1
|
Ding X, Xie S, Zhang W, Zhu Y, Xu D, Xian S, Sun H, Guo X, Li Y, Lu J, Tong X, Huang R, Ji S, Xia Z. Current application of tissue-engineered dermal scaffolds mimicking the extracellular matrix microenvironment in wound healing. Regen Ther 2025; 28:371-382. [PMID: 39896445 PMCID: PMC11786805 DOI: 10.1016/j.reth.2024.12.018] [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: 10/20/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
With the continuous advancement of materials science, cell biology, and biotechnology, tissue engineering has introduced novel solutions to traditional wound healing approaches, particularly demonstrating significant potential in addressing complex or non-healing wounds. One of the key technologies in this field, dermal scaffolds, serve as wound coverage materials that mimic the structural framework of the dermis. They primarily assume the function of extracellular matrix, providing space for cell attachment, migration, and proliferation, thus supporting cellular growth and regulating multiple biological processes in healing. Tissue engineering utilizes combinations of natural or synthetic scaffolds, seeded cells, or growth factors to induce distinct effects in angiogenesis, extracellular matrix deposition, and functional recovery. Therefore, various bioengineered dermal scaffolds hold significant potential for clinical translation in wound healing. This review outlines various extracellular matrix molecules utilized in the development of dermal scaffolds, emphasizes recent progress in cell- and growth factor-modified scaffolds, and discusses the challenges and future perspectives in this evolving field.
Collapse
Affiliation(s)
| | | | | | - Yushu Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Dayuan Xu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shuyuan Xian
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Hanlin Sun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Yixu Li
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
2
|
Huang J, Deng Q, Tsang LL, Chang G, Guo J, Ruan YC, Wang CC, Li G, Chan HF, Zhang X, Jiang X. Mesenchymal stem cells from perinatal tissues promote diabetic wound healing via PI3K/AKT activation. Stem Cell Res Ther 2025; 16:59. [PMID: 39923118 PMCID: PMC11807333 DOI: 10.1186/s13287-025-04141-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/13/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Diabetic foot ulcers (DFUs) represent a major complication of diabetes, often leading to poor healing outcomes with conventional treatments. Mesenchymal stem cell (MSC) therapies have emerged as a promising alternative, given their potential to modulate various pathways involved in wound healing. This study evaluates and compares the therapeutic potential of MSCs derived from perinatal tissues-human umbilical cord MSCs (hUCMSCs), human chorionic villi MSCs (hCVMSCs), and human decidua basalis MSCs (hDCMSCs)-in a diabetic wound healing model. METHODS We performed in vitro and in vivo studies to compare the efficacy of hUCMSCs, hCVMSCs, and hDCMSCs. Mass spectrometry was used to analyze the secreted proteins of the MSCs. We incorporated the MSCs into a polyethylene glycol diacrylate (PEGDA) and sodium alginate (SA) hydrogel matrix with collagen I (Col-I) to evaluate their effects on wound healing. RESULTS All three types of MSCs promoted wound healing, with hUCMSCs and hCVMSCs showing stronger effects compared to hDCMSCs. Both hUCMSCs and hCVMSCs demonstrated robust wound healing kinetics, with enhanced keratinocyte proliferation (KRT14+/Ki67+ cells), maturation (KRT10/KRT14 ratio), and angiogenesis. In vitro studies demonstrated that the MSC-derived secretome enhanced keratinocyte proliferation and migration, endothelial cell function and stem cell recruitment, indicating robust paracrine effects. Mass spectrometry revealed a conserved set of proteins including THBS1 (thrombospondin 1), SERPINE1 (serpin family E member 1), ANXA1 (annexin A1), LOX (lysyl oxidase), and ITGB1 (integrin beta-1) which are involved in extracellular matrix (ECM) organization and wound healing, with the PI3K/AKT signaling pathway playing a central role. The PEGDA/SA/Col-I hydrogel demonstrated a unique balance of mechanical and biological properties and an optimal environment for MSC viability and function. Application of either hUCMSC- or hCVMSC-laden hydrogels resulted in accelerated wound closure, improved re-epithelialization, increased collagen deposition, and enhanced vascularization in vivo. CONCLUSIONS MSCs From perinatal tissues particularly hUCMSCs and hCVMSCs significantly enhance diabetic wound healing through PI3K/AKT pathway activation while hDCMSCs exhibited weaker efficacy. The PEGDA/SA/Col-I hydrogel supports MSC viability and function offering a promising scaffold for DFU treatment. These findings underscore the potential of specific perinatal MSCs and optimized hydrogel formulations in advancing diabetic wound care.
Collapse
Affiliation(s)
- Jiawei Huang
- School of Biomedical Sciences, Faculty of Medicine; CUHK-GIBH CAS Joint Research Laboratory On Stem Cell and Regenerative Medicine; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qingwen Deng
- School of Biomedical Sciences, Faculty of Medicine; CUHK-GIBH CAS Joint Research Laboratory On Stem Cell and Regenerative Medicine; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lai Ling Tsang
- School of Biomedical Sciences, Faculty of Medicine; CUHK-GIBH CAS Joint Research Laboratory On Stem Cell and Regenerative Medicine; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Guozhu Chang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jinghui Guo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, China
| | - Ye Chun Ruan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong; Reproduction and Development, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiaohu Zhang
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Xiaohua Jiang
- School of Biomedical Sciences, Faculty of Medicine; CUHK-GIBH CAS Joint Research Laboratory On Stem Cell and Regenerative Medicine; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, The Chinese University of Hong Kong, Hong Kong SAR, China.
- The Chinese University of Hong Kong, Shenzhen Research Institute, Shenzhen, 518000, China.
| |
Collapse
|
3
|
Shen J, Wu L, Shi X, Chen G, Liu T, Xu F, Xu X, Kou X, Zhao Y, Wang H, Wang C, Gao S, Xu S. Transplantation of the LRP1 high subpopulation of human umbilical cord-derived mesenchymal stem cells improves ovarian function in mice with premature ovarian failure and aged mice. Stem Cell Res Ther 2024; 15:64. [PMID: 38438896 PMCID: PMC10913679 DOI: 10.1186/s13287-024-03660-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Premature ovarian failure (POF) has a profound impact on female reproductive and psychological health. In recent years, the transplantation of umbilical cord-derived mesenchymal stem cells (UC-MSCs) has demonstrated unprecedented potential in the treatment of POF. However, the heterogeneity of human UC-MSCs remains a challenge for their large-scale clinical application. Therefore, it is imperative to identify specific subpopulations within UC-MSCs that possess the capability to improve ovarian function, with the aim of reducing the uncertainty arising from the heterogeneity while achieving more effective treatment of POF. METHODS 10 × Genomics was performed to investigate the heterogeneity of human UC-MSCs. We used LRP1 as a marker and distinguished the potential therapeutic subpopulation by flow cytometry, and determined its secretory functions. Unsorted UC-MSCs, LRP1high and LRP1low subpopulation was transplanted under the ovarian capsules of aged mice and CTX-induced POF mice, and therapeutic effects was evaluated by assessing hormone levels, estrous cycles, follicle counts, and embryo numbers. RNA sequencing on mouse oocytes and granulosa cells after transplantation was performed to explore the mechanism of LRP1high subpopulation on mouse oocytes and granulosa cells. RESULTS We identified three distinct functional subtypes, including mesenchymal stem cells, multilymphoid progenitor cells and trophoblasts. Additionally, we identified the LRP1high subpopulation, which improved ovarian function in aged and POF mice. We elucidated the unique secretory functions of the LRP1high subpopulation, capable of secreting various chemokines, cytokines, and growth factors. Furthermore, LRP1 plays a crucial role in regulating the ovarian microenvironment, including tissue repair and extracellular matrix remodeling. Consistent with its functions, the transcriptomes of oocytes and granulosa cells after transplantation revealed that the LRP1high subpopulation improves ovarian function by modulating the extracellular matrix of oocytes, NAD metabolism, and mitochondrial function in granulosa cells. CONCLUSION Through exploration of the heterogeneity of UC-MSCs, we identified the LRP1high subpopulation capable of improving ovarian function in aged and POF mice by secreting various factors and remodeling the extracellular matrix. This study provides new insights into the targeted exploration of human UC-MSCs in the precise treatment of POF.
Collapse
Affiliation(s)
- Jiacheng Shen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Li Wu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Xiaoying Shi
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Science and Technology, Tongji University, Tongji, 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Gang Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Tingwei Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fangfang Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiaocui Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiaochen Kou
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yanhong Zhao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hong Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Science and Technology, Tongji University, Tongji, 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shaorong Gao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Shaohua Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
4
|
Yaja K, Aungsuchawan S, Narakornsak S, Pothacharoen P, Pantan R, Tancharoen W. Combination of human platelet lysate and 3D gelatin scaffolds to enhance osteogenic differentiation of human amniotic fluid derived mesenchymal stem cells. Heliyon 2023; 9:e18599. [PMID: 37576189 PMCID: PMC10413082 DOI: 10.1016/j.heliyon.2023.e18599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023] Open
Abstract
Bone disorders are major health issues requiring specialized care; however, the traditional bone grafting method had several limitations. Thus, bone tissue engineering has become a potential alternative. In therapeutic treatments, using fetal bovine serum (FBS) as a culture supplement may result in the risk of contamination and host immunological response; therefore, human platelet lysate (hPL) has been considered a viable alternative source. This study attempted to compare the effectiveness and safety of different culture supplements, either FBS or hPL, on the osteoblastic differentiation potential of mesenchymal stem cells derived from human amniotic fluid (hAF-MSCs) under a three-dimensional gelatin scaffold. The results indicate that hAF-MSCs have the potential to be used in clinical applications as they meet the criteria for mesenchymal stem cells based on their morphology, the expression of a particular surface antigen, their proliferation ability, and their capacity for multipotent differentiation. After evaluation by MTT and Alamar blue proliferation assay, 10% of hPL was selected. The osteogenic differentiation of hAF-MSCs under three-dimensional gelatin scaffold using osteogenic-induced media supplemented with hPL was achievable and markedly stimulated osteoblast differentiation. Moreover, the expressions of osteoblastogenic related genes, including OCN, ALP, and COL1A1, exhibited the highest degree of expression under hPL-supplemented circumstances when compared with the control and the FBS-supplemented group. The induced cells under hPL-supplemented conditions also presented the highest ALP activity level and the greatest degree of calcium accumulation. These outcomes would indicate that hPL is a suitable substitute for animal derived serum. Importantly, osteogenic differentiation of human amniotic fluid derived mesenchymal stem cells using hPL-supplemented media and three-dimensional scaffolds may open the door to developing an alternative construct for repairing bone defects.
Collapse
Affiliation(s)
- Kantirat Yaja
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sirinda Aungsuchawan
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Suteera Narakornsak
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Peraphan Pothacharoen
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Thailand
| | - Rungusa Pantan
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Waleephan Tancharoen
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
5
|
Shirbaghaee Z, Heidari Keshel S, Rasouli M, Valizadeh M, Hashemi Nazari SS, Hassani M, Soleimani M. Report of a phase 1 clinical trial for safety assessment of human placental mesenchymal stem cells therapy in patients with critical limb ischemia (CLI). Stem Cell Res Ther 2023; 14:174. [PMID: 37408043 PMCID: PMC10324209 DOI: 10.1186/s13287-023-03390-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/29/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is associated with increased risk of tissue loss, leading to significant morbidity and mortality. Therapeutic angiogenesis using cell-based treatments, notably mesenchymal stem cells (MSCs), is essential for enhancing blood flow to ischemic areas in subjects suffering from CLI. The objective of this study was to evaluate the feasibility of using placenta-derived mesenchymal stem cells (P-MSCs) in patients with CLI. METHODS This phase I dose-escalation study investigated P-MSCs in nine CLI patients who were enrolled into each of the two dosage groups (20 × 106 and 60 × 106 cells), delivered intramuscularly twice, two months apart. The incidence of treatment-related adverse events was the primary endpoint. The decrease in inflammatory cytokines, improvement in the ankle-brachial pressure index (ABI), maximum walking distance, vascular collateralization, alleviation of rest pain, healing of ulceration, and avoidance of major amputation in the target leg were the efficacy outcomes. RESULTS All dosages of P-MSCs, including the highest tested dose of 60 × 106 cells, were well tolerated. During the 6-month follow-up period, there was a statistically significant decrease in IL-1 and IFN-γ serum levels following P-MSC treatment. The blood lymphocyte profile of participants with CLI did not significantly differ, suggesting that the injection of allogeneic cells did not cause T-cell proliferation in vivo. We found clinically substantial improvement in rest pain, ulcer healing, and maximum walking distance after P-MSC implantation. In patients with CLI, we performed minor amputations rather than major amputations. Angiography was unable to demonstrate new small vessels formation significantly. CONCLUSION The observations from this phase I clinical study indicate that intramuscular administration of P-MSCs is considered safe and well tolerated and may dramatically improve physical performance and minimize inflammatory conditions in patients with CLI. TRIAL REGISTRATION IRCT, IRCT20210221050446N1. Registered May 09, 2021.
Collapse
Affiliation(s)
- Zeinab Shirbaghaee
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Heidari Keshel
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Rasouli
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Valizadeh
- Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Saeed Hashemi Nazari
- Prevention of Cardiovascular Disease Research Center, Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassani
- Department of Vascular and Endovascular Surgery, Ayatollah Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Velenjak St., Shahid Chamran Highway, Tehran, Iran.
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Applied Cell Sciences and Hematology Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
6
|
Rosner M, Horer S, Feichtinger M, Hengstschläger M. Multipotent fetal stem cells in reproductive biology research. Stem Cell Res Ther 2023; 14:157. [PMID: 37287077 DOI: 10.1186/s13287-023-03379-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023] Open
Abstract
Due to the limited accessibility of the in vivo situation, the scarcity of the human tissue, legal constraints, and ethical considerations, the underlying molecular mechanisms of disorders, such as preeclampsia, the pathological consequences of fetomaternal microchimerism, or infertility, are still not fully understood. And although substantial progress has already been made, the therapeutic strategies for reproductive system diseases are still facing limitations. In the recent years, it became more and more evident that stem cells are powerful tools for basic research in human reproduction and stem cell-based approaches moved into the center of endeavors to establish new clinical concepts. Multipotent fetal stem cells derived from the amniotic fluid, amniotic membrane, chorion leave, Wharton´s jelly, or placenta came to the fore because they are easy to acquire, are not associated with ethical concerns or covered by strict legal restrictions, and can be banked for autologous utilization later in life. Compared to adult stem cells, they exhibit a significantly higher differentiation potential and are much easier to propagate in vitro. Compared to pluripotent stem cells, they harbor less mutations, are not tumorigenic, and exhibit low immunogenicity. Studies on multipotent fetal stem cells can be invaluable to gain knowledge on the development of dysfunctional fetal cell types, to characterize the fetal stem cells migrating into the body of a pregnant woman in the context of fetomaternal microchimerism, and to obtain a more comprehensive picture of germ cell development in the course of in vitro differentiation experiments. The in vivo transplantation of fetal stem cells or their paracrine factors can mediate therapeutic effects in preeclampsia and can restore reproductive organ functions. Together with the use of fetal stem cell-derived gametes, such strategies could once help individuals, who do not develop functional gametes, to conceive genetically related children. Although there is still a long way to go, these developments regarding the usage of multipotent fetal stem cells in the clinic should continuously be accompanied by a wide and detailed ethical discussion.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Stefanie Horer
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | | | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| |
Collapse
|
7
|
Gudauskaitė G, Kairienė I, Ivaškienė T, Rascon J, Mobasheri A. Therapeutic Perspectives for the Clinical Application of Umbilical Cord Hematopoietic and Mesenchymal Stem Cells: Overcoming Complications Arising After Allogeneic Hematopoietic Stem Cell Transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1409:111-126. [PMID: 35995905 DOI: 10.1007/5584_2022_726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
This review focuses on the therapeutic features of umbilical cord blood (UCB) cells as a source for allogeneic hematopoietic stem cell transplantation (aHSCT) in adult and child populations to treat malignant and nonmalignant hematologic diseases, genetic disorders, or pathologies of the immune system, when standard treatment (e.g., chemotherapy) is not effective or clinically contraindicated. In this article, we summarize the immunological properties and the advantages and disadvantages of using UCB stem cells and discuss a variety of treatment outcomes using different sources of stem cells from different donors both in adults and pediatric population. We also highlight the critical properties (total nucleated cell dose depending on HLA compatibility) of UCB cells that reach better survival rates, reveal the advantages of double versus single cord blood unit transplantation, and present recommendations from the most recent studies. Moreover, we summarize the mechanism of action and potential benefit of mesenchymal umbilical cord cells and indicate the most common posttransplantation complications.
Collapse
Affiliation(s)
- Greta Gudauskaitė
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Ignė Kairienė
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Tatjana Ivaškienė
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Jelena Rascon
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ali Mobasheri
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium.
| |
Collapse
|
8
|
Corsini M, Mitola S. Alternative In Vivo Models to Study Teratoma. Methods Mol Biol 2023; 2572:181-189. [PMID: 36161417 DOI: 10.1007/978-1-0716-2703-7_14] [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] [Indexed: 06/16/2023]
Abstract
Embryonic stem cells give rise to teratomas when injected in vivo in experimental animal models. The characterization, the manipulation, and the breaking off of this specific characteristic are doubtlessly the last frontier for the applications of stem cells in translational medicine. Moreover, the urgency to adapt to new scientific demands drives the researcher to find alternative and faster models for testing the teratogenic properties of embryonic stem cells. Here, we compare the emerging model of the chick embryo chorioallantoic membrane (CAM) to the murine model, which represents the gold standard procedure for teratogenesis.
Collapse
Affiliation(s)
- Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
9
|
da Silva CG, Martins CF. Stem Cells as Nuclear Donors for Mammalian Cloning. Methods Mol Biol 2023; 2647:105-119. [PMID: 37041331 DOI: 10.1007/978-1-0716-3064-8_5] [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: 04/13/2023]
Abstract
Mammals are routinely cloned by introducing somatic nuclei into enucleated oocytes. Cloning contributes to propagating desired animals, to germplasm conservation efforts, among other applications. A challenge to more broader use of this technology is the relatively low cloning efficiency, which inversely correlates with donor cell differentiation status. Emerging evidence suggests that adult multipotent stem cells improve cloning efficiency, while the greater potential of embryonic stem cells for cloning remains restricted to the mouse. The derivation of pluripotent or totipotent stem cells from livestock and wild species and their association with modulators of epigenetic marks in donor cells should increase cloning efficiency.
Collapse
Affiliation(s)
- Carolina Gonzales da Silva
- Federal Institute of Education, Science and Technology of Bahia, Campus Xique-Xique, Xique-Xique, Bahia, Brazil
| | - Carlos Frederico Martins
- Brazilian Agricultural Research Corporation (Embrapa Cerrados), Brasília, Federal District, Brazil.
| |
Collapse
|
10
|
Shirbaghaee Z, Hassani M, Heidari Keshel S, Soleimani M. Emerging roles of mesenchymal stem cell therapy in patients with critical limb ischemia. Stem Cell Res Ther 2022; 13:462. [PMID: 36068595 PMCID: PMC9449296 DOI: 10.1186/s13287-022-03148-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/19/2022] [Indexed: 11/25/2022] Open
Abstract
Critical limb ischemia (CLI), the terminal stage of peripheral arterial disease (PAD), is characterized by an extremely high risk of amputation and vascular issues, resulting in severe morbidity and mortality. In patients with severe limb ischemia with no alternative therapy options, such as endovascular angioplasty or bypass surgery, therapeutic angiogenesis utilizing cell-based therapies is vital for increasing blood flow to ischemic regions. Mesenchymal stem cells (MSCs) are currently considered one of the most encouraging cells as a regenerative alternative for the surgical treatment of CLI, including restoring tissue function and repairing ischemic tissue via immunomodulation and angiogenesis. The regenerative treatments for limb ischemia based on MSC therapy are still considered experimental. Despite recent advances in preclinical and clinical research studies, it is not recommended for regular clinical use. In this study, we review the immunomodulatory features of MSC besides the current understanding of different sources of MSC in the angiogenic treatment of CLI subjects and their potential applications as therapeutic agents. Specifically, this paper concentrates on the most current clinical application issues, and several recommendations are provided to improve the efficacy of cell therapy for CLI patients.
Collapse
Affiliation(s)
- Zeinab Shirbaghaee
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassani
- Department of Vascular and Endovascular Surgery, Ayatollah Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Heidari Keshel
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Applied Cell Science and Hematology Department, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
11
|
Moonshi SS, Adelnia H, Wu Y, Ta HT. Placenta‐Derived Mesenchymal Stem Cells for Treatment of Diseases: A Clinically Relevant Source. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Shehzahdi S. Moonshi
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
| | - Hossein Adelnia
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia Queensland 4072 Australia
| | - Yuao Wu
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
| | - Hang T. Ta
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
- Bioscience Discipline School of Environment and Science Griffith University Nathan Queensland 4111 Australia
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia Queensland 4072 Australia
| |
Collapse
|
12
|
Zhang Y, Zhong Y, Zou L, Liu X. Significance of Placental Mesenchymal Stem Cell in Placenta Development and Implications for Preeclampsia. Front Pharmacol 2022; 13:896531. [PMID: 35721156 PMCID: PMC9198303 DOI: 10.3389/fphar.2022.896531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/16/2022] [Indexed: 12/29/2022] Open
Abstract
The well-developed placentation is fundamental for the reproductive pregnancy while the defective placental development is the pathogenetic basis of preeclampsia (PE), a dangerous complication of pregnancy comprising the leading causes of maternal and perinatal morbidity and mortality. Placenta-derived mesenchymal stem cells (PMSCs) are a group of multipotent stem cells that own a potent capacity of differentiating into constitutive cells of vessel walls. Additionally, with the paracrine secretion of various factors, PMSCs inextricably link and interact with other component cells in the placenta, collectively improving the placental vasculature, uterine spiral artery remolding, and uteroplacental interface immunoregulation. Recent studies have further indicated that preeclamptic PMSCs, closely implicated in the abnormal crosstalk between other ambient cells, disturb the homeostasis and development in the placenta. Nevertheless, PMSCs transplantation or PMSCs exosome therapies tend to improve the placental vascular network and trophoblastic functions in the PE model, suggesting PMSCs may be a novel and putative therapeutic strategy for PE. Herein, we provide an overview of the multifaceted contributions of PMSCs in early placental development. Thereinto, the intensive interactions between PMSCs and other component cells in the placenta were particularly highlighted and further extended to the implications in the pathogenesis and therapeutic strategies of PE.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanqi Zhong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Walentowicz P, Sadlecki P, Walentowicz-Sadlecka M, Bajek A, Grabiec M, Drewa T. Human amniotic fluid as a source of stem cells. Open Med (Wars) 2022; 17:648-660. [PMID: 35434378 PMCID: PMC8982042 DOI: 10.1515/med-2022-0468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Human amniotic fluid collected during amniocentesis contains a heterogeneous population of differentiated and undifferentiated cells. Properties and number of these cells vary depending on the gestational age and the presence of potential fetal pathologies. The aim of this study was to analyze the effects of maternal, fetal, and environmental factors on the success rates of amniotic fluid stem cell cultures, the number of human amniotic fluid stem cells (hAFSC), their growth rates in primary cultures, and the number of cell passages. The study included 355 patients qualified for genetic amniocentesis at the Prenatal Genetic Unit, Department of Obstetrics, Gynecology and Oncologic Gynecology, Nicolaus Copernicus University Medical College in Bydgoszcz in 2011–2017. The mean age of the study participants was 34 ± 6.2 years, and mean gravidity amounted to 2.48 ± 1.4. Amniotic fluid sample volume turned out to be a highly significant (p < 0.01) predictor of culture success, and the relationship was particularly evident in women older than 40 years. Another highly significant predictor of culture success was the presence of two cell populations in the sample (p < 0.01). The likelihood of culture success correlated significantly (p < 0.05) with the season of the year at the time of amniocentesis. The number of cell passages differed significantly depending on the maternal age (p < 0.01). The number of passages also showed a highly significant relationship with the season of the year the sample was obtained (p < 0.01). Younger maternal age was identified as a determinant of high passage number (≥3), and another highly significant determinant of high passage number was the presence of two cell populations in the amniotic fluid sample (p < 0.01). Percentage of successfully established hAFSC cultures and the number of passages depended on amniotic fluid volume, the presence of two cell populations within the sample, and the season of the year. Individual characteristics of the donors, such as age and gravidity, did not exert a significant effect on the number of isolated hAFSCs and the rate of their growth. Patients’ place of residence, fetal karyotype, transportation time, and purity of the samples did not affect the success rates for primary cultures and the number of passages.
Collapse
Affiliation(s)
- Pawel Walentowicz
- Department of Obstetrics and Gynecology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University , Bydgoszcz 85-168 , Poland
- Department of Obstetrics, Gynecology and Oncological Gynecology, Regional Polyclinical Hospital , 87-100 Torun , Poland
| | - Pawel Sadlecki
- Department of Obstetrics and Gynecology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University , Bydgoszcz 85-168 , Poland
- Department of Obstetrics, Gynecology and Oncological Gynecology, Regional Polyclinical Hospital , 87-100 Torun , Poland
| | - Malgorzata Walentowicz-Sadlecka
- Department of Obstetrics and Gynecology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University , Bydgoszcz 85-168 , Poland
- 2nd Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education , 01-809 Warsaw , Poland
| | - Anna Bajek
- Department of Tissue Engineering, Nicolaus Copernicus University , Bydgoszcz 85-092 , Poland
| | - Marek Grabiec
- Department of Obstetrics and Gynecology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University , Bydgoszcz 85-168 , Poland
| | - Tomasz Drewa
- Department of Regenerative Medicine, Cell and Tissue Bank, Chair of Urology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Medical College in Bydgoszcz , 85-094 , Poland
| |
Collapse
|
14
|
Li C, Zhao H, Cheng L, Wang B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci 2021; 11:187. [PMID: 34727974 PMCID: PMC8561357 DOI: 10.1186/s13578-021-00698-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapeutics is already available for treatment of a range of diseases or medical conditions. Autologous or allogeneic MSCs obtained from self or donors have their own advantages and disadvantages in their medical practice. Therapeutic benefits of using autologous vs. allogeneic MSCs are inconclusive. Transplanted MSCs within the body interact with their physical microenvironment or niche, physiologically or pathologically, and such cells in a newly established tissue microenvironment may be impacted by the pathological harmful environmental factors to alter their unique biological behaviors. Meanwhile, a temporary microenvironment/niche may be also altered by the resident or niche-surrounding MSCs. Therefore, the functional plasticity and heterogeneity of MSCs caused by different donors and subpopulations of MSCs may result in potential uncertainty in their safe and efficacious medical practice. Acknowledging a connection between MSCs' biology and their existing microenvironment, donor-controlled clinical practice for the long-term therapeutic benefit is suggested to further consider minimizing MSCs potential harm for MSC-based individual therapies. In this review, we summarize the advantages and disadvantages of autologous vs. allogeneic MSCs in their therapeutic applications. Among other issues, we highlight the importance of better understanding of the various microenvironments that may affect the properties of niche-surrounding MSCs and discuss the clinical applications of MSCs within different contexts for treatment of different diseases including cardiomyopathy, lupus and lupus nephritis, diabetes and diabetic complications, bone and cartilage repair, cancer and tissue fibrosis.
Collapse
Affiliation(s)
- Chenghai Li
- Stem Cell Program of Clinical Research Center, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| | - Hua Zhao
- Institute of Reproductive Medicine, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Linna Cheng
- Institute of Hematology, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Bin Wang
- Department of Neurosurgery, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| |
Collapse
|
15
|
Majumder D. An Analysis of Structure-function Co-relation between GLI Oncoprotein and HLA Immune-gene Transcriptional Regulation through Molecular Docking. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394717666210805115050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
GLI proteins play a significant role in the transduction of the Hedgehog
(Hh) signaling pathway. A variety of human cancers, including the brain, gastrointestinal, lung,
breast, and prostate cancers, demonstrate inappropriate activation of this pathway. GLI helps in proliferation
and has an inhibitory role in the differentiation of hematopoietic stem cells. Malignancies
may have a defect in differentiation. Different types of malignancies and undifferentiated cells
have a low level of HLA expression on their cell surface.
Objective:
Human Leukocytic Antigen (HLA) downregulation is frequently observed in cancer
cells. This work is aimed to hypothesize whether this downregulation of HLA molecules is GLI oncoprotein
mediated or not. To understand the roles of different types of GLI oncoproteins on different
classes of HLA transcriptional machinery was carried out through structure-based modeling
and molecular docking studies.
Methods:
To investigate the role of GLI in HLA expression /downregulation is Hh-GLI mediated
or not, molecular docking based computational interaction studies were performed between different
GLI proteins (GLI1, GLI2, and GLI3) with TATA box binding protein (TBP) and compare the
binding efficiencies of different HLA gene (both HLA class I and –II) regulating transcription factors
(RelA, RFX5, RFXAP, RFXANK, CIITA, CREB1, and their combinations) with TBP. Due to
unavailability of 3D protein structures of GLI2 and cyclin D2 (a natural ligand of GLI1) were modelled
followed by structural validation by Ramachandran plot analysis.
Results:
GLI proteins especially, GLI1 and GLI2, have almost similar binding energy of RFX5-RFXANK-
RFXAP and CIITA multi-protein complex to TBP but has lower binding energy between
RelA to TBP.
Conclusion:
This study suggests that HLA class I may not be downregulated by GLI; however,
over-expression of GLI1 is may be responsible for HLA class II downregulation. Thus this protein
may be responsible for the maintenance of the undifferentiated state of malignant cells. This study
also suggests the implicative role of GLI1 in the early definitive stage of hematopoiesis.
Collapse
Affiliation(s)
- Durjoy Majumder
- Department of Physiology, West Bengal State University, Berunanpukuria, Malikapur, Barasat, 700 126 Kolkata,India
| |
Collapse
|
16
|
Mukherjee S, Yadav G, Kumar R. Recent trends in stem cell-based therapies and applications of artificial intelligence in regenerative medicine. World J Stem Cells 2021; 13:521-541. [PMID: 34249226 PMCID: PMC8246250 DOI: 10.4252/wjsc.v13.i6.521] [Citation(s) in RCA: 12] [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: 02/16/2021] [Revised: 03/22/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cells are undifferentiated cells that can self-renew and differentiate into diverse types of mature and functional cells while maintaining their original identity. This profound potential of stem cells has been thoroughly investigated for its significance in regenerative medicine and has laid the foundation for cell-based therapies. Regenerative medicine is rapidly progressing in healthcare with the prospect of repair and restoration of specific organs or tissue injuries or chronic disease conditions where the body’s regenerative process is not sufficient to heal. In this review, the recent advances in stem cell-based therapies in regenerative medicine are discussed, emphasizing mesenchymal stem cell-based therapies as these cells have been extensively studied for clinical use. Recent applications of artificial intelligence algorithms in stem cell-based therapies, their limitation, and future prospects are highlighted.
Collapse
Affiliation(s)
- Sayali Mukherjee
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow 226028, Uttar Pradesh, India
| | - Garima Yadav
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow 226028, Uttar Pradesh, India
| | - Rajnish Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow 226028, Uttar Pradesh, India
| |
Collapse
|
17
|
Stem Cells: A Historical Review about Biological, Religious, and Ethical Issues. Stem Cells Int 2021; 2021:9978837. [PMID: 34012469 PMCID: PMC8105090 DOI: 10.1155/2021/9978837] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/08/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023] Open
Abstract
Stem cells can be used to replace damaged cells or regenerate organs and have broadened our knowledge of the development and progression of certain diseases. Despite significant advances in understanding stem cell biology, several problems limit their use. These problems are related not only to the growth of tumors in animal models and their rejection in transplant cases but also to ethical and social issues about the use of embryonic cells. The ethical-scientific debate on this type of cells has taken on great interest both for their application in regenerative medicine and for the potential possibilities in the field of cell and gene therapy. Different points of view often have the expression of a perception that depends on scientific goals or opportunities or on religious traditions and beliefs. Therefore, as the questions and doubts about when life begins, so do the answers for the use of these cells as therapy or otherwise. So, in addition to the origin of stem cells, there are currently some social bioethical (such as political and legislative issues) and religious dilemmas. The purpose of the study is aimed at being a narrative on the history of stem cells and the evolution of their use to date, as well as to clarify the bioethical position of the various religions today in comparison with the social ones regarding the research and use of embryonic and adult ones. Hence, their biological hypostasis regarding the concepts of “conception” and “fertilization” and their development and therapeutic use compared to those of the main theological doctrines.
Collapse
|
18
|
Hashemi SS, Mohammadi AA, Moshirabadi K, Zardosht M. Effect of dermal fibroblasts and mesenchymal stem cells seeded on an amniotic membrane scaffold in skin regeneration: A case series. J Cosmet Dermatol 2021; 20:4040-4047. [PMID: 33656768 DOI: 10.1111/jocd.14043] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/25/2021] [Accepted: 02/15/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Skin wound healing has always been a challenging subject as it involves the coordinated functioning of various cells and molecules. Any disorder in wound healing can cause healing failure and result in chronic wounds. In this study, we hypothesized that co-cultured dermal fibroblasts (DFs) and Wharton's jelly mesenchymal stem cells (WJ-MSCs) seeded on an acellular amniotic membrane scaffold could be used to promote skin regeneration in chronic ulcers. MATERIALS AND METHODS In this case series, the chronic wounds of five diabetic patients aged between 30 and 60 years were treated with co-cultured WJ-MSCs and DFs seeded on an acellular amniotic membrane. Treatment was applied and the wound healing process was evaluated every three days for nine days, with the patients being subsequently followed up for one month. The wound healing percentage, time taken for the wound to heal, and wound size were monitored. RESULTS The mean wound healing rate (WHR) increased progressively in all lesions. The mean percentage of wound healing after transplantation of the biological scaffold enriched with WJ-MSCs and autologous DFs after treatment was 93.92%, respectively. The healing percentage significantly increased after three days; significant decreases in wound size and healing time were recorded after six and nine days of treatment, respectively (p < 0.002); and total skin regeneration and re-epithelialization were achieved by the ninth day of treatment. There were no side effects or complications. CONCLUSION Given the current problems and complications presented by chronic wounds, Novel Clinical approaches involving cell therapy and tissue engineering can be regarded as an attractive therapeutic option for the treatment of chronic and difficult-to-heal wounds.
Collapse
Affiliation(s)
- Seyedeh-Sara Hashemi
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Akbar Mohammadi
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Division of Plastic and Reconstructive Surgery, Department of Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kasra Moshirabadi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mitra Zardosht
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
19
|
Chieregato K, Bernardi M, Alghisi A, Giordano R, Lazzari L, Perbellini O, Rassu M, Ruggeri M, Astori G. A flow cytometric assay for the quantification of MSC lysis by peripheral blood mononucleated cells. Heliyon 2021; 7:e06036. [PMID: 33553772 PMCID: PMC7856420 DOI: 10.1016/j.heliyon.2021.e06036] [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: 10/23/2020] [Revised: 12/31/2020] [Accepted: 01/14/2021] [Indexed: 11/03/2022] Open
Abstract
Mesenchymal stromal cells (MSC) are attractive candidates for the treatment of acute graft versus host disease (aGvHD) or autoimmune disorders. However, mechanisms of MSC recognition remain unclear and there are evidences that MSC are not totally immunoprivileged. Data suggest that MSC undergo apoptosis after infusion in presence of cytotoxic cells and their death could drive immunosuppression. In GvHD patients, that activity was associated with clinical response. It is mandatory to develop an in vitro potency testing predictor of the "in vivo" response to the therapy. We describe a flow cytometric assay based on differential immunostaining of target and effector cells where BM MSC are enumerated with fluorospheres to determine the loss of target cells after co-culture with PB MNC. 6/13 (46%) of BM MSC lots were lysed by PB MNC and the lysis was proportional to the E/T cell ratio. The method overcomes the problems linked to the use of dyes or radioactive, evidencing the limitations linked to the use of a single vital dye and proposing a precise gating strategy based on absolute cell counts where cells are left untouched. The assay is easy and could be used to predict the response of the patients to the therapy.
Collapse
Affiliation(s)
- Katia Chieregato
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy.,CORIS Veneto - Consorzio per la Ricerca Sanitaria, Padova, Italy
| | - Martina Bernardi
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy.,CORIS Veneto - Consorzio per la Ricerca Sanitaria, Padova, Italy
| | - Alberta Alghisi
- Immunohematology and Transfusion Medicine Service, Vicenza Hospital, Italy
| | - Rosaria Giordano
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine - Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Omar Perbellini
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy
| | - Mario Rassu
- Department of Microbiology, Vicenza Hospital, Italy
| | - Marco Ruggeri
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Haematology Unit, Vicenza Hospital, Italy
| |
Collapse
|
20
|
Yadav P, Vats R, Bano A, Bhardwaj R. Hematopoietic Stem Cells Culture, Expansion and Differentiation: An Insight into Variable and Available Media. Int J Stem Cells 2020; 13:326-334. [PMID: 32840223 PMCID: PMC7691860 DOI: 10.15283/ijsc19157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
Owing to differentiation and self-renewal capacity, hematopoietic stem cells clasp potentiality to engender all blood cell types, leading to their immense competence to play a diverse role in therapeutic applications. Although these stem cells are the most investigated and exploited until now, further research is still essential to comprehend their nature, fate, and potential. Enhanced usage of hematopoietic stem cells in research and therapeutics intensified the requirement of expansion and differentiation of hematopoietic stem cells under in vitro conditions. Since these cells remain in senescence for a prolonged period before isolation, selection of appropriate growth medium along with supplements and culture conditions are crucial to initiate their cell division and to designate their destiny. The precise equilibrium between self-renewal and differentiation of stem cells sustained by exclusive medium along with special growth or differentiation factors is accountable for generating diverse cell lineages. Maintenance of hematopoietic stem and progenitor cell lines along with the advancement of research work generate an inexorable demand for production and commercialization of specialized stem cell culture media, with or without serum along with specific growth factors and supplements. Media commercialization for precise stem cell types, culturing and differentiation is a cost-effective developing field. Here in this review, we are assembling various types of hematopoietic stem cell self-renewal, expansion and differentiation media along with supplements and culture conditions, either developed and used by various scientists or are available commercially.
Collapse
Affiliation(s)
- Pooja Yadav
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, India
| | - Ravina Vats
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, India
| | - Afsareen Bano
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, India
| | - Rashmi Bhardwaj
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
21
|
Mesenchymal stem/stromal cells: Developmental origin, tumorigenesis and translational cancer therapeutics. Transl Oncol 2020; 14:100948. [PMID: 33190044 PMCID: PMC7672320 DOI: 10.1016/j.tranon.2020.100948] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/27/2020] [Accepted: 11/05/2020] [Indexed: 12/20/2022] Open
Abstract
While a large and growing body of research has demonstrated that mesenchymal stem/stromal cells (MSCs) play a dual role in tumor growth and inhibition, studies exploring the capability of MSCs to contribute to tumorigenesis are rare. MSCs are key players during tumorigenesis and cancer development, evident in their faculty to increase cancer stem cells (CSCs) population, to generate the precursors of certain forms of cancer (e.g. sarcoma), and to induce epithelial-mesenchymal transition to create the CSC-like state. Indeed, the origin and localization of the native MSCs in their original tissues are not known. MSCs are identified in the primary tumor sites and the fetal and extraembryonic tissues. Acknowledging the developmental origin of MSCs and tissue-resident native MSCs is essential for better understanding of MSC contributions to the cellular origin of cancer. This review stresses that the plasticity of MSCs can therefore instigate further risk in select therapeutic strategies for some patients with certain forms of cancer. Towards this end, to explore the safe and effective MSC-based anti-cancer therapies requires a strong understanding of the cellular and molecular mechanisms of MSC action, ultimately guiding new strategies for delivering treatment. While clinical trial efforts using MSC products are currently underway, this review also provides new insights on the underlying mechanisms of MSCs to tumorigenesis and focuses on the approaches to develop MSC-based anti-cancer therapeutic applications.
Collapse
|
22
|
Gao Y, Jin SZ. Strategies for treating oesophageal diseases with stem cells. World J Stem Cells 2020; 12:488-499. [PMID: 32742566 PMCID: PMC7360987 DOI: 10.4252/wjsc.v12.i6.488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/02/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
There is a wide range of oesophageal diseases, the most general of which are inflammation, injury and tumours, and treatment methods are constantly being developed and updated. With an increasingly comprehensive understanding of stem cells and their characteristics of multilineage differentiation, self-renewal and homing as well as the combination of stem cells with regenerative medicine, tissue engineering and gene therapy, stem cells are playing an important role in the treatment of a variety of diseases. Mesenchymal stem cells have many advantages and are most commonly applied; however, most of these applications have been in experimental studies, with few related clinical trials for comparison. Therefore, the methods, positive significance and limitations of stem cells in the treatment of oesophageal diseases remain incompletely understood. Thus, the purpose of this paper is to review the current literature and summarize the efficacy of stem cells in the treatment of oesophageal diseases, including oesophageal ulceration, acute radiation-induced oesophageal injury, corrosive oesophageal injury, oesophageal stricture formation after endoscopic submucosal dissection and oesophageal reconstruction, as well as gene therapy for oesophageal cancer.
Collapse
Affiliation(s)
- Yang Gao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| | - Shi-Zhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang Province, China
| |
Collapse
|
23
|
Mathew SA, Naik C, Cahill PA, Bhonde RR. Placental mesenchymal stromal cells as an alternative tool for therapeutic angiogenesis. Cell Mol Life Sci 2020; 77:253-265. [PMID: 31468060 PMCID: PMC11104823 DOI: 10.1007/s00018-019-03268-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/24/2019] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
Dysregulation of angiogenesis is a phenomenon observed in several disorders such as diabetic foot, critical limb ischemia and myocardial infarction. Mesenchymal stromal cells (MSCs) possess angiogenic potential and have recently emerged as a powerful tool for cell therapy to promote angiogenesis. Although bone marrow-derived MSCs are the primary cell of choice, obtaining them has become a challenge. The placenta has become a popular alternative as it is a highly vascular organ, easily available and ethically more favorable with a rich supply of MSCs. Comparatively, placenta-derived MSCs (PMSCs) are clinically promising due to their proliferative, migratory, clonogenic and immunomodulatory properties. PMSCs release a plethora of cytokines and chemokines key to angiogenic signaling and facilitate the possibility of delivering PMSC-derived exosomes as a targeted therapy to promote angiogenesis. However, there still remains the challenge of heterogeneity in the isolated populations, questions on the maternal or fetal origin of these cells and the diversity in previously reported isolation and culture conditions. Nonetheless, the growing rate of clinical trials using PMSCs clearly indicates a shift in favor of PMSCs. The overall aim of the review is to highlight the importance of this rather poorly understood cell type and emphasize the need for further investigations into their angiogenic potential as an alternative source for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Suja Ann Mathew
- School of Regenerative Medicine, Manipal Academy of Higher Education, MAHE, Allalasandra, Near Royal Orchid, Yellahanka, Bangalore, 560 065, India.
| | - Charuta Naik
- School of Regenerative Medicine, Manipal Academy of Higher Education, MAHE, Allalasandra, Near Royal Orchid, Yellahanka, Bangalore, 560 065, India
| | - Paul A Cahill
- School of Biotechnology, Faculty of Science and Health, Dublin City University, Glasnevin Dublin 9, Ireland
| | - Ramesh R Bhonde
- Dr. D.Y. Patil Vidyapeeth (DPU), Pimpri, Pune, 411018, India.
| |
Collapse
|
24
|
Marino L, Castaldi MA, Rosamilio R, Ragni E, Vitolo R, Fulgione C, Castaldi SG, Serio B, Bianco R, Guida M, Selleri C. Mesenchymal Stem Cells from the Wharton's Jelly of the Human Umbilical Cord: Biological Properties and Therapeutic Potential. Int J Stem Cells 2019; 12:218-226. [PMID: 31022994 PMCID: PMC6657936 DOI: 10.15283/ijsc18034] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 09/23/2018] [Accepted: 11/30/2018] [Indexed: 12/15/2022] Open
Abstract
Wharton's jelly mesenchymal stem cells (WJ-MSCs) are a class of stem cells with high differentiative potential, an immuno-privileged status and easy access for collection, which raise no legal or ethical issues. WJ-MSCs exhibit several features of embryonic stem cells, both in the phenotypic and genetic aspects, with only a few differences, such as a shorter doubling time and a more extensive ex vivo expansion capacity. WJ-MSCs have immunomodulatory properties, involving both innate and adaptive immune responses. This review focuses on the role of WJ-MSCs in the management of graft-versus-host disease (GvHD), a life-threatening complication of the allogenic transplantation of hematopoietic stem cells. Different studies documented the beneficial effect of the infusion of WJ-MSCs, even when not fully HLA identical, in patients with severe GvHD, refractory to standard treatment. Finally, we summarized current ongoing clinical trials with WJ-MSCs and their potential in regenerative medicine.
Collapse
Affiliation(s)
- Luigi Marino
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Maria Antonietta Castaldi
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy.,Department of Obstetrics and Gynecology, Azienda Ospedaliera di Rilievo Nazionale e di Alta Specialità "San Giuseppe Moscati", Avellino, Italy
| | - Rosa Rosamilio
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Enrico Ragni
- Orthopedic Biotechnology Laboratory, Galeazzi Orthopedic Institute, Milan, Italy
| | - Rosa Vitolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Caterina Fulgione
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | | | - Bianca Serio
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Rosario Bianco
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Maurizio Guida
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| |
Collapse
|
25
|
Bone Tissue Engineering Using Human Cells: A Comprehensive Review on Recent Trends, Current Prospects, and Recommendations. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9010174] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of proper cells for bone tissue engineering remains a major challenge worldwide. Cells play a pivotal role in the repair and regeneration of the bone tissue in vitro and in vivo. Currently, a large number of differentiated (somatic) and undifferentiated (stem) cells have been used for bone reconstruction alone or in combination with different biomaterials and constructs (e.g., scaffolds). Although the results of the cell transplantation without any supporting or adjuvant material have been very effective with regard to bone healing. Recent advances in bone scaffolding are now becoming new players affecting the osteogenic potential of cells. In the present study, we have critically reviewed all the currently used cell sources for bone reconstruction and discussed the new horizons that are opening up in the context of cell-based bone tissue engineering strategies.
Collapse
|
26
|
Arjmand B, Goodarzi P, Aghayan HR, Payab M, Rahim F, Alavi-Moghadam S, Mohamadi-jahani F, Larijani B. Co-transplantation of Human Fetal Mesenchymal and Hematopoietic Stem Cells in Type 1 Diabetic Mice Model. Front Endocrinol (Lausanne) 2019; 10:761. [PMID: 31781036 PMCID: PMC6856665 DOI: 10.3389/fendo.2019.00761] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Introduction: Cell therapy can overcome the limitation of conventional treatments (including different medications and β cell replacement) for type 1 diabetes. Based- on several studies human fetal mesenchymal and hematopoietic stem cells are ideal candidates for stem cell therapy. On the other hand, co-transplantation of them can improve their effects. Accordingly, the aim of this research is co-transplantation of human fetal mesenchymal and hematopoietic stem cells in type 1 diabetes. Materials and Methods: The liver of legally aborted fetus was harvested. Then, mononuclear cells were isolated and extracted mesenchymal stromal cells and CD34+ hematopoietic stem cells were cultured. Expression of pluripotency markers were evaluated. For molecular imaging, mesenchymal stromal cells were labeled using GFP- vector. BALB/c inbred male mice were modeled by injection a single dose of Streptozotocin. Diabetic animals were received stem cells. After stem cell transplantation, in vivo imaging was performed and blood glucose levels were measured weekly. Results: Fetal mesenchymal stromal cells were demonstrated differentiation potential. Expression of pluripotency markers were positive. The mean of blood glucose levels were reduced in mixed mesenchymal and hematopoietic stem cells transplantation. A lot of GFP-labeled mesenchymal stem cells were engrafted in the pancreas of animal models that received a mixed suspension of hematopoietic and mesenchymal stromal cells. Conclusions: Human fetal stem cells are valuable source for cell therapy and co-transplantation of mesenchymal stromal cells can improve therapeutic effects of hematopoietic stem cells.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia and Hemoglobinopathies Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sepideh Alavi-Moghadam
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Mohamadi-jahani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Bagher Larijani ;
| |
Collapse
|
27
|
Silva CGD, Martins CF, Bessler HC, da Fonseca Neto ÁM, Cardoso TC, Franco MM, Mendonça ADS, Leme LDO, Borges JRJ, Malaquias JV, Báo SN. Use of trichostatin A alters the expression of HDAC3 and KAT2 and improves in vitro development of bovine embryos cloned using less methylated mesenchymal stem cells. Reprod Domest Anim 2018; 54:289-299. [PMID: 30317681 DOI: 10.1111/rda.13360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022]
Abstract
The aim of this work was to investigate the methylation and hydroxymethylation status of mesenchymal stem cells (MSC) from amniotic fluid (MSC-AF), adipose tissue (MSC-AT) and fibroblasts (FIB-control) and to verify the effect of trichostatin A (TSA) on gene expression and development of cloned bovine embryos produced using these cells. Characterization of MSC from two animals (BOV1 and BOV2) was performed by flow cytometry, immunophenotyping and analysis of cellular differentiation genes expression. The cells were used in the nuclear transfer in the absence or presence of 50 nM TSA for 20 hr in embryo culture. Expression of HDAC1, HDAC3 and KAT2A genes was measured in embryos by qRT-PCR. Methylation results showed difference between animals, with MSC from BOV2 demonstrating lower methylation rate than BOV1. Meanwhile, MSC-AF were less hydroxymethylated for both animals. MSC-AF from BOV2 produced 44.92 ± 8.88% of blastocysts when embryos were exposed to TSA and similar to embryo rate of MSC-AT also treated with TSA (37.96 ± 15.80%). However, when methylation was lower in FIB compared to MSC, as found in BOV1, the use of TSA was not sufficient to increase embryo production. MSC-AF embryos expressed less HDAC3 when treated with TSA, and expression of KAT2A was higher in embryos produced with all MSC and treated with TSA than embryos produced with FIB. The use of MSC less methylated and more hydroxymethylated in combination with embryo incubation with TSA can induce lower expression of HDAC3 and higher expression of KAT2A in the embryos and consequently improve bovine embryo production.
Collapse
Affiliation(s)
- Carolina Gonzales da Silva
- University of Brasília, Brasília, Distrito Federal, Brazil.,Embrapa Cerrados, Brasília, Distrito Federal, Brazil
| | | | | | | | - Tereza Cristina Cardoso
- Laboratory of Animal Virology and Cell Culture, UNESP - University of São Paulo State, Araçatuba, São Paulo, Brazil
| | | | | | | | | | | | - Sônia Nair Báo
- University of Brasília, Brasília, Distrito Federal, Brazil
| |
Collapse
|
28
|
Dynamics of Telomere Length and Telomerase Activity in the Human Fetal Liver at 5-12 Weeks of Gestation. Stem Cells Int 2018; 2018:1385903. [PMID: 30057621 PMCID: PMC6051073 DOI: 10.1155/2018/1385903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
Fetal stem cell- (FSC-) based therapy is a promising treatment option for many diseases. The differentiation potential of FSCs is greater than that in adult stem cells, and they are more tissue-specific and have lower immunogenicity and better intrinsic homing than embryonic ones. Embryonic stem cells have higher proliferative potential than FSCs but can cause teratomas. Therefore, an evaluation of this potential represents an important biomedical challenge. Since regulation of telomere length (TL) is one mechanism governing cellular proliferation, TL is a useful surrogate marker for cell replicative potential. The prenatal dynamics of TL, however, has never been comprehensively studied. In the present study, dynamics of TL and telomerase activity in the human fetal liver during 5–12 weeks of gestation is examined. Both TL and telomerase activity were positively correlated with week of gestation. For both parameters studied, the trend to increase was evident up to 10th week of gestation. After that, they reached a plateau and remained stable. These findings indicate that telomerase activity remains high during the fetal stage, suggesting high replicative capacity of FSCs and their considerable potential for transplantation therapies. These findings, however, are preliminary only due to small sample size and require further evaluation.
Collapse
|
29
|
Kim HW, Lee HS, Kang JM, Bae SH, Kim C, Lee SH, Schwarz J, Kim GJ, Kim JS, Cha DH, Kim J, Chang SW, Lee TH, Moon J. Dual Effects of Human Placenta-Derived Neural Cells on Neuroprotection and the Inhibition of Neuroinflammation in a Rodent Model of Parkinson's Disease. Cell Transplant 2018; 27:814-830. [PMID: 29871515 PMCID: PMC6047269 DOI: 10.1177/0963689718766324] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common age-related neurodegenerative disease
in the elderly and the patients suffer from uncontrolled movement disorders due to loss of
dopaminergic (DA) neurons on substantia nigra pars compacta (SNpc). We previously reported
that transplantation of human fetal midbrain-derived neural precursor cells restored the
functional deficits of a 6-hydroxy dopamine (6-OHDA)-treated rodent model of PD but its
low viability and ethical issues still remain to be solved. Albeit immune privilege and
neural differentiation potentials suggest mesenchymal stem cells (MSCs) from various
tissues including human placenta MSCs (hpMSCs) for an alternative source, our
understanding of their therapeutic mechanisms is still limited. To expand our knowledge on
the MSC-mediated PD treatment, we here investigated the therapeutic mechanism of hpMSCs
and hpMSC-derived neural phenotype cells (hpNPCs) using a PD rat model. Whereas both
hpMSCs and hpNPCs protected DA neurons in the SNpc at comparable levels, the hpNPC
transplantation into 6-OHDA treated rats exhibited longer lasting recovery in motor
deficits than either the saline or the hpMSC treated rats. The injected hpNPCs induced
delta-like ligand (DLL)1 and neurotrophic factors, and influenced environments prone to
neuroprotection. Compared with hpMSCs, co-cultured hpNPCs more efficiently protected
primary neural precursor cells from midbrain against 6-OHDA as well as induced their
differentiation into DA neurons. Further experiments with conditioned media from hpNPCs
revealed that the secreted factors from hpNPCs modulated immune responses and neural
protection. Taken together, both DLL1-mediated contact signals and paracrine factors play
critical roles in hpNPC-mediated improvement. First showing here that hpMSCs and their
neural derivative hpNPCs were able to restore the PD-associated deficits via dual
mechanisms, neuroprotection and immunosuppression, this study expanded our knowledge of
therapeutic mechanisms in PD and other age-related diseases.
Collapse
Affiliation(s)
- Han Wool Kim
- 1 General Medical Research Institute, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Hyun-Seob Lee
- 1 General Medical Research Institute, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Jun Mo Kang
- 1 General Medical Research Institute, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Sang-Hun Bae
- 1 General Medical Research Institute, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Korea.,2 Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Chul Kim
- 1 General Medical Research Institute, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Sang-Hun Lee
- 3 Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea
| | - Johannes Schwarz
- 4 German Center for Neurodegenerative Diseases (DZNE), Technical University Munich, Munich, Germany
| | - Gi Jin Kim
- 5 Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Jin-Su Kim
- 6 Molecular Imaging Research Center, Korea Institute Radiological and Medical Sciences, Seoul, Korea
| | - Dong Hyun Cha
- 7 Deparment of Ob and Gyn, CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Joopyung Kim
- 8 Department of Neurosurgery, Bundang CHA Hospital, CHA University School of Medicine, Seongnam-si, Korea
| | - Sung Woon Chang
- 9 Department of Ob and Gyn, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Tae Hee Lee
- 10 Formulae Pharmacology Department, School of Oriental Medicine, Gachon University, Gyeonggi, Korea
| | - Jisook Moon
- 1 General Medical Research Institute, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, Korea.,2 Department of Biotechnology, CHA University, Seongnam-si, Gyeonggi-do, Korea
| |
Collapse
|
30
|
Ventura Ferreira MS, Bienert M, Müller K, Rath B, Goecke T, Opländer C, Braunschweig T, Mela P, Brümmendorf TH, Beier F, Neuss S. Comprehensive characterization of chorionic villi-derived mesenchymal stromal cells from human placenta. Stem Cell Res Ther 2018; 9:28. [PMID: 29402304 PMCID: PMC5800083 DOI: 10.1186/s13287-017-0757-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/29/2017] [Accepted: 12/19/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Studies in which mesenchymal stromal cells (MSC) from the placenta are compared with multiple MSC types from other sources are rare. The chorionic plate of the human placenta is mainly composed of fetal blood vessels embedded in fetal stroma tissue, lined by trophoblastic cells and organized into chorionic villi (CV) structures. METHODS We comprehensively characterized human MSC collected from postnatal human chorionic villi of placenta (CV-MSC) by analyzing their growth and proliferation potential, differentiation, immunophenotype, extracellular matrix production, telomere length, aging phenotype, and plasticity. RESULTS Immunophenotypic characterization of CV-MSC confirmed the typical MSC marker expression as defined by the International Society for Cellular Therapy. The surface marker profile was consistent with increased potential for proliferation, vascular localization, and early myogenic marker expression. CV-MSC retained multilineage differentiation potential and extracellular matrix remodeling properties. They have undergone reduced telomere loss and delayed onset of cellular senescence as they aged in vitro compared to three other MSC sources. We present evidence that increased human telomerase reverse transcriptase gene expression could not explain the exceptional telomere maintenance and senescence onset delay in cultured CV-MSC. Our in-vitro tumorigenesis detection assay suggests that CV-MSC are not prone to undergo malignant transformation during long-term in-vitro culture. Besides SOX2 expression, no other pluripotency features were observed in early and late passages of CV-MSC. CONCLUSIONS Our work brings forward two remarkable characteristics of CV-MSC, the first being their extended life span as a result of delayed replicative senescence and the second being a delayed aged phenotype characterized by improved telomere length maintenance. MSC from human placenta are very attractive candidates for stem cell-based therapy applications.
Collapse
Affiliation(s)
- Mónica S. Ventura Ferreira
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
- 0000 0001 0728 696Xgrid.1957.aDepartment of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| | - Michaela Bienert
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
- 0000 0001 0728 696Xgrid.1957.aHelmholtz Institute for Biomedical Engineering, Biointerface Group, RWTH Aachen University, Aachen, Germany
| | - Katrin Müller
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Björn Rath
- 0000 0001 0728 696Xgrid.1957.aDepartment of Orthopedic Surgery, RWTH Aachen University, Aachen, Germany
| | - Tamme Goecke
- 0000 0001 0728 696Xgrid.1957.aDepartment for Gynecology, RWTH Aachen University, Aachen, Germany
| | - Christian Opländer
- 0000 0000 9024 6397grid.412581.bDepartment of Translational Wound Research, Centre for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Till Braunschweig
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Petra Mela
- 0000 0001 0728 696Xgrid.1957.aDepartment of Tissue Engineering and Textile Implants, Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Tim H. Brümmendorf
- 0000 0001 0728 696Xgrid.1957.aDepartment of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| | - Fabian Beier
- 0000 0001 0728 696Xgrid.1957.aDepartment of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| | - Sabine Neuss
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
- 0000 0001 0728 696Xgrid.1957.aHelmholtz Institute for Biomedical Engineering, Biointerface Group, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
31
|
Amati E, Perbellini O, Rotta G, Bernardi M, Chieregato K, Sella S, Rodeghiero F, Ruggeri M, Astori G. High-throughput immunophenotypic characterization of bone marrow- and cord blood-derived mesenchymal stromal cells reveals common and differentially expressed markers: identification of angiotensin-converting enzyme (CD143) as a marker differentially expressed between adult and perinatal tissue sources. Stem Cell Res Ther 2018; 9:10. [PMID: 29338788 PMCID: PMC5771027 DOI: 10.1186/s13287-017-0755-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSC) are a heterogeneous population of multipotent progenitors used in the clinic because of their immunomodulatory properties and their ability to differentiate into multiple mesodermal lineages. Although bone marrow (BM) remains the most common MSC source, cord blood (CB) can be collected noninvasively and without major ethical concerns. Comparative studies comprehensively characterizing the MSC phenotype across several tissue sources are still lacking. This study provides a 246-antigen immunophenotypic analysis of BM- and CB-derived MSC aimed at identifying common and strongly expressed MSC markers as well as the existence of discriminating markers between the two sources. METHODS BM-MSC (n = 4) were expanded and analyzed as bulk (n = 6) or single clones isolated from the bulk culture (n = 3). CB-MSC (n = 6) were isolated and expanded as single clones in 5/6 samples. The BM-MSC and CB-MSC phenotype was investigated by flow cytometry using a panel of 246 monoclonal antibodies. To define the markers common to both sources, those showing the smallest variation between samples (coefficient of variation of log2 fold increase ≤ 0.5, n = 59) were selected for unsupervised hierarchical cluster analysis (HCL). Differentially expressed markers were identified by directly comparing the expression of all 246 antigens between BM-MSC and CB-MSC. RESULTS Based on HCL, 18 markers clustered as strongly expressed in BM-MSC and CB-MSC, including alpha-smooth muscle antigen (SMA), beta-2-microglobulin, CD105, CD13, CD140b, CD147, CD151, CD276, CD29, CD44, CD47, CD59, CD73, CD81, CD90, CD98, HLA-ABC, and vimentin. All except CD140b and alpha-SMA were suitable for the specific identification of ex-vivo expanded MSC. Notably, only angiotensin-converting enzyme (CD143) was exclusively expressed on BM-MSC. CD143 expression was tested on 10 additional BM-MSC and CB-MSC and on 10 umbilical cord- and adipose tissue-derived MSC samples, confirming that its expression is restricted to adult sources. CONCLUSIONS This is the first study that has comprehensively compared the phenotype of BM-MSC and CB-MSC. We have identified markers that could complement the minimal panel proposed for the in-vitro MSC definition, being shared and strongly expressed by BM- and CB-derived MSC. We have also identified CD143 as a marker exclusively expressed on MSC derived from adult tissue sources. Further studies will elucidate the biological role of CD143 and its potential association with tissue-specific MSC features.
Collapse
Affiliation(s)
- Eliana Amati
- Advanced Cellular Therapy Laboratory, Hematology Unit, S. Bortolo Hospital, ULSS 8 Berica, Contra' San Francesco 41, 36100, Vicenza, Italy
| | - Omar Perbellini
- Advanced Cellular Therapy Laboratory, Hematology Unit, S. Bortolo Hospital, ULSS 8 Berica, Contra' San Francesco 41, 36100, Vicenza, Italy
| | | | - Martina Bernardi
- Advanced Cellular Therapy Laboratory, Hematology Unit, S. Bortolo Hospital, ULSS 8 Berica, Contra' San Francesco 41, 36100, Vicenza, Italy.,Hematology Project Foundation, Vicenza, Italy
| | - Katia Chieregato
- Advanced Cellular Therapy Laboratory, Hematology Unit, S. Bortolo Hospital, ULSS 8 Berica, Contra' San Francesco 41, 36100, Vicenza, Italy.,Hematology Project Foundation, Vicenza, Italy
| | - Sabrina Sella
- Advanced Cellular Therapy Laboratory, Hematology Unit, S. Bortolo Hospital, ULSS 8 Berica, Contra' San Francesco 41, 36100, Vicenza, Italy
| | | | - Marco Ruggeri
- Advanced Cellular Therapy Laboratory, Hematology Unit, S. Bortolo Hospital, ULSS 8 Berica, Contra' San Francesco 41, 36100, Vicenza, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Hematology Unit, S. Bortolo Hospital, ULSS 8 Berica, Contra' San Francesco 41, 36100, Vicenza, Italy.
| |
Collapse
|
32
|
Abbasalizadeh S, Pakzad M, Cabral JMS, Baharvand H. Allogeneic cell therapy manufacturing: process development technologies and facility design options. Expert Opin Biol Ther 2017; 17:1201-1219. [PMID: 28699788 DOI: 10.1080/14712598.2017.1354982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Currently, promising outcomes from clinical trials of allogeneic cells, especially allogeneic mesenchymal stromal cells, fibroblasts, keratinocytes, and human cardiac stem cells, have encouraged research institutions, small and medium enterprises (SMEs), and big pharmaceutical companies to invest and focus on developing allogeneic cell therapy products. Commercial and large-scale production of allogeneic cell therapy products requires unique capabilities to develop technologies that generate safe and effective allogeneic cells/cell lines and their fully characterized master/working banks. In addition, it is necessary to design robust upstream and downstream manufacturing processes, and establish integrated, well-designed manufacturing facilities to produce high quality affordable products in accordance with current GMP regulations for the production of cell therapy products. Areas covered: The authors highlight: the recent advances in the development of allogeneic products, the available options to develop robust manufacturing processes, and facility design considerations. Expert opinion: Currently, there are multiple challenges in development of allogeneic cell therapy products. Indeed, the field is still in its infancy; with technologies and regulations still under development, as is our understanding of the mechanisms of action in the body and their interaction with the host immune system. Their characterization and testing is also an emerging and very complex area.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran.,b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Mohammad Pakzad
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Joaquim M S Cabral
- b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Hossein Baharvand
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran.,c Department of Developmental Biology , University of Science and Culture , Tehran , Iran
| |
Collapse
|
33
|
Characteristics and Cardiomyogenic Potential of Rat Fetal Cardiac Progenitor Cells at Different Developmental Stage. Tissue Eng Regen Med 2017; 14:253-265. [PMID: 30603482 DOI: 10.1007/s13770-016-0016-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/19/2016] [Accepted: 04/19/2016] [Indexed: 12/19/2022] Open
Abstract
In recent years, several kinds of cardiac progenitor cells have been identified and isolated from heart tissue. These cells showed differentiation potential into cardiomyocytes, smooth muscle cells, and endothelial cells in vitro and in vivo. Morphogenetic events are tightly regulated during development to determine cell destiny and reshape the embryonic lineage. In this study, we directly compared the characteristics of rat fetal cardiac progenitor cells (rFCPCs) isolated from the chamber formation stage at embryonic day 12 (E12) and at the septation stage of E15. Both kinds of rFCPCs expressed mesenchymal stem cell markers (CD105, CD73, and CD29) but not CD34 and CD45. The E12 rFCPCs expressed a high level of Oct4 compared to E15 until passage 5 and showed a steep decline of Nkx2.5 expression at passage 5. However, Nkx2.5 expression at E15 was maintained until passage 5 and Oct4 expression slightly increased at passage 5. We also detected an intense staining for Oct4 antibody in E12 heart tissue sections. The average doubling time of the E12 rFCPCs from passage 3 to passage 15 was about 5 hours longer than E15. These cells could also be induced into cardiomyocytes expressing α-MHC, cTnT, cTnC, and Cx43 under cardiomyogenic culture conditions and rFCPCs at E15 showed more intense staining of α-MHC than cells at E12 by immunocytochemistry. Taken together, our results show that developmental differences between E12 and E15 may influence their properties and differentiation. Furthermore those differences should be considered when deciding on the optimal cell source for cell replacement therapy in cardiovascular regeneration.
Collapse
|
34
|
Sardesai VS, Shafiee A, Fisk NM, Pelekanos RA. Avoidance of Maternal Cell Contamination and Overgrowth in Isolating Fetal Chorionic Villi Mesenchymal Stem Cells from Human Term Placenta. Stem Cells Transl Med 2017; 6:1070-1084. [PMID: 28205414 PMCID: PMC5442838 DOI: 10.1002/sctm.15-0327] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 07/05/2016] [Accepted: 07/28/2016] [Indexed: 12/16/2022] Open
Abstract
Human placenta is rich in mesenchymal stem/stromal cells (MSC), with their origin widely presumed fetal. Cultured placental MSCs are confounded by a high frequency of maternal cell contamination. Our recent systematic review concluded that only a small minority of placental MSC publications report fetal/maternal origin, and failed to discern a specific methodology for isolation of fetal MSC from term villi. We determined isolation conditions to yield fetal and separately maternal MSC during ex vivo expansion from human term placenta. MSCs were isolated via a range of methods in combination; selection from various chorionic regions, different commercial media, mononuclear cell digest and/or explant culture. Fetal and maternal cell identities were quantitated in gender‐discordant pregnancies by XY chromosome fluorescence in situ hybridization. We first demonstrated reproducible maternal cell contamination in MSC cultures from all chorionic anatomical locations tested. Cultures in standard media rapidly became composed entirely of maternal cells despite isolation from fetal villi. To isolate pure fetal cells, we validated a novel isolation procedure comprising focal dissection from the cotyledonary core, collagenase/dispase digestion and explant culture in endothelial growth media that selected, and provided a proliferative environment, for fetal MSC. Comparison of MSC populations within the same placenta confirmed fetal to be smaller, more osteogenic and proliferative than maternal MSC. We conclude that in standard media, fetal chorionic villi‐derived MSC (CV‐MSC) do not grow readily, whereas maternal MSC proliferate to result in maternal overgrowth during culture. Instead, fetal CV‐MSCs require isolation under specific conditions, which has implications for clinical trials using placental MSC. Stem Cells Translational Medicine2017;6:1070–1084
Collapse
Affiliation(s)
- Varda S Sardesai
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia
| | - Abbas Shafiee
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Nicholas M Fisk
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia.,Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - Rebecca A Pelekanos
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia
| |
Collapse
|
35
|
Muiños-López E, Hermida-Gómez T, Fuentes-Boquete I, de Toro-Santos J, Blanco FJ, Díaz-Prado SM. * Human Amniotic Mesenchymal Stromal Cells as Favorable Source for Cartilage Repair. Tissue Eng Part A 2017; 23:901-912. [PMID: 28073305 DOI: 10.1089/ten.tea.2016.0422] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Localized trauma-derived breakdown of the hyaline articular cartilage may progress toward osteoarthritis, a degenerative condition characterized by total loss of articular cartilage and joint function. Tissue engineering technologies encompass several promising approaches with high therapeutic potential for the treatment of these focal defects. However, most of the research in tissue engineering is focused on potential materials and structural cues, while little attention is directed to the most appropriate source of cells endowing these materials. In this study, using human amniotic membrane (HAM) as scaffold, we defined a novel static in vitro model for cartilage repair. In combination with HAM, four different cell types, human chondrocytes, human bone marrow-derived mesenchymal stromal cells (hBMSCs), human amniotic epithelial cells, and human amniotic mesenchymal stromal cells (hAMSCs) were assessed determining their therapeutic potential. MATERIAL AND METHODS A chondral lesion was drilled in human cartilage biopsies simulating a focal defect. A pellet of different cell types was implanted inside the lesion and covered with HAM. The biopsies were maintained for 8 weeks in culture. Chondrogenic differentiation in the defect was analyzed by histology and immunohistochemistry. RESULTS HAM scaffold showed good integration and adhesion to the native cartilage in all groups. Although all cell types showed the capacity of filling the focal defect, hBMSCs and hAMSCs demonstrated higher levels of new matrix synthesis. However, only the hAMSCs-containing group presented a significant cytoplasmic content of type II collagen when compared with chondrocytes. More collagen type I was identified in the new synthesized tissue of hBMSCs. In accordance, hBMSCs and hAMSCs showed better International Cartilage Research Society scoring although without statistical significance. CONCLUSION HAM is a useful material for articular cartilage repair in vitro when used as scaffold. In combination with hAMSCs, HAM showed better potential for cartilage repair with similar reparation capacity than chondrocytes.
Collapse
Affiliation(s)
- Emma Muiños-López
- 1 Tissue Bioengineering and Cell Therapy Unit (GBTTC-CHUAC), CIBER-BBN/ISCIII, Rheumatology Group, Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC) , Galician Health Service (SERGAS), A Coruña, Spain
| | - Tamara Hermida-Gómez
- 1 Tissue Bioengineering and Cell Therapy Unit (GBTTC-CHUAC), CIBER-BBN/ISCIII, Rheumatology Group, Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC) , Galician Health Service (SERGAS), A Coruña, Spain
| | - Isaac Fuentes-Boquete
- 2 Cell Therapy and Regenerative Medicine Unit, CIBER-BBN/ISCIII, Rheumatology Group, Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), Department of Medicine, Faculty of Health Sciences, University of A Coruña , A Coruña, Spain
| | - Javier de Toro-Santos
- 3 Reumathology Service, Cell Therapy and Regenerative Medicine Unit, CIBER-BBN/ISCIII, Rheumatology Group, Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), Department of Medicine, Faculty of Health Sciences, University of A Coruña , A Coruña, Spain
| | - Francisco Javier Blanco
- 4 Reumathology Service, Tissue Bioengineering and Cell Therapy Unit (GBTTC-CHUAC), CIBER-BBN/ISCIII, Rheumatology Group, Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), Faculty of Medicine, University of Santiago de Compostela , A Coruña, Spain
| | - Silvia María Díaz-Prado
- 2 Cell Therapy and Regenerative Medicine Unit, CIBER-BBN/ISCIII, Rheumatology Group, Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), Department of Medicine, Faculty of Health Sciences, University of A Coruña , A Coruña, Spain
| |
Collapse
|
36
|
Amati E, Sella S, Perbellini O, Alghisi A, Bernardi M, Chieregato K, Lievore C, Peserico D, Rigno M, Zilio A, Ruggeri M, Rodeghiero F, Astori G. Generation of mesenchymal stromal cells from cord blood: evaluation of in vitro quality parameters prior to clinical use. Stem Cell Res Ther 2017; 8:14. [PMID: 28115021 PMCID: PMC5260040 DOI: 10.1186/s13287-016-0465-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 12/13/2016] [Accepted: 12/21/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Increasing evidence suggests the safety and efficacy of mesenchymal stromal cells (MSC) as advanced therapy medicinal products because of their immunomodulatory properties and supportive role in hematopoiesis. Although bone marrow remains the most common source for obtaining off-the-shelf MSC, cord blood (CB) represents an alternative source, which can be collected noninvasively and without major ethical concerns. However, the low estimated frequency and inconsistency of successful isolation represent open challenges for the use of CB-derived MSC in clinical trials. This study explores whether CB may represent a suitable source of MSC for clinical use and analyzes several in vitro parameters useful to better define the quality of CB-derived MSC prior to clinical application. METHODS CB units (n = 50) selected according to quality criteria (CB volume ≥ 20 ml, time from collection ≤ 24 h) were cultured using a standardized procedure for CB-MSC generation. MSC were analyzed for their growth potential and secondary colony-forming capacity. Immunophenotype and multilineage differentiation potential of culture-expanded CB-MSC were assessed to verify MSC identity. The immunomodulatory activity at resting conditions and after inflammatory priming (IFN-γ-1b and TNF-α for 48 hours) was explored to assess the in vitro potency of CB-MSC prior to clinical application. Molecular karyotyping was used to assess the genetic stability after prolonged MSC expansion. RESULTS We were able to isolate MSC colonies from 44% of the processed units. Our results do not support a role of CB volume in determining the outcome of the cultures, in terms of both isolation and proliferative capacity of CB-MSC. Particularly, we have confirmed the existence of two different CB-MSC populations named short- and long-living (SL- and LL-) CBMSC, clearly diverging in their growth capacity and secondary colony-forming efficiency. Only LL-CBMSC were able to expand consistently and to survive for longer periods in vitro, while preserving genetic stability. Therefore, they may represent interesting candidates for therapeutic applications. We have also observed that LL-CBMSC were not equally immunosuppressive, particularly after inflammatory priming and despite upregulating priming-inducible markers. CONCLUSIONS This work supports the use of CB as a potential MSC source for clinical applications, remaining more readily available compared to conventional sources. We have provided evidence that not all LL-CBMSC are equally immunosuppressive in an inflammatory environment, suggesting the need to include the assessment of potency among the release criteria for each CB-MSC batch intended for clinical use, at least for the treatment of immune disorders as GvHD.
Collapse
Affiliation(s)
- Eliana Amati
- Advanced Cellular Therapy Laboratory - Hematology Unit, S. Bortolo Hospital - ULSS 6, Contra' San Francesco 41, 36100, Vicenza, Italy
| | - Sabrina Sella
- Advanced Cellular Therapy Laboratory - Hematology Unit, S. Bortolo Hospital - ULSS 6, Contra' San Francesco 41, 36100, Vicenza, Italy
| | - Omar Perbellini
- Advanced Cellular Therapy Laboratory - Hematology Unit, S. Bortolo Hospital - ULSS 6, Contra' San Francesco 41, 36100, Vicenza, Italy
| | | | - Martina Bernardi
- Advanced Cellular Therapy Laboratory - Hematology Unit, S. Bortolo Hospital - ULSS 6, Contra' San Francesco 41, 36100, Vicenza, Italy.,Hematology Project Foundation, Vicenza, Italy
| | - Katia Chieregato
- Advanced Cellular Therapy Laboratory - Hematology Unit, S. Bortolo Hospital - ULSS 6, Contra' San Francesco 41, 36100, Vicenza, Italy.,Hematology Project Foundation, Vicenza, Italy
| | - Chiara Lievore
- Transfusion Medicine, S. Bortolo Hospital, Vicenza, Italy
| | - Denise Peserico
- Advanced Cellular Therapy Laboratory - Hematology Unit, S. Bortolo Hospital - ULSS 6, Contra' San Francesco 41, 36100, Vicenza, Italy
| | - Manuela Rigno
- Transfusion Medicine, S. Bortolo Hospital, Vicenza, Italy
| | - Anna Zilio
- Genetics and Molecular Biology, Transfusion Medicine, S. Bortolo Hospital, Vicenza, Italy
| | - Marco Ruggeri
- Advanced Cellular Therapy Laboratory - Hematology Unit, S. Bortolo Hospital - ULSS 6, Contra' San Francesco 41, 36100, Vicenza, Italy
| | | | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory - Hematology Unit, S. Bortolo Hospital - ULSS 6, Contra' San Francesco 41, 36100, Vicenza, Italy.
| |
Collapse
|
37
|
Kil K, Choi MY, Kong JS, Kim WJ, Park KH. Regenerative efficacy of mesenchymal stromal cells from human placenta in sensorineural hearing loss. Int J Pediatr Otorhinolaryngol 2016; 91:72-81. [PMID: 27863646 DOI: 10.1016/j.ijporl.2016.10.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 10/07/2016] [Accepted: 10/09/2016] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Hearing loss is a common chronic disorder characterized by decline of auditory function. The global population have suffered from deafness and the transplantation of stem cells is regarded as a therapeutic strategy for this disease. METHODS We collected placenta from a total of 13 samples of full term pregnant women and isolated MSCs derived from human placenta and transplanted MSCs on deaf animal model. The normal group and the sensorineural hearing loss (SNHL) group and the experimental (transplanted MSCs) group were compared and estimated hearing level using auditory brainstem response (ABR) recordings and the otoacoustic emission (OAE) test. RESULTS ABR threshold value and DPOAE level showed that MSCs transplantation groups was improved than the SNHL group. And the number of spiral ganglion neurons were increased in all turn of the cochlea. And there was no evidence of acute immunological rejection and inflammation response was not observed. DISCUSSION This study is to evaluate regenerative efficacy of hearing loss by transplanting mesenchymal stromal cells (MSCs) derived from human placenta (amnion and chorion) in deaf animal model. We identified that MSCs transplantation restored auditory impairment and promoted cell regeneration. We hope to overcome sensorineural hearing loss by transplanting stem cells such as mesenchymal stromal cells (MSCs) from easily accessible adult stem cell source in placenta.
Collapse
Affiliation(s)
- Kicheol Kil
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, South Korea
| | - Mi Young Choi
- Department of Medical Cell Biology, The Catholic University of Korea School of Medicine, Seoul, South Korea; Department of Otolaryngology-Head & Neck Surgery, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, South Korea
| | - Ji Sun Kong
- Department of Otolaryngology-Head & Neck Surgery, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, South Korea
| | - Woo Jin Kim
- Department of Otolaryngology-Head & Neck Surgery, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, South Korea
| | - Kyoung Ho Park
- Department of Otolaryngology-Head & Neck Surgery, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, South Korea.
| |
Collapse
|
38
|
Lange-Consiglio A, Perrini C, Bertero A, Esposti P, Cremonesi F, Vincenti L. Isolation, molecular characterization, and in vitro differentiation of bovine Wharton jelly-derived multipotent mesenchymal cells. Theriogenology 2016; 89:338-347. [PMID: 28341078 DOI: 10.1016/j.theriogenology.2016.09.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/21/2016] [Accepted: 09/23/2016] [Indexed: 12/16/2022]
Abstract
Extrafetal tissues are a noncontroversial and inexhaustible source of mesenchymal stem cells that can be harvested noninvasively at low cost. In the veterinary field, as in man, stem cells derived from extrafetal tissues express plasticity, reduced immunogenicity, and have high anti-inflammatory potential making them promising candidates for treatment of many diseases. Umbilical cord mesenchymal cells have been isolated and characterized in different species and have recently been investigated as potential candidates in regenerative medicine. In this study, cells derived from bovine Wharton jelly (WJ) were isolated for the first time by enzymatic methods, frozen/thawed, cultivated for at least 10 passages, and characterized. Wharton jelly-derived cells readily attached to plastic culture dishes displaying typical fibroblast-like morphology and, although their proliferative capacity decreased to the seventh passage, these cells showed a mean doubling time of 34.55 ± 6.33 hours and a mean frequency of one colony-forming unit fibroblast like for every 221.68 plated cells. The results of molecular biology studies and flow cytometry analyses revealed that WJ-derived cells showed the typical antigen profile of mesenchymal stem cells and were positive for CD29, CD44, CD105, CD166, Oct-4, and c-Myc. They were negative for CD34 and CD14. Remarkably, WJ-derived cells showed differentiation ability. After culture in induced media, WJ-derived cells were able to differentiate into osteogenic, adipogenic, chondrogenic, and neurogenic lines as shown by positive staining and expression of specific markers. On polymerase chain reaction analysis, these cells were negative for MHC-II and positive for MHC-I, thus reinforcing the role of extrafetal tissue as an allogenic source for bovine cell-based therapies. These results provide evidence that bovine WJ-derived cells may have the potential to differentiate to repair damaged tissues and reinforce the importance of extrafetal tissues as stem cell sources in veterinary regenerative medicine. A more detailed evaluation of their immunologic properties is necessary to better understand their potential role in cellular therapy.
Collapse
Affiliation(s)
- Anna Lange-Consiglio
- Reproduction Unit, Large Animal Hospital, Università degli Studi di Milano, Lodi, Italy
| | - Claudia Perrini
- Reproduction Unit, Large Animal Hospital, Università degli Studi di Milano, Lodi, Italy
| | - Alessia Bertero
- Department of Animal Science, Università degli Studi di Torino, Torino, Italy
| | - Paola Esposti
- Reproduction Unit, Large Animal Hospital, Università degli Studi di Milano, Lodi, Italy
| | - Fausto Cremonesi
- Reproduction Unit, Large Animal Hospital, Università degli Studi di Milano, Lodi, Italy.
| | - Leila Vincenti
- Department of Animal Science, Università degli Studi di Torino, Torino, Italy
| |
Collapse
|
39
|
Vitrenko Y, Kostenko I, Kulebyakina K, Duda A, Klunnyk M, Sorochynska K. Fetal Tissues Tested for Microbial Sterility by Culture- and PCR-Based Methods Can be Safely Used in Clinics. Cell Transplant 2016; 26:339-350. [PMID: 27501947 DOI: 10.3727/096368916x692735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cell preparations to be used in clinical practice must be free of infectious agents. Safety concerns are especially elevated upon the use of human fetal tissues, which are otherwise highly advantageous in cell therapy. We demonstrate that treating fetal samples with antibiotic, extensive washing, and homogenization prior to cryoconservation efficiently removes microbes in general. Screening a large collection by an automatic culture system showed that 89.2% fetal tissue samples were sterile, while contamination was detected in 10.8% samples. Liver and chorion were contaminated more than the brain, kidney, lung, and soft tissues. Broad-range PCR from the bacterial 16s rRNA gene was adopted as a confirmatory assay; however, the concordance between the culture-based and PCR assays was weak. Taxonomic identification was done for contaminated samples by bacteriological methods and sequencing 16s rRNA PCR products. The two approaches revealed different spectra of taxonomic groups sharing only Lactobacillus, the most frequently found genus. In addition, other representatives of vaginal microbiota were detected by culture-based identification, while PCR product sequencing has also revealed a subset of nosocomial microorganisms. Importantly, species known to cause sepsis were identified by both techniques, arguing for their indispensability and mutual complementarity. We suggest that most contaminations are taken up during collection of fetal material rather than originating from an in utero infection. In conclusion, a rigorous microbiological control by culture and PCR is a prerequisite for safe clinical use of fetal tissue suspensions.
Collapse
|
40
|
Wei Y, Fang J, Cai S, Lv C, Zhang S, Hua J. Primordial germ cell-like cells derived from canine adipose mesenchymal stem cells. Cell Prolif 2016; 49:503-11. [PMID: 27374854 PMCID: PMC6496567 DOI: 10.1111/cpr.12271] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/30/2016] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES Previous studies have shown that adipose mesenchymal stem cells (AMSCs) share the potency of typical bone marrow mesenchymal stem cells (MSCs); however, there is little information concerning characteristics of canine AMSCs (CAMSCs); it has not previously been made clear whether CAMSCs would be able to differentiate into other cell types. MATERIALS AND METHODS In this study, typical AMSC lines were established, and their characteristics including morphology, typical markers and differentiation potentiality were tested. RESULTS The cells exhibited typical MSC morphology and were positive for CD90, CD44 and CD166, considered to be MSCs surface markers. They were negative for CD34 and CD45. The CAMSCs also exhibited embryonic stem cell (ESC) markers, including Oct4 and Sox2, at passage 2. In an appropriate microenvironment, CAMSCs differentiated into EBs and were able to produce cells of the three germ layers. These results indicate that established cells were putative adipocyte-derived MSCs, which also displayed properties of ESCs. Moreover, when the CAMSCs were induced by bone morphogenetic protein 4 (BMP4), they differentiated into PGC-like cells (PGCLCs) and male germ-like cells, which were positive for PR domain-containing 1 (Prdm1), PR domain-containing 14 (Prdm14), doublesex and mab-3 related transcription factor (Dmrt1), as well as for promyelocytic leukaemia zinc finger (Plzf). Quantitative real-time PCR (qRT-PCR) and western blotting analysis verified higher expression levels of these markers. CONCLUSION This study provides an efficient approach to study germ cell development using CAMSCs.
Collapse
Affiliation(s)
- Yudong Wei
- College of Veterinary MedicineShaanxi Centre of Stem Cells Engineering & TechnologyNorthwest A&F UniversityYangling Shaanxi 712100China
| | - Jia Fang
- College of Veterinary MedicineShaanxi Centre of Stem Cells Engineering & TechnologyNorthwest A&F UniversityYangling Shaanxi 712100China
| | - Shufang Cai
- College of Veterinary MedicineShaanxi Centre of Stem Cells Engineering & TechnologyNorthwest A&F UniversityYangling Shaanxi 712100China
| | - Changrong Lv
- College of Veterinary MedicineShaanxi Centre of Stem Cells Engineering & TechnologyNorthwest A&F UniversityYangling Shaanxi 712100China
| | - Shiqiang Zhang
- College of Veterinary MedicineShaanxi Centre of Stem Cells Engineering & TechnologyNorthwest A&F UniversityYangling Shaanxi 712100China
| | - Jinlian Hua
- College of Veterinary MedicineShaanxi Centre of Stem Cells Engineering & TechnologyNorthwest A&F UniversityYangling Shaanxi 712100China
| |
Collapse
|
41
|
Bajek A, Olkowska J, Walentowicz-Sadłecka M, Walentowicz P, Sadłecki P, Grabiec M, Bodnar M, Marszałek A, Dębski R, Porowińska D, Czarnecka J, Kaźmierski Ł, Drewa T. High Quality Independent From a Donor: Human Amniotic Fluid Derived Stem Cells-A Practical Analysis Based on 165 Clinical Cases. J Cell Biochem 2016; 118:116-126. [DOI: 10.1002/jcb.25618] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/03/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Anna Bajek
- Department of Tissue Engineering; Nicolaus Copernicus University; Bydgoszcz 85-092 Poland
| | - Joanna Olkowska
- Department of Tissue Engineering; Nicolaus Copernicus University; Bydgoszcz 85-092 Poland
| | | | - Paweł Walentowicz
- Department of Obstetrics and Gynecology; Nicolaus Copernicus University; Bydgoszcz 85-168 Poland
| | - Paweł Sadłecki
- Department of Obstetrics and Gynecology; Nicolaus Copernicus University; Bydgoszcz 85-168 Poland
| | - Marek Grabiec
- Department of Obstetrics and Gynecology; Nicolaus Copernicus University; Bydgoszcz 85-168 Poland
| | - Magdalena Bodnar
- Department of Clinical Pathomorphology; Nicolaus Copernicus University; Bydgoszcz 85-094 Poland
| | - Andrzej Marszałek
- Department of Clinical Pathomorphology; Nicolaus Copernicus University; Bydgoszcz 85-094 Poland
| | - Robert Dębski
- Department of Experimental Oncology; Nicolaus Copernicus University; Bydgoszcz 85-094 Poland
| | - Dorota Porowińska
- Department of Biochemistry; Nicolaus Copernicus University; Toruń 87-100 Poland
| | - Joanna Czarnecka
- Department of Biochemistry; Nicolaus Copernicus University; Toruń 87-100 Poland
| | - Łukasz Kaźmierski
- Department of Tissue Engineering; Nicolaus Copernicus University; Bydgoszcz 85-092 Poland
| | - Tomasz Drewa
- Department of Tissue Engineering; Nicolaus Copernicus University; Bydgoszcz 85-092 Poland
- Department of Urology; Nicolaus Copernicus Hospital; Toruń 87-100 Poland
| |
Collapse
|
42
|
Mezouar D, Merzouk H, Merzouk AS, Merzouk SA, Belarbi B, Narce M. In vitro effects of vitamins C and E, n-3 and n-6 PUFA and n-9 MUFA on placental cell function and redox status in type 1 diabetic pregnant women. Placenta 2016; 42:114-21. [PMID: 27238721 DOI: 10.1016/j.placenta.2016.04.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/22/2016] [Accepted: 04/14/2016] [Indexed: 10/21/2022]
Abstract
The aim of this investigation was to determine the in vitro effects of vitamin C and E, n-3 and n-6 PUFA and n-9 MUFA on placental cell proliferation and function in type 1 diabetes. Placenta tissues were collected from 30 control healthy and 30 type 1 diabetic women at delivery. Placental cells were isolated and were cultured in RPMI medium supplemented with vitamin C (50 μM), vitamin E (50 μM), n-3 PUFA (100 μM), n-6 PUFA (100 μM) or n-9 MUFA (100 μM). Cell proliferation, cell glucose uptake and intracellular oxidative status were investigated. Our results showed that basal placental cell proliferation, glucose uptake, malondialdehyde (MDA) and carbonyl proteins were higher while intracellular reduced glutathione (GSH) levels and catalase activities were lower in placentas from diabetic women as compared to controls. Vitamins C and E induced a modulation of placental cell proliferation and glucose consumption without affecting intracellular redox status in both diabetic and control groups. N-3 and n-6 PUFA diminished placental cell proliferation and enhanced intracellular oxidative stress while n-9 MUFA had no effects in the two groups. Co-administration of n-3 or n-6 PUFA and vitamin C or E were capable of reversing back the PUFA-decreased cell proliferation and normalizing placental cell function and redox status especially in diabetes. In conclusion, PUFA and antioxidant vitamin combinations may be beneficial in improving placenta function and in reducing placental oxidative stress in type 1 diabetic pregnancy.
Collapse
Affiliation(s)
- Djamila Mezouar
- Laboratory of Physiology, Physiopathology and Biochemistry of Nutrition, Department of Biology, Faculty of Natural and Life Sciences, Earth and Universe, University Abou-Bekr Belkaïd, Tlemcen 13000, Algeria
| | - Hafida Merzouk
- Laboratory of Physiology, Physiopathology and Biochemistry of Nutrition, Department of Biology, Faculty of Natural and Life Sciences, Earth and Universe, University Abou-Bekr Belkaïd, Tlemcen 13000, Algeria.
| | - Amel Saidi Merzouk
- Laboratory of Physiology, Physiopathology and Biochemistry of Nutrition, Department of Biology, Faculty of Natural and Life Sciences, Earth and Universe, University Abou-Bekr Belkaïd, Tlemcen 13000, Algeria
| | - Sid Ahmed Merzouk
- Department of Technical Sciences, Faculty of Engineering, University Abou-Bekr Belkaïd, Tlemcen 13000, Algeria
| | - Boumediene Belarbi
- Gynecology and Obstetrics Department, Mother and Infant Hospital Center, University Abou-Bekr Belkaïd, Tlemcen 13000, Algeria
| | - Michel Narce
- INSERM UMR866, "Lipids Nutrition Cancer," Faculty of Life, Earth and Environment Sciences, University of Burgundy, Dijon 21000, France
| |
Collapse
|
43
|
The Immunomodulatory Effects of Mesenchymal Stem Cells in Prevention or Treatment of Excessive Scars. Stem Cells Int 2015; 2016:6937976. [PMID: 26839566 PMCID: PMC4709788 DOI: 10.1155/2016/6937976] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/22/2015] [Accepted: 09/17/2015] [Indexed: 12/12/2022] Open
Abstract
Excessive scars, including keloids and hypertrophic scars, result from aberrations in the process of physiologic wound healing. An exaggerated inflammatory process is one of the main pathophysiological contributors. Scars may cause pain, and pruritis, limit joint mobility, and cause a range of cosmetic deformities that affect the patient's quality of life. Extensive research has been done on hypertrophic scar and keloid formation that has resulted in the plethora of treatment and prevention methods practiced today. Mesenchymal stem cells, among their multifunctional roles, are known regulators of inflammation and have been receiving attention as a major candidate for cell therapy to treat or prevent excessive scars. This paper extensively reviews the body of research examining the mechanism and potential of stem cell therapy in the treatment of excessive scars.
Collapse
|
44
|
Gothard D, Cheung K, Kanczler JM, Wilson DI, Oreffo ROC. Regionally-derived cell populations and skeletal stem cells from human foetal femora exhibit specific osteochondral and multi-lineage differentiation capacity in vitro and ex vivo. Stem Cell Res Ther 2015; 6:251. [PMID: 26684339 PMCID: PMC4683700 DOI: 10.1186/s13287-015-0247-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 11/25/2014] [Accepted: 11/25/2015] [Indexed: 01/08/2023] Open
Abstract
Background Adult skeletal stem cells (SSCs) often exhibit limited in vitro expansion with undesirable phenotypic changes and loss of differentiation capacity. Foetal tissues offer an alternative cell source, providing SSCs which exhibit desirable differentiation capacity over prolonged periods, ideal for extensive in vitro and ex vivo investigation of fundamental bone biology and skeletal development. Methods We have examined the derivation of distinct cell populations from human foetal femora. Regionally isolated populations including epiphyseal and diaphyseal cells were carefully dissected. Expression of the SSC marker Stro-1 was also found in human foetal femora over a range of developmental stages and subsequently utilised for immuno-selection. Results Regional populations exhibited chondrogenic (epiphyseal) and osteogenic (diaphyseal) phenotypes following in vitro and ex vivo characterisation and molecular analysis, indicative of native SSC maturation during skeletal development. However, each population exhibited potential for induced multi-lineage differentiation towards bone (bone nodule formation), cartilage (proteoglycan and mucopolysaccharide deposition) and fat (lipid deposition), suggesting the presence of a shared stem cell sub-population. This shared sub-population may be comprised of Stro-1+ cells, which were later identified and immuno-selected from whole foetal femora exhibiting multi-lineage differentiation capacity in vitro and ex vivo. Conclusions Distinct populations were isolated from human foetal femora expressing osteochondral differentiation capacity. Stro-1 immuno-selected SSCs were isolated from whole femora expressing desirable multi-lineage differentiation capacity over prolonged in vitro expansion, superior to their adult-derived counterparts, providing a valuable cell source with which to study bone biology and skeletal development. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0247-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David Gothard
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, School of Medicine, Institute of DevelopmentalSciences, Mail Point 887, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK.
| | - Kelvin Cheung
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, School of Medicine, Institute of DevelopmentalSciences, Mail Point 887, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK.
| | - Janos M Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, School of Medicine, Institute of DevelopmentalSciences, Mail Point 887, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK.
| | - David I Wilson
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, School of Medicine, Institute of Developmental Sciences, Mail Point 887, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK. .,University Hospital Southampton NHS Foundation Trust, Tremona Road, SO16 6YD, Southampton, UK.
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, University of Southampton, School of Medicine, Institute of DevelopmentalSciences, Mail Point 887, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK.
| |
Collapse
|
45
|
Shivakumar SB, Bharti D, Jang SJ, Hwang SC, Park JK, Shin JK, Byun JH, Park BW, Rho GJ. Cryopreservation of Human Wharton's Jelly-derived Mesenchymal Stem Cells Following Controlled Rate Freezing Protocol Using Different Cryoprotectants; A Comparative Study. Int J Stem Cells 2015; 8:155-69. [PMID: 26634064 PMCID: PMC4651280 DOI: 10.15283/ijsc.2015.8.2.155] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES To compare the effect of three different cryoprotectants on basic stem cell characteristics for the possibility of using well defined, dimethyl sulfoxide (DMSO) and serum free freezing solutions to cryopreserve human Wharton's jelly-derived mesenchymal stem cells (WJMSCs) following controlled rate freezing protocol. METHODS The mesenchymal stem cells isolated from human Wharton's jelly were cryopreserved using 10% DMSO, 10% polyvinylpyrrolidone (PVP) and a cocktail solution comprising of 0.05 M glucose, 0.05 M sucrose and 1.5 M ethylene glycol following controlled rate freezing protocol. We investigated the post-thaw cell viability, morphology, proliferation capacity, basic stem cell characteristics, in vitro differentiation potential and apoptosis-related gene expression profile before and after cryopreservation. RESULTS The cryoprotectant 10% DMSO has shown higher post-thaw cell viability of 81.2±0.58% whereas 10% PVP and cocktail solution have shown 62.87±0.35% and 72.2±0.23%, respectively at 0 h immediately thawing. The cell viability was further reduced in all the cryopreserved groups at 24 h later post-thaw culture. Further, the complete elimination of FBS in cryoprotectants has resulted in drastic reduction in cell viability. Cryopreservation did not alter the basic stem cell characteristics, plasticity and multipotency except proliferation rate. The expression of pro-apoptotic BAX and p53 genes were higher whilst p21 was lower in all the cryopreserved groups when compare to the control group of WJMSCs. CONCLUSION Although 10% DMSO has shown higher post-thaw cell viability compare to 10% PVP and cocktail solution, the present study indicates the feasibility of developing a well-defined DMSO free cryosolution which can improve storage and future broad range applications of WJMSCs in regenerative medicine without losing their basic stem cell characteristics.
Collapse
Affiliation(s)
- Sharath Belame Shivakumar
- Department of Vet OBS/Theriogenology and Biotechnology, Gyeongsang National University, Jinju, Korea
| | - Dinesh Bharti
- Department of Vet OBS/Theriogenology and Biotechnology, Gyeongsang National University, Jinju, Korea
| | - Si-Jung Jang
- Department of Vet OBS/Theriogenology and Biotechnology, Gyeongsang National University, Jinju, Korea
| | - Sun-Chul Hwang
- Department of Orthopaedic Surgery, School of Medicine, Gyeongsang National University, Jinju, Korea
| | - Ji-Kwon Park
- Department of Obstetrics and Gynaecology, School of Medicine, Gyeongsang National University, Jinju, Korea
| | - Jeong-Kyu Shin
- Department of Obstetrics and Gynaecology, School of Medicine, Gyeongsang National University, Jinju, Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, School of Medicine, Gyeongsang National University, Jinju, Korea
| | - Bong-Wook Park
- Department of Oral and Maxillofacial Surgery, School of Medicine, Gyeongsang National University, Jinju, Korea
| | - Gyu-Jin Rho
- Department of Vet OBS/Theriogenology and Biotechnology, Gyeongsang National University, Jinju, Korea ; Research Institute of Life Sciences, Gyeongsang National University, Jinju, Korea
| |
Collapse
|
46
|
Jhala D, Vasita R. A Review on Extracellular Matrix Mimicking Strategies for an Artificial Stem Cell Niche. POLYM REV 2015. [DOI: 10.1080/15583724.2015.1040552] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
47
|
Alrefaei GI, Al-Karim S, Ayuob NN, Ali SS. Does the maternal age affect the mesenchymal stem cell markers and gene expression in the human placenta? What is the evidence? Tissue Cell 2015; 47:406-19. [DOI: 10.1016/j.tice.2015.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 12/01/2022]
|
48
|
Rutigliano L, Valentini L, Martino NA, Pizzi F, Zanghì A, Dell'Aquila ME, Minervini F. Ochratoxin A at low concentrations inhibits in vitro growth of canine umbilical cord matrix mesenchymal stem cells through oxidative chromatin and DNA damage. Reprod Toxicol 2015; 57:121-9. [PMID: 26055943 DOI: 10.1016/j.reprotox.2015.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 05/22/2015] [Accepted: 05/28/2015] [Indexed: 12/12/2022]
Abstract
Ochratoxin A (OTA) exposure during pregnancy in laboratory animals induces delayed/abnormal embryo development. Foetal adnexa-derived mesenchymal stem cells (MSCs) could help evaluate the developmental risk of exposure to chemicals in advanced gestational age. We tested the effects of OTA at concentrations ranging from 2.5×10(-4) to 25nM on growth parameters of canine umbilical cord matrix (UCM)-derived MSCs. The hypothesis that oxidative chromatin and DNA damage could underlie OTA-mediated cell toxicity was also investigated. After in vitro exposure, OTA significantly decreased cell density and increased doubling time in a passage- and concentration-dependent manner and no exposed cells survived beyond passage 5. Significantly higher rates of cells showed condensed and fragmented chromatin and oxidized DNA, as assessed by OxyDNA assay. These findings showed that in vitro exposure to OTA, at picomolar levels, perturbs UCM-MSC growth parameters through oxidative chromatin and DNA damage, suggesting possible consequences on canine foetal development.
Collapse
Affiliation(s)
- Lucia Rutigliano
- Veterinary Clinics and Animal Production Section, Department of Emergency and Organ Transplantations (DETO), University of Bari Aldo Moro, Str. Prov. Casamassima km 3, 70010 Valenzano (BA), Italy.
| | - Luisa Valentini
- Veterinary Clinics and Animal Production Section, Department of Emergency and Organ Transplantations (DETO), University of Bari Aldo Moro, Str. Prov. Casamassima km 3, 70010 Valenzano (BA), Italy.
| | - Nicola Antonio Martino
- Istituto Zooprofilattico Sperimentale Puglia e Basilicata, Via Manfredonia 20, 71121 Foggia, Italy; Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica (DBBB), Università degli Studi di Bari Aldo Moro, Str. Prov. Casamassima Km 3, 70010 Valenzano (BA), Italy.
| | - Flavia Pizzi
- Istituto di Biologia e Biotecnologia Agraria (IBBA) Consiglio Nazionale delle Ricerche (CNR), Unità Org. di Supporto di Lodi-c/o Parco Tecnologico Padano, via Einstein, 26500 Lodi, Italy.
| | - Antonina Zanghì
- Department of Veterinary Sciences, University of Messina, Polo Universitario SS Annunziata, 98168 Messina, Italy.
| | - Maria Elena Dell'Aquila
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica (DBBB), Università degli Studi di Bari Aldo Moro, Str. Prov. Casamassima Km 3, 70010 Valenzano (BA), Italy.
| | - Fiorenza Minervini
- Institute of Sciences of Food Production (ISPA), National Research Council of Italy (CNR), Via G. Amendola 122/O, 70125 Bari, Italy.
| |
Collapse
|
49
|
Pipino C, Pandolfi A. Osteogenic differentiation of amniotic fluid mesenchymal stromal cells and their bone regeneration potential. World J Stem Cells 2015; 7:681-690. [PMID: 26029340 PMCID: PMC4444609 DOI: 10.4252/wjsc.v7.i4.681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/16/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
Abstract
In orthopedics, tissue engineering approach using stem cells is a valid line of treatment for patients with bone defects. In this context, mesenchymal stromal cells of various origins have been extensively studied and continue to be a matter of debate. Although mesenchymal stromal cells from bone marrow are already clinically applied, recent evidence suggests that one may use mesenchymal stromal cells from extra-embryonic tissues, such as amniotic fluid, as an innovative and advantageous resource for bone regeneration. The use of cells from amniotic fluid does not raise ethical problems and provides a sufficient number of cells without invasive procedures. Furthermore, they do not develop into teratomas when transplanted, a consequence observed with pluripotent stem cells. In addition, their multipotent differentiation ability, low immunogenicity, and anti-inflammatory properties make them ideal candidates for bone regenerative medicine. We here present an overview of the features of amniotic fluid mesenchymal stromal cells and their potential in the osteogenic differentiation process. We have examined the papers actually available on this regard, with particular interest in the strategies applied to improve in vitro osteogenesis. Importantly, a detailed understanding of the behavior of amniotic fluid mesenchymal stromal cells and their osteogenic ability is desirable considering a feasible application in bone regenerative medicine.
Collapse
|
50
|
Larijani B, Aghayan HR, Goodarzi P, Arjmand B. GMP-grade human fetal liver-derived mesenchymal stem cells for clinical transplantation. Methods Mol Biol 2015; 1283:123-136. [PMID: 25092054 DOI: 10.1007/7651_2014_101] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Stem cell therapy seems a promising avenue in regenerative medicine. Within various stem cells, mesenchymal stem cells have progressively used for cellular therapy. Because of the age-related decreasing in the frequency and differentiating capacity of adult MSCs, fetal tissues such as fetal liver, lung, pancreas, spleen, etc. have been introduced as an alternative source of MSCs for cellular therapy. On the other hand, using stem cells as advanced therapy medicinal products, must be performed in compliance with cGMP as a quality assurance system to ensure the safety, quality, and identity of cell products during translation from the basic stem cell sciences into clinical cell transplantation. In this chapter the authors have demonstrated the manufacturing of GMP-grade human fetal liver-derived mesenchymal stem cells.
Collapse
Affiliation(s)
- Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, 14114, Iran
| | | | | | | |
Collapse
|