1
|
Wu W, An X, Gong W, Yang L, Liu N, Liu B, Guo B, Jiang Q, Li L. ShK-modified UCMSCs Inhibit M1-Like Macrophage Polarization and Alleviate Osteoarthritis Progression via PI3K/Akt Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2406822. [PMID: 39721037 DOI: 10.1002/advs.202406822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/15/2024] [Indexed: 12/28/2024]
Abstract
The potassium channel Kv1.3 plays an important role in regulating immune cell functions in many inflammatory diseases whereas rarely in osteoarthritis (OA). Here, it is demonstrated that the Kv1.3 of macrophages is upregulated in response to LPS stimulation, as well as in human OA synovium samples than non-OA. Administration of Stichodactyla toxin (ShK), a Kv1.3 blocker, significantly inhibited cartilage degeneration and synovial inflammation in animal models of OA in vivo by inhibiting M1 macrophage polarization and reducing the production of inflammatory factors. In this study, a transgenically engineered human umbilical cord mesenchymal stem cell (UCMSC) delivery system is developed that secreted a peptide ShK, a Kv1.3 potassium blocker, into the knee articular cavity. Collectively, the results identified Kv1.3 as a potential therapeutic target for OA and demonstrated the efficacy of using ShK transgenic engineered UCMSCs as a delivery for the peptide in OA treatment.
Collapse
Affiliation(s)
- Wenshu Wu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, 211166, P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting, 321 Zhongshan Road, Nanjing, 210000, P. R. China
| | - Xueying An
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, 211166, P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting, 321 Zhongshan Road, Nanjing, 210000, P. R. China
| | - Wang Gong
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, 211166, P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting, 321 Zhongshan Road, Nanjing, 210000, P. R. China
| | - Lin Yang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, 211166, P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting, 321 Zhongshan Road, Nanjing, 210000, P. R. China
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, P.R. China
| | - Na Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, 211166, P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting, 321 Zhongshan Road, Nanjing, 210000, P. R. China
| | - Bin Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, 211166, P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting, 321 Zhongshan Road, Nanjing, 210000, P. R. China
| | - Baosheng Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, 211166, P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting, 321 Zhongshan Road, Nanjing, 210000, P. R. China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, 211166, P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting, 321 Zhongshan Road, Nanjing, 210000, P. R. China
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, P.R. China
| | - Lan Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
- Institute of Medical 3D Printing, Nanjing University, Nanjing, 211166, P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting, 321 Zhongshan Road, Nanjing, 210000, P. R. China
| |
Collapse
|
2
|
Guda T, Stukel Shah JM, Lundquist BD, Macaitis JM, Pérez ML, Pfau-Cloud MR, Beltran FO, Schmitt CW, Corbin EM, Grunlan MA, Lien W, Wang HC, Burdette AJ. An In Vivo Assessment of Different Mesenchymal Stromal Cell Tissue Types and Their Differentiation State on a Shape Memory Polymer Scaffold for Bone Regeneration. J Biomed Mater Res B Appl Biomater 2024; 112:e35516. [PMID: 39607370 DOI: 10.1002/jbm.b.35516] [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: 01/26/2024] [Revised: 10/16/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024]
Abstract
A combined biomaterial and cell-based solution to heal critical size bone defects in the craniomaxillofacial area is a promising alternative therapeutic option to improve upon autografting, the current gold standard. A shape memory polymer (SMP) scaffold, composed of biodegradable poly(ε-caprolactone) and coated with bioactive polydopamine, was evaluated with mesenchymal stromal cells (MSCs) derived from adipose (ADSC), bone marrow (BMSC), or umbilical cord (UCSC) tissue in their undifferentiated state or pre-differentiated toward osteoblasts for bone healing in a rat calvarial defect model. Pre-differentiating ADSCs and UCSCs resulted in higher new bone volume fraction (15.69% ± 1.64%) compared to empty (i.e., untreated) defects and scaffold-only (i.e., unseeded) groups (4.41% ± 1.11%). Notably, only differentiated UCSCs exhibited a significant increase in new bone volume, surpassing both undifferentiated UCSCs and unseeded scaffolds. Further, differentiated ADSCs and UCSCs had significantly higher trabecular numbers than their undifferentiated counterparts, unseeded scaffolds, and untreated defects. Although the mineral density regenerated within the unseeded scaffold surpassed that achieved with cell seeding, the connectivity of this bone was diminished, as the regenerated tissue confined itself to the spherical morphology of the scaffold pores. The SMP scaffold alone, with undifferentiated BMSCs, with undifferentiated and differentiated ADSCs, and differentiated UCSCs (29.72 ± 1.49 N) demonstrated significant osseointegration compared to empty defects (14.34 ± 2.21 N) after 12 weeks of healing when assessed by mechanical push-out testing. Based on these results and tissue availability to obtain the cells, pre-differentiated ADSCs and UCSCs emerge as particularly promising candidates when paired with the SMP scaffold for repairing critical size bone defects in the craniofacial skeleton.
Collapse
Affiliation(s)
- Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas San Antonio, San Antonio, Texas, USA
| | | | | | | | - Mística Lozano Pérez
- Department of Biomedical Engineering and Chemical Engineering, University of Texas San Antonio, San Antonio, Texas, USA
| | - Michaela R Pfau-Cloud
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Felipe O Beltran
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas, USA
| | - Connie W Schmitt
- Air Force Research Laboratory, 711th Human Performance Wing, Airman Systems Directorate, Bioeffects Division, Veterinary Science Branch, San Antonio, Texas, USA
| | - Emily M Corbin
- Air Force Research Laboratory, 711th Human Performance Wing, Airman Systems Directorate, Bioeffects Division, Veterinary Science Branch, San Antonio, Texas, USA
| | - Melissa A Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas, USA
- Department of Chemistry, Texas A&M University, College Station, Texas, USA
| | - Wen Lien
- USAF Dental Research & Consultation Service, San Antonio, USA
| | - Heuy-Ching Wang
- Naval Medical Research Unit San Antonio, San Antonio, Texas, USA
| | | |
Collapse
|
3
|
Ismail MT, Anggrahini DW, Haryana SM, Setianto BY. HUVECs-derived exosomes increase neovascularization and decrease limb necrosis in hindlimb ischemia. NARRA J 2024; 4:e1358. [PMID: 39816111 PMCID: PMC11731677 DOI: 10.52225/narra.v4i3.1358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/20/2024] [Indexed: 01/18/2025]
Abstract
Chronic limb-threatening ischemia (CLTI) is the most severe manifestation of peripheral arterial disease (PAD) and imposes a significantly high burden due to its high risk of mortality and amputation. Revascularization is the first-line treatment for CLTI; however, the amputation rate remains high, and approximately one-third of patients are not eligible for this treatment. Therefore, there is an urgent need for more effective therapeutic strategies. The aim of this study was to investigate the effects and mechanisms of human umbilical vein endothelial cells (HUVECs)-derived exosomes on neovascularization and the degree of necrosis in a hindlimb ischemia model and to study the biological processes underlying their mechanisms. This is an in vivo experimental study with a post-test-only control group design. Forty BALB/c mice were randomized to receive injections of exosomes, conditioned media, and phosphate-buffered saline (PBS) one day after unilateral double ligation. A sham-operated group was also included as a control. Capillary density, arteriole lumen diameter, and histopathological necrosis were measured after seven days, while clinical necrosis was observed daily. MicroRNA profiling, in silico analysis, and transcriptomic analysis of vascular endothelial growth factor (VEGF) mRNA expression were performed to determine the possible biological processes. No amputation was found in the exosome group, as well as in the conditioned media and sham-operated groups, compared to three out of seven mice (43%) in the PBS group. The capillary density was higher in the exosome than in the PBS group (p = 0.026). The arteriole lumen diameter in the exosome group was larger than in the PBS (p = 0.033) and sham-operated (p = 0.034) groups. The scores of clinical necrosis and histopathological necrosis in the exosome group were lower than the PBS group (p = 0.005), while the histopathological necrosis scores were also lower but statistically insignificant. In silico analysis showed improvement in neovascularization and necrosis, possibly through energy regulation, PI3 K/AKT and TGF-β activation, the ubiquitin-proteasome system, and tyrosine kinases receptors. HUVEC exosomes were associated with lower VEGF mRNA expression, which may indicate a more effective compensatory mechanism under ischemic conditions. The exosome group had the lowest VEGF mRNA expression compared to other groups, although the difference was not statistically significant. This study highlights that HUVECs-derived exosomes improve neovascularization and decrease necrosis in a hindlimb ischemia mice model, potentially by modulating several possible mechanisms.
Collapse
Affiliation(s)
- Muhamad T. Ismail
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Dyah W. Anggrahini
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Sofia M. Haryana
- Department of Cell Histology and Biology Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Budi Y. Setianto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
4
|
Jin P, Liu H, Chen X, Liu W, Jiang T. From Bench to Bedside: The Role of Extracellular Vesicles in Cartilage Injury Treatment. Biomater Res 2024; 28:0110. [PMID: 39583872 PMCID: PMC11582190 DOI: 10.34133/bmr.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/20/2024] [Accepted: 10/26/2024] [Indexed: 11/26/2024] Open
Abstract
Cartilage repair is the key to the treatment of joint-related injury. However, because cartilage lacks vessels and nerves, its self-repair ability is extremely low. Extracellular vesicles (EVs) are bilayer nanovesicles with membranes mainly composed of ceramides, cholesterol, phosphoglycerides, and long-chain free fatty acids, containing DNA, RNA, and proteins (such as integrins and enzymes). For mediating intercellular communication and regulating mechanisms, EVs have been shown by multiple studies to be effective treatment options for cartilage repair. This review summarizes recent findings of different sources (mammals, plants, and bacteria) and uses of EVs in cartilage repair, mechanisms of EVs captured by injured chondrocytes, and quantification and storage of EVs, which may provide scientific guidance for promoting the development of EVs in the field of cartilage injury treatment.
Collapse
Affiliation(s)
- Pan Jin
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Huan Liu
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Xichi Chen
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Wei Liu
- Health Science Center,
Yangtze University, Jingzhou 434023, Hubei, China
| | - Tongmeng Jiang
- Key Laboratory of Emergency and Trauma of Ministry of Education, Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, The First Affiliated Hospital of Hainan Medical University,
Hainan Medical University, Haikou 571199, China
- Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma; Hainan Provincial Stem Cell Research Institute; Hainan Academy of Medical Sciences,
Hainan Medical University, Haikou 571199, China
| |
Collapse
|
5
|
Fu YS, Tsai SW, Tong ZJ, Yeh CC, Chen TH, Chen CF. Wharton's jelly of the umbilical cord serves as a natural biomaterial to promote osteogenesis. Biomater Sci 2024. [PMID: 39415619 DOI: 10.1039/d3bm02137h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Various factors can contribute to bone damage or loss, presenting challenges for bone regeneration. Our study explores the potential clinical applications of two processed forms of Wharton's jelly of the human umbilical cord for treating bone loss. Wharton's jelly from fresh umbilical cords underwent two distinct processes: (1) frozen Wharton's jelly (WJF), preserved with cryoprotective agents, and (2) decellularized Wharton's jelly matrix (WJD), prepared only via lyophilization without cryoprotectants. Both WJD and WJF are rich in collagen, hyaluronan, and polysaccharide proteins. Notably, WJD exhibited a porous structure lacking nuclei from human umbilical cord mesenchymal stem cells, unlike WJF. In direct contact experiments, WJD stimulated osteoblast migration, enhanced osteoblast maturation, and promoted calcium deposition for bone formation when administered to cultured rat osteoblasts. Furthermore, in transwell co-culture experiments, both WJD and WJF increased the rat osteoblast expression of RUNX2 and OPN genes, elevated alkaline phosphatase levels, and enhanced extracellular calcium precipitation, indicating their role in osteoblast maturation and new bone formation. Hyaluronic acid, one of the ingredients from WJD and WJF, was identified as a key component triggering osteogenesis. In vivo experiments involved creating circular bone defects in the calvarias of rats, where WJD and WJF were separately implanted and monitored over five months using micro-computerized tomography. Our results demonstrated that both WJD and WJF enhanced angiogenesis, collagen formation, osteoblast maturation, and bone growth within the bone defects. In summary, WJD and WJF, natural biomaterials with biocompatibility and nontoxicity, act not only as effective scaffolds but also promote osteoblast adhesion and differentiation, and accelerate osteogenesis.
Collapse
Affiliation(s)
- Yu-Show Fu
- Department of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| | - Shang-Wen Tsai
- Division of Joint Reconstruction, Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China.
- Department of Orthopaedics, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| | - Zhen-Jie Tong
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| | - Chang-Ching Yeh
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Obstetrics and Gynecology, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- Department of Nurse-Midwifery and Women Health, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Tien-Hua Chen
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung City, Taiwan
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
- Trauma Center, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Cheng-Fong Chen
- Division of Joint Reconstruction, Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China.
- Department of Orthopaedics, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Republic of China
| |
Collapse
|
6
|
Wang H, Zhang Q, Wu S, Pan D, Ning Y, Wang C, Guo J, Gu Y. Mesenchymal stem cell therapy in eosinophilic granulomatosis with polyangiitis-related lower limb gangrene: a case report. Stem Cell Res Ther 2024; 15:307. [PMID: 39285456 PMCID: PMC11406883 DOI: 10.1186/s13287-024-03924-9] [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: 01/30/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Eosinophilic granulomatosis with polyangiitis (EGPA), a rare but life-threatening systemic vasculitis, is distinguished by marked eosinophilia and presents with diverse symptoms, including asthma, cutaneous purpura, ecchymosis, skin necrosis, cardiac lesions, peripheral neuropathy, and necrotizing vasculitis. The etiology of EGPA involves a complex interaction among humoral, adaptive, innate, and allergic immune responses. Standard treatment employs prolonged high-dose glucocorticoid therapy, which is critical for survival; however, some patients' symptoms cannot be relieved. CASE REPORT This case report details the medical management of an 11-year-old patient with EGPA, who was at risk of bilateral lower limb amputation due to differential arterial occlusion and severe, necrotizing vasculitis-induced gangrene in both feet. Treatment modalities administered included systemic infusion of Umbilical Cord Mesenchymal Stem Cells (UC-MSCs), targeted gastrocnemius muscle injections, and application of a Placenta-Derived Mesenchymal Stem Cells (PD-MSCs) hydrogel. RESULTS After receiving a four-month regimen of allogeneic mesenchymal stem cell therapy via intravenous and local administration, the patient showed normalized eosinophil counts, reestablished blood flow in the dorsal arteries, and marked improvement in foot ulcerations. CONCLUSION Mesenchymal stem cell therapy is a promising option for severe EGPA cases refractory to glucocorticoids.
Collapse
Affiliation(s)
- Hui Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
- ShangRao Jingkai Health-Biotech United Hospital, ShangRao, 334000, Jiangxi, China
| | - Qian Zhang
- ShangRao Jingkai Health-Biotech United Hospital, ShangRao, 334000, Jiangxi, China
- Shangrao Normal University, ShangRao, 334000, Jiangxi, China
| | - Sensen Wu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
| | - Dikang Pan
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
| | - Yachan Ning
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
| | - Cong Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
| | - Jianming Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China.
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China.
| |
Collapse
|
7
|
Eivazi Zadeh Z, Nour S, Kianersi S, Jonidi Shariatzadeh F, Williams RJ, Nisbet DR, Bruggeman KF. Mining human clinical waste as a rich source of stem cells for neural regeneration. iScience 2024; 27:110307. [PMID: 39156636 PMCID: PMC11326931 DOI: 10.1016/j.isci.2024.110307] [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] [Indexed: 08/20/2024] Open
Abstract
Neural diseases are challenging to treat and are regarded as one of the major causes of disability and morbidity in the world. Stem cells can provide a solution, by offering a mechanism to replace damaged circuitry. However, obtaining sufficient cell sources for neural regeneration remains a significant challenge. In recent years, waste-derived stem(-like) cells (WDS-lCs) extracted from both prenatal and adult clinical waste tissues/products, have gained increasing attention for application in neural tissue repair and remodeling. This often-overlooked pool of cells possesses favorable characteristics; including self-renewal, neural differentiation, secretion of neurogenic factors, cost-effectiveness, and low ethical concerns. Here, we offer a perspective regarding the biological properties, extraction protocols, and preclinical and clinical treatments where prenatal and adult WDS-lCs have been utilized for cell replacement therapy in neural applications, and the challenges involved in optimizing these approaches toward patient led therapies.
Collapse
Affiliation(s)
- Zahra Eivazi Zadeh
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Shirin Nour
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
- Polymer Science Group, Department of Chemical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sogol Kianersi
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences, University of Galway, Galway, Ireland
| | | | - Richard J. Williams
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
- iMPACT, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - David R. Nisbet
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU College of Health & Medicine, Canberra, ACT, Australia
- Research School of Chemistry, ANU College of Science, Canberra, ACT, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
- Founder and Scientific Advisory of Nano Status, Building 137, Sullivans Creek Rd, ANU, Acton, Canberra, ACT, Australia
| | - Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials Research, School of Engineering, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
8
|
Lau CS, Park SY, Ethiraj LP, Singh P, Raj G, Quek J, Prasadh S, Choo Y, Goh BT. Role of Adipose-Derived Mesenchymal Stem Cells in Bone Regeneration. Int J Mol Sci 2024; 25:6805. [PMID: 38928517 PMCID: PMC11204188 DOI: 10.3390/ijms25126805] [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: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Bone regeneration involves multiple factors such as tissue interactions, an inflammatory response, and vessel formation. In the event of diseases, old age, lifestyle, or trauma, bone regeneration can be impaired which could result in a prolonged healing duration or requiring an external intervention for repair. Currently, bone grafts hold the golden standard for bone regeneration. However, several limitations hinder its clinical applications, e.g., donor site morbidity, an insufficient tissue volume, and uncertain post-operative outcomes. Bone tissue engineering, involving stem cells seeded onto scaffolds, has thus been a promising treatment alternative for bone regeneration. Adipose-derived mesenchymal stem cells (AD-MSCs) are known to hold therapeutic value for the treatment of various clinical conditions and have displayed feasibility and significant effectiveness due to their ease of isolation, non-invasive, abundance in quantity, and osteogenic capacity. Notably, in vitro studies showed AD-MSCs holding a high proliferation capacity, multi-differentiation potential through the release of a variety of factors, and extracellular vesicles, allowing them to repair damaged tissues. In vivo and clinical studies showed AD-MSCs favoring better vascularization and the integration of the scaffolds, while the presence of scaffolds has enhanced the osteogenesis potential of AD-MSCs, thus yielding optimal bone formation outcomes. Effective bone regeneration requires the interplay of both AD-MSCs and scaffolds (material, pore size) to improve the osteogenic and vasculogenic capacity. This review presents the advances and applications of AD-MSCs for bone regeneration and bone tissue engineering, focusing on the in vitro, in vivo, and clinical studies involving AD-MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Chau Sang Lau
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - So Yeon Park
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Lalith Prabha Ethiraj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Priti Singh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Grace Raj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Jolene Quek
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT 06269, USA;
| | - Yen Choo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Bee Tin Goh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
9
|
Mello DB, Mesquita FCP, Silva dos Santos D, Asensi KD, Dias ML, Campos de Carvalho AC, Goldenberg RCDS, Kasai-Brunswick TH. Mesenchymal Stromal Cell-Based Products: Challenges and Clinical Therapeutic Options. Int J Mol Sci 2024; 25:6063. [PMID: 38892249 PMCID: PMC11173248 DOI: 10.3390/ijms25116063] [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: 05/02/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Mesenchymal stromal cell (MSC)-based advanced therapy medicinal products (ATMPs) are being tried in a vast range of clinical applications. These cells can be isolated from different donor tissues by using several methods, or they can even be derived from induced pluripotent stem cells or embryonic stem cells. However, ATMP heterogeneity may impact product identity and potency, and, consequently, clinical trial outcomes. In this review, we discuss these topics and the need to establish minimal criteria regarding the manufacturing of MSCs so that these innovative therapeutics may be better positioned to contribute to the advancement of regenerative medicine.
Collapse
Affiliation(s)
- Debora B. Mello
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
| | | | - Danúbia Silva dos Santos
- Center of Cellular Technology, National Institute of Cardiology, INC, Rio de Janeiro 22240-002, Brazil;
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
| | - Karina Dutra Asensi
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Marlon Lemos Dias
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Antonio Carlos Campos de Carvalho
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Regina Coeli dos Santos Goldenberg
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Tais Hanae Kasai-Brunswick
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
10
|
Zhu Y, Huang C, Zheng L, Li Q, Ge J, Geng S, Zhai M, Chen X, Yuan H, Li Y, Jia W, Sun K, Li Y, Ye T, Zhao Z, Liu H, Liu Z, Jiang H. Safety and efficacy of umbilical cord tissue-derived mesenchymal stem cells in the treatment of patients with aging frailty: a phase I/II randomized, double-blind, placebo-controlled study. Stem Cell Res Ther 2024; 15:122. [PMID: 38679727 PMCID: PMC11057094 DOI: 10.1186/s13287-024-03707-2] [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: 01/08/2024] [Accepted: 03/24/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) hold a great promise for cell-based therapy in the field of regenerative medicine. In this study, we aimed to evaluate the safety and efficacy of intravenous infusion of human umbilical cord-derived MSCs (HUC-MSCs) in patients with aging frailty. METHODS In this randomized, double-blind, placebo-controlled trial, participants diagnosed with aging frailty were randomly assigned to receive intravenous administrations of HUC-MSCs or placebo. All of serious adverse events and AEs were monitored to evaluate the safety of treatment during the 6-month follow-up. The primary efficacy endpoint was alteration of physical component scores (PCS) of SF-36 qualities of life at 6 months. The secondary outcomes including physical performance tests and pro-inflammatory cytokines, were also observed and compared at each follow-up visits. All evaluations were performed at 1 week, 1, 2, 3 and 6 months following the first intravenous infusion of HUC-MSCs. RESULTS In the MSCs group, significant improvements in PCS of SF-36 were observed from first post-treatment visit and sustained throughout the follow-up period, with greater changes compared to the placebo group (p = 0.042). EQ-VAS scores of MSCs group improved significantly at 2 month (p = 0.023) and continued until the end of the 6-month visit (p = 0.002) in comparison to the placebo group. The timed up and go (TUG) physical performance test revealed significant group difference and showed continual enhancements over 6 months (p < 0.05). MSC transplantation improved the function of 4-m walking test (4MWT) compared with the placebo group with a decrease of 2.05 s at 6 months of follow-up (p = 0.21). The measurement of grip strength revealed group difference with MSCs group demonstrating better performance, particularly at 6 months (p = 0.002). Inflammatory cytokines (TNF-α, IL-17) exhibited declines in MSCs group at 6 months compared to the placebo group (p = 0.034 and 0.033, respectively). There was no difference of incidence of AEs between the two groups. CONCLUSION Intravenous transplantation of HUC-MSCs is a safe and effective therapeutic approach on aging frailty. The positive outcomes observed in improving quality of life, physical performance, and reducing chronic inflammation, suggest that HUC-MSC therapy may be a promising potential treatment option for aging frailty. TRIAL REGISTRATION Clinicaltrial.gov; NCT04314011; https://clinicaltrials.gov/ct2/show/NCT04314011 .
Collapse
Affiliation(s)
- Yingqian Zhu
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Ce Huang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai Municipality, 200032, China
| | - Liang Zheng
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qingqing Li
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jianli Ge
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - ShaSha Geng
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Miaomiao Zhai
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xin Chen
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Huixiao Yuan
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yang Li
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Wenwen Jia
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Keping Sun
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yan Li
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Tong Ye
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhengmei Zhao
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hailiang Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Zhongmin Liu
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, 200120, China.
| | - Hua Jiang
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Department of General Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
11
|
Hu Z, Li D, Wu S, Pei K, Fu Z, Yang Y, Huang Y, Yang J, Liu C, Hu J, Cai C, Liao Y. Unveiling the functional heterogeneity of cytokine-primed human umbilical cord mesenchymal stem cells through single-cell RNA sequencing. Cell Biosci 2024; 14:40. [PMID: 38532459 DOI: 10.1186/s13578-024-01219-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/13/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) hold immense promise for use in immunomodulation and regenerative medicine. However, their inherent heterogeneity makes it difficult to achieve optimal therapeutic outcomes for a specific clinical disease. Primed MSCs containing a certain cytokine can enhance their particular functions, thereby increasing their therapeutic potential for related diseases. Therefore, understanding the characteristic changes and underlying mechanisms of MSCs primed by various cytokines is highly important. RESULTS In this study, we aimed to reveal the cellular heterogeneity, functional subpopulations, and molecular mechanisms of MSCs primed with IFN-γ, TNF-α, IL-4, IL-6, IL-15, and IL-17 using single-cell RNA sequencing (scRNA-seq). Our results demonstrated that cytokine priming minimized the heterogeneity of the MSC transcriptome, while the expression of MSC surface markers exhibited only slight changes. Notably, compared to IL-6, IL-15, and IL-17; IFN-γ, TNF-α, and IL-4 priming, which stimulated a significantly greater number of differentially expressed genes (DEGs). Functional analysis, which included Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, indicated that IFN-γ, TNF-α, and IL-4-primed hUC-MSCs are involved in interferon-mediated immune-related processes, leukocyte migration, chemotaxis potential, and extracellular matrix and cell adhesion, respectively. Moreover, an investigation of various biological function scores demonstrated that IFN-γ-primed hUC-MSCs exhibit strong immunomodulatory ability, TNF-α-primed hUC-MSCs exhibit high chemotaxis potential, and IL-4-primed hUC-MSCs express elevated amounts of collagen. Finally, we observed that cytokine priming alters the distribution of functional subpopulations of MSCs, and these subpopulations exhibit various potential biological functions. Taken together, our study revealed the distinct regulatory effects of cytokine priming on MSC heterogeneity, biological function, and functional subpopulations at the single-cell level. CONCLUSIONS These findings contribute to a comprehensive understanding of the inflammatory priming of MSCs, paving the way for their precise treatment in clinical applications.
Collapse
Affiliation(s)
- Zhiwei Hu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Duanduan Li
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Shiduo Wu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Ke Pei
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Zeqin Fu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Yulin Yang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Yinfu Huang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Jian Yang
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Chuntao Liu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
| | - Junyuan Hu
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China
- Shenzhen Beike Biotechnology Research Institute, Shenzhen, 518054, China
| | - Cheguo Cai
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Yan Liao
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, 518054, China.
- Shenzhen Beike Biotechnology Research Institute, Shenzhen, 518054, China.
| |
Collapse
|
12
|
Esmaeili A, Eteghadi A, Landi FS, Yavari SF, Taghipour N. Recent approaches in regenerative medicine in the fight against neurodegenerative disease. Brain Res 2024; 1825:148688. [PMID: 38042394 DOI: 10.1016/j.brainres.2023.148688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Neurodegenerative diseases arise due to slow and gradual loss of structure and/or function of neurons and glial cells and cause different degrees of loss of cognition abilities and sensation. The little success in developing effective treatments imposes a high and regressive economic impact on society, patients and their families. In recent years, regenerative medicine has provided a great opportunity to research new innovative strategies with strong potential to treatleva these diseases. These effects are due to the ability of suitable cells and biomaterials to regenerate damaged nerves with differentiated cells, creating an appropriate environment for recovering or preserving existing healthy neurons and glial cells from destruction and damage. Ultimately, a better understanding and thus a further investigation of stem cell technology, tissue engineering, gene therapy, and exosomes allows progress towards practical and effective treatments for neurodegenerative diseases. Therefore, in this review, advances currently being developed in regenerative medicine using animal models and human clinical trials in neurological disorders are summarized.
Collapse
Affiliation(s)
- Ali Esmaeili
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefeh Eteghadi
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzaneh Saeedi Landi
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shadnaz Fakhteh Yavari
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloofar Taghipour
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Jamali F, Aldughmi M, Atiani S, Al-Radaideh A, Dahbour S, Alhattab D, Khwaireh H, Arafat S, Jaghbeer JA, Rahmeh R, Abu Moshref K, Bawaneh H, Hassuneh MR, Hourani B, Ababneh O, Alghwiri A, Awidi A. Human Umbilical Cord-Derived Mesenchymal Stem Cells in the Treatment of Multiple Sclerosis Patients: Phase I/II Dose-Finding Clinical Study. Cell Transplant 2024; 33:9636897241233045. [PMID: 38450623 PMCID: PMC10921855 DOI: 10.1177/09636897241233045] [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: 09/12/2023] [Revised: 01/08/2024] [Accepted: 01/31/2024] [Indexed: 03/08/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic neuro-inflammatory disease resulting in disabilities that negatively impact patients' life quality. While current treatment options do not reverse the course of the disease, treatment using mesenchymal stromal/stem cells (MSC) is promising. There has yet to be a consensus on the type and dose of MSC to be used in MS. This work aims to study the safety and efficacy of two treatment protocols of MSCs derived from the umbilical cord (UC-MSCs) and their secretome. The study included two groups of MS patients; Group A received two intrathecal doses of UC-MSCs, and Group B received a single dose. Both groups received UC-MSCs conditioned media 3 months post-treatment. Adverse events in the form of a clinical checklist and extensive laboratory tests were performed. Whole transcriptome analysis was performed on patients' cells at baseline and post-treatment. Results showed that all patients tolerated the cellular therapy without serious adverse events. The general disability scale improved significantly in both groups at 6 months post-treatment. Examining specific aspects of the disease revealed more parameters that improved in Group A compared to Group B patients, including a significant increase in the (CD3+CD4+) expressing lymphocytes at 12 months post-treatment. In addition, better outcomes were noted regarding lesion load, cortical thickness, manual dexterity, and information processing speed. Both protocols impacted the transcriptome of treated participants with genes, transcription factors, and microRNAs (miRNAs) differentially expressed compared to baseline. Inflammation-related and antigen-presenting (HLA-B) genes were downregulated in both groups. In contrast, TNF-alpha, TAP-1, and miR142 were downregulated only in Group A. The data presented indicate that both protocols are safe. Furthermore, it suggests that administering two doses of stem cells can be more beneficial to MS patients. Larger multisite studies should be initiated to further examine similar or higher doses of MSCs.
Collapse
Affiliation(s)
- Fatima Jamali
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Mayis Aldughmi
- Department of Physical Therapy, School of Rehabilitation Sciences, The University of Jordan, Amman, Jordan
| | - Serin Atiani
- Data Science Department, Princess Sumaya University for Technology, Amman, Jordan
| | - Ali Al-Radaideh
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Jordan University Hospital, The University of Jordan, Amman, Jordan
- Laboratory of Nanomedicine, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Said Dahbour
- Department of Medical Imaging, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Dana Alhattab
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Department of Medical Radiography, School of Health Sciences, University of Doha for Science and Technology, Doha, Qatar
| | - Hind Khwaireh
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Sally Arafat
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Joud Al Jaghbeer
- Department of Physical Therapy, School of Rehabilitation Sciences, The University of Jordan, Amman, Jordan
| | - Reem Rahmeh
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | | | - Hisham Bawaneh
- Hematology Department, Jordan University Hospital, Amman, Jordan
| | - Mona R. Hassuneh
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biology, Faculty of Sciences, The University of Jordan, Amman, Jordan
| | - Bayan Hourani
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Osameh Ababneh
- Department of Ophthalmology, Jordan University Hospital, School of Medicine, The University of Jordan, Amman, Jordan
| | - Alia Alghwiri
- Department of Physical Therapy, School of Rehabilitation Sciences, The University of Jordan, Amman, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Hematology Department, Jordan University Hospital, Amman, Jordan
- Department of Internal Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
14
|
Azizsoltani A, Hatami B, Zali MR, Mahdavi V, Baghaei K, Alizadeh E. Obeticholic acid-loaded exosomes attenuate liver fibrosis through dual targeting of the FXR signaling pathway and ECM remodeling. Biomed Pharmacother 2023; 168:115777. [PMID: 37913732 DOI: 10.1016/j.biopha.2023.115777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023] Open
Abstract
End-stage of liver fibrosis as a precancerous state could lead to cirrhosis and hepatocellular carcinoma which liver transplantation is the only effective treatment. Previous studies have indicated that farnesoid X receptor (FXR) agonists, such as obeticholic acid (OCA) protect against hepatic injuries. However, free OCA administration results in side effects in clinical trials that could be alleviated by applying bio carriers such as MSC-derived exosomes (Exo) with the potential to mimic the biological regenerative effect of their parent cells, as proposed in this study. Loading OCA into the Exo was conducted via water bath sonication. Ex vivo bio distribution studies validated the Exo-loaded OCA more permanently accumulated in the liver. Using CCL4-induced liver fibrosis, we proposed whether Exo isolated from human Warton's Jelly mesenchymal stem cells loaded with a minimal dosage of OCA can facilitate liver recovery. Notably, Exo-loaded OCA exerted additive anti-fibrotic efficacy on histopathological features in CCL4-induced fibrotic mice. Compared to baseline, Exo-mediated delivery OCA results in marked improvements in the fibrotic-related indicators as well as serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) concentrations. Accordingly, the synergistic impact of Exo-loaded OCA as a promising approach is associated with the inactivation of hepatic stellate cells (HSCs), extracellular matrix (ECM) remodeling, and Fxr-Cyp7a1 cascade on CCL4-induced liver fibrosis mice. In conclusion, our data confirmed the additive protective effects of Exo-loaded OCA in fibrotic mice, which suggests a valuable therapeutic strategy to combat liver fibrosis. Furthermore, the use of Exo for accurate drug delivery to the liver tissue can be inspiring.
Collapse
Affiliation(s)
- Arezou Azizsoltani
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahideh Mahdavi
- Iranian Research Institute of Plant Protection (IRIPP), Agricultural Research, Education and Extension Organization (AREEO), Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Effat Alizadeh
- Drug Applied Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Öztürk TM, Özyazgan İ, Sezer G, Yalçın B, Göç R, Ülger M, Özocak H, Yakan B. Investigation of the effects of umbilical cord-derived mesenchymal stem cells and curcumin on Achilles tendon healing - can they act synergistically? ULUS TRAVMA ACIL CER 2023; 29:1218-1227. [PMID: 37889023 PMCID: PMC10771245 DOI: 10.14744/tjtes.2023.04203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/26/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND It is known that curcumin and umbilical cord-derived mesenchymal stem cells (UC-MSCs) positively affect experi-mental tendon injury healing. This study investigated individual effects and potential synergistic effects of using curcumin and UC-MSCs alone and together. METHODS Eighty female Wistar albino rats were randomly divided into five groups: Control, curcumin, sesame oil, MSCs, and Curcumin+MSCs groups. In all rats, punch tendon defect was created in both right and left Achilles tendons. While no additional treatment was applied to the control group, curcumin, sesame oil used as a solvent for curcumin, MSCs, and MSCs and curcumin com-bination were applied locally to the injury site, respectively, in the other groups. Curcumin was solved in sesame oil before application. In each group, half of the animals were euthanized in the post-operative 2nd week while the other half were euthanized in the post-operative 4th week. The right Achilles was used for biomechanical testing, while the left Achilles was used for histological evaluation and immunohistochemical analysis of type I, Type III collagen, and tenomodulin. RESULTS Histologically, significant improvement was observed in the curcumin, MSCs, and Curcumin+ MSCs groups compared to the control Group in the 2nd week. In the 2nd and 4th weeks, Type III collagen was significantly increased in the curcumin group com-pared to the control group. In week 4, tenomodulin increased significantly in the curcumin and MSCs groups compared to the control group. Tendon tensile strength increased significantly in MSCs and Curcumin+MSCs groups compared to the control group in the 4th week. No superiority was observed between the treatment groups regarding their positive effects on recovery. CONCLUSION Locally used curcumin and UC-MSCs showed positive effects that were not superior to each other in the healing of injury caused by a punch in the Achilles tendons of rats. However, synergistic effects on healing were not observed when they were applied together.
Collapse
Affiliation(s)
| | - İrfan Özyazgan
- Department of Plastic Reconstructive and Aesthetic Surgery, Erciyes University, Faculty of Medicine, Kayseri-Türkiye
| | - Gülay Sezer
- Department of Pharmacology, Erciyes University, Faculty of Medicine, Kayseri-Türkiye
| | - Betül Yalçın
- Department of Histology and Embryology, Adıyaman University, Faculty of Medicine, Adıyaman-Türkiye
| | - Rümeysa Göç
- Department of Histology and Embryology, Cumhuriyet University, Faculty of Medicine, Sivas-Türkiye
| | - Menekşe Ülger
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, Kayseri-Türkiye
| | | | - Birkan Yakan
- Department of Histology and Embryology, Erciyes University, Faculty of Medicine, Kayseri-Türkiye
| |
Collapse
|
16
|
Arfianti A, Ulfah, Hutabarat LS, Agnes Ivana G, Budiarti AD, Sahara NS, Saputra NP. Hipoxia modulates the secretion of growth factors of human umbilical cord-derived mesenchymal stem cells. Biomedicine (Taipei) 2023; 13:49-56. [PMID: 37937056 PMCID: PMC10627211 DOI: 10.37796/2211-8039.1416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 03/08/2023] [Indexed: 11/09/2023] Open
Abstract
Background Mesenchymal stem cell (MSC) has great potential as therapies due its ability to regenerate tissue damage and promote tissue homeostasis. Preconditioning of MSC in low oxygen concentration has been shown to affect the therapeutic potential of these cells. This study aimed to compare the characteristic and secretion of trophic factors of MSCs cultured under hypoxia and normoxia. Methods MSCs were isolated from Wharton's jelly of human umbilical cord (UC) tissue by explant method and characterized by flow cytometry. Following 24 h of CoCl2-induced hypoxic culture, the viability and metabolic activity of MSC were analyzed by trypan blue exclusion test and methyl thiazolyl tetrazolium (MTT) assay, respectively. The secretion of hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF) was assessed in conditioned medium using enzyme-linked immunosorbent assay (ELISA) method. Results Flow cytometry analysis showed >99% of the population of MSCs cells were positive for CD73 and CD90 and > 62% were positive for CD105. While the cell viability of MSC was not affected by hypoxic cultured condition, the metabolic activity rate of these cells was decreased under hypoxic conditioning. In line with reduced metabolic activity, hypoxic human UC-derived MSC produced less HGF than normoxic counterpart. Compared to normoxic MSC, hypoxic preconditioned MSC secreted higher level of VEGF in the conditioned medium (p < 0.05). Conclusions Hypoxia decreased the metabolic activity of MSCs associated with the modulation of HGF and VEGF secretions. It is suggested that hypoxia may also affect the therapeutic capacity of MSC cells.
Collapse
Affiliation(s)
- Arfianti Arfianti
- Department of Medical Biology, Faculty of Medicine, Universitas Riau, Pekanbaru, 28133,
Indonesia
| | - Ulfah
- Department of Anatomy, Faculty of Medicine, Universitas Riau, Pekanbaru, 28133,
Indonesia
| | - Leopold S. Hutabarat
- Undergraduate Program, Faculty of Medicine, Universitas Riau, Pekanbaru, 28133,
Indonesia
| | - G Agnes Ivana
- Undergraduate Program, Faculty of Medicine, Universitas Riau, Pekanbaru, 28133,
Indonesia
| | - Anisa D. Budiarti
- Undergraduate Program, Faculty of Medicine, Universitas Riau, Pekanbaru, 28133,
Indonesia
| | - Nabilla S. Sahara
- LONTAR Laboratory, Faculty of Medicine, Universitas Riau, Pekanbaru, 28133,
Indonesia
| | - Nicko P.K. Saputra
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Riau, Pekanbaru, 28133,
Indonesia
| |
Collapse
|
17
|
Zhang Y, Yang Y, Gao X, Gao W, Zhang L. Research progress on mesenchymal stem cells and their exosomes in systemic sclerosis. Front Pharmacol 2023; 14:1263839. [PMID: 37693906 PMCID: PMC10485262 DOI: 10.3389/fphar.2023.1263839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease with an unknown etiology. Clinically, it is characterized by localized or diffuse skin thickening and fibrosis. The pathogenesis of SSc includes microvascular injury, autoimmune-mediated inflammation, and fibroblast activation. These processes interact and contribute to the diverse clinicopathology and presentation of SSc. Given the limited effectiveness and substantial side effects of traditional treatments, the treatment strategy for SSc has several disadvantages. Mesenchymal stem cells (MSCs) are expected to serve as effective treatment options owing to their significant immunomodulatory, antifibrotic, and pro-angiogenic effects. Exosomes, secreted by MSCs via paracrine signaling, mirror the effect of MSCs as well as offer the benefit of targeted delivery, minimal immunogenicity, robust reparability, good safety and stability, and easy storage and transport. This enables them to circumvent the limitations of the MSCs. When using exosomes, it is crucial to consider preparation methods, quality standards, and suitable drug delivery systems, among other technical issues. Therefore, this review aims to summarize the latest research progress on MSCs and exosomes in SSc, offering novel ideas for treating SSc.
Collapse
Affiliation(s)
| | | | | | | | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
18
|
Rebelatto CLK, Boldrini-Leite LM, Daga DR, Marsaro DB, Vaz IM, Jamur VR, de Aguiar AM, Vieira TB, Furman BP, Aguiar CO, Brofman PRS. Quality Control Optimization for Minimizing Security Risks Associated with Mesenchymal Stromal Cell-Based Product Development. Int J Mol Sci 2023; 24:12955. [PMID: 37629136 PMCID: PMC10455270 DOI: 10.3390/ijms241612955] [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: 06/27/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have been considered a therapeutic strategy in regenerative medicine because of their regenerative and immunomodulatory properties. The translation of MSC-based products has some challenges, such as regulatory and scientific issues. Quality control should be standardized and optimized to guarantee the reproducibility, safety, and efficacy of MSC-based products to be administered to patients. The aim of this study was to develop MSC-based products for use in clinical practice. Quality control assays include cell characterization, cell viability, immunogenicity, and cell differentiation; safety tests such as procoagulant tissue factor (TF), microbiological, mycoplasma, endotoxin, genomic stability, and tumorigenicity tests; and potency tests. The results confirm that the cells express MSC markers; an average cell viability of 96.9%; a low expression of HLA-DR and costimulatory molecules; differentiation potential; a high expression of TF/CD142; an absence of pathogenic microorganisms; negative endotoxins; an absence of chromosomal abnormalities; an absence of genotoxicity and tumorigenicity; and T-lymphocyte proliferation inhibition potential. This study shows the relevance of standardizing the manufacturing process and quality controls to reduce variability due to the heterogeneity between donors. The results might also be useful for the implementation and optimization of new analytical techniques and automated methods to improve safety, which are the major concerns related to MSC-based therapy.
Collapse
Affiliation(s)
- Carmen Lúcia Kuniyoshi Rebelatto
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Lidiane Maria Boldrini-Leite
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Debora Regina Daga
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Daniela Boscaro Marsaro
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Isadora May Vaz
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Valderez Ravaglio Jamur
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Alessandra Melo de Aguiar
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute—Fiocruz-Paraná, Curitiba 81350-010, Brazil;
| | - Thalita Bastida Vieira
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
| | - Bianca Polak Furman
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
| | - Cecília Oliveira Aguiar
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
| | - Paulo Roberto Slud Brofman
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| |
Collapse
|
19
|
Stefańska K, Nemcova L, Blatkiewicz M, Żok A, Kaczmarek M, Pieńkowski W, Mozdziak P, Piotrowska-Kempisty H, Kempisty B. Expression Profile of New Marker Genes Involved in Differentiation of Human Wharton's Jelly-Derived Mesenchymal Stem Cells into Chondrocytes, Osteoblasts, Adipocytes and Neural-like Cells. Int J Mol Sci 2023; 24:12939. [PMID: 37629120 PMCID: PMC10455417 DOI: 10.3390/ijms241612939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Wharton's jelly (WJ) contains mesenchymal stem cells (MSCs) exhibiting broad immunomodulatory properties and differentiation capacity, which makes them a promising tool for cellular therapies. Although the osteogenic, chondrogenic and adipogenic differentiation is a gold standard for proper identification of MSCs, it is important to elucidate the exact molecular mechanisms governing these processes to develop safe and efficient cellular therapies. Umbilical cords were collected from healthy, full-term deliveries, for subsequent MSCs (WJ-MSCs) isolation. WJ-MSCs were cultivated in vitro for osteogenic, chondrogenic, adipogenic and neurogenic differentiation. The RNA samples were isolated and the transcript levels were evaluated using NovaSeq platform, which led to the identification of differentially expressed genes. Expression of H19 and SLPI was enhanced in adipocytes, chondrocytes and osteoblasts, and NPPB was decreased in all analyzed groups compared to the control. KISS1 was down-regulated in adipocytes, chondrocytes, and neural-like cells compared to the control. The most of identified genes were already implicated in differentiation of MSCs; however, some genes (PROK1, OCA2) have not yet been associated with initiating final cell fate. The current results indicate that both osteo- and adipo-induced WJ-MSCs share many similarities regarding the most overexpressed genes, while the neuro-induced WJ-MSCs are quite distinctive from the other three groups. Overall, this study provides an insight into the transcriptomic changes occurring during the differentiation of WJ-MSCs and enables the identification of novel markers involved in this process, which may serve as a reference for further research exploring the role of these genes in physiology of WJ-MSCs and in regenerative medicine.
Collapse
Affiliation(s)
- Katarzyna Stefańska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Cellivia 3 S.A., 61-623 Poznan, Poland
| | - Lucie Nemcova
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic
| | - Małgorzata Blatkiewicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Agnieszka Żok
- Division of Philosophy of Medicine and Bioethics, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Gene Therapy Laboratory, Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Wojciech Pieńkowski
- Division of Perinatology and Women’s Diseases, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Paul Mozdziak
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 60177 Brno, Czech Republic
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
20
|
Huang H, Liu X, Wang J, Suo M, Zhang J, Sun T, Zhang W, Li Z. Umbilical cord mesenchymal stem cells for regenerative treatment of intervertebral disc degeneration. Front Cell Dev Biol 2023; 11:1215698. [PMID: 37601097 PMCID: PMC10439242 DOI: 10.3389/fcell.2023.1215698] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
Intervertebral disc degeneration is thought to be a major contributor to low back pain, the etiology of which is complex and not yet fully understood. To compensate for the lack of drug and surgical treatment, mesenchymal stem cells have been proposed for regenerative treatment of intervertebral discs in recent years, and encouraging results have been achieved in related trials. Mesenchymal stem cells can be derived from different parts of the body, among which mesenchymal stem cells isolated from the fetal umbilical cord have excellent performance in terms of difficulty of acquisition, differentiation potential, immunogenicity and ethical risk. This makes it possible for umbilical cord derived mesenchymal stem cells to replace the most widely used bone marrow-derived and adipose tissue derived mesenchymal stem cells as the first choice for regenerating intervertebral discs. However, the survival of umbilical cord mesenchymal stem cells within the intervertebral disc is a major factor affecting their regenerative capacity. In recent years biomaterial scaffolds in tissue engineering have aided the survival of umbilical cord mesenchymal stem cells by mimicking the natural extracellular matrix. This seems to provide a new idea for the application of umbilical cord mesenchymal stem cells. This article reviews the structure of the intervertebral disc, disc degeneration, and the strengths and weaknesses of common treatment methods. We focus on the cell source, cell characteristics, mechanism of action and related experiments to summarize the umbilical cord mesenchymal stem cells and explore the feasibility of tissue engineering technology of umbilical cord mesenchymal stem cells. Hoping to provide new ideas for the treatment of disc degeneration.
Collapse
Affiliation(s)
- Huagui Huang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Liu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinzuo Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Moran Suo
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jing Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tianze Sun
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wentao Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhonghai Li
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| |
Collapse
|
21
|
Hu Z, Jiang Z, Meng S, Liu R, Yang K. Research Progress on the Osteogenesis-Related Regulatory Mechanisms of Human Umbilical Cord Mesenchymal Stem Cells. Stem Cell Rev Rep 2023; 19:1252-1267. [PMID: 36917312 DOI: 10.1007/s12015-023-10521-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2023] [Indexed: 03/16/2023]
Abstract
In recent years, research on human umbilical cord mesenchymal stem cells (hUCMSCs) derived from human umbilical cord tissue has accelerated and entered clinical application research. Compared with mesenchymal stem cells (MSCs) from other sources, hUCMSCs can be extracted from different parts of umbilical cord or from the whole umbilical cord. It has the characteristics of less ethical controversy, high differentiation potential, strong proliferation ability, efficient expansion in vitro, avoiding immune rejection and immune privilege, and avoids the limitations of lack of embryonic stem cells, heterogeneity, ethical and moral constraints. hUCMSCs avoid the need for embryonic stem cell sources, heterogeneity, and ethical and moral constraints. Bone defects are very common in clinical practice, but completely effective bone tissue regeneration treatment is challenging. Currently, autologous bone transplantation and allogeneic bone transplantation are main treatment approaches in clinical work, but each has different shortcomings, such as limited sources, invasiveness, immune rejection and insufficient osteogenic ability. Therefore, to solve the bottleneck of bone tissue regeneration and repair, a great amount of research has been carried out to explore the clinical advantages of hUCMSCs as seed cells to promote osteogenesis.However, the regulation of osteogenic differentiation of hUCMSCs is an extremely complex process. Although a large number of studies have demonstrated that the role of hUCMSCs in enhancing local bone regeneration and repair through osteogenic differentiation and transplantation into the body involves multiple signaling pathways, there is no relevant article that summarize the findings. This article discusses the osteogenesis-related regulatory mechanisms of hUCMSCs, summarizes the currently known related mechanisms, and speculates on the possible signals.
Collapse
Affiliation(s)
- Zhengqi Hu
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Zhiliang Jiang
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Shengzi Meng
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Rong Liu
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Kun Yang
- Department of Periodontology, Hospital of Stomatology, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
22
|
Romano IR, D'Angeli F, Vicario N, Russo C, Genovese C, Lo Furno D, Mannino G, Tamburino S, Parenti R, Giuffrida R. Adipose-Derived Mesenchymal Stromal Cells: A Tool for Bone and Cartilage Repair. Biomedicines 2023; 11:1781. [PMID: 37509421 PMCID: PMC10376676 DOI: 10.3390/biomedicines11071781] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
The osteogenic and chondrogenic differentiation ability of adipose-derived mesenchymal stromal cells (ASCs) and their potential therapeutic applications in bone and cartilage defects are reported in this review. This becomes particularly important when these disorders can only be poorly treated by conventional therapeutic approaches, and tissue engineering may represent a valuable alternative. Being of mesodermal origin, ASCs can be easily induced to differentiate into chondrocyte-like and osteocyte-like elements and used to repair damaged tissues. Moreover, they can be easily harvested and used for autologous implantation. A plethora of ASC-based strategies are being developed worldwide: they include the transplantation of freshly harvested cells, in vitro expanded cells or predifferentiated cells. Moreover, improving their positive effects, ASCs can be implanted in combination with several types of scaffolds that ensure the correct cell positioning; support cell viability, proliferation and migration; and may contribute to their osteogenic or chondrogenic differentiation. Examples of these strategies are described here, showing the enormous therapeutic potential of ASCs in this field. For safety and regulatory issues, most investigations are still at the experimental stage and carried out in vitro and in animal models. Clinical applications have, however, been reported with promising results and no serious adverse effects.
Collapse
Affiliation(s)
- Ivana Roberta Romano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Floriana D'Angeli
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Cristina Russo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Carlo Genovese
- Faculty of Medicine and Surgery, "Kore" University of Enna, 94100 Enna, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Giuliana Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy
| | - Serena Tamburino
- Chi.Pla Chirurgia Plastica, Via Suor Maria Mazzarello, 54, 95128 Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| |
Collapse
|
23
|
Russo E, Corrao S, Di Gaudio F, Alberti G, Caprnda M, Kubatka P, Kruzliak P, Miceli V, Conaldi PG, Borlongan CV, La Rocca G. Facing the Challenges in the COVID-19 Pandemic Era: From Standard Treatments to the Umbilical Cord-Derived Mesenchymal Stromal Cells as a New Therapeutic Strategy. Cells 2023; 12:1664. [PMID: 37371134 DOI: 10.3390/cells12121664] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which counts more than 650 million cases and more than 6.6 million of deaths worldwide, affects the respiratory system with typical symptoms such as fever, cough, sore throat, acute respiratory distress syndrome (ARDS), and fatigue. Other nonpulmonary manifestations are related with abnormal inflammatory response, the "cytokine storm", that could lead to a multiorgan disease and to death. Evolution of effective vaccines against SARS-CoV-2 provided multiple options to prevent the infection, but the treatment of the severe forms remains difficult to manage. The cytokine storm is usually counteracted with standard medical care and anti-inflammatory drugs, but researchers moved forward their studies on new strategies based on cell therapy approaches. The perinatal tissues, such as placental membranes, amniotic fluid, and umbilical cord derivatives, are enriched in mesenchymal stromal cells (MSCs) that exert a well-known anti-inflammatory role, immune response modulation, and tissue repair. In this review, we focused on umbilical-cord-derived MSCs (UC-MSCs) used in in vitro and in vivo studies in order to evaluate the weakening of the severe symptoms, and on recent clinical trials from different databases, supporting the favorable potential of UC-MSCs as therapeutic strategy.
Collapse
Affiliation(s)
- Eleonora Russo
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| | - Simona Corrao
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy
| | | | - Giusi Alberti
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University, University Hospital Bratislava, 81499 Bratislava, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03649 Martin, Slovakia
| | - Peter Kruzliak
- Research and Development Services, Pradlacka 18, 61300 Brno, Czech Republic
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy
| | - Cesario Venturina Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Giampiero La Rocca
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
24
|
Stefańska K, Nemcova L, Blatkiewicz M, Pieńkowski W, Ruciński M, Zabel M, Mozdziak P, Podhorska-Okołów M, Dzięgiel P, Kempisty B. Apoptosis Related Human Wharton's Jelly-Derived Stem Cells Differentiation into Osteoblasts, Chondrocytes, Adipocytes and Neural-like Cells-Complete Transcriptomic Assays. Int J Mol Sci 2023; 24:10023. [PMID: 37373173 DOI: 10.3390/ijms241210023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) exhibit multilineage differentiation potential, adhere to plastic, and express a specific set of surface markers-CD105, CD73, CD90. Although there are relatively well-established differentiation protocols for WJ-MSCs, the exact molecular mechanisms involved in their in vitro long-term culture and differentiation remain to be elucidated. In this study, the cells were isolated from Wharton's jelly of umbilical cords obtained from healthy full-term deliveries, cultivated in vitro, and differentiated towards osteogenic, chondrogenic, adipogenic and neurogenic lineages. RNA samples were isolated after the differentiation regimen and analyzed using an RNA sequencing (RNAseq) assay, which led to the identification of differentially expressed genes belonging to apoptosis-related ontological groups. ZBTB16 and FOXO1 were upregulated in all differentiated groups as compared to controls, while TGFA was downregulated in all groups. In addition, several possible novel marker genes associated with the differentiation of WJ-MSCs were identified (e.g., SEPTIN4, ITPR1, CNR1, BEX2, CD14, EDNRB). The results of this study provide an insight into the molecular mechanisms involved in the long-term culture in vitro and four-lineage differentiation of WJ-MSCs, which is crucial to utilize WJ-MSCs in regenerative medicine.
Collapse
Affiliation(s)
- Katarzyna Stefańska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Cellivia 3 S.A., 61-623 Poznan, Poland
| | - Lucie Nemcova
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic
| | - Małgorzata Blatkiewicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Wojciech Pieńkowski
- Division of Perinatology and Women's Diseases, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Marcin Ruciński
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Paul Mozdziak
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Marzenna Podhorska-Okołów
- Division of Ultrastructural Research, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 60177 Brno, Czech Republic
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
25
|
Truong NC, Phan TNM, Huynh NT, Pham KD, Van Pham P. Interferon-Gamma Increases the Immune Modulation of Umbilical Cord-Derived Mesenchymal Stem Cells but Decreases Their Chondrogenic Potential. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023. [PMID: 37291444 DOI: 10.1007/5584_2023_776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
INTRODUCTION The pro-inflammatory cytokine interferon-gamma (IFN-γ) is reported to be an agent that boosts the immune modulation of mesenchymal stem cells (MSCs). However, the effects of IFN-γ on the chondrogenic potential of treated MSCs have not been evaluated in depth. This study aimed to evaluate the effects of IFN-γ on the immune modulation and chondrogenic potential of human umbilical cord-derived MSCs (hUC-MSCs). METHODS UC-MSCs were isolated and expanded following published protocols. They were characterized as MSCs before their use in further experiments. The UC-MSCs were treated with IFN-γ at 10 ng/mL for 48 h. Changes in phenotype were investigated based on changes in MSC markers, immunomodulatory genes (TGF-β, IL-4, and IDO) for immune modulation, and cartilage-related genes during the induction of differentiation (Col1a2, Col2a1, Sox9, Runx2, and Acan) for chondrogenic potential. RESULTS IFN-γ-treated UC-MSCs maintained MSC markers and exhibited decreased expression of transcriptional regulatory factors in chondrogenesis (Sox9 and Runx2) and the extracellular matrix-specific genes Col1a2 and Acan but not Col2a1 compared to non-treated cells (p < 0.05). Furthermore, the immunomodulatory capability of IFN-γ-treated UC-MSCs was clearly revealed through their increased expression of IDO and IL-4 and decreased expression of TGF-β compared to non-treated cells (p < 0.05). CONCLUSION This study demonstrated that UC-MSCs treated with IFN-γ at 10 ng/mL had reduced expression of chondrocyte-specific genes; however, they maintained multi-lineage differentiation and exhibited immunomodulatory properties.
Collapse
Affiliation(s)
- Nhat Chau Truong
- Stem Cell Institute, University of Science, Ho Chi Minh City, Viet Nam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Thu Ngoc-Minh Phan
- Stem Cell Institute, University of Science, Ho Chi Minh City, Viet Nam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Nhi Thao Huynh
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam
- Laboratory of Stem Cell Research and Application, University of Science, Ho Chi Minh City, Viet Nam
| | - Khuong Duy Pham
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam
- Laboratory of Stem Cell Research and Application, University of Science, Ho Chi Minh City, Viet Nam
| | - Phuc Van Pham
- Stem Cell Institute, University of Science, Ho Chi Minh City, Viet Nam.
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam.
- Laboratory of Cancer Research, University of Science, Ho Chi Minh City, Viet Nam.
| |
Collapse
|
26
|
Teoh PL, Mohd Akhir H, Abdul Ajak W, Hiew VV. Human Mesenchymal Stromal Cells Derived from Perinatal Tissues: Sources, Characteristics and Isolation Methods. Malays J Med Sci 2023; 30:55-68. [PMID: 37102047 PMCID: PMC10125235 DOI: 10.21315/mjms2023.30.2.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 04/22/2022] [Indexed: 04/28/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) derived from perinatal tissues have become indispensable sources for clinical applications due to their superior properties, ease of accessibility and minimal ethical concerns. MSCs isolated from different placenta (PL) and umbilical cord (UC) compartments exhibit great potential for stem cell-based therapies. However, their biological activities could vary due to tissue origins and differences in differentiation potentials. This review provides an overview of MSCs derived from various compartments of perinatal tissues, their characteristics and current isolation methods. Factors affecting the yield and purity of MSCs are also discussed as they are important to ensure consistent and unlimited supply for regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Peik Lin Teoh
- Biotechnology Research Institute, Universiti Malaysia Sabah, Sabah, Malaysia
| | | | - Warda Abdul Ajak
- Biotechnology Research Institute, Universiti Malaysia Sabah, Sabah, Malaysia
| | - Vun Vun Hiew
- Biotechnology Research Institute, Universiti Malaysia Sabah, Sabah, Malaysia
| |
Collapse
|
27
|
Intravenously Administered Human Umbilical Cord-Derived Mesenchymal Stem Cell (HucMSC) Improves Cardiac Performance following Infarction via Immune Modulation. Stem Cells Int 2023; 2023:6256115. [PMID: 36970596 PMCID: PMC10038737 DOI: 10.1155/2023/6256115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/19/2023] Open
Abstract
Overactive inflammatory responses contribute to progressive cardiac dysfunction after myocardial infarction (MI). Mesenchymal stem cell (MSC) has generated significant interest as potent immune modulators that can regulate excessive immune responses. We hypothesized that intravenous (iv) administration of human umbilical cord-derived MSC (HucMSC) exerts systemic and local anti-inflammation effects, leading to improved heart function after MI. In murine MI models, we confirmed that single iv administration of HucMSC (
) improved cardiac performance and prevented adverse remodeling after MI. A small proportion of HucMSC is trafficked to the heart, preferentially in the infarcted region. HucMSC administration increased CD3+ T cell proportion in the periphery while decreased T cell proportion in both infarcted heart and mediastinal lymph nodes (med-LN) at 7-day post-MI, indicating a systematic and local T cell interchange mediated by HucMSC. The inhibitory effects of HucMSC on T cell infiltration in the infarcted heart and med-LN sustained to 21-day post-MI. Our findings suggested that iv administration of HucMSC fostered systemic and local immunomodulatory effects that contributed to the improvement of cardiac performance after MI.
Collapse
|
28
|
Nagai T. Systemic Transfusion of Human Umbilical Cord-Derived Mesenchymal Stromal Cells - Toward Less Invasive and More Effective Cell Therapy for Severe Peripheral Arterial Disease. Circ J 2023; 87:421-423. [PMID: 36273916 DOI: 10.1253/circj.cj-22-0631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Toshio Nagai
- Department of Cardiology, International University of Health and Welfare, Narita Hospital
| |
Collapse
|
29
|
Soleimani M, Masoumi A, Momenaei B, Cheraqpour K, Koganti R, Chang AY, Ghassemi M, Djalilian AR. Applications of mesenchymal stem cells in ocular surface diseases: sources and routes of delivery. Expert Opin Biol Ther 2023; 23:509-525. [PMID: 36719365 PMCID: PMC10313829 DOI: 10.1080/14712598.2023.2175605] [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: 09/26/2022] [Accepted: 01/30/2023] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) are novel, promising agents for treating ocular surface disorders. MSCs can be isolated from several tissues and delivered by local or systemic routes. They produce several trophic factors and cytokines, which affect immunomodulatory, transdifferentiating, angiogenic, and pro-survival pathways in their local microenvironment via paracrine secretion. Moreover, they exert their therapeutic effect through a contact-dependent manner. AREAS COVERED In this review, we discuss the characteristics, sources, delivery methods, and applications of MSCs in ocular surface disorders. We also explore the potential application of MSCs to inhibit senescence at the ocular surface. EXPERT OPINION Therapeutic application of MSCs in ocular surface disorders are currently under investigation. One major research area is corneal epitheliopathies, including chemical or thermal burns, limbal stem cell deficiency, neurotrophic keratopathy, and infectious keratitis. MSCs can promote corneal epithelial repair and prevent visually devastating sequelae of non-healing wounds. However, the optimal dosages and delivery routes have yet to be determined and further clinical trials are needed to address these fundamental questions.
Collapse
Affiliation(s)
- Mohammad Soleimani
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Masoumi
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bita Momenaei
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Kasra Cheraqpour
- Eye Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Arthur Y Chang
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Mahmoud Ghassemi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
30
|
Mokhtari T, Shayan M, Rezaei Rashnudi A, Hassanzadeh G, Mehran Nia K. Wharton's jelly mesenchymal stem cells attenuate global hypoxia-induced learning and memory impairment via preventing blood-brain barrier breakdown. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:1053-1060. [PMID: 37605722 PMCID: PMC10440140 DOI: 10.22038/ijbms.2023.70137.15250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/30/2023] [Indexed: 08/23/2023]
Abstract
Objectives Intracerebroventricular (ICV) injections of mesenchymal stem cells (MSCs) may improve the function and structure of blood-brain barrier (BBB), possibly by preserving the BBB integrity. This study examined the impact of Wharton's jelly (WJ)-MSCs on cognitive dysfunction and BBB disruption following a protracted hypoxic state. Materials and Methods Twenty-four male Wistar rats were randomly studied in four groups: Control (Co): Healthy animals, Sham (Sh): Rats were placed in the cage without hypoxia induction and with ICV injection of vehicle, Hypoxic (Hx)+vehicle: Hypoxic rats with ICV injection of vehicle (5 μl of PBS), and Hx+MSCs: Hypoxic rats with ICV injection of MSCs. Spatial learning and memory were evaluated one week after WJ-MSCs injection, and then animals were sacrificed for molecular research. Results Hypoxia increased latency and lowered the time and distance required reaching the target quarter, according to the findings. Furthermore, hypoxic rats had lower gene expression and protein levels of hippocampus vascular endothelial (VE)-cadherin, claudin 5, and tricellulin gene expression than Co and Sh animals (P<0.05). Finally, administering WJ-MSCs after long-term hypoxia effectively reversed the cognitive deficits and prevented the BBB breakdown via the upregulation of VE-cadherin, claudin 5, and tricellulin genes (P<0.05). Conclusion These findings suggest that prolonged hypoxia induces spatial learning and memory dysfunction and increases BBB disruption, the potential mechanism of which might be via reducing VE-cadherin, claudin 5, and tricellulin genes. Hence, appropriate treatment with WJ-MSCs could reverse ischemia adverse effects and protect the BBB integrity following prolonged hypoxia.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| | - Maryam Shayan
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kobra Mehran Nia
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Jiang Z, Wang J, Sun G, Feng M. BDNF-modified human umbilical cord mesenchymal stem cells-derived dopaminergic-like neurons improve rotation behavior of Parkinson's disease rats through neuroprotection and anti-neuroinflammation. Mol Cell Neurosci 2022; 123:103784. [PMID: 36228967 DOI: 10.1016/j.mcn.2022.103784] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease still without any cure. Brain-derived neurotrophic factor (BDNF) has shown therapeutic potential in PD, which is limited by its short half-life and inability to penetrate the blood-brain barrier. Stem cells not only present migration, differentiation and neurotrophy characteristics, but also can be used as delivery vectors for BDNF. This study aimed to investigate the therapeutic effects and possible mechanisms of BDNF-modified human umbilical cord mesenchymal stem cells (hUC-MSCs)-derived dopaminergic (DAergic)-like neurons in the PD rats. Results showed that transplantation of BDNF-modified hUC-MSCs-derived DAergic-like neurons improved the apomorphine induced rotation behavior of PD rats, increased the dopamine concentration and the expression of glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule-1 (Iba-1) in the striatum, promoted the expression of tyrosine hydroxylase (TH), nuclear receptor-related factor 1 (Nurr1), pituitary homeobox 3 (Pitx3), BDNF, tyrosine kinase B (TrkB), phosphatidylinositol-3-hydroxykinase (PI3K), phosphorylated protein kinase B (p-Akt), heat shock protein 60 (Hsp60), toll-like receptor 4 (TLR4) and myeloid differentiation factor 88 (MyD88) and inhibited the neural apoptosis in the substantia nigra (SN) and striatum. Results suggest that BDNF-modified hUC-MSCs-derived DAergic-like neurons improve the rotation of PD rats might through neuroprotection and anti-neuroinflammation by regulating the BDNF-TrkB-PI3K/Akt and Hsp60-TLR4/MyD88 signaling pathways, respectively.
Collapse
Affiliation(s)
- Zhi Jiang
- Department of Geriatrics, The Second Affiliated Hospital, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing 210011, China; Department of Neurology, The Second People's Hospital of NanTong, Nantong 226006, China
| | - Jie Wang
- Department of Geriatrics, The Second Affiliated Hospital, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing 210011, China; Department of Neurology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China
| | - Gaohui Sun
- Department of Geriatrics, The Second Affiliated Hospital, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing 210011, China
| | - Meijiang Feng
- Department of Geriatrics, The Second Affiliated Hospital, Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing 210011, China.
| |
Collapse
|
32
|
Yudintceva N, Mikhailova N, Fedorov V, Samochernych K, Vinogradova T, Muraviov A, Shevtsov M. Mesenchymal Stem Cells and MSCs-Derived Extracellular Vesicles in Infectious Diseases: From Basic Research to Clinical Practice. Bioengineering (Basel) 2022; 9:662. [PMID: 36354573 PMCID: PMC9687734 DOI: 10.3390/bioengineering9110662] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/30/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are attractive in various fields of regenerative medicine due to their therapeutic potential and complex unique properties. Basic stem cell research and the global COVID-19 pandemic have given impetus to the development of cell therapy for infectious diseases. The aim of this review was to systematize scientific data on the applications of mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (MSC-EVs) in the combined treatment of infectious diseases. Application of MSCs and MSC-EVs in the treatment of infectious diseases has immunomodulatory, anti-inflammatory, and antibacterial effects, and also promotes the restoration of the epithelium and stimulates tissue regeneration. The use of MSC-EVs is a promising cell-free treatment strategy that allows solving the problems associated with the safety of cell therapy and increasing its effectiveness. In this review, experimental data and clinical trials based on MSCs and MSC-EVs for the treatment of infectious diseases are presented. MSCs and MSC-EVs can be a promising tool for the treatment of various infectious diseases, particularly in combination with antiviral drugs. Employment of MSC-derived EVs represents a more promising strategy for cell-free treatment, demonstrating a high therapeutic potential in preclinical studies.
Collapse
Affiliation(s)
- Natalia Yudintceva
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Natalia Mikhailova
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
| | - Viacheslav Fedorov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Konstantin Samochernych
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Vinogradova
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Health of the Russian Federation, St. Petersburg 191036, Russia
| | - Alexandr Muraviov
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Health of the Russian Federation, St. Petersburg 191036, Russia
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| |
Collapse
|
33
|
Garg K, Zilate S. Umbilical Cord-Derived Mesenchymal Stem Cells for the Treatment of Infertility Due to Premature Ovarian Failure. Cureus 2022; 14:e30529. [PMID: 36415442 PMCID: PMC9674197 DOI: 10.7759/cureus.30529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
Females belonging to the reproductive age group may face challenges regarding infertility or miscarriage due to conditions such as premature ovarian failure (POF). It is the condition that happens when a female's ovaries stop working before she is 40. The majority of the causes of POF cases are idiopathic. Other reasons include genetic disorders (Turner's syndrome, bone morphogenetic protein 15 (BMP15) mutation, galactosemia, mutation of forkhead box protein L2 (FOXL2), growth differentiation factor-9 (GDF9), mutation of luteinizing hormone (LH) and follicle-stimulating hormone receptors (FSHR), etc.), enzymatic mutation such as aromatase, autoimmune disorders (Addison's disease, vitiligo, systemic lupus erythematosus, myasthenia gravis, autoimmune thyroiditis, autoimmune polyglandular syndrome, etc.), vaccination, and environmental factors (cigarette smoking, toxins, and infections). Many attempts have been made to treat POF by various methods. Some of the methods of treatment include hormone replacement therapy (HRT), melatonin therapy, dehydroepiandrosterone (DHEA) therapy, and stem cell therapy. Stem cell therapy has proven to be the most efficient form for treating POF as compared to all other options. Umbilical cord-derived mesenchymal stem cells (UC-MSCs) are the best among the other sources of mesenchymal stem cells (MSCs) for the treatment of POF as they have a painless extraction procedure. They have a tremendous capacity for self-repair and regeneration, which helps them in restoring degenerated ovaries. This review includes information on the causes of POF, its efficacious therapeutic approaches, and the impact of transplantation of human umbilical cord mesenchymal stem cells (hUCMSCs) as an option for the therapy of POF. Numerous studies conducted on stem cell therapy prove that it is an effective approach for the treatment of sterility.
Collapse
|
34
|
Effects of Human Deciduous Dental Pulp-Derived Mesenchymal Stem Cell-Derived Conditioned Medium on the Metabolism of HUVECs, Osteoblasts, and BMSCs. Cells 2022; 11:cells11203222. [PMID: 36291089 PMCID: PMC9600042 DOI: 10.3390/cells11203222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we assessed the effects of human deciduous dental pulp-derived mesenchymal stem cell-derived conditioned medium (SHED-CM) on the properties of various cell types. The effects of vascular endothelial growth factor (VEGF) in SHED-CM on the luminal architecture, proliferative ability, and angiogenic potential of human umbilical vein endothelial cells (HUVECs) were determined. We also investigated the effects of SHED-CM on the proliferation of human-bone-marrow mesenchymal stem cells (hBMSCs) and mouse calvarial osteoblastic cells (MC3T3-E1) as well as the expression of ALP, OCN, and RUNX2. The protein levels of ALP were examined using Western blot analysis. VEGF blockade in SHED-CM suppressed the proliferative ability and angiogenic potential of HUVECs, indicating that VEGF in SHED-CM contributes to angiogenesis. The culturing of hBMSCs and MC3T3-E1 cells with SHED-CM accelerated cell growth and enhanced mRNA expression of bone differentiation markers. The addition of SHED-CM enhanced ALP protein expression in hBMSCs and MT3T3-E1 cells compared with that of the 0% FBS group. Furthermore, SHED-CM promoted the metabolism of HUVECs, MC3T3-E1 cells, and hBMSCs. These findings indicate the potential benefits of SHED-CM in bone tissue regeneration.
Collapse
|
35
|
Shin JY, Kim DY, Lee J, Shin YJ, Kim YS, Lee PH. Priming mesenchymal stem cells with α-synuclein enhances neuroprotective properties through induction of autophagy in Parkinsonian models. STEM CELL RESEARCH & THERAPY 2022; 13:483. [PMID: 36153562 PMCID: PMC9509608 DOI: 10.1186/s13287-022-03139-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 08/14/2022] [Indexed: 11/30/2022]
Abstract
Background Mesenchymal stem cells (MSCs) may be one of candidates for disease-modifying therapy in Parkinsonian diseases. As knowledge regarding the therapeutic properties of MSCs accumulates, some obstacles still remain to be overcome, especially, successful clinical translation requires the development of culture systems that mimic the natural MSC niche, while allowing clinical-scale cell expansion without compromising quality and function of the cells. In recent years, priming approaches using bioactive peptide or complement components have been investigated to enhance the therapeutic potential of MSCs. Methods We investigated an innovative priming strategy by conditioning the MSCs with α-synuclein (α-syn). To induce priming, MSCs were treated with different concentrations of α-syn and various time course. We evaluated whether α-syn enhances stemness properties of MSCs and priming MSCs with α-syn would modulate autophagy-related gene expression profiles. Results Treatment of naïve MSCs with α-syn upregulated transcriptional factors responsible for regulation of stemness, which was associated with the elevated expression of genes involved in glycolysis and cell re-programming. Primed MSCs with α-syn enhanced the expression of autophagy-regulating miRNA, and exosomes derived from primed MSCs were packed with autophagy-associated miRNA. In α-syn-overexpressing neuronal cells, primed MSCs with α-syn enhanced neuronal viability relative to naïve MSCs, through the induction of autophagy and lysosome activity. Animal study using an α-syn-overexpressing mice showed that the pro-survival effect of MSCs on dopaminergic neurons was more prominent in primed MSC-treated mice compared with that in naïve MSC-treated mice. Conclusions The present data suggest that MSC priming with α-syn exerts neuroprotective effects through augmented stemness and possibly the enhancement of autophagy-mediated α-syn modulation in Parkinsonian models. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03139-w.
Collapse
|
36
|
Manufacture and Quality Control of Human Umbilical Cord-Derived Mesenchymal Stem Cell Sheets for Clinical Use. Cells 2022; 11:cells11172732. [PMID: 36078137 PMCID: PMC9454431 DOI: 10.3390/cells11172732] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Human umbilical cord-derived mesenchymal stem cell (UC−MSC) sheets have attracted much attention in cell therapy. However, the culture media and coating matrix used for the preparation of UC−MSC sheets have not been safe enough to comply with current clinical drug standards. Moreover, the UC−MSC sheet preservation systems developed before did not comply with Good Manufacturing Practice (GMP) regulations. In this study, the culture medium and coating matrix were developed for UC−MSC sheet production to comply with clinical drug standards. Additionally, the GMP-compliant preservation solution and method for the UC−MSC sheet were developed. Then, quality standards of the UC−MSC sheet were formulated according to national and international regulations for drugs. Finally, the production process of UC−MSC sheets on a large scale was standardized, and three batches of trial production were conducted and tested to meet the established quality standards. This research provides the possibility for clinical trials of UC−MSC sheet products in the development stage of new drugs and lays the foundation for industrial large-scale production after the new drug is launched.
Collapse
|
37
|
Lee AY, Jang KH, Jo CH. Minimal Cube Explant Provides Optimal Isolation Condition of Mesenchymal Stem Cells from Umbilical Cord. Tissue Eng Regen Med 2022; 19:793-807. [PMID: 35325405 PMCID: PMC9294096 DOI: 10.1007/s13770-022-00440-w] [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: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Enzymatic digestion and explant method have been widely used for isolating umbilical cord-derived mesenchymal stem cells (UC MSCs), although there is still a strong need for robust protocols for optimal isolation for large-scale stem cell banks. This study aims to establish an explant method for clinical scale production of MSCs from human UC tissue and to characterize UC MSCs isolated and cultured with the explant method. METHODS UC MSCs were isolated by enzymatic digestion, minimal cube explant (MCE) 1-2, MCE 2-4, and MCE 10 and cultured, respectively. Also, human antibody array and basic fibroblast growth factor (bFGF) secretion in conditioned medium (CM) was analyzed. The cells were evaluated initial cell number, colony forming unit-fibroblast (CFU-F), proliferation capacity, CD marker expression, and multi-lineage differentiation. SA-β-gal assay as well as expression of p16, p21 and p53 was performed by RT-PCR. RESULTS MCE 2-4 is the most optimized method for isolation of small umbilical cord-derived fast proliferating cells (smumf cells) with the greatest number. MCE 2-4 had the highest secretion of various bioactive factors including bFGF. The MCE 2-4 provided significantly higher CD146 expression than enzymatic digestion, and that expression was maintained until P20. The gene expression of p16, p21, and p53 of smumf cells did not change until P10 and SA-β-gal activity did not increase until P14. CONCLUSION This study demonstrated that MCE 2-4 provided an optimal environment to isolate MSCs with quantity and quality from human whole UC tissue through secretion of various bioactive factors inherent to UC.
Collapse
Affiliation(s)
- Ah-Young Lee
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea
| | - Kwi-Hoon Jang
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea
| | - Chris Hyunchul Jo
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea.
- Department of Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
38
|
Mirza A, Khan I, Qazi REM, Salim A, Husain M, Herzig JW. Role of Wnt/β-catenin pathway in cardiac lineage commitment of human umbilical cord mesenchymal stem cells by zebularine and 2'-deoxycytidine. Tissue Cell 2022; 77:101850. [PMID: 35679684 DOI: 10.1016/j.tice.2022.101850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 12/26/2022]
Abstract
Wnt/β-catenin, a highly conserved signaling pathway, is involved in determining cell fate. During heart development, Wnt signaling controls specification, proliferation and differentiation of cardiac cells. This study is aimed to investigate the role of Wnt/β-catenin signaling in cardiac lineage commitment of human umbilical cord mesenchymal stem cells (hUCMSCs) after treatment with demethylating agents, zebularine and 2'-deoxycytidine (2-DC). hUCMSCs were treated with 20 µM zebularine or 2-DC for 24 h and cultured for 14 days. Control and treated MSCs were analyzed for cardiac lineage commitment at gene and protein levels. Significant upregulation of early and late cardiac markers, GATA4, Nkx2.5, cardiac myosin heavy chain (cMHC), α-actinin, cardiac troponin T (cTnT) and cardiac troponin I (cTnI) was observed in treated MSCs as compared to the untreated control. We also analyzed gene expression of key Wnt/β-catenin signaling molecules in cultures of treated and untreated hUCMSCs at 24 h, and days 3, 7 and 14. The pattern of mRNA gene expression showed that Wnt/β-catenin signaling is regulated during cardiac lineage commitment of hUCMSCs in a time-dependent manner, with the pathway being activated early but inhibited later in cardiac development. Findings of this study can lead us to identify more specific and effective strategies for cardiac lineage commitment.
Collapse
Affiliation(s)
- Amber Mirza
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Rida-E-Maria Qazi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | | | | |
Collapse
|
39
|
Wu M, Liu F, Yan L, Huang R, Hu R, Zhu J, Li S, Long C. MiR-145-5p restrains chondrogenic differentiation of synovium-derived mesenchymal stem cells by suppressing TLR4. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2022; 41:625-642. [PMID: 35403567 DOI: 10.1080/15257770.2022.2057535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Osteoarthritis (OA) is a progressive degeneration of articular cartilage with involvement of synovial membrane, and subchondral bone. Recently, cell-based therapies, including the application of stem cells such as mesenchymal stem cells (MSCs), have been introduced for restoration of the articular cartilage. Toll-like receptors (TLRs) were reported to participate in OA progression and MSC chondrogenesis. Here, the role and molecular mechanism of toll like receptor 4 (TLR4) in chondrogenic differentiation of synovium-derived MSCs (SMSCs) were investigated. Molecular markers (CD44, CD90, CD45 and CD14) on SMSC surfaces were identified by flow cytometry. Multi-potential differentiation capacities of SMSCs for chondrogenesis, adipogenesis and osteogenesis were examined by Alcian blue, oil red O and Alizarin red staining, respectively. TLR4 and miR-145-5p levels in SMSCs were assessed using RT-qPCR. The protein expression of TGFB3, Col II, SOX9 and Aggrecan in SMSCs was tested by western blotting. Cytokine secretions were analyzed with ELISA for IL-1β and IL-6. Intracellular NAD+ content and NAD+/NADH ratio were assessed. The interaction between miR-145-5p and TLR4 was confirmed by RNA pulldown and luciferase reporter assays. In this study, SMSCs were identified to have immunophenotypic characteristics of MSCs. TLR4 knockdown inhibited chondrogenic and osteogenic differentiation of SMSCs. Mechanistically, TLR4 was targeted by miR-145-5p in SMSCs. Moreover, TLR4 elevation offset the inhibitory impact of miR-145-5p upregulation on chondrogenic differentiation of SMSCs. Overall, miR-145-5p restrains chondrogenesis of SMSCs by suppressing TLR4.
Collapse
Affiliation(s)
- Mingzheng Wu
- Department of Orthopedics, Wuhan Fourth Hospital (Wuhan Puai Hospital), Wuhan, Hubei, China
| | - Feng Liu
- Department of Orthopedics, Wuhan Fourth Hospital (Wuhan Puai Hospital), Wuhan, Hubei, China
| | - Li Yan
- Department of Orthopedics, Wuhan Fourth Hospital (Wuhan Puai Hospital), Wuhan, Hubei, China
| | - Ruokun Huang
- Department of Orthopedics, Wuhan Fourth Hospital (Wuhan Puai Hospital), Wuhan, Hubei, China
| | - Rui Hu
- Department of Orthopedics, Wuhan Fourth Hospital (Wuhan Puai Hospital), Wuhan, Hubei, China
| | - Jin Zhu
- Department of Orthopedics, Wuhan Fourth Hospital (Wuhan Puai Hospital), Wuhan, Hubei, China
| | - Shanqing Li
- Department of Orthopedics, Wuhan Fourth Hospital (Wuhan Puai Hospital), Wuhan, Hubei, China
| | - Chao Long
- Department of Radiology, Wuhan Fourth Hospital (Wuhan Puai Hospital), Wuhan, Hubei, China
| |
Collapse
|
40
|
Sharma A, Kulkarni R, Sane H, Awad N, Bopardikar A, Joshi A, Baweja S, Joshi M, Vishwanathan C, Gokulchandran N, Badhe P, Khan M, Paranjape A, Kulkarni P, Methal AK. Phase 1 clinical trial for intravenous administration of mesenchymal stem cells derived from umbilical cord and placenta in patients with moderate COVID-19 virus pneumonia: results of stage 1 of the study. AMERICAN JOURNAL OF STEM CELLS 2022; 11:37-55. [PMID: 35873716 PMCID: PMC9301142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/10/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE Mesenchymal stem cells can serve as a therapeutic option for COVID-19. Their immunomodulatory and anti-inflammatory properties can regulate the exaggerated inflammatory response and promote recovery of lung damage. METHOD Phase-1, single-centre open-label, prospective clinical trial was conducted to evaluate the safety and efficacy of intravenous administration of mesenchymal stem cells derived from umbilical cord and placenta in moderate COVID-19. The study was done in 2 stages with total 20 patients. Herein, the results of stage 1 including first 10 patients receiving 100 million cells on day 1 and 4 with a follow up of 6 months have been discussed. RESULTS No adverse events were recorded immediately after the administration of MSCs or on follow up. There was no deterioration observed in clinical, laboratory and radiological parameters. All symptoms of the study group resolved within 10 days. Levels of inflammatory biomarkers such as NLR, CRP, IL6, ferritin and D-dimer improved in all patients after intervention along with improved oxygenation demonstrated by improvement in the SpO2/FiO2 ratio and PaO2/FiO2 ratio. None of the patients progressed to severe stage. 9 out of 10 patients were discharged within 9 days of their admission. Improvements were noted in chest x-ray and chest CT scan scores at day 7 in most patients. No post-covid fibrosis was observed on chest CT 28 days after intervention and Chest X ray after 6 months of the intervention. CONCLUSION Administration of 100 million mesenchymal stem cells in combination with standard treatment was found to be safe and resulted in prevention of the cytokine storm, halting of the disease progression and acceleration of recovery in moderate COVID-19. This clinical trial has been registered with the Clinical Trial Registry- India (CTRI) as CTRI/2020/08/027043. http://www.ctri.nic.in/Clinicaltrials/pmaindet2.php?trialid=43175.
Collapse
Affiliation(s)
- Alok Sharma
- Department of Neurosurgery, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
- Department of Medical Services and Clinical Research, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | | | - Hemangi Sane
- Department of Research & Development, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | - Nilkanth Awad
- Department of Pulmonary Medicine, LTMG Hospital and LTM Medical CollegeSion, Mumbai, Maharashtra, India
| | | | - Anagha Joshi
- Department of Radiology, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
| | - Sujata Baweja
- Department of Microbiology, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
| | - Mohan Joshi
- Dean, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
| | | | - Nandini Gokulchandran
- Department of Medical Services and Clinical Research, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | - Prerna Badhe
- Department of Regenerative Laboratory, NeuroGen Brain and Spine InstituteSeawoods, Navi Maharashtra, India
| | - Mazhar Khan
- Department of Neurosurgery, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
| | - Amruta Paranjape
- Department of Research & Development, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | - Pooja Kulkarni
- Department of Research & Development, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | - Arjun K Methal
- Department of Research & Development, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| |
Collapse
|
41
|
Piao L, Huang Z, Inoue A, Kuzuya M, Cheng XW. Human umbilical cord-derived mesenchymal stromal cells ameliorate aging-associated skeletal muscle atrophy and dysfunction by modulating apoptosis and mitochondrial damage in SAMP10 mice. Stem Cell Res Ther 2022; 13:226. [PMID: 35659361 PMCID: PMC9166592 DOI: 10.1186/s13287-022-02895-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background Skeletal muscle mass and function losses in aging individuals are associated with quality of life deterioration and disability. Mesenchymal stromal cells exert immunomodulatory and anti-inflammatory effects and could yield beneficial effects in aging-related degenerative disease. Methods and results We investigated the efficacy of umbilical cord-derived mesenchymal stromal cells (UC-MSCs) on sarcopenia-related skeletal muscle atrophy and dysfunction in senescence-accelerated mouse prone 10 (SAMP10) mice. We randomly assigned 24-week-old male SAMP10 mice to a UC-MSC treatment group and control group. At 12 weeks post-injection, the UC-MSC treatment had ameliorated sarcopenia-related muscle changes in performance, morphological structures, and mitochondria biogenesis, and it enhanced the amounts of proteins or mRNAs for myosin heavy chain, phospho-AMP-activated protein kinase, phospho-mammalian target of rapamycin, phospho-extracellular signal-regulated kinase1/2, peroxisome proliferator-activated receptor-γ coactivator, GLUT-4, COX-IV, and hepatocyte growth factor in both gastrocnemius and soleus muscles, and it reduced the levels of proteins or mRNAs for cathepsin K, cleaved caspase-3/-8, tumor necrosis factor-α, monocyte chemoattractant protein-1, and gp91phox mRNAs. The UC-MSC treatment retarded mitochondria damage, cell apoptosis, and macrophage infiltrations, and it enhanced desmin/laminin expression and proliferating and CD34+/Integrin α7+ cells in both types of skeletal muscle of the SAMP10 mice. In vitro, we observed increased levels of HGF, PAX-7, and MoyD mRNAs at the 4th passage of UC-MSCs. Conclusions Our results suggest that UC-MSCs can improve sarcopenia-related skeletal muscle atrophy and dysfunction via anti-apoptosis, anti-inflammatory, and mitochondrial biogenesis mechanisms that might be mediated by an AMPK-PGC1-α axis, indicating that UC-MSCs may provide a promising treatment for sarcopenia/muscle diseases.
Collapse
Affiliation(s)
- Limei Piao
- Department of Human Life Cord Applied Cell Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Aichi-ken, 466-8550, Japan.,Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Zhe Huang
- Department of Human Life Cord Applied Cell Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Aichi-ken, 466-8550, Japan.
| | - Aiko Inoue
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Masafumi Kuzuya
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Xian Wu Cheng
- Department of Human Life Cord Applied Cell Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Aichi-ken, 466-8550, Japan. .,Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
| |
Collapse
|
42
|
Wu Y, Zhang H, Wang S, Li L, Wang R, Jiang S. Human umbilical cord-derived stem cell sheets improve left ventricular function in rat models of ischemic heart failure. Eur J Pharmacol 2022; 925:174994. [PMID: 35513020 DOI: 10.1016/j.ejphar.2022.174994] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/06/2022] [Accepted: 04/28/2022] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Human umbilical cord-derived mesenchymal stem cells (UC-MSCs) are among the most promising cell therapy sources used to treat ischemic heart disease. Cell sheet engineering has been used to transplant stem cells and improve their therapeutic effectiveness. We aimed to evaluate the effectiveness of UC-MSC sheets in the treatment of chronic ischemic heart failure. METHODS AND RESULTS Flow cytometric analysis showed that UC-MSCs were positive for CD73, CD90, and CD105. UC-MSC sheets were produced from UC-MSCs using temperature-responsive culture dishes. Afterward, these sheets were transplanted onto the epicardial surface at the infarct heart in rat models of chronic ischemic heart failure. At four weeks after the transplantation, echocardiography analysis revealed that the cardiac function of the UC-MSC sheets group was significantly better than that of the suspension and myocardial infarction (MI) only groups. Furthermore, histological examinations revealed that the left ventricular remodeling was attenuated compared with the suspension and MI-only groups. In the UC-MSC slice group, the neovascular den and cell size in the infarct margin region were was significantly improved than in the suspension and MI-only groups. Also, the UC-MSC sheets inhibited the PI3K/AKT/mTOR signaling pathway in chronic ischemic heart failure. CONCLUSIONS UC-MSC sheets can maintain cardiac function and attenuate ventricular remodeling in chronic ischemic heart failure, indicating that this strategy would be a promising therapeutic option in the clinical scenario.
Collapse
Affiliation(s)
- Yuanbin Wu
- Medical School of Chinese PLA, Beijing, 100853, China; Division of Adult Cardiac Surgery, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Huajun Zhang
- Division of Adult Cardiac Surgery, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Shuling Wang
- Division of Adult Cardiac Surgery, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Libing Li
- Division of Adult Cardiac Surgery, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Rong Wang
- Division of Adult Cardiac Surgery, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Shengli Jiang
- Division of Adult Cardiac Surgery, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
43
|
Nilforoushzadeh MA, Afzali H, Raoofi A, Nouri M, Naser R, Gholami O, Nasiry D, Mohammadnia A, Razzaghi Z, Alimohammadi A, Naraghi ZS, Peyrovan A, Jahangiri F, Khodaverdi Darian E, Rustamzadeh A, Zare S. Topical spray of Wharton's jelly mesenchymal stem cells derived from umbilical cord accelerates diabetic wound healing. J Cosmet Dermatol 2022; 21:5156-5167. [PMID: 35478316 DOI: 10.1111/jocd.15022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND & AIM Cell-based therapy utilizing mesenchymal stem cells (MSCs) is currently being investigated as a therapeutic agent for chronic wounds. There is no evidence regarding effectiveness of the spray and local transfer of this cellular product in diabetic wound healing. Accordingly, the present study, using clinical, pathological and biometric parameters, investigated the effectiveness of the spray of these cells in the healing of diabetic wounds in rats. METHODS Three days after the induction of diabetes (50 mg/kg single dose of streptozotocin) a circular excision was created on the back of rats. Diabetic rats were divided into two groups (n=21): Control and WJ-MSCs group. Sampling of the studied groups was performed on days 7, 14 and 21 after wounding. Histological, ultrasound imaging of dermis and epidermis in the wound area for thickness and density measurement and skin elasticity were evaluated. RESULTS Our results on days 7, 14, and 21 after wounding showed that the wound closure, thickness and density of new epidermis and dermis, as well as skin elasticity in healed wound were significantly higher in WJ-MSCs group compared to the Control group. CONCLUSION Application of WJ-MSCs suspension spray on the wound area can accelerate healing in diabetic wounds. Our findings may potentially provide a helpful therapeutic strategy for patients with a diabetic wound.
Collapse
Affiliation(s)
- Mohammad Ali Nilforoushzadeh
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Jordan Dermatology and Hair Transplantation Center, Tehran, Iran
| | - Hamideh Afzali
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Raoofi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Maryam Nouri
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Naser
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Gholami
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Davood Nasiry
- Amol Faculty of Paramedicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdolreza Mohammadnia
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Razzaghi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alimohammad Alimohammadi
- Forensic medicine specialist, Research center of legal medicine organization of Iran, Tehran, Iran
| | - Zahra Safaie Naraghi
- Department of Pathology, Razi Skin Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Aisan Peyrovan
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Faeze Jahangiri
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Khodaverdi Darian
- Department of Medical Biotechnology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.,Biotechnology Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Auob Rustamzadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Coccini T, Spinillo A, Roccio M, Lenta E, Valsecchi C, De Simone U. Human Umbilical Cord Mesenchymal Stem Cell-Based in vitro Model for Neurotoxicity Testing. Curr Protoc 2022; 2:e423. [PMID: 35471597 DOI: 10.1002/cpz1.423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neurotoxicity (NT) testing for regulatory purposes is based on in vivo animal testing. There is general consensus, however, about the need for the development of alternative methodologies to allow researchers to more rapidly and cost effectively screen large numbers of chemicals for their potential to cause NT, or to investigate their mode of action. In vitro assays are considered an important source of information for making regulatory decisions, and human cell-based systems are recommended as one of the most relevant models in toxicity testing, to reduce uncertainty in the extrapolation of results from animal-based models. Human neuronal models range from various neuroblastoma cell lines to stem cell-derived systems, including those derived from mesenchymal stem/stromal cells (hMSC). hMSCs exhibit numerous advantages, including the fact that they can be obtained in high yield from healthy human adult tissues, can be cultured with a minimal laboratory setup and without genetic manipulations, are able of continuous and repeated self-renewal, are nontumorigenic, and can form large populations of stably differentiated cells representative of different tissues, including neuronal cells. hMSCs derived from human umbilical cord (hUC) in particular possess several prominent advantages, including a painless, non-invasive, and ethically acceptable collection procedure, simple and convenient preparation, and high proliferation capacity. In addition, hMSCs can be efficiently differentiated into neuron-like cells (hNLCs), which can then be used for the assessment of neuronal toxicity of potential neurotoxic compounds in humans. Here, we describe a step-by-step procedure to use hMSCs from the umbilical cord for in vitro neurotoxicity testing. First, we describe how to isolate, amplify, and store hMSCs derived from the umbilical cord. We then outline the steps to transdifferentiate these cells into hNLCs, and then use the hNLCs for neurotoxicity testing by employing multiple common cytotoxicity assays after treatment with test compounds. The approach follows the most updated guidance on using human cell-based systems. These protocols will allow investigators to implement an alternative system for obtaining primary NLCs of human origin, and support advancement in neurotoxicity research. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Isolation and maintenance of human mesenchymal stem/stromal cells (hMSCs) obtained from the umbilical cord lining membrane Basic Protocol 2: Transdifferentiation of hMSCs into neuron-like cells (hNLCs) and basic neurotoxicity assessment.
Collapse
Affiliation(s)
- Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Arsenio Spinillo
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Marianna Roccio
- Department of Obstetrics and Gynecology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Elisa Lenta
- Immunology and Transplantation Laboratory, Pediatric Hematology Oncology Unit, Cell Factory, Department of Maternal and Children's Health, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chiara Valsecchi
- Immunology and Transplantation Laboratory, Pediatric Hematology Oncology Unit, Cell Factory, Department of Maternal and Children's Health, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Uliana De Simone
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Centre-National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
45
|
Chouw A, Sartika CR, Milanda T, Faried A. Interleukins Profiling in Umbilical Cord Mesenchymal Stem Cell-Derived Secretome. Stem Cells Cloning 2022; 15:1-9. [PMID: 35444427 PMCID: PMC9015105 DOI: 10.2147/sccaa.s356763] [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: 01/18/2022] [Accepted: 04/01/2022] [Indexed: 11/26/2022] Open
Abstract
Background Umbilical cord mesenchymal stem cells (UC-MSCs)-derived secretome is currently used in regenerative therapy. MSCs are believed to secrete a wide spectrum of bioactive molecules which give paracrine effects in immunomodulation and regenerative capacities. One group that was found in secretome is interleukins (ILs), a cytokine that plays an essential role in the process of proliferation, differentiation, maturation, migration, and adhesion of immune cells. However, as there are many types of ILs, the profile of ILs in the UC-MSCs-derived secretome has been limitedly reported. Therefore, in this study, we would like to profile and detect the interleukin concentration secreted by UC-MSCs. Methods UC-MSCs-derived secretome was collected from UC-MSCs passage 5 after 24- and 48-hour incubation (n=9). Secretome was filtered using 0.2 µm and stored at -80°C for further detection. All samples were normalized before the interleukin (IL-2, IL-4, IL-6, IL-9, IL-10, IL-12, IL-17A) detection using a MACSPlex Cytokine Kit. Results The IL-6 has the highest concentration among other interleukins in both groups and increases significantly (p<0.003) after incubation for 48 hours. The pro-inflammatory factors are decreasing while anti-inflammatory factors are increasing after 48-hour incubation. Discussion Our studies show that the UC-MSCs secrete pro- and anti-inflammatory interleukins. The concentration of anti-inflammatory interleukins shows to be increasing, while the pro-inflammatory interleukins are decreasing within the longer incubation time, but this not be applicable for IL-10 and IL-6. IL-6 has the highest concentration among other ILs. These results may provide important clues regarding when is the right time for secretome to be used in therapy patients, because all the molecules in the secretome can lead to many clinical manifestations.
Collapse
Affiliation(s)
- Angliana Chouw
- Doctoral Program, Faculty of Pharmacy Universitas Padjadjaran, Jatinangor, West Java, Indonesia
- Research and Development, Prodia StemCell Indonesia, Jakarta, DKI, Jakarta, Indonesia
| | - Cynthia Retna Sartika
- Research and Development, Prodia StemCell Indonesia, Jakarta, DKI, Jakarta, Indonesia
| | - Tiana Milanda
- Department of Biological Pharmacy, Faculty of Pharmacy Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Ahmad Faried
- Stem Cell Working Group, Faculty of Medicine Universitas Padjadjaran, Dr. Hasan Sadikin Hospital, Bandung, West Java, Indonesia
| |
Collapse
|
46
|
Cao Q, Huang C, Chen XM, Pollock CA. Mesenchymal Stem Cell-Derived Exosomes: Toward Cell-Free Therapeutic Strategies in Chronic Kidney Disease. Front Med (Lausanne) 2022; 9:816656. [PMID: 35386912 PMCID: PMC8977463 DOI: 10.3389/fmed.2022.816656] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic kidney disease (CKD) is rising in global prevalence and has become a worldwide public health problem, with adverse outcomes of kidney failure, cardiovascular disease, and premature death. However, current treatments are limited to slowing rather than reversing disease progression or restoring functional nephrons. Hence, innovative strategies aimed at kidney tissue recovery hold promise for CKD therapy. Mesenchymal stem cells (MSCs) are commonly used for regenerative therapy due to their potential for proliferation, differentiation, and immunomodulation. Accumulating evidence suggests that the therapeutic effects of MSCs are largely mediated by paracrine secretion of extracellular vesicles (EVs), predominantly exosomes. MSC-derived exosomes (MSC-Exos) replicate the functions of their originator MSCs via delivery of various genetic and protein cargos to target cells. More recently, MSC-Exos have also been utilized as natural carriers for targeted drug delivery. Therapeutics can be effectively incorporated into exosomes and then delivered to diseased tissue. Thus, MSC-Exos have emerged as a promising cell-free therapy in CKD. In this paper, we describe the characteristics of MSC-Exos and summarize their therapeutic efficacy in preclinical animal models of CKD. We also discuss the potential challenges and strategies in the use of MSC-Exos-based therapies for CKD in the future.
Collapse
Affiliation(s)
- Qinghua Cao
- Renal Medicine, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Chunling Huang
- Renal Medicine, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Xin-Ming Chen
- Renal Medicine, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Carol A Pollock
- Renal Medicine, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
47
|
Fazekas B, Alagesan S, Watson L, Ng O, Conroy CM, Català C, Andres MV, Negi N, Gerlach JQ, Hynes SO, Lozano F, Elliman SJ, Griffin MD. Comparison of Single and Repeated Dosing of Anti-Inflammatory Human Umbilical Cord Mesenchymal Stromal Cells in a Mouse Model of Polymicrobial Sepsis. Stem Cell Rev Rep 2022; 18:1444-1460. [PMID: 35013938 PMCID: PMC8747454 DOI: 10.1007/s12015-021-10323-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cells (MSCs) ameliorate pre-clinical sepsis and sepsis-associated acute kidney injury (SA-AKI) but clinical trials of single-dose MSCs have not indicated robust efficacy. This study investigated immunomodulatory effects of a novel MSC product (CD362-selected human umbilical cord-derived MSCs [hUC-MSCs]) in mouse endotoxemia and polymicrobial sepsis models. Initially, mice received intra-peritoneal (i.p.) lipopolysaccharide (LPS) followed by single i.p. doses of hUC-MSCs or vehicle. Next, mice underwent cecal ligation and puncture (CLP) followed by intravenous (i.v.) doses of hUC-MSCs at 4 h or 4 and 28 h. Analyses included serum/plasma assays of biochemical indices, inflammatory mediators and the AKI biomarker NGAL; multi-color flow cytometry of peritoneal macrophages (LPS) and intra-renal immune cell subpopulations (CLP) and histology/immunohistochemistry of kidney (CLP). At 72 h post-LPS injections, hUC-MSCs reduced serum inflammatory mediators and peritoneal macrophage M1/M2 ratio. Repeated, but not single, hUC-MSC doses administered at 48 h post-CLP resulted in lower serum concentrations of inflammatory mediators, lower plasma NGAL and reversal of sepsis-associated depletion of intra-renal T cell and myeloid cell subpopulations. Hierarchical clustering analysis of all 48-h serum/plasma analytes demonstrated partial co-clustering of repeated-dose hUC-MSC CLP animals with a Sham group but did not reveal a distinct signature of response to therapy. It was concluded that repeated doses of CD362-selected hUC-MSCs are required to modulate systemic and local immune/inflammatory events in polymicrobial sepsis and SA-AKI. Inter-individual variability and lack of effect of single dose MSC administration in the CLP model are consistent with observations to date from early-phase clinical trials.
Collapse
Affiliation(s)
- Barbara Fazekas
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | | | | | - Olivia Ng
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Orbsen Therapeutics Ltd., Galway, Ireland
| | - Callum M Conroy
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Orbsen Therapeutics Ltd., Galway, Ireland
| | - Cristina Català
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Neema Negi
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Jared Q Gerlach
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway, Ireland
| | - Sean O Hynes
- Discipline of Pathology, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Histopathology, Galway University Hospitals, Galway, Ireland
| | - Francisco Lozano
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona, Spain
- Department de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | | | - Matthew D Griffin
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland.
- Department of Nephrology, Saolta University Health Care Group, Galway University Hospitals, Galway, Ireland.
- National University of Ireland Galway, REMEDI, Biomedical Sciences, Corrib Village, Dangan, Galway, H91 TK33, Ireland.
| |
Collapse
|
48
|
Regeneration of Chronic Rotator Cuff Tear in a Rabbit Model: Synergetic Benefits of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells, Polydeoxyribonucleotides, and Microcurrent Therapy. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6496773. [PMID: 35342750 PMCID: PMC8941538 DOI: 10.1155/2022/6496773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/06/2022] [Accepted: 02/24/2022] [Indexed: 11/22/2022]
Abstract
Objective To investigate synergic therapeutic effects of combined injection of intralesional mesenchymal stem cells derived from human umbilical cord blood (UCB-MSCs) and polydeoxyribonucleotide (PDRN) combined with microcurrent therapy (MIC) on full thickness rotator cuff tendon tear (FTRCTT) in rabbit models. Methods Thirty-two rabbit models were assigned to 4 different groups. FTRCTT in the supraspinatus tendon was created. After 6 weeks, 4 types of procedures (0.2 mL normal saline injection, group 1 (G1-NS); 0.2 mL SC injection, group 2 (G2-MSC); 0.2 mL SC and weekly four injections of 0.2 mL PDRN with sham MIC, group 3 (G3-MSC+PDRN+sham MIC); and 0.2 mL SC and weekly four injections of 0.2 mL PDRN with MIC for four weeks, group 4 (G4-MSC+PDRN+MIC)) were performed in FTRCTT. Gross morphologic and histological changes of proliferating cell nuclear antigen (PCNA), vascular endothelial growth factor (VEGF) and platelet endothelial cell adhesion molecule (PECAM-1) and motion analysis were performed. Results There was a significant difference in gross morphologic changes between baseline and week 4 posttreatment in group 4 compared to the other three groups (p = 0.01). In groups 3 and 4, all parameters of histochemical and motion analysis have been found to be significantly greater than the ones in groups 1 and 2 (p < 0.05). In group 4, PCNA-, VEGF-, and PECAM-1-stained cells, as well as walking distance, were significantly greater than the ones in group 3 (p < 0.05). Conclusion The treatment with UCB-MSCs and PDRN combined with MIC might be the most effective in rabbit models' traumatic FRTCTT.
Collapse
|
49
|
Abbasi B, Shamsasenjan K, Ahmadi M, Beheshti SA, Saleh M. Mesenchymal stem cells and natural killer cells interaction mechanisms and potential clinical applications. Stem Cell Res Ther 2022; 13:97. [PMID: 35255980 PMCID: PMC8900412 DOI: 10.1186/s13287-022-02777-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/09/2021] [Indexed: 12/29/2022] Open
Abstract
Natural killer cells (NK cells) are innate immune cells that are activated to fight tumor cells and virus-infected cells. NK cells also play an important role in the graft versus leukemia response. However, they can over-develop inflammatory reactions by secreting inflammatory cytokines and increasing Th1 differentiation, eventually leading to tissue damage. Today, researchers have attributed some autoimmune diseases and GVHD to NK cells. On the other hand, it has been shown that mesenchymal stem cells (MSCs) can modulate the activity of NK cells, while some researchers have shown that NK cells can cause MSCs to lysis. Therefore, we considered it is necessary to investigate the effect of these two cells and their signaling pathway in contact with each other, also their clinical applications.
Collapse
Affiliation(s)
- Batol Abbasi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedeh Ameneh Beheshti
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahshid Saleh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Nguyen Thanh L, Hoang VT, Le Thu H, Nguyen PAT, Hoang DM, Ngo DV, Cao Vu H, Nguyen Thi Bich V, Heke M. Human Umbilical Cord Mesenchymal Stem Cells for Severe Neurological Sequelae due to Anti- N-Methyl-d-Aspartate Receptor Encephalitis: First Case Report. Cell Transplant 2022; 31:9636897221110876. [PMID: 35815930 PMCID: PMC9277426 DOI: 10.1177/09636897221110876] [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] [Indexed: 11/16/2022] Open
Abstract
Anti-N-methyl-d-aspartate (NMDA) receptor encephalitis is caused by altered patient immune reactions. This study reports the first patient with severe neurologic sequelae after NMDA receptor encephalitis treated with allogeneic umbilical cord-derived mesenchymal stem/stromal cells (UC-MSCs). A 5-year-old girl was diagnosed with NMDA receptor encephalitis and treated with immunosuppressive medicaments and intravenous immunoglobulin (IVIG). Despite intensive therapy, the patient's condition worsened so that allogenic UC-MSC therapy was contemplated. The patient received three intrathecal infusions of xeno- and serum-free cultured UC-MSCs at a dose of 106 cells/kg. At baseline and after each UC-MSC administration, the patient was examined by the German Coma Recovery Scale (CRS), the Gross Motor Function Classification System (GMFCS), the Gross Motor Function Measure-88 (GMFM-88), the Manual Ability Classification System (MACS), the Modified Ashworth Scale, and the Denver II test. Before cell therapy, she was in a permanent vegetative state with diffuse cerebral atrophy. Her cognition and motor functions improved progressively after three UC-MSC infusions. At the last visit, she was capable of walking, writing, and counting numbers. Control of urinary and bowel functions was completely recovered. Cerebral atrophy was reduced on brain magnetic resonance imaging (MRI). Overall, the outcomes of this patient suggest a potential cell therapy for autoimmune encephalitis and its neurological consequences.
Collapse
Affiliation(s)
- Liem Nguyen Thanh
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hanoi, Vietnam.,College of Health Science, VinUniversity, Hanoi, Vietnam
| | - Van T Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hanoi, Vietnam
| | | | | | - Duc M Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hanoi, Vietnam
| | | | - Hung Cao Vu
- Vietnam National Children's Hospital, Hanoi, Vietnam
| | | | - Michael Heke
- Department of Biology, Stanford University, Stanford, CA, USA
| |
Collapse
|