetes. TGF-beta1 and exendin-4 enhance MSCs differentiation into pancreatic beta cells. Pancreatic lineage is evaluated by gene expressions of insulin-1, insulin-2. Pancreatic differentiation is evaluated by expressions of PDX-1, PAX-4 and NeuroD. Differentiated MSCs have more therapeutic potentials than undifferentiated MSCs.
Collapse
|
30
|
Wang Y, Hai T, Liu L, Liu Z, Zhou Q. Cell therapy in diabetes: current progress and future prospects. Sci Bull (Beijing) 2015. [DOI: 10.1007/s11434-015-0844-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
31
|
Akhmanova M, Osidak E, Domogatsky S, Rodin S, Domogatskaya A. Physical, Spatial, and Molecular Aspects of Extracellular Matrix of In Vivo Niches and Artificial Scaffolds Relevant to Stem Cells Research. Stem Cells Int 2015; 2015:167025. [PMID: 26351461 PMCID: PMC4553184 DOI: 10.1155/2015/167025] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/07/2015] [Accepted: 06/24/2015] [Indexed: 12/27/2022] Open
Abstract
Extracellular matrix can influence stem cell choices, such as self-renewal, quiescence, migration, proliferation, phenotype maintenance, differentiation, or apoptosis. Three aspects of extracellular matrix were extensively studied during the last decade: physical properties, spatial presentation of adhesive epitopes, and molecular complexity. Over 15 different parameters have been shown to influence stem cell choices. Physical aspects include stiffness (or elasticity), viscoelasticity, pore size, porosity, amplitude and frequency of static and dynamic deformations applied to the matrix. Spatial aspects include scaffold dimensionality (2D or 3D) and thickness; cell polarity; area, shape, and microscale topography of cell adhesion surface; epitope concentration, epitope clustering characteristics (number of epitopes per cluster, spacing between epitopes within cluster, spacing between separate clusters, cluster patterns, and level of disorder in epitope arrangement), and nanotopography. Biochemical characteristics of natural extracellular matrix molecules regard diversity and structural complexity of matrix molecules, affinity and specificity of epitope interaction with cell receptors, role of non-affinity domains, complexity of supramolecular organization, and co-signaling by growth factors or matrix epitopes. Synergy between several matrix aspects enables stem cells to retain their function in vivo and may be a key to generation of long-term, robust, and effective in vitro stem cell culture systems.
Collapse
Affiliation(s)
| | - Egor Osidak
- Imtek Limited, 3 Cherepkovskaya 15, Moscow 21552, Russia
- Gamaleya Research Institute of Epidemiology and Microbiology Federal State Budgetary Institution, Ministry of Health of the Russian Federation, Gamalei 18, Moscow 123098, Russia
| | - Sergey Domogatsky
- Imtek Limited, 3 Cherepkovskaya 15, Moscow 21552, Russia
- Russian Cardiology Research and Production Center Federal State Budgetary Institution, Ministry of Health of the Russian Federation, 3 Cherepkovskaya 15, Moscow 21552, Russia
| | - Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Anna Domogatskaya
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| |
Collapse
|
32
|
Stem Cells and Regenerative Medicine: Myth or Reality of the 21th Century. Stem Cells Int 2015; 2015:734731. [PMID: 26300923 PMCID: PMC4537770 DOI: 10.1155/2015/734731] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/22/2015] [Accepted: 05/24/2015] [Indexed: 02/07/2023] Open
Abstract
Since the 1960s and the therapeutic use of hematopoietic stem cells of bone marrow origin, there has been an increasing interest in the study of undifferentiated progenitors that have the ability to proliferate and differentiate into various tissues. Stem cells (SC) with different potency can be isolated and characterised. Despite the promise of embryonic stem cells, in many cases, adult or even fetal stem cells provide a more interesting approach for clinical applications. It is undeniable that mesenchymal stem cells (MSC) from bone marrow, adipose tissue, or Wharton's Jelly are of potential interest for clinical applications in regenerative medicine because they are easily available without ethical problems for their uses. During the last 10 years, these multipotent cells have generated considerable interest and have particularly been shown to escape to allogeneic immune response and be capable of immunomodulatory activity. These properties may be of a great interest for regenerative medicine. Different clinical applications are under study (cardiac insufficiency, atherosclerosis, stroke, bone and cartilage deterioration, diabetes, urology, liver, ophthalmology, and organ's reconstruction). This review focuses mainly on tissue and organ regeneration using SC and in particular MSC.
Collapse
|