1
|
Cheng S, Zhong L, Yin J, Duan H, Xie Q, Luo W, Jie W. Controllable digital and analog resistive switching behavior of 2D layered WSe 2 nanosheets for neuromorphic computing. NANOSCALE 2023; 15:4801-4808. [PMID: 36779310 DOI: 10.1039/d2nr06580k] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Memristors with controllable resistive switching (RS) behavior have been considered as promising candidates for synaptic devices in next-generation neuromorphic computing. In this work, two-terminal memristors with controllable digital and analog RS behavior are fabricated based on two-dimensional (2D) WSe2 nanosheets. Under a relatively high operating voltage of 4 V, the memristor demonstrates stable and reliable non-volatile bipolar digital RS with a high switching ratio of 6.3 × 104. On the other hand, under a relatively low operation voltage, the memristor exhibits analog RS with a series of tunable resistance states. The fabricated memristors can work as an artificial synapse with fundamental synaptic functions, such as long-term potentiation (LTP) and depression (LTD) as well as paired-pulse facilitation (PPF). More importantly, the memristor demonstrates high conductance modulation linearity with the calculated nonlinear parameter for conductance as -0.82 in the LTP process, which is beneficial to improving the accuracy of neuromorphic computing. Furthermore, the neuromorphic computing of file types and image recognition can be emulated based on a constructed three-layer artificial neural network (ANN) with a recognition accuracy that can reach up to 95.9% for small digits. In addition, memristors can be used to emulate the learning-forgetting experience of the human brain. Consequently, the memristor based on 2D WSe2 nanosheets not only exhibits controllable RS behavior but also simulates synaptic functions and is expected to be a potential candidate for future neuromorphic computing applications.
Collapse
Affiliation(s)
- Siqi Cheng
- College of Chemistry and Materials Science, Sichuan Normal University, Chengdu, 610066, China.
| | - Lun Zhong
- College of Chemistry and Materials Science, Sichuan Normal University, Chengdu, 610066, China.
| | - Jinxiang Yin
- College of Chemistry and Materials Science, Sichuan Normal University, Chengdu, 610066, China.
| | - Huan Duan
- College of Chemistry and Materials Science, Sichuan Normal University, Chengdu, 610066, China.
| | - Qin Xie
- State Key Laboratory of Electronic Thin Films and Integrated Devices, School of electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Wenbo Luo
- State Key Laboratory of Electronic Thin Films and Integrated Devices, School of electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Wenjing Jie
- College of Chemistry and Materials Science, Sichuan Normal University, Chengdu, 610066, China.
| |
Collapse
|
2
|
Xue G, Zhang J, Wu L, Sun S, Wu H, Hou Y, Wang J. Differentiation of umbilical cord mesenchymal stem cells into hepatocytes with CYP450 metabolic enzyme activity induced by a liver injury microenvironment. Biochem Biophys Res Commun 2023; 647:47-54. [PMID: 36716645 DOI: 10.1016/j.bbrc.2023.01.065] [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: 01/06/2023] [Revised: 01/15/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
The aim of this study was to observe the effect of a simulated liver tissue injury microenvironment on the directed differentiation of umbilical cord mesenchymal stem cells into hepatocytes with CYP450 metabolic activity in vitro, and to explore the mechanisms underlying this directed differentiation. Normal and damaged liver tissue homogenate supernatants (LHS and CCl4-LHS, respectively) were used as induction fluids. After induction for different durations, Western blot and RT-PCR were used to measure the protein and gene expression of the hepatocellular proteins AFP, CK18, ALB, and the CYP450 family. Simultaneously, the metabolic activity of CYP450 in hepatocytes was determined. Compared with the LHS and CCl4-LHS controls, the LHS and CCl4-LHS induction groups showed a significantly elevated protein and gene expression of AFP, CK18, ALB, CYP1A1/2, CYP2A6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 (P < 0.05). The metabolic activity of CYP450 in hepatocytes was increased (P < 0.05). In addition, compared with the LHS group, the CCl4-LHS group induced cell differentiation more rapidly and with a higher efficiency. The results suggested that a liver injury microenvironment is conducive for the directed differentiation of umbilical cord mesenchymal stem cells into hepatocytes with metabolic enzyme activity.
Collapse
Affiliation(s)
- Gai Xue
- Department of Sports Human Science, Shijiazhuang University, Shijiazhuang, 050035, Hebei province, China
| | - JinHu Zhang
- Hebei Academy of Chinese Medicine Sciences, Hebei University of Chinese Medicine, Shijiazhuang, 050030, Hebei Province, China
| | - LiYing Wu
- Department of Pharmacy, The 980th Hospital of the People's Liberation Army Joint Logistics Support Force, Shijiazhuang, 050082, Hebei Province, China
| | - Shuang Sun
- Department of Sports Human Science, Shijiazhuang University, Shijiazhuang, 050035, Hebei province, China
| | - HongHai Wu
- Department of Pharmacy, The 980th Hospital of the People's Liberation Army Joint Logistics Support Force, Shijiazhuang, 050082, Hebei Province, China
| | - YanNing Hou
- Department of Pharmacy, The 980th Hospital of the People's Liberation Army Joint Logistics Support Force, Shijiazhuang, 050082, Hebei Province, China
| | - JinXuan Wang
- Department of Sports Human Science, Shijiazhuang University, Shijiazhuang, 050035, Hebei province, China.
| |
Collapse
|
3
|
Narayan G, Ronima K R, Thummer RP. Direct Reprogramming of Somatic Cells into Induced β-Cells: An Overview. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1410:171-189. [PMID: 36515866 DOI: 10.1007/5584_2022_756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The persistent shortage of insulin-producing islet mass or β-cells for transplantation in the ever-growing diabetic population worldwide is a matter of concern. To date, permanent cure to this medical complication is not available and soon after the establishment of lineage-specific reprogramming, direct β-cell reprogramming became a viable alternative for β-cell regeneration. Direct reprogramming is a straightforward and powerful technique that can provide an unlimited supply of cells by transdifferentiating terminally differentiated cells toward the desired cell type. This approach has been extensively used by multiple groups to reprogram non-β-cells toward insulin-producing β-cells. The β-cell identity has been achieved by various studies via ectopic expression of one or more pancreatic-specific transcription factors in somatic cells, bypassing the pluripotent state. This work highlights the importance of the direct reprogramming approaches (both integrative and non-integrative) in generating autologous β-cells for various applications. An in-depth understanding of the strategies and cell sources could prove beneficial for the efficient generation of integration-free functional insulin-producing β-cells for diabetic patients lacking endogenous β-cells.
Collapse
Affiliation(s)
- Gloria Narayan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Ronima K R
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.
| |
Collapse
|
4
|
Khazaei M, Khazaei F, Niromand E, Ghanbari E. Tissue engineering approaches and generation of insulin-producing cells to treat type 1 diabetes. J Drug Target 2023; 31:14-31. [PMID: 35896313 DOI: 10.1080/1061186x.2022.2107653] [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: 01/05/2023]
Abstract
Tissue engineering (TE) has become a new effective solution to a variety of medical problems, including diabetes. Mesenchymal stem cells (MSCs), which have the ability to differentiate into endodermal and mesodermal cells, appear to be appropriate for this function. The purpose of this review was to evaluate the outcomes of various researches on the insulin-producing cells (IPCs) generation from MSCs with TE approaches to increase efficacy of type 1 diabetes treatments. The search was performed in PubMed/Medline, Scopus and Embase databases until 2021. Studies revealed that MSCs could also differentiate into IPCs under certain conditions. Therefore, a wide range of protocols have been used for this differentiation, but their effectiveness is very different. Scaffolds can provide a microenvironment that enhances the MSCs to IPCs differentiation, improves their metabolic activity and up-regulate pancreatic-specific transcription factors. They also preserve IPCs architecture and enhance insulin production as well as protect against cell death. This systematic review offers a framework for prospective research based on data. In vitro and in vivo evidence suggests that scaffold-based TE can improve the viability and function of IPCs.
Collapse
Affiliation(s)
- Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Khazaei
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Niromand
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Ghanbari
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
5
|
Dubey A, Saini S, Sharma V, Malik H, Kumar D, De AK, Bhattacharya D, Malakar D. Deducing Insulin-Producing Cells from Goat Adipose Tissue-Derived Mesenchymal Stem Cells. Cell Reprogram 2022; 24:195-203. [PMID: 35787695 DOI: 10.1089/cell.2022.0029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mesenchymal stem cell is a potent tool for regenerative medicine against control of incurable diseases in human and animals. Diabetes mellitus is one such condition marked with the blood glucose is high due to lack of insulin (INS) hormone secreted by the pancreatic cells. Rare, but sporadic, cases of dysfunctional pancreatic cells in goat as well as the promises of stem cell therapy as an off-the-shelf medicine prompted us to explore the potential of adipose-derived goat mesenchymal stem cells (AD-MSCs) to transdifferentiate into pancreatic islet-like cells. We isolated, in vitro cultured, and characterized the AD-MSCs by expression of MSC-specific markers and differentiation into multiple mesodermal lineage cells. The characterized AD-MSCs were in vitro transdifferentiated into INS-producing islet-like cells using a cocktail of glucose, nicotinamide, activin-A, exendin-4, pentagastrin, retinoic acid, and mercaptoethanol in 3 weeks. The transdifferentiated islet-like cells demonstrated the expression of pancreatic endoderm-specific transcripts PDX1, NGN3, PAX6, PAX4, ISL1, and GLUT2 as well as protein expression of pancreatic and duodenal homeobox 1 (PDX1), INS, and Islets 1 (ISL1). The islet-like cells also demonstrated the significant glucose-dependent INS release with respect to the course of transdifferentiation regime. The study envisaged to create the building material for basic research into mechanism of glucose homeostasis, which may pave road for developments in diabetes drug discovery and regenerative therapies.
Collapse
Affiliation(s)
- Amit Dubey
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Sikander Saini
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Vishal Sharma
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Hrudananda Malik
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Dinesh Kumar
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Arun Kumar De
- Animal Science Division, ICAR-Central Island Agricultural Research Institute, Port Blair, India
| | - Debasis Bhattacharya
- Animal Science Division, ICAR-Central Island Agricultural Research Institute, Port Blair, India
| | - Dhruba Malakar
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
6
|
Azam MA, Khan KB, Salahuddin S, Rehman E, Khan SA, Khan MA, Kadry S, Gandomi AH. A review on multimodal medical image fusion: Compendious analysis of medical modalities, multimodal databases, fusion techniques and quality metrics. Comput Biol Med 2022; 144:105253. [PMID: 35245696 DOI: 10.1016/j.compbiomed.2022.105253] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/20/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND OBJECTIVES Over the past two decades, medical imaging has been extensively apply to diagnose diseases. Medical experts continue to have difficulties for diagnosing diseases with a single modality owing to a lack of information in this domain. Image fusion may be use to merge images of specific organs with diseases from a variety of medical imaging systems. Anatomical and physiological data may be included in multi-modality image fusion, making diagnosis simpler. It is a difficult challenge to find the best multimodal medical database with fusion quality evaluation for assessing recommended image fusion methods. As a result, this article provides a complete overview of multimodal medical image fusion methodologies, databases, and quality measurements. METHODS In this article, a compendious review of different medical imaging modalities and evaluation of related multimodal databases along with the statistical results is provided. The medical imaging modalities are organized based on radiation, visible-light imaging, microscopy, and multimodal imaging. RESULTS The medical imaging acquisition is categorized into invasive or non-invasive techniques. The fusion techniques are classified into six main categories: frequency fusion, spatial fusion, decision-level fusion, deep learning, hybrid fusion, and sparse representation fusion. In addition, the associated diseases for each modality and fusion approach presented. The quality assessments fusion metrics are also encapsulated in this article. CONCLUSIONS This survey provides a baseline guideline to medical experts in this technical domain that may combine preoperative, intraoperative, and postoperative imaging, Multi-sensor fusion for disease detection, etc. The advantages and drawbacks of the current literature are discussed, and future insights are provided accordingly.
Collapse
Affiliation(s)
- Muhammad Adeel Azam
- Department of Advanced Robotics, Istituto Italiano di Tecnologia, Genova, Italy; Department of Informatics, Bioengineering, Robotics, and System Engineering, University of Genoa, Italy
| | - Khan Bahadar Khan
- Department of Information and Communication Engineering, Faculty of Engineering, The Islamia University of Bahawalpur, Pakistan
| | - Sana Salahuddin
- Institute of Physics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Eid Rehman
- Department of Software Engineering, Foundation University Islamabad, 44000, Pakistan
| | - Sajid Ali Khan
- Department of Software Engineering, Foundation University Islamabad, 44000, Pakistan
| | | | - Seifedine Kadry
- Department of Applied Data Science, Noroff University College, Kristiansand, Norway
| | - Amir H Gandomi
- Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
7
|
Sun YL, Shang LR, Liu RH, Li XY, Zhang SH, Ren YK, Fu K, Cheng HB, Yahaya BH, Liu YL, Lin JT. Therapeutic effects of menstrual blood-derived endometrial stem cells on mouse models of streptozotocin-induced type 1 diabetes. World J Stem Cells 2022; 14:104-116. [PMID: 35126831 PMCID: PMC8788184 DOI: 10.4252/wjsc.v14.i1.104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/20/2021] [Accepted: 12/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D), a chronic metabolic and autoimmune disease, seriously endangers human health. In recent years, mesenchymal stem cell (MSC) transplantation has become an effective treatment for diabetes. Menstrual blood-derived endometrial stem cells (MenSC), a novel MSC type derived from the decidual endometrium during menstruation, are expected to become promising seeding cells for diabetes treatment because of their noninvasive collection procedure, high proliferation rate and high immunomodulation capacity.
AIM To comprehensively compare the effects of MenSC and umbilical cord-derived MSC (UcMSC) transplantation on T1D treatment, to further explore the potential mechanism of MSC-based therapies in T1D, and to provide support for the clinical application of MSC in diabetes treatment.
METHODS A conventional streptozotocin-induced T1D mouse model was established, and the effects of MenSC and UcMSC transplantation on their blood glucose and serum insulin levels were detected. The morphological and functional changes in the pancreas, liver, kidney, and spleen were analyzed by routine histological and immunohistochemical examinations. Changes in the serum cytokine levels in the model mice were assessed by protein arrays. The expression of target proteins related to pancreatic regeneration and apoptosis was examined by western blot.
RESULTS MenSC and UcMSC transplantation significantly improved the blood glucose and serum insulin levels in T1D model mice. Immunofluorescence analysis revealed that the numbers of insulin+ and CD31+ cells in the pancreas were significantly increased in MSC-treated mice compared with control mice. Subsequent western blot analysis also showed that vascular endothelial growth factor (VEGF), Bcl2, Bcl-xL and Proliferating cell nuclear antigen in pancreatic tissue was significantly upregulated in MSC-treated mice compared with control mice. Additionally, protein arrays indicated that MenSC and UcMSC transplantation significantly downregulated the serum levels of interferon γ and tumor necrosis factor α and upregulated the serum levels of interleukin-6 and VEGF in the model mice. Additionally, histological and immunohistochemical analyses revealed that MSC transplantation systematically improved the morphologies and functions of the liver, kidney, and spleen in T1D model mice.
CONCLUSION MenSC transplantation significantly improves the symptoms in T1D model mice and exerts protective effects on their main organs. Moreover, MSC-mediated angiogenesis, antiapoptotic effects and immunomodulation likely contribute to the above improvements. Thus, MenSC are expected to become promising seeding cells for clinical diabetes treatment due to their advantages mentioned above.
Collapse
Affiliation(s)
- Yu-Liang Sun
- Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Kepala Batas 13200, Penang, Malaysia
| | - Ling-Rui Shang
- Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
| | - Rui-Hong Liu
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
| | - Xin-Yi Li
- Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
| | - Sheng-Hui Zhang
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
| | - Ya-Kun Ren
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
| | - Kang Fu
- Department of Technical, Henan Intercell Biotechnology co. LTD, Xinxiang 453000, Henan Province, China
| | - Hong-Bin Cheng
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
| | - Badrul Hisham Yahaya
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Kepala Batas 13200, Penang, Malaysia
| | - Yan-Li Liu
- Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
| | - Jun-Tang Lin
- Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
- College of Biomedical Engineering, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
| |
Collapse
|
8
|
Chen T, Wang H, Jiang C, Lu Y. PKD1 alleviates oxidative stress-inhibited osteogenesis of rat bone marrow-derived mesenchymal stem cells through TAZ activation. J Cell Biochem 2021; 122:1715-1725. [PMID: 34407229 PMCID: PMC9292359 DOI: 10.1002/jcb.30124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/07/2021] [Accepted: 07/22/2021] [Indexed: 01/03/2023]
Abstract
Oxidative stress is known to inhibit osteogenesis and PKD1 is implicated in bone remodeling and skeletogenesis. In the present study, we explored the role of PKD1 in osteogenesis under oxidative stress. H2 O2 was used to induce oxidative stress in rat bone marrow (BM)-mesenchymal stem cells (MSCs) during osteoblast differentiation. Alkaline phosphatase (ALP) activity, calcium deposits, and the RUNX2 marker were assayed to determine osteogenic differentiation. The correlation of PKD1, Sirt1, c-MYC, and TAZ was further confirmed by chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assay. We found that H2 O2 induced the downregulation of PKD1 expression and the upregulation of c-MYC, and Sirt1 was accompanied by decreasing cell viability in BM-MSCs. During osteogenic differentiation, the expression of PKD1 was upregulated significantly whereas Sirt1 tended to be upregulated mildly under normal conditions. Both PKD1 and Sirt1 were upregulated upon oxidative stress. The positive correlation of PKD1 expression with osteogenic differentiation under normal conditions might be hindered by oxidative stress and PKD1 could interact with TAZ under oxidative stress to regulate osteogenic differentiation. Our results suggest that PKD1 may alleviate oxidative stress-inhibited osteogenesis of rat BM-MSCs through TAZ activation.
Collapse
Affiliation(s)
- Tongtong Chen
- Department of Radiology, Ruijin Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Hanqi Wang
- Department of Radiology, Ruijin Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Chaoyin Jiang
- Department of Orthopedic SurgeryShanghai Jiaotong University Affiliated Sixth People's HospitalShanghaiChina
- Department of Orthopedic SurgeryHaikou Orthopedic and Diabetes Hospital of Shanghai Sixth People's HospitalHainanChina
| | - Yong Lu
- Department of Radiology, Ruijin Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
- Department of Radiology, Ruijin Hospital Luwan Branch, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| |
Collapse
|
9
|
Wang J, Dai P, Zou T, Lv Y, Zhao W, Zhang X, Zhang Y. Transcriptome analysis of the transdifferentiation of canine BMSCs into insulin producing cells. BMC Genomics 2021; 22:134. [PMID: 33632121 PMCID: PMC7905582 DOI: 10.1186/s12864-021-07426-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/05/2021] [Indexed: 12/31/2022] Open
Abstract
Background Bone marrow mesenchymal stem cells are a potential resource for the clinical therapy of certain diseases. Canine, as a companion animal, living in the same space with human, is an ideal new model for human diseases research. Because of the high prevalence of diabetes, alternative transplantation islets resource (i.e. insulin producing cells) for diabetes treatment will be in urgent need, which makes our research on the transdifferentiation of Bone marrow mesenchymal stem cells into insulin producing cells become more important. Result In this study, we completed the transdifferentiation process and achieved the transcriptome profiling of five samples with two biological duplicates, namely, “BMSCs”, “islets”, “stage 1”, “stage 2” and “stage 3”, and the latter three samples were achieved on the second, fifth and eighth day of induction. A total of 11,530 differentially expressed transcripts were revealed in the profiling data. The enrichment analysis of differentially expressed genes revealed several signaling pathways that are essential for regulating proliferation and transdifferentiation, including focal adhesion, ECM-receptor interaction, tight junction, protein digestion and absorption, and the Rap1 signaling pathway. Meanwhile, the obtained protein–protein interaction network and functional identification indicating involvement of three genes, SSTR2, RPS6KA6, and VIP could act as a foundation for further research. Conclusion In conclusion, to the best of our knowledge, this is the first survey of the transdifferentiation of canine BMSCs into insulin-producing cells according with the timeline using next-generation sequencing technology. The three key genes we pick out may regulate decisive genes during the development of transdifferentiation of insulin producing cells. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07426-3.
Collapse
Affiliation(s)
- Jinglu Wang
- The College of Veterinary Medicine of the Northwest Agriculture and Forestry University, No.3 Taicheng Road, Yangling, 712100, Shaanxi, P. R. China
| | - Pengxiu Dai
- The College of Veterinary Medicine of the Northwest Agriculture and Forestry University, No.3 Taicheng Road, Yangling, 712100, Shaanxi, P. R. China
| | - Tong Zou
- The College of Veterinary Medicine of the Northwest Agriculture and Forestry University, No.3 Taicheng Road, Yangling, 712100, Shaanxi, P. R. China
| | - Yangou Lv
- The College of Veterinary Medicine of the Northwest Agriculture and Forestry University, No.3 Taicheng Road, Yangling, 712100, Shaanxi, P. R. China
| | - Wen Zhao
- The College of Veterinary Medicine of the Northwest Agriculture and Forestry University, No.3 Taicheng Road, Yangling, 712100, Shaanxi, P. R. China
| | - Xinke Zhang
- The College of Veterinary Medicine of the Northwest Agriculture and Forestry University, No.3 Taicheng Road, Yangling, 712100, Shaanxi, P. R. China
| | - Yihua Zhang
- The College of Veterinary Medicine of the Northwest Agriculture and Forestry University, No.3 Taicheng Road, Yangling, 712100, Shaanxi, P. R. China.
| |
Collapse
|
10
|
Kadkhodaeian HA. Mesenchymal Stem Cells: Signaling Pathways in Transdifferentiation Into Retinal Progenitor Cells. Basic Clin Neurosci 2021; 12:29-42. [PMID: 33995925 PMCID: PMC8114861 DOI: 10.32598/bcn.9.10.510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/25/2018] [Accepted: 02/02/2020] [Indexed: 11/29/2022] Open
Abstract
Several signaling pathways and transcription factors control the cell fate in its in vitro development and differentiation. The orchestrated use of these factors results in cell specification. In coculture methods, many of these factors secrete from host cells but control the process. Today, transcription factors required for retinal progenitor cells are well known, but the generation of these cells from mesenchymal stem cells is an ideal goal. The purpose of the paper is to review novel methods for retinal progenitor cell production and selecting a set of signaling molecules in the presence of adult retinal pigment epithelium and extraocular mesenchyme acting as inducers of retinal cell differentiation.
Collapse
|
11
|
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: 31] [Impact Index Per Article: 6.2] [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
|
12
|
Sávio-Silva C, Beyerstedt S, Soinski-Sousa PE, Casaro EB, Balby-Rocha MTA, Simplício-Filho A, Alves-Silva J, Rangel ÉB. Mesenchymal Stem Cell Therapy for Diabetic Kidney Disease: A Review of the Studies Using Syngeneic, Autologous, Allogeneic, and Xenogeneic Cells. Stem Cells Int 2020; 2020:8833725. [PMID: 33505469 PMCID: PMC7812547 DOI: 10.1155/2020/8833725] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022] Open
Abstract
Diabetic kidney disease (DKD) is a microvascular complication of diabetes mellitus (DM) and comprises multifactorial pathophysiologic mechanisms. Despite current treatment, around 30-40% of individuals with type 1 and type 2 DM (DM1 and DM2) have progressive DKD, which is the most common cause of end-stage chronic kidney disease worldwide. Mesenchymal stem cell- (MSC-) based therapy has important biological and therapeutic implications for curtailing DKD progression. As a chronic disease, DM may impair MSC microenvironment, but there is compelling evidence that MSC derived from DM1 individuals maintain their cardinal properties, such as potency, secretion of trophic factors, and modulation of immune cells, so that both autologous and allogeneic MSCs are safe and effective. Conversely, MSCs derived from DM2 individuals are usually dysfunctional, exhibiting higher rates of senescence and apoptosis and a decrease in clonogenicity, proliferation, and angiogenesis potential. Therefore, more studies in humans are needed to reach a conclusion if autologous MSCs from DM2 individuals are effective for treatment of DM-related complications. Importantly, the bench to bedside pathway has been constructed in the last decade for assessing the therapeutic potential of MSCs in the DM setting. Laboratory research set the basis for establishing further translation research including preclinical development and proof of concept in model systems. Phase I clinical trials have evaluated the safety profile of MSC-based therapy in humans, and phase II clinical trials (proof of concept in trial participants) still need to answer important questions for treating DKD, yet metabolic control has already been documented. Therefore, randomized and controlled trials considering the source, optimal cell number, and route of delivery in DM patients are further required to advance MSC-based therapy. Future directions include strategies to reduce MSC heterogeneity, standardized protocols for isolation and expansion of those cells, and the development of well-designed large-scale trials to show significant efficacy during a long follow-up, mainly in individuals with DKD.
Collapse
Affiliation(s)
- Christian Sávio-Silva
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Stephany Beyerstedt
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Poliana E. Soinski-Sousa
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Expedito B. Casaro
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | - Antônio Simplício-Filho
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Jamille Alves-Silva
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Érika B. Rangel
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Nephrology Division, Federal University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
13
|
Meta-Analysis of the Therapeutic Effect of Shenqi Jiangtang Granule on Type 2 Diabetes Mellitus. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5754823. [PMID: 33062014 PMCID: PMC7542533 DOI: 10.1155/2020/5754823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/01/2020] [Accepted: 07/23/2020] [Indexed: 11/21/2022]
Abstract
Objective To systematically evaluate the effectiveness of Shenqi Jiangtang granule (SQJT) in the treatment of type 2 diabetes. Methods We searched CNKI, Wanfang Data, VIP, and PubMed databases to collect randomized controlled trials (RCT) of Shenqi Jiangtang granules in the treatment of type 2 diabetes. The search time was from January 2014 to the present. Data were extracted, and quality was evaluated. Metadata analysis of the extracted data was carried out using RevMa5.2 software. The final results are expressed in relative risk (RR), mean difference (MD), and 95% CI. Results This study included a total of 13 studies, 1160 subjects. Meta-analysis results showed that the test group was better than the control group (RR = 1.26, 95% CI 1.18–1.34, P < 0.00001). The fasting blood glucose, postprandial blood glucose, and glycated hemoglobin of the test group were also significantly better than those of the control group. Conclusion Shenqi Jiangtang granules have a certain clinical effect and low adverse reaction rate for the treatment or adjuvant treatment of type 2 diabetes. At present, the drug has been widely used in clinical practice, but a large number of large-sample clinical trials are needed to further verify its specific efficacy and safety.
Collapse
|
14
|
Oh I, Raymundo B, Jung SA, Kim HJ, Park J, Kim C. Extremely
Low‐Frequency
Electromagnetic Field Altered
PPARγ
and
CCL2
Levels and Suppressed
CD44
+
/
CD24
−
Breast Cancer Cells Characteristics. B KOREAN CHEM SOC 2020. [DOI: 10.1002/bkcs.12072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- In‐Rok Oh
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| | - Bernardo Raymundo
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| | - Sung A Jung
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| | - Hyun Jung Kim
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| | - Jung‐Keug Park
- Dongguk University Biomedi CampusDongguk University Goyang Korea
| | - Chan‐Wha Kim
- College of Life Sciences and BiotechnologyKorea University Seoul 136‐701 Korea
| |
Collapse
|
15
|
Aali E, Madjd Z, Tekiyehmaroof N, Sharifi AM. Control of Hyperglycemia Using Differentiated and Undifferentiated Mesenchymal Stem Cells in Rats with Type 1 Diabetes. Cells Tissues Organs 2020; 209:13-25. [PMID: 32634811 DOI: 10.1159/000507790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/07/2020] [Indexed: 11/19/2022] Open
Abstract
Due to their ability in self-renewing and differentiation into a wide variety of tissues, mesenchymal stem cells (MSCs) exhibit outstanding potential for regenerative medicine. This study was aimed at investigating different aspects of MSC therapy in controlling hyperglycemia in streptozotocin-induced diabetes rats. Using an islet cell differentiation protocol, bone marrow (BM) MSCs were differentiated into insulin-producing cells (IPCs). The differentiation process was evaluated by immunocytochemistry, reverse transcriptase PCR, and dithizone staining. Diabetic animals in 4 diabetic individual groups received normal saline, BM-MSCs, coadministration of BM-MSCs with supernatant, and IPCs. Blood glucose and insulin levels were monitored during the experiment. Immunohistochemical analysis of the pancreas was performed at the end of the experiment. Administration of BM-MSCs could not reverse glucose and insulin levels in experimental animals as efficiently as cotransplantation of BM-MSCs with supernatant. The effect of coadministration of BM-MSCs with supernatant and transplantation of IPCs on controlling hyperglycemia is comparable. Immunohistochemical analysis showed that number and size of islets per section were significantly increased in groups receiving IPCs and BM-MSC-supernatant compared to the MSC group of animals. In conclusion, coadministration of BM-MSCs with supernatant could be used as efficiently as IPC transplantation in controlling hyperglycemia in diabetic rats.
Collapse
Affiliation(s)
- Ehsan Aali
- Department of Pharmacology, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Zahra Madjd
- Department of Pathology, Oncology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Tekiyehmaroof
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Sharifi
- Razi Drug Research Center and Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran,
| |
Collapse
|
16
|
PRDX6 Promotes the Differentiation of Human Mesenchymal Stem (Stromal) Cells to Insulin-Producing Cells. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7103053. [PMID: 32051828 PMCID: PMC6995490 DOI: 10.1155/2020/7103053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) can be differentiated in vitro to form insulin-producing cells (IPCs). However, the proportion of induced cells is modest. Extracts from injured pancreata of rodents promoted this differentiation, and three upregulated proteins were identified in these extracts. The aim of this study was to evaluate the potential benefits of adding these proteins to the differentiation medium alone or in combination. Our results indicate that the proportion of IPCs among the protein(s)-supplemented samples was significantly higher than that in the samples with no added proteins. The yield from samples supplemented with PRDX6 alone was 4-fold higher than that from samples without added protein. These findings were also supported by the results of fluorophotometry. Gene expression profiles revealed higher levels among protein-supplemented samples. Significantly higher levels of GGT, SST, Glut-2, and MafB expression were noted among PRDX6-treated samples. There was a stepwise increase in the release of insulin and c-peptide, as a function of increasing glucose concentrations, indicating that the differentiated cells were glucose sensitive and insulin responsive. PRDX6 exerts its beneficial effects as a result of its biological antioxidant properties. Considering its ease of use as a single protein, PRDX6 is now routinely used in our differentiation protocols.
Collapse
|
17
|
El-Sherbiny M, Eladl MA, Ranade AV, Guimei M, Gabr H. Functional beta-cells derived from umbilical cord blood mesenchymal stem cells for curing rats with streptozotocin-induced diabetes mellitus. Singapore Med J 2019; 61:39-45. [PMID: 31535156 DOI: 10.11622/smedj.2019120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION This study aimed to investigate the therapeutic response to injected human umbilical cord blood mesenchymal stem cells (UCBMSCs) among albino rats with streptozotocin (STZ)-induced diabetes mellitus. METHODS Control group (GI; n = 25) rats were fed with standard rat diet. Rats with STZ-induced diabetes mellitus without (GII; n = 25) and with (GIII; n = 25) differentiated human UCBMSCs implantation were the test groups. Rats were sacrificed in Week 11 following implantation. Liver biopsies were sectioned and stained in order to highlight both the presence and function of impregnated cells in the liver tissue. RESULTS Haematoxylin and eosin-stained sections in GI and GII rats showed normal liver architecture while GIII rats showed presence of cell clusters inside the liver tissue and around the central veins. Cell clusters with blue cytoplasm were present in sections in GIII rats but absent in GI and GII rats, indicating the presence of injected differentiated human UCBMSCs. The anti-human insulin immunostaining of GIII rats showed clusters of cells within the liver parenchyma and around central veins, indicating that these cells were active and secreting insulin. CONCLUSION UCBMSCs are proficient in differentiating into insulin-producing cells in vivo under specific conditions and, when transplanted into the liver of albino rats with STZ-induced diabetes mellitus, were able to secrete insulin and partially control the status of diabetes mellitus in rats.
Collapse
Affiliation(s)
| | - Mohamed Ahmed Eladl
- Department of Basic Medical Sciences, Medical College, University of Sharjah, Sharjah, United Arab Emirates
| | - Anu Vinod Ranade
- Department of Basic Medical Sciences, Medical College, University of Sharjah, Sharjah, United Arab Emirates
| | - Maha Guimei
- Department of Clinical Sciences, Medical College, University of Sharjah, Sharjah, United Arab Emirates.,Department of Pathology, University of Alexandria, Alexandria, Egypt
| | - Hala Gabr
- Faculty of Medicine, Department of Clinical Pathology, Cairo University, Cairo, Egypt
| |
Collapse
|
18
|
Rashed S, Gabr M, Abdel-Aziz AA, Zakaria M, Khater S, Ismail A, Fouad A, Refaie A. Differentiation Potential of Nestin (+) and Nestin (-) Cells Derived from Human Bone Marrow Mesenchymal Stem Cells into Functional Insulin Producing Cells. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 8:1-13. [PMID: 32195201 DOI: 10.22088/ijmcm.bums.8.1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/13/2019] [Indexed: 01/09/2023]
Abstract
The feasibility of isolating and manipulating mesenchymal stem cells (MSCs) from human patients provides hope for curing numerous diseases and disorders. Recent phenotypic analysis has shown heterogeneity of MSCs. Nestin progenitor cell is a subpopulation within MSCs which plays a role in pancreas regeneration during embryogenesis. This study aimed to separate nestin (+) cells from human bone marrow MSCs, and differentiate these cells into functional insulin producing cells (IPCs) compared with nestin (-) cells. Manual magnetic separation was performed to obtain nestin (+) cells from MSCs. Approximately 91±3.3% of nestin (+) cells were positive for anti-nestin antibody. Pluripotent genes were overexpressed in nestin (+) cells compared with nestin (-) cells as revealed by quantitative real time-PCR (qRT-PCR). Following in vitro differentiation, flow cytometric analysis showed that 2.7±0.5% of differentiated nestin (+) cells were positive for anti-insulin antibody in comparison with 0.08±0.02% of nestin (-) cells. QRT-PCR showed higher expression of insulin and other endocrine genes in comparison with nestin (-) cells. While immunofluorescence technique showed the presence of insulin and C-peptide granules in nestin (+) cells. Therefore, our results introduced nestin (+) cells as a pluripotent subpopulation within human MSCs which is capable to differentiate and produce functional IPCs.
Collapse
Affiliation(s)
- Sahar Rashed
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Mahmoud Gabr
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Abdel-Aziz Abdel-Aziz
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Mahmoud Zakaria
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Sherry Khater
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Amani Ismail
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ali Fouad
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ayman Refaie
- Nephrology Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| |
Collapse
|
19
|
A simple method for the generation of insulin producing cells from bone marrow mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2019; 55:462-471. [PMID: 31111346 DOI: 10.1007/s11626-019-00358-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 04/11/2019] [Indexed: 10/26/2022]
Abstract
To produce insulin-producing cells (IPCs) from bone marrow mesenchymal stem cells (BM-MSCs) using a simple and cost effective method. During the initial 7 days of three-dimensional (3D) culture, BM-MSCs were cultured on 1% agar or agarose to form multicellular spheroids. Spheroids and spheroid-derived single cells (SS and SSC, respectively) were cultured in the absence of any proteinaceous growth factor in a simple specific medium for a further 7 d. The insulin content of the differentiated cells was evaluated at the mRNA and protein levels. Furthermore, the expression of pancreatic beta cells-related genes other than INS as well as the in vitro responses of IPCs to different glucose concentrations were investigated. Cellular clusters generated on agar and SS conditions (agar+SS-IPCs) stained better with beta cell specific stains and were more reactive to serum-containing insulin reactive antibodies compared with agarose-SS-IPCs. Gene expression analysis revealed that in comparison to agarose + SS-IPCs, agar+SS-IPCs expressed significantly higher levels of INS-1, INS-2, PDX-1, NKX6.1, and XBP-1. Of interest, agar+SS-IPCs expressed 2215.3 ± 120.8-fold more INS-1 gene compared to BM-MSCs. The expression of β-cell associated genes was also higher in agar+SS-IPCs compared to the agar+SSC-IPCs. Moreover, the expression of INS-1 gene was significantly higher in agar+SS-IPCs compared with agar+SSC-IPCs after culture in media with high concentration of glucose. Compared to the most expensive and time-consuming protocols, 3D culture of MSCs on agar followed by 2D culture of cellular clusters in a minimally supplemented high glucose media produced highly potent IPCs which may pay the way to the treatment of diabetic patients.
Collapse
|
20
|
Xiong XR, Lan DL, Li J, Yin S, Xiong Y, Zi XD. Effects of Cellular Extract on Epigenetic Reprogramming. Cell Reprogram 2019; 21:115-121. [PMID: 31084436 DOI: 10.1089/cell.2018.0074] [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
Functional reprogramming of a differentiated cell toward pluripotent cell may have long-term applications in numerous aspects, especially in regenerative medicine. Evidences accumulating from recent studies suggest that cellular extracts from stem cells or pluripotent cells can induce epigenetic reprogramming and facilitate pluripotency in otherwise highly differentiated cell types. Epigenetic reprogramming using cellular extracts has gained increasing attention and applied to recognize the functional factors, acquire the target cell types, and explain the mechanism of reprogramming. Now, more and more researches have proved that cellular extract treatment is an important strategy of cellular reprogramming. Thus, this review mainly focused on the progresses and potential mechanisms in epigenetic reprogramming using cellular extracts.
Collapse
Affiliation(s)
- Xian-Rong Xiong
- 1 College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Dao-Liang Lan
- 2 Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Chengdu, China
| | - Jian Li
- 1 College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Shi Yin
- 2 Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Ministry of Education, Chengdu, China
| | - Yan Xiong
- 1 College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Xiang-Dong Zi
- 1 College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| |
Collapse
|
21
|
Navaei-Nigjeh M, Moloudizargari M, Baeeri M, Gholami M, Lotfibakhshaiesh N, Soleimani M, Vasheghani-Farahani E, Ai J, Abdollahi M. Reduction of marginal mass required for successful islet transplantation in a diabetic rat model using adipose tissue-derived mesenchymal stromal cells. Cytotherapy 2018; 20:1124-1142. [PMID: 30068495 DOI: 10.1016/j.jcyt.2018.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/10/2018] [Accepted: 06/06/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Adipose tissue-derived mesenchymal stromal cells (AT-MSCs), widely known as multipotent progenitors, release several cytokines that support cell survival and repair. There are in vitro and in vivo studies reporting the regenerative role of AT-MSCs possibly mediated by their protective effects on functional islet cells as well as their capacity to differentiate into insulin-producing cells (IPCs). METHODS On such a basis, our goal in the present study was to use three different models including direct and indirect co-cultures and islet-derived conditioned medium (CM) to differentiate AT-MSCs into IPCs and to illuminate the molecular mechanisms of the beneficial impact of AT-MSCs on pancreatic islet functionality. Furthermore, we combined in vitro co-culture of islets and AT-MSCs with in vivo assessment of islet graft function to assess whether co-transplantation of islets with AT-MSCs can reduce marginal mass required for successful islet transplantation and prolong graft function in a diabetic rat model. RESULTS Our findings demonstrated that AT-MSCs are suitable for creating a microenvironment favorable for the repair and longevity of the pancreas β cells through the improvement of islet survival and maintenance of cell morphology and insulin secretion due to their potent properties in differentiation. Most importantly, hybrid transplantation of islets with AT-MSCs significantly promoted survival, engraftment and insulin-producing function of the graft and reduced the islet mass required for reversal of diabetes. CONCLUSIONS This strategy might be of therapeutic potential solving the problem of donor islet material loss that currently limits the application of allogeneic islet transplantation as a more widespread therapy for type 1 diabetes.
Collapse
Affiliation(s)
- Mona Navaei-Nigjeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Science, Tehran, Iran
| | - Milad Moloudizargari
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Baeeri
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Science, Tehran, Iran
| | - Mahdi Gholami
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Science, Tehran, Iran
| | - Nasrin Lotfibakhshaiesh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Science, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Miladpour B, Rasti M, Owji AA, Mostafavipour Z, Khoshdel Z, Noorafshan A, Zal F. Quercetin potentiates transdifferentiation of bone marrow mesenchymal stem cells into the beta cells in vitro. J Endocrinol Invest 2017; 40:513-521. [PMID: 28000178 DOI: 10.1007/s40618-016-0592-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/27/2016] [Indexed: 01/17/2023]
Abstract
PURPOSE Type 1 diabetes is an autoimmune disease caused by the destruction of β-cells in the pancreas. Bone marrow mesenchymal stem cells are multipotent and easy accessible adult stem cells that may provide options in the treatment of type 1 diabetes. Injured pancreatic extract can promote the differentiation of rat bone marrow mesenchymal stem cells into β-cells. We aimed to observe the effect of quercetin in differentiation and insulin secretion in β-cells. METHODS Bone marrow mesenchymal stem cells were obtained from the tibiae of rats. Cell surface markers were analyzed by flow cytometry. The cells were treated with rat injured pancreatic extract and quercetin for 2 weeks. Insulin secretion was measured by ELISA. Insulin expression and some islet factors were evaluated by RT-PCR. PDX1, a marker for β-cell function and differentiation, was evaluated by both immunocytochemistry and Western blot. β-cell count was determined by stereology and cell count assay. RESULTS ELISA showed significant differences in insulin secretion in the cells treated with RIPE + 20 μM quercetin (0.55 ± 0.01 µg/L) compared with the cells treated with RIPE alone (0.48 ± 0.01 µg/L) (P = 0.026). RT-PCR results confirmed insulin expression in both groups. PDX1 protein was detected in both groups by Western blot and immunocytochemistry. Stereology results showed a significant increase in β-cell number in the RIPE + quercetin-treated cells (47 ± 2.0) when compared with RIPE treatment alone (44 ± 2.5) (P = 0.015). CONCLUSIONS Quercetin has a strengthening effect on the differentiation of rat bone marrow mesenchymal stem cells into β-cells and increases insulin secretion from the differentiated β-cells in vitro.
Collapse
Affiliation(s)
- B Miladpour
- Biochemistry Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - M Rasti
- Biochemistry Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - A A Owji
- Biochemistry Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Z Mostafavipour
- Biochemistry Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Z Khoshdel
- Biochemistry Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - A Noorafshan
- Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - F Zal
- Biochemistry Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran.
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
23
|
Joukes J, Yu Y, Victor JD, Krekelberg B. Recurrent Network Dynamics; a Link between Form and Motion. Front Syst Neurosci 2017; 11:12. [PMID: 28360844 PMCID: PMC5350104 DOI: 10.3389/fnsys.2017.00012] [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: 11/24/2016] [Accepted: 02/21/2017] [Indexed: 11/28/2022] Open
Abstract
To discriminate visual features such as corners and contours, the brain must be sensitive to spatial correlations between multiple points in an image. Consistent with this, macaque V2 neurons respond selectively to patterns with well-defined multipoint correlations. Here, we show that a standard feedforward model (a cascade of linear–non-linear filters) does not capture this multipoint selectivity. As an alternative, we developed an artificial neural network model with two hierarchical stages of processing and locally recurrent connectivity. This model faithfully reproduced neurons’ selectivity for multipoint correlations. By probing the model, we gained novel insights into early form processing. First, the diverse selectivity for multipoint correlations and complex response dynamics of the hidden units in the model were surprisingly similar to those observed in V1 and V2. This suggests that both transient and sustained response dynamics may be a vital part of form computations. Second, the model self-organized units with speed and direction selectivity that was correlated with selectivity for multipoint correlations. In other words, the model units that detected multipoint spatial correlations also detected space-time correlations. This leads to the novel hypothesis that higher-order spatial correlations could be computed by the rapid, sequential assessment and comparison of multiple low-order correlations within the receptive field. This computation links spatial and temporal processing and leads to the testable prediction that the analysis of complex form and motion are closely intertwined in early visual cortex.
Collapse
Affiliation(s)
- Jeroen Joukes
- Center for Molecular and Behavioral Neuroscience, Rutgers University, NewarkNJ, USA; Behavioral and Neural Sciences Graduate Program, Rutgers University, NewarkNJ, USA
| | - Yunguo Yu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York NY, USA
| | - Jonathan D Victor
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York NY, USA
| | - Bart Krekelberg
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark NJ, USA
| |
Collapse
|
24
|
Paulini J, Higuti E, Bastos RMC, Gomes SA, Rangel ÉB. Mesenchymal Stem Cells as Therapeutic Candidates for Halting the Progression of Diabetic Nephropathy. Stem Cells Int 2016; 2016:9521629. [PMID: 28058051 PMCID: PMC5187468 DOI: 10.1155/2016/9521629] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/08/2016] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess pleiotropic properties that include immunomodulation, inhibition of apoptosis, fibrosis and oxidative stress, secretion of trophic factors, and enhancement of angiogenesis. These properties provide a broad spectrum for their potential in a wide range of injuries and diseases, including diabetic nephropathy (DN). MSCs are characterized by adherence to plastic, expression of the surface molecules CD73, CD90, and CD105 in the absence of CD34, CD45, HLA-DR, and CD14 or CD11b and CD79a or CD19 surface molecules, and multidifferentiation capacity in vitro. MSCs can be derived from many tissue sources, consistent with their broad, possibly ubiquitous distribution. This article reviews the existing literature and knowledge of MSC therapy in DN, as well as the most appropriate rodent models to verify the therapeutic potential of MSCs in DN setting. Some preclinical relevant studies are highlighted and new perspectives of combined therapies for decreasing DN progression are discussed. Hence, improved comprehension and interpretation of experimental data will accelerate the progress towards clinical trials that should assess the feasibility and safety of this therapeutic approach in humans. Therefore, MSC-based therapies may bring substantial benefit for patients suffering from DN.
Collapse
Affiliation(s)
- Janaina Paulini
- Sociedade Beneficente Albert Einstein, Albert Einstein Hospital, 05652 São Paulo, SP, Brazil
| | - Eliza Higuti
- Sociedade Beneficente Albert Einstein, Albert Einstein Hospital, 05652 São Paulo, SP, Brazil
| | - Rosana M. C. Bastos
- Sociedade Beneficente Albert Einstein, Albert Einstein Hospital, 05652 São Paulo, SP, Brazil
| | - Samirah A. Gomes
- Sociedade Beneficente Albert Einstein, Albert Einstein Hospital, 05652 São Paulo, SP, Brazil
- University of São Paulo, 01246 São Paulo, SP, Brazil
| | - Érika B. Rangel
- Sociedade Beneficente Albert Einstein, Albert Einstein Hospital, 05652 São Paulo, SP, Brazil
- Federal University of São Paulo, 04023 São Paulo, SP, Brazil
| |
Collapse
|
25
|
Xun Q, Wang H, He F. Bone marrow mesenchymal stem cells for treatment of pancreatic diseases: Research status and prospects. Shijie Huaren Xiaohua Zazhi 2016; 24:3232-3237. [DOI: 10.11569/wcjd.v24.i21.3232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The incidence of pancreatic diseases is increasing year by year. Current treatments for pancreatic diseases are mainly symptomatic, and the research on the repair and restoration of function of the pancreatic cells progresses slowly. Stem cells have been widely used in the treatment of diseases in recent years because of their ability of multi-directional differentiation and repair of cell damage caused by disease and injury. Numerous studies confirm that pancreatic stem cells after transplantation can differentiate into pancreatic cells and play an important role in the recovery of external secretory function and repair of the damaged pancreatic cells. Particularly, both in vivo and in vitro studies show that bone marrow mesenchymal stem cells have achieved remarkable results in the treatment of pancreatic diseases, laying a theoretical and practical basis for clinical treatment of pancreatic diseases with stem cells. This article outlines the progress in treatment of acute pancreatitis, chronic pancreatitis and pancreatic cancer with bone marrow mesenchymal stem cells, demonstrating that stem cells are expected to become one of new methods for the treatment of pancreatic diseases.
Collapse
|
26
|
Mehrfarjam Z, Esmaeili F, Shabani L, Ebrahimie E. Induction of pancreatic β cell gene expression in mesenchymal stem cells. Cell Biol Int 2016; 40:486-500. [DOI: 10.1002/cbin.10567] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Zahra Mehrfarjam
- Razi Herbal Medicines Research Center; Lorestan University of Medical Sciences; P.O. Box 681499468 Khorramabad Iran
| | - Fariba Esmaeili
- Faculty of Basic Sciences; Department of Biology; University of Isfahan; P.O. Box 8174673441 Isfahan Iran
- Research Institute of Biotechnology; Shahrekord University; P.O. Box 115 Shahrekord Iran
| | - Leila Shabani
- Research Institute of Biotechnology; Shahrekord University; P.O. Box 115 Shahrekord Iran
| | - Esmaeil Ebrahimie
- Institute of Biotechnology; Shiraz University; Shiraz Iran
- Division of Information Technology, Engineering & Environment; School of Information Technology and Mathematical Sciences; University of South Australia; Adelaide Australia
- Department of Genetics and Evolution; The University of Adelaide; Adelaide Australia
- Faculty of Science and Engineering; School of Biological Sciences; Flinders University; Adelaide Australia
| |
Collapse
|
27
|
Kim MJ, Koo JE, Han GY, Kim B, Lee YS, Ahn C, Kim CW. Dual-Blocking of PI3K and mTOR Improves Chemotherapeutic Effects on SW620 Human Colorectal Cancer Stem Cells by Inducing Differentiation. J Korean Med Sci 2016; 31:360-70. [PMID: 26955235 PMCID: PMC4779859 DOI: 10.3346/jkms.2016.31.3.360] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/19/2015] [Indexed: 12/28/2022] Open
Abstract
Cancer stem cells (CSCs) have tumor initiation, self-renewal, metastasis and chemo-resistance properties in various tumors including colorectal cancer. Targeting of CSCs may be essential to prevent relapse of tumors after chemotherapy. Phosphatidylinositol-3-kinase (PI3K) and mammalian target of rapamycin (mTOR) signals are central regulators of cell growth, proliferation, differentiation, and apoptosis. These pathways are related to colorectal tumorigenesis. This study focused on PI3K and mTOR pathways by inhibition which initiate differentiation of SW620 derived CSCs and investigated its effect on tumor progression. By using rapamycin, LY294002, and NVP-BEZ235, respectively, PI3K and mTOR signals were blocked independently or dually in colorectal CSCs. Colorectal CSCs gained their differentiation property and lost their stemness properties most significantly in dual-blocked CSCs. After treated with anti-cancer drug (paclitaxel) on the differentiated CSCs cell viability, self-renewal ability and differentiation status were analyzed. As a result dual-blocking group has most enhanced sensitivity for anti-cancer drug. Xenograft tumorigenesis assay by using immunodeficiency mice also shows that dual-inhibited group more effectively increased drug sensitivity and suppressed tumor growth compared to single-inhibited groups. Therefore it could have potent anti-cancer effects that dual-blocking of PI3K and mTOR induces differentiation and improves chemotherapeutic effects on SW620 human colorectal CSCs.
Collapse
Affiliation(s)
- Min-Jung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
- Ministry of Food and Drug Safety, Cheongju, Korea
| | - Jeong-Eun Koo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Gi-Yeon Han
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Buyun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Yoo-Sun Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Chiyoung Ahn
- Ministry of Food and Drug Safety, Cheongju, Korea
| | - Chan-Wha Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| |
Collapse
|
28
|
Effect of Microenvironment on Differentiation of Human Umbilical Cord Mesenchymal Stem Cells into Hepatocytes In Vitro and In Vivo. BIOMED RESEARCH INTERNATIONAL 2016; 2016:8916534. [PMID: 27088093 PMCID: PMC4764721 DOI: 10.1155/2016/8916534] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 12/13/2022]
Abstract
Human umbilical cord-derived mesenchymal stem cells (hUCMSCs) are considered to be an ideal cell source for cell therapy of many diseases. The aim of this study was to investigate the contribution of the microenvironment to the hepatic differentiation potential of hUCMSCs in vitro and in vivo and to explore their therapeutic use in acute liver injury in rats. We established a new model to simulate the liver tissue microenvironment in vivo using liver homogenate supernatant (LHS) in vitro. This induced environment could drive hUCMSCs to differentiate into hepatocyte-like cells within 7 days. The differentiated cells expressed hepatocyte-specific markers and demonstrated hepatocellular functions. We also injected hUCMSCs into rats with CCl4-induced acute hepatic injury. The hUCMSCs were detected in the livers of recipient rats and expressed the human hepatocyte-specific markers, suggesting that hUCMSCs could differentiate into hepatocyte-like cells in vivo in the liver tissue microenvironment. Levels of biochemistry markers improved significantly after transplantation of hUCMSCs compared with the nontransplantation group (P < 0.05). In conclusion, this study demonstrated that the liver tissue microenvironment may contribute to the differentiation of hUCMSCs into hepatocytes both in vitro and in vivo.
Collapse
|
29
|
Xin Y, Jiang X, Wang Y, Su X, Sun M, Zhang L, Tan Y, Wintergerst KA, Li Y, Li Y. Insulin-Producing Cells Differentiated from Human Bone Marrow Mesenchymal Stem Cells In Vitro Ameliorate Streptozotocin-Induced Diabetic Hyperglycemia. PLoS One 2016; 11:e0145838. [PMID: 26756576 PMCID: PMC4710504 DOI: 10.1371/journal.pone.0145838] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 12/09/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The two major obstacles in the successful transplantation of islets for diabetes treatment are inadequate supply of insulin-producing tissue and immune rejection. Induction of the differentiation of human bone marrow-derived mesenchymal stem cells (hMSCs) into insulin-producing cells (IPCs) for autologous transplantation may alleviate those limitations. METHODS hMSCs were isolated and induced to differentiate into IPCs through a three-stage differentiation protocol in a defined media with high glucose, nicotinamide, and exendin-4. The physiological characteristics and functions of IPCs were then evaluated. Next, about 3 × 10(6) differentiated cells were transplanted into the renal sub-capsular space of streptozotocin (STZ)-induced diabetic nude mice. Graft survival and function were assessed by immunohistochemistry, TUNEL staining and measurements of blood glucose levels in the mice. RESULTS The differentiated IPCs were characterized by Dithizone (DTZ) positive staining, expression of pancreatic β-cell markers, and human insulin secretion in response to glucose stimulation. Moreover, 43% of the IPCs showed L-type Ca2+ channel activity and similar changes in intracellular Ca2+ in response to glucose stimulation as that seen in pancreatic β-cells in the process of glucose-stimulated insulin secretion. Transplantation of functional IPCs into the renal subcapsular space of STZ-induced diabetic nude mice ameliorated the hyperglycemia. Immunofluorescence staining revealed that transplanted IPCs sustainably expressed insulin, c-peptide, and PDX-1 without apparent apoptosis in vivo. CONCLUSIONS IPCs derived from hMSCs in vitro can ameliorate STZ-induced diabetic hyperglycemia, which indicates that these hMSCs may be a promising approach to overcome the limitations of islet transplantation.
Collapse
Affiliation(s)
- Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
- Department of Pediatrics, Division of Endocrinology, University of Louisville, Wendy L. Novak Diabetes Care Center, Louisville, Kentucky, United States of America
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Xuejin Su
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Meiyu Sun
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Lihong Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yi Tan
- Department of Pediatrics, Division of Endocrinology, University of Louisville, Wendy L. Novak Diabetes Care Center, Louisville, Kentucky, United States of America
| | - Kupper A. Wintergerst
- Department of Pediatrics, Division of Endocrinology, University of Louisville, Wendy L. Novak Diabetes Care Center, Louisville, Kentucky, United States of America
| | - Yan Li
- Department of Orthopedic Surgery, Karolinska University Hospital, Stockholm, Sweden
- * E-mail: (Yan Li); (Yulin Li)
| | - Yulin Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
- * E-mail: (Yan Li); (Yulin Li)
| |
Collapse
|
30
|
Abstract
Regenerative medicine using patient's own stem cells (SCs) to repair dysfunctional tissues is an attractive approach to complement surgical and pharmacological treatments for aging and degenerative disorders. Recently, dental SCs have drawn much attention owing to their accessibility, plasticity and applicability for regenerative use not only for dental, but also other body tissues. In ophthalmology, there has been increasing interest to differentiate dental pulp SC and periodontal ligament SC (PDLSC) towards ocular lineage. Both can commit to retinal fate expressing eye field transcription factors and generate rhodopsin-positive photoreceptor-like cells. This proposes a novel therapeutic alternative for retinal degeneration diseases. Moreover, as PDLSC shares similar cranial neural crest origin and proteoglycan secretion with corneal stromal keratoctyes and corneal endothelial cells, this offers the possibility of differentiating PDLSC to these corneal cell types. The advance could lead to a shift in the medical management of corneal opacities and endothelial disorders from highly invasive corneal transplantation using limited donor tissue to cell therapy utilizing autologous cells. This article provides an overview of dental SC research and the perspective of utilizing dental SCs for ocular regenerative medicine.
Collapse
|
31
|
Sahraneshin Samani F, Ebrahimi M, Zandieh T, Khoshchehreh R, Baghaban Eslaminejad M, Aghdami N, Baharvand H. In Vitro Differentiation of Human Umbilical Cord Blood CD133(+)Cells into Insulin Producing Cells in Co-Culture with Rat Pancreatic Mesenchymal Stem Cells. CELL JOURNAL 2015. [PMID: 26199900 PMCID: PMC4503835 DOI: 10.22074/cellj.2016.3717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Objective Pancreatic stroma plays an important role in the induction of pancreatic cells
by the use of close range signaling. In this respect, we presume that pancreatic mesenchymal cells (PMCs) as a fundamental factor of the stromal niche may have an effective
role in differentiation of umbilical cord blood cluster of differentiation 133+ (UCB-CD133+)
cells into newly-formed β-cells in vitro.
Materials and Methods This study is an experimental research. The UCB-CD133+cells
were purified by magnetic activated cell sorting (MACS) and differentiated into insulin
producing cells (IPCs) in co-culture, both directly and indirectly with rat PMCs. Immunocytochemistry and enzyme linked immune sorbent assay (ELISA) were used to determine
expression and production of insulin and C-peptide at the protein level.
Results Our results demonstrated that UCB-CD133+differentiated into IPCs. Cells in
islet-like clusters with (out) co-cultured with rat pancreatic stromal cells produced insulin
and C-peptide and released them into the culture medium at the end of the induction protocol. However they did not respond well to glucose challenges.
Conclusion Rat PMCs possibly affect differentiation of UCB-CD133+cells into IPCs by
increasing the number of immature β-cells.
Collapse
Affiliation(s)
- Fazel Sahraneshin Samani
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran ; Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran ; Department of Regenerative Biomedicine at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tahereh Zandieh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reyhaneh Khoshchehreh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran ; Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Biomedicine at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
32
|
Zhong L, Gou J, Deng N, Shen H, He T, Zhang BQ. Three-dimensional co-culture of hepatic progenitor cells and mesenchymal stem cells in vitro and in vivo. Microsc Res Tech 2015; 78:688-96. [PMID: 26031767 DOI: 10.1002/jemt.22526] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 03/24/2015] [Accepted: 04/30/2015] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Here we co-cultured hepatic progenitor cells (HPCs) and mesenchymal stem cells (MSCs) to investigate whether the co-culture environments could increase hepatocytes form. METHODS Three-dimensional (3D) co-culture model of HPCs and MSCs was developed and morphological features of cells were continuously observed. Hepatocyte specific markers Pou5f1/Oct4, AFP, CK-18 and Alb were analyzed to confirm the differentiation of HPCs. The mRNA expression of CK-18 and Alb was analyzed by RT-PCR to investigate the influence of co-culture model to the terminal differentiation process of mature hepatocytes. The functional properties of hepatocyte-like cells were detected by continuously monitoring the albumin secretion using Gaussia luciferase assays. Scaffolds with HPCs and MSCs were implanted into nude mouse subcutaneously to set up the in vivo co-culture model. RESULTS Although two groups formed smooth spheroids and high expressed of CK-18 and Alb, hybrid spheroids had more regular structures and higher cell density. CK-18 and Alb mRNA were at a relatively higher expression level in co-culture system during the whole cultivation time (P < 0.05). Albumin secretion rates in the hybrid spheroids had been consistently higher than that in the mono-culture spheroids (P < 0.05). In vivo, the hepatocyte-like cells were consistent with the morphological features of mature hepatocytes and more well-differentiated hepatocyte-like cells were observed in the co-culture group. CONCLUSIONS HPCs and MSCs co-culture system is an efficient way to form well-differentiated hepatocyte-like cells, hence, may be helpful to the cell therapy of hepatic tissues and alleviate the problem of hepatocytes shortage.
Collapse
Affiliation(s)
- Li Zhong
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| | - Juhua Gou
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| | - Nian Deng
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| | - Hao Shen
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| | - Tongchuan He
- Department of Surgery, Molecular Oncology Laboratory, the University of Chicago Medical Center, Chicago, Illinois, 60637
| | - Bing-Qiang Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Chongqing, 400016, China
| |
Collapse
|
33
|
Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells into Insulin-Producing Cells: Evidence for Further Maturation In Vivo. BIOMED RESEARCH INTERNATIONAL 2015; 2015:575837. [PMID: 26064925 PMCID: PMC4443784 DOI: 10.1155/2015/575837] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/11/2015] [Indexed: 12/24/2022]
Abstract
The aim of this study was to provide evidence for further in vivo maturation of insulin-producing cells (IPCs) derived from human bone marrow-derived mesenchymal stem cells (HBM-MSCs). HBM-MSCs were obtained from three insulin-dependent type 2 diabetic volunteers. Following expansion, cells were differentiated according to a trichostatin-A/GLP protocol. One million cells were transplanted under the renal capsule of 29 diabetic nude mice. Blood glucose, serum human insulin and c-peptide levels, and glucose tolerance curves were determined. Mice were euthanized 1, 2, 4, or 12 weeks after transplantation. IPC-bearing kidneys were immunolabeled, number of IPCs was counted, and expression of relevant genes was determined. At the end of in vitro differentiation, all pancreatic endocrine genes were expressed, albeit at very low values. The percentage of IPCs among transplanted cells was small (≤3%). Diabetic animals became euglycemic 8 ± 3 days after transplantation. Thereafter, the percentage of IPCs reached a mean of ~18% at 4 weeks. Relative gene expression of insulin, glucagon, and somatostatin showed a parallel increase. The ability of the transplanted cells to induce euglycemia was due to their further maturation in the favorable in vivo microenvironment. Elucidation of the exact mechanism(s) involved requires further investigation.
Collapse
|
34
|
Hefei W, Yu R, Haiqing W, Xiao W, Jingyuan W, Dongjun L. Morphological characteristics and identification of islet-like cells derived from rat adipose-derived stem cells cocultured with pancreas adult stem cells. Cell Biol Int 2015; 39:253-63. [PMID: 25262665 DOI: 10.1002/cbin.10387] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 07/31/2014] [Indexed: 12/14/2022]
Abstract
Diabetes is a significant public health problem that can be treated with insulin therapy; however, therapies designed to cure diabetes are limited. The goal of the current study was to assess the potential for curative treatment of diabetes using adipose-derived stem cells (ADSCs). To achieve this goal, the differentiation of rat ADSCs into pancreatic islet-like cells induced by coculture with pancreatic adult stem cells (PASCs) was characterized. Differentiation of ADSCs into islet-like cells induced by coculturing was determined morphologically, as well as by the assessment of islet cell markers using dithizone staining, immunohistochemistry, RT-PCR, qPCR, and western blotting. The results showed that ADSCs formed islet-like round cell masses after coculture with PASCs. These differentiated cells were shown to be positive for islet cell markers, including dithizone incorporation; PDX1, CK19 and Nestin by immunohistochemistry, and insulin, PDX1 and glucagon expression by RT-PCR. Differentiated ADSCs induced by coculturing also expressed insulin at the mRNA and protein level, with the level of insulin mRNA expression in cocultured ADSCs being 0.05 times greater than that of PASCs (P < 0.05). Taken together, our results demonstrate that ADSCs can be induced to differentiate into islet-like cells by coculture with PASCs; thus these cells can be used for transplantation, providing a theoretical foundation for the treatment of diabetes using this approach.
Collapse
Affiliation(s)
- Wang Hefei
- National Research Center for Animal Transgenic Bio-technology, Inner Mongolia University, 24 Zhao Jun Road, Hohhot, 010070, China
| | | | | | | | | | | |
Collapse
|
35
|
Caruso M, Parolini O. Multipotent Mesenchymal Stromal Cell-Based Therapies: Regeneration Versus Repair. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
36
|
Hashemian SJ, Kouhnavard M, Nasli-Esfahani E. Mesenchymal Stem Cells: Rising Concerns over Their Application in Treatment of Type One Diabetes Mellitus. J Diabetes Res 2015; 2015:675103. [PMID: 26576437 PMCID: PMC4630398 DOI: 10.1155/2015/675103] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 01/17/2015] [Accepted: 01/18/2015] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disorder that leads to beta cell destruction and lowered insulin production. In recent years, stem cell therapies have opened up new horizons to treatment of diabetes mellitus. Among all kinds of stem cells, mesenchymal stem cells (MSCs) have been shown to be an interesting therapeutic option based on their immunomodulatory properties and differentiation potentials confirmed in various experimental and clinical trial studies. In this review, we discuss MSCs differential potentials in differentiation into insulin-producing cells (IPCs) from various sources and also have an overview on currently understood mechanisms through which MSCs exhibit their immunomodulatory effects. Other important issues that are provided in this review, due to their importance in the field of cell therapy, are genetic manipulations (as a new biotechnological method), routes of transplantation, combination of MSCs with other cell types, frequency of transplantation, and special considerations regarding diabetic patients' autologous MSCs transplantation. At the end, utilization of biomaterials either as encapsulation tools or as scaffolds to prevent immune rejection, preparation of tridimensional vascularized microenvironment, and completed or ongoing clinical trials using MSCs are discussed. Despite all unresolved concerns about clinical applications of MSCs, this group of stem cells still remains a promising therapeutic modality for treatment of diabetes.
Collapse
Affiliation(s)
- Seyed Jafar Hashemian
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Seyed Jafar Hashemian:
| | - Marjan Kouhnavard
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Nasli-Esfahani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Ren L, Chen L, Qi H, Li F, Gong F. In vitro differentiation of human adipose tissue-derived stem cells into islet-like clusters promoted by islet neogenesis-associated protein pentadecapeptide. Cells Tissues Organs 2014; 199:329-41. [PMID: 25471531 DOI: 10.1159/000362500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2014] [Indexed: 11/19/2022] Open
Abstract
Human adipose tissue-derived stem cells (hASCs) are considered an ideal tool for the supply of insulin-producing cells to treat diabetes mellitus, with high differentiation efficiency. Islet neogenesis-associated protein (INGAP) is an initiator of islet neogenesis, and the peptide sequence comprising amino acids 104-118, named INGAP pentadecapeptide (INGAP-PP), has been shown to increase β-cell mass in animals and human pathological states. Here, we report a novel 4-step method to promote hASCs to differentiate into islet-like clusters (ILCs) more efficiently by adding INGAP-PP. The hASCs were isolated, purified and differentiated using a 4-step protocol including trichostatin A, INGAP-PP/scrambled peptide (Scrambled-P), dexamethasone, nicotinamide, glucagon-like peptide-1, transforming growth factor β1 and exendin-4. Results showed that ILCs in the INGAP-PP group were more similar to the fresh islets with regard to both size and morphology and expressed significantly higher levels of both insulin and C-peptide than those in the Scrambled-P group. Moreover, the ILCs from the INGAP-PP group secreted higher levels of insulin and C-peptide than those from the Scrambled-P group in response to both a low (5.6 mM) and high (25 mM) glucose challenge and secreted 6 times more hormones under the high-glucose challenge. Real-time PCR and immunocytochemistry showed that ILCs of the INGAP-PP group expressed human pancreatic endocrine hormones and transcription factors. Transplantation of ILCs into diabetic rats partially reversed diabetes and prolonged their life span. In conclusion, the INGAP-PP protocol can efficiently induce hASCs to differentiate into ILCs in vitro, and thus hASCs could be a promising source of cells for transplantation to treat diabetes mellitus.
Collapse
Affiliation(s)
- Lili Ren
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | |
Collapse
|
38
|
Jung IS, Kim HJ, Noh R, Kim SC, Kim CW. Effects of extremely low frequency magnetic fields on NGF induced neuronal differentiation of PC12 cells. Bioelectromagnetics 2014; 35:459-69. [DOI: 10.1002/bem.21861] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 04/25/2014] [Indexed: 12/21/2022]
Affiliation(s)
- In-Soo Jung
- College of Life Sciences and Biotechnology; Korea University; Seoul Korea
| | - Hyun-Jung Kim
- College of Life Sciences and Biotechnology; Korea University; Seoul Korea
| | - Ran Noh
- College of Life Sciences and Biotechnology; Korea University; Seoul Korea
| | - Soo-Chan Kim
- Department of Electrical Electronic and Control Engineering; Hankyong National University; Anseong-si Kyonggi-do Korea
| | - Chan-Wha Kim
- College of Life Sciences and Biotechnology; Korea University; Seoul Korea
| |
Collapse
|
39
|
Orlando G, Gianello P, Salvatori M, Stratta RJ, Soker S, Ricordi C, Domínguez-Bendala J. Cell replacement strategies aimed at reconstitution of the β-cell compartment in type 1 diabetes. Diabetes 2014; 63:1433-44. [PMID: 24757193 DOI: 10.2337/db13-1742] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Emerging technologies in regenerative medicine have the potential to restore the β-cell compartment in diabetic patients, thereby overcoming the inadequacies of current treatment strategies and organ supply. Novel approaches include: 1) Encapsulation technology that protects islet transplants from host immune surveillance; 2) stem cell therapies and cellular reprogramming, which seek to regenerate the depleted β-cell compartment; and 3) whole-organ bioengineering, which capitalizes on the innate properties of the pancreas extracellular matrix to drive cellular repopulation. Collaborative efforts across these subfields of regenerative medicine seek to ultimately produce a bioengineered pancreas capable of restoring endocrine function in patients with insulin-dependent diabetes.
Collapse
|
40
|
Contribution of murine bone marrow mesenchymal stem cells to pancreas regeneration after partial pancreatectomy in mice. Cell Biol Int 2014; 36:823-31. [PMID: 22574754 DOI: 10.1042/cbi20110680] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The implantation of BMSCs (bone marrow mesenchymal stem cells) has emerged as a potential method of treating tissue damage, but the in vivo differentiation of BMSCs in an injured pancreas and its therapeutic effects have not been determined. Our aim has been to investigate the potential of BMSCs to contribute to the parenchyma and mesenchymal components of the pancreas during rapid regeneration, with preliminary exploration of the molecular mechanisms of this process. GFP(+) (green fluorescent protein(+) ) BMSCs were intravenously infused into the tail veins of mice that had received a 65-70% partial pancreatectomy, while mice that had only received a partial pancreatectomy and mice that had only been injected with BMSCs served as controls. Four weeks later, the injected GFP(+) BMSCs were diffusely engrafted in the pancreatic parenchyma and mesenchyma of the recipient mice with pancreatic injuries and had differentiated into pancreatic ductal epithelial cells (accounting for 1.7±0.3%), vascular endothelial cells (3.2±0.6%) and PSCs (pancreatic stellate cells) (5.2±1.6%), but no β or neural cells. Significantly, more engrafted and differentiated GFP(+) BMSCs were observed in the regenerating pancreas than in the normal pancreas. For the mice that received a partial pancreatectomy, the pancreatic weight/body weight of the mice with BMSC treatment was greater than mice without BMSC treatment (P<0.05). In addition, real-time RT-PCR (reverse transcription-PCR) showed that the expression levels of miR-9 (microRNA 9) and miR-204 in the engrafted BMSCs (5.2- and 2.6-fold, P<0.05, respectively) were increased compared with wild-type BMSCs. We also observed a significant reduction in the expression of miR-375 (0.71-fold, P<0.05) in engrafted GFP(+) BMSCs compared with wild-type BMSCs. BMSCs can therefore be a potential cell bank for treating pancreatic injuries by contributing to a variety of cell types. This process might be related to the expression of miR-9, miR-204 and miR-375.
Collapse
|
41
|
Ebrahimie M, Esmaeili F, Cheraghi S, Houshmand F, Shabani L, Ebrahimie E. Efficient and simple production of insulin-producing cells from embryonal carcinoma stem cells using mouse neonate pancreas extract, as a natural inducer. PLoS One 2014; 9:e90885. [PMID: 24614166 PMCID: PMC3948699 DOI: 10.1371/journal.pone.0090885] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/03/2014] [Indexed: 01/15/2023] Open
Abstract
An attractive approach to replace the destroyed insulin-producing cells (IPCs) is the generation of functional β cells from stem cells. Embryonal carcinoma (EC) stem cells are pluripotent cells which can differentiate into all cell types. The present study was carried out to establish a simple nonselective inductive culture system for generation of IPCs from P19 EC cells by 1–2 weeks old mouse pancreas extract (MPE). Since, mouse pancreatic islets undergo further remodeling and maturation for 2–3 weeks after birth, we hypothesized that the mouse neonatal MPE contains essential factors to induce in vitro differentiation of pancreatic lineages. Pluripotency of P19 cells were first confirmed by expression analysis of stem cell markers, Oct3/4, Sox-2 and Nanog. In order to induce differentiation, the cells were cultured in a medium supplemented by different concentrations of MPE (50, 100, 200 and 300 µg/ml). The results showed that P19 cells could differentiate into IPCs and form dithizone-positive cell clusters. The generated P19-derived IPCs were immunoreactive to proinsulin, insulin and insulin receptor beta. The expression of pancreatic β cell genes including, PDX-1, INS1 and INS2 were also confirmed. The peak response at the 100 µg/ml MPE used for investigation of EP300 and CREB1 gene expression. When stimulated with glucose, these cells synthesized and secreted insulin. Network analysis of the key transcription factors (PDX-1, EP300, CREB1) during the generation of IPCs resulted in introduction of novel regulatory candidates such as MIR17, and VEZF1 transcription factors, as well as MORN1, DKFZp761P0212, and WAC proteins. Altogether, we demonstrated the possibility of generating IPCs from undifferentiated EC cells, with the characteristics of pancreatic β cells. The derivation of pancreatic cells from EC cells which are ES cell siblings would provide a valuable experimental tool in study of pancreatic development and function as well as rapid production of IPCs for transplantation.
Collapse
Affiliation(s)
- Marzieh Ebrahimie
- Department of Biology, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Fariba Esmaeili
- Department of Biology, Faculty of Basic Sciences, University of Isfahan, Isfahan, Iran
- Research Institute of Biotechnology, Shahrekord University, Shahrekord, Iran
| | - Somayeh Cheraghi
- Department of Biology, Faculty of Basic Sciences, Azad Islamic University of Shahrekord, Shahrekord, Iran
| | - Fariba Houshmand
- Department of Physiology, Faculty of Medical Sciences, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Leila Shabani
- Department of Biology, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
- Research Institute of Biotechnology, Shahrekord University, Shahrekord, Iran
| | - Esmaeil Ebrahimie
- Institute of Biotechnology, Shiraz University, Shiraz, Iran
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, Australia
- * E-mail:
| |
Collapse
|
42
|
Tsai PJ, Wang HS, Lin CH, Weng ZC, Chen TH, Shyu JF. Intraportal injection of insulin-producing cells generated from human bone marrow mesenchymal stem cells decreases blood glucose level in diabetic rats. Endocr Res 2014; 39:26-33. [PMID: 23772634 DOI: 10.3109/07435800.2013.797432] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We studied the process of trans-differentiation of human bone marrow mesenchymal stem cells (hBM-MSCs) into insulin-producing cells. Streptozotocin (STZ)-induced diabetic rat model was used to study the effect of portal vein transplantation of these insulin-producing cells on blood sugar levels. The BM-MSCs were differentiated into insulin-producing cells under defined conditions. Real-time PCR, immunocytochemistry and glucose challenge were used to evaluate in vitro differentiation. Flow cytometry showed that hBM-MSCs were strongly positive for CD44, CD105 and CD73 and negative for hematopoietic markers CD34, CD38 and CD45. Differentiated cells expressed C-peptide as well as β-cells specific genes and hormones. Glucose stimulation increased C-peptide secretion in these cells. The insulin-producing, differentiated cells were transplanted into the portal vein of STZ-induced diabetic rats using a Port-A catheter. The insulin-producing cells were localized in the liver of the recipient rat and expressed human C-peptide. Blood glucose levels were reduced in diabetic rats transplanted with insulin-producing cells. We concluded that hBM-MSCs could be trans-differentiated into insulin-producing cells in vitro. Portal vein transplantation of insulin-producing cells alleviated hyperglycemia in diabetic rats.
Collapse
Affiliation(s)
- Pei-Jiun Tsai
- Institute of Clinical Medicine, National Yang Ming University, Taipei , Taiwan , R.O.C
| | | | | | | | | | | |
Collapse
|
43
|
Reduction of fibrosis in dibutyltin dichloride-induced chronic pancreatitis using rat umbilical mesenchymal stem cells from Wharton's jelly. Pancreas 2013; 42:1291-302. [PMID: 24152954 DOI: 10.1097/mpa.0b013e318296924e] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES The objective of this study was to investigate the effects of rat umbilical cord mesenchymal stem cells (UCMSCs) from Wharton's jelly on dibutyltin dichloride (DBTC)-induced chronic pancreatitis (CP) and subsequent pancreatic fibrosis in rats. METHODS A rat model of CP induced by DBTC was used. Male Sprague-Dawley rats were randomly divided into 4 groups: the control, DBTC, DBTC + UCMSCs, and control + UCMSC groups. Umbilical cord mesenchymal stem cells were administered intravenously on day 5 after the administration of DBTC. On days 14 and 28, the rats were evaluated morphologically and biochemically. The expression levels of inflammatory cytokines and chemokines in the pancreatic tissues of different groups were evaluated using quantitative real-time polymerase chain reaction. The activation of pancreatic stellate cells was estimated by immunochemistry and Western blot analysis of α-smooth muscle actin. RESULTS Umbilical cord mesenchymal stem cells were detected in inflamed pancreatic tissues. Umbilical cord mesenchymal stem cell treatment improved the histological scores and alleviated the fibrosis of pancreas samples, The expression of cytokines in the DBTC + UCMSC group was significantly lower than that in the DBTC group. Also, pancreatic stellate cell activation was inhibited by UCMSC treatment. CONCLUSIONS Xenogeneic transplantation of UCMSCs is a novel approach for the treatment of CP and subsequent fibrosis. Umbilical cord mesenchymal stem cells may be a promising therapeutic intervention for human CP in the future.
Collapse
|
44
|
Are Mesenchymal Cells Indeed Pluripotent Stem Cells or Just Stromal Cells? OCT-4 and VSELs Biology Has Led to Better Understanding. Stem Cells Int 2013; 2013:547501. [PMID: 24187558 PMCID: PMC3800663 DOI: 10.1155/2013/547501] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/06/2013] [Accepted: 08/26/2013] [Indexed: 12/24/2022] Open
Abstract
Stem cells have excited researchers because of their potential to regenerate. However, which stem cells will be the best candidate for regenerative medicine remains an enigma. Compared to pluripotent stem cells with associated risks of immune rejection and teratoma formation, adult stem cells especially the mesenchymal stem cells (MSCs) are hyped to be a suitable alternate since they also exhibit pluripotent properties. This review shows that there is a subpopulation of pluripotent very small embryonic-like stem cells (VSELs) among MSCs culture. The two populations differ from each other in expression pattern of OCT-4. VSELs exhibit nuclear OCT-4A, whereas the MSCs have cytoplasmic OCT-4B, similar to our earlier findings in testis and ovary. Pluripotent VSELs with nuclear OCT-4A exist in various adult body organs, and the immediate progenitors express cytoplasmic OCT-4B which is eventually lost as the cell differentiates further. To conclude it is essential to discriminate between nuclear and cytoplasmic OCT-4 expression and also to acknowledge the presence of VSELs.
Collapse
|
45
|
Xie H, Wang Y, Zhang H, Qi H, Zhou H, Li FR. Role of injured pancreatic extract promotes bone marrow-derived mesenchymal stem cells efficiently differentiate into insulin-producing cells. PLoS One 2013; 8:e76056. [PMID: 24058711 PMCID: PMC3776851 DOI: 10.1371/journal.pone.0076056] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 08/24/2013] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be successfully induced to differentiate into insulin-producing cells (IPCs) by a variety of small molecules and cytokines in vitro. However, problems remain, such as low transdifferentiation efficiency and poor maturity of trans-differentiated cells. The damaged pancreatic cells secreted a large amount of soluble proteins, which were able to promote pancreative islet regeneration and MSCs differentiation. In this study, we utilized the rat injured pancreatic tissue extract to modulate rat bone marrow-derived MSCs differentiation into IPCs by the traditional two-step induction. Our results showed that injured pancreatic tissue extract could effectively promote the trans-differentiation efficiency and maturity of IPCs by the traditional induction. Moreover, IPCs were able to release more insulin in a glucose-dependent manner and ameliorate better the diabetic conditions of streptozotocin (STZ)-treated rats. Our study provides a new strategy to induce an efficient and directional differentiation of MSCs into IPCs.
Collapse
Affiliation(s)
- Hongbin Xie
- The Key Laboratory of stem cell and cellular therapy, the Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University, Shenzhen, China
| | - Yunshuai Wang
- The Key Laboratory of stem cell and cellular therapy, the Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University, Shenzhen, China
| | - Hui Zhang
- Laboratory of Cancer Cell Proteomics, Nevada Cancer Institute, Las Vegas, Nevada, United States of America
| | - Hui Qi
- The Key Laboratory of stem cell and cellular therapy, the Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University, Shenzhen, China
| | - Hanxin Zhou
- Department of General Surgery, First Hospital (Shenzhen second People’s Hospital) of Shenzhen University, Shenzhen, China
| | - Fu-Rong Li
- The Key Laboratory of stem cell and cellular therapy, the Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University, Shenzhen, China
- Shenzhen Institute of Gerontology, Shenzhen, China
- * E-mail:
| |
Collapse
|
46
|
Anisimov SV, Zemelko VI, Grinchuk TM, Nikolsky NN. Menstrual blood stem cells as a potential source for cell therapy. ACTA ACUST UNITED AC 2013. [DOI: 10.1134/s1990519x13030024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
47
|
Mesenchymal stem cell therapy in diabetes mellitus: progress and challenges. J Nucleic Acids 2013; 2013:194858. [PMID: 23762531 PMCID: PMC3666198 DOI: 10.1155/2013/194858] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 04/18/2013] [Indexed: 02/07/2023] Open
Abstract
Advanced type 2 diabetes mellitus is associated with significant morbidity and mortality due to cardiovascular, nervous, and renal complications. Attempts to cure diabetes mellitus using islet transplantation have been successful in providing a source for insulin secreting cells. However, limited donors, graft rejection, the need for continued immune suppression, and exhaustion of the donor cell pool prompted the search for a more sustained source of insulin secreting cells. Stem cell therapy is a promising alternative for islet transplantation in type 2 diabetic patients who fail to control hyperglycemia even with insulin injection. Autologous stem cell transplantation may provide the best outcome for those patients, since autologous cells are readily available and do not entail prolonged hospital stays or sustained immunotoxic therapy. Among autologous adult stem cells, mesenchymal stem cells (MSCs) therapy has been applied with varying degrees of success in both animal models and in clinical trials. This review will focus on the advantages of MSCs over other types of stem cells and the possible mechanisms by which MSCs transplant restores normoglycemia in type 2 diabetic patients. Sources of MSCs including autologous cells from diabetic patients and the use of various differentiation protocols in relation to best transplant outcome will be discussed.
Collapse
|
48
|
Expression of the chitinase family glycoprotein YKL-40 in undifferentiated, differentiated and trans-differentiated mesenchymal stem cells. PLoS One 2013; 8:e62491. [PMID: 23671604 PMCID: PMC3650021 DOI: 10.1371/journal.pone.0062491] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 03/22/2013] [Indexed: 12/19/2022] Open
Abstract
The glycoprotein YKL-40 (CHI3L1) is a secreted chitinase family protein that induces angiogenesis, cell survival, and cell proliferation, and plays roles in tissue remodeling and immune regulation. It is expressed primarily in cells of mesenchymal origin, is overexpressed in numerous aggressive carcinomas and sarcomas, but is rarely expressed in normal ectodermal tissues. Bone marrow-derived mesenchymal stem cells (MSCs) can be induced to differentiate into various mesenchymal tissues and trans-differentiate into some non-mesenchymal cell types. Since YKL-40 has been used as a mesenchymal marker, we followed YKL-40 expression as undifferentiated MSCs were induced to differentiate into bone, cartilage, and neural phenotypes. Undifferentiated MSCs contain significant levels of YKL-40 mRNA but do not synthesize detectable levels of YKL-40 protein. MSCs induced to differentiate into chondrocytes and osteocytes soon began to express and secrete YKL-40 protein, as do ex vivo cultured chondrocytes and primary osteocytes. In contrast, MSCs induced to trans-differentiate into neurons did not synthesize YKL-40 protein, consistent with the general absence of YKL-40 protein in normal CNS parenchyma. However, these trans-differentiated neurons retained significant levels of YKL-40 mRNA, suggesting the mechanisms which prevented YKL-40 translation in undifferentiated MSCs remained in place, and that these trans-differentiated neurons differ in at least this way from neurons derived from neuronal stem cells. Utilization of a differentiation protocol containing β-mercaptoethanol resulted in cells that expressed significant amounts of intracellular YKL-40 protein that was not secreted, which is not seen in normal cells. Thus the synthesis of YKL-40 protein is a marker for MSC differentiation into mature mesenchymal phenotypes, and the presence of untranslated YKL-40 mRNA in non-mesenchymal cells derived from MSCs reflects differences between differentiated and trans-differentiated phenotypes.
Collapse
|
49
|
Karaoz E, Okcu A, Ünal ZS, Subasi C, Saglam O, Duruksu G. Adipose tissue-derived mesenchymal stromal cells efficiently differentiate into insulin-producing cells in pancreatic islet microenvironment both in vitro and in vivo. Cytotherapy 2013; 15:557-70. [DOI: 10.1016/j.jcyt.2013.01.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 11/07/2012] [Accepted: 01/07/2013] [Indexed: 12/16/2022]
|
50
|
Ricordi C, Inverardi L, Domínguez-Bendala J. From cellular therapies to tissue reprogramming and regenerative strategies in the treatment of diabetes. Regen Med 2012; 7:41-8. [DOI: 10.2217/rme.12.70] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Diabetes mellitus represents a global epidemic affecting over 350 million patients worldwide and projected by the WHO to surpass the 500 million patient mark within the next two decades. Besides Type 1 and Type 2 diabetes mellitus, the study of the endocrine compartment of the pancreas is of great translational interest, as strategies aimed at restoring its mass could become therapies for glycemic dysregulation, drug-related diabetes following diabetogenic therapies, or hyperglycemic disturbances following the treatment of cancer and nesidioblastosis. Such strategies generally fall under one of the ‘three Rs’: replacement (islet transplantation and stem cell differentiation); reprogramming (e.g., from the exocrine compartment of the pancreas); and regeneration (replication and induction of endogenous stem cells). As the latter has been extensively reviewed in recent months by us and others, this article focuses on emerging reprogramming and replacement approaches.
Collapse
Affiliation(s)
- Camillo Ricordi
- University of Miami Cell Transplant Center and Diabetes Research Institute, Miami, FL, USA
| | - Luca Inverardi
- University of Miami Cell Transplant Center and Diabetes Research Institute, Miami, FL, USA
| | - Juan Domínguez-Bendala
- University of Miami Cell Transplant Center and Diabetes Research Institute, Miami, FL, USA
| |
Collapse
|