1
|
Wu KC, Chang YH, Ding DC, Lin SZ. Mesenchymal Stromal Cells for Aging Cartilage Regeneration: A Review. Int J Mol Sci 2024; 25:12911. [PMID: 39684619 DOI: 10.3390/ijms252312911] [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: 11/06/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Cartilage degeneration is a key feature of aging and osteoarthritis, characterized by the progressive deterioration of joint function, pain, and limited mobility. Current treatments focus on symptom relief, not cartilage regeneration. Mesenchymal stromal cells (MSCs) offer a promising therapeutic option due to their capability to differentiate into chondrocytes, modulate inflammation, and promote tissue regeneration. This review explores the potential of MSCs for cartilage regeneration, examining their biological properties, action mechanisms, and applications in preclinical and clinical settings. MSCs derived from bone marrow, adipose tissue, and other sources can self-renew and differentiate into multiple cell types. In aging cartilage, they aid in tissue regeneration by secreting growth factors and cytokines that enhance repair and modulate immune responses. Recent preclinical studies show that MSCs can restore cartilage integrity, reduce inflammation, and improve joint function, although clinical translation remains challenging due to limitations such as cell viability, scalability, and regulatory concerns. Advancements in MSC delivery, including scaffold-based approaches and engineered exosomes, may improve therapeutic effectiveness. Potential risks, such as tumorigenicity and immune rejection, are also discussed, emphasizing the need for optimized treatment protocols and large-scale clinical trials to develop effective, minimally invasive therapies for cartilage regeneration.
Collapse
Affiliation(s)
- Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| | - Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
- Institute of Medical Sciences, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
2
|
Lee H, Lim Y, Lee SH. Rapid-acting pain relief in knee osteoarthritis: autologous-cultured adipose-derived mesenchymal stem cells outperform stromal vascular fraction: a systematic review and meta-analysis. Stem Cell Res Ther 2024; 15:446. [PMID: 39568086 PMCID: PMC11580442 DOI: 10.1186/s13287-024-04034-2] [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: 09/02/2024] [Accepted: 10/29/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Knee osteoarthritis (OA) is a leading cause of disability, with current treatment options often falling short of providing satisfactory outcomes. Autologous-cultured adipose-derived mesenchymal stem cells (ADMSCs) and stromal vascular fractions (SVFs) have emerged as potential regenerative therapies. METHODS A comprehensive search was conducted among multiple databases for studies up to June 2023. The risk of bias was assessed in randomized and non-randomized studies, adhering to PRISMA guidelines. The study has been registered with PROSPERO (CRD 42023433160). RESULTS Our analysis encompassed 31 studies involving 1,406 patients, of which, 19 studies with 958 patients were included in a meta-analysis, examining both SVF and autologous-cultured ADMSC methods. Significant pain reduction was observed with autologous-cultured ADMSCs starting at 3 months (MD = -2.43, 95% CI, -3.99, -0.86), whereas significant pain mitigation in response to SVF therapy was found to start at 12 months (MD = -2.13, 95% CI, -3.06, -1.21). Both autologous-cultured ADMSCs and SVF provided significant improvement in knee function starting at 12 months (MD = -9.19, 95% CI, -12.48, -5.90 vs. MD = -9.09, 95% CI, -12.67, -5.51, respectively). We found no evidence of severe adverse events linked directly to ADMSC therapy. CONCLUSION Autologous-cultured ADMSCs offer a promising alternative for more rapid pain relief in knee OA, with both ADMSCs and SVF demonstrating substantial long-term benefits in joint function and cartilage regeneration, in the absence of any severe ADMSC-related adverse events.
Collapse
Affiliation(s)
- Haneul Lee
- Department of Physical Therapy, College of Medical Science, Gachon University, Incheon, Korea
| | - Youngeun Lim
- Department of Physical Therapy, College of Medical Science, Gachon University, Incheon, Korea
| | - Seon-Heui Lee
- College of Nursing, Research Institute of AI and Nursing Science, Gachon University, Incheon, Korea.
| |
Collapse
|
3
|
Baek A, Baek D, Kim SH, Kim J, Notario GR, Lee DW, Kim HJ, Cho SR. Polydeoxyribonucleotide ameliorates IL-1β-induced impairment of chondrogenic differentiation in human bone marrow-derived mesenchymal stem cells. Sci Rep 2024; 14:26076. [PMID: 39478005 PMCID: PMC11525668 DOI: 10.1038/s41598-024-77264-2] [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: 01/24/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative disease of the joints, prevalent worldwide. Polydeoxyribonucleotide (PDRN) is used for treating knee OA. However, the role of PDRN in IL-1β-induced inflammatory responses in human bone marrow-derived mesenchymal stem cells (hBMSCs) remains unknown. Here, we investigated the role of PDRN in IL-1β-induced impairment of chondrogenic differentiation in hBMSCs. hBMSCs treated with PDRN showed a large micromass, enhanced safranin O and alcian blue staining intensity, and increased expression of chondrogenic genes in IL-1β-induced inflammatory responses, in addition to regulation of catabolic and anabolic genes. In addition, PDRN treatment suppressed the expression of inflammatory cytokines and mitigated IL-1β-induced apoptosis in hBMSCs. Mechanistically, PDRN treatment increased the formation of cyclic adenosine monophosphate (cAMP) and upregulated the phosphorylation of cAMP-dependent protein kinase A (PKA)/cAMP response element binding protein (CREB) through the adenosine A2A receptor in hBMSCs and thus blocked the nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) signaling pathway. Thus, IL-1β-induced expression of inflammatory cytokines in hBMSCs was directly reduced by adenosine A2A receptor activation. Based on our results, we suggest that PDRN may be a promising MSC-based therapeutic agent for OA.
Collapse
Affiliation(s)
- Ahreum Baek
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Dawoon Baek
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Forensic DNA Division, National Forensic Service, Daegu, Republic of Korea
| | - Sung Hoon Kim
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Jinyoung Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Geneva Rose Notario
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Do-Won Lee
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Kim
- Department of Rehabilitation Medicine, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, Republic of Korea.
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Zeng L, Yang K, Yu G, Chen J, Long Z, Xiang W, Liu S, Zheng Y, Yan Y, Hao M, Sun L. Efficacy and safety of culture-expanded mesenchymal stromal cell therapy in the treatment of 4 types of inflammatory arthritis: A systematic review and meta-analysis of 36 randomized controlled trials. Semin Arthritis Rheum 2024; 68:152498. [PMID: 38970896 DOI: 10.1016/j.semarthrit.2024.152498] [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: 03/22/2024] [Revised: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 07/08/2024]
Abstract
OBJECTIVE This study aims to assess the effectiveness and safety of mesenchymal stem cell (MSC) transplantation in the treatment of inflammatory arthritis. METHODS Two researchers conducted a comprehensive search of Chinese and English databases from their inception until July 2023. The literature screening and data extraction were then performed. Statistical analysis was carried out using RevMan 5.4 software. RESULTS A total of 36 relevant RCTs, involving 2,076 participants, were ultimately included in this study. These RCTs encompassed four types of inflammatory arthritis, namely rheumatoid arthritis (RA), osteoarthritis (OA), ankylosing spondylitis (AS), and systemic sclerosis (SSc). The results demonstrated that MSC therapy exhibited improvements in the Visual Analog Scale (VAS) for pain in OA patients (bone marrow: SMD=-0.95, 95 % CI: -1.55 to -0.36, P = 0.002; umbilical cord: SMD=-2.03, 95 % CI: -2.99 to -1.07, P < 0.0001; adipose tissue: SMD=-1.26, 95 % CI: -1.99 to -0.52, P = 0.0009). Specifically, MSCs sourced from adipose tissue showed enhancements in Western Ontario and McMaster Universities Arthritis Index (WOMAC) pain (P = 0.0001), WOMAC physical function (P = 0.001), and total WOMAC scores (P = 0.0003). As for MSC therapy in RA, AS, and SSc, the current systematic review suggests a potential therapeutic effect of MSCs on these inflammatory arthritic conditions. Safety assessments indicated that MSC therapy did not increase the incidence of adverse events. CONCLUSION MSCs have the potential to alleviate joint pain and improve joint function in patients with inflammatory arthritis. Moreover, MSC therapy appears to be relatively safe and could be considered as a viable alternative treatment option for inflammatory arthritis.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China; Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China.
| | - Ganpeng Yu
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Junpeng Chen
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China; Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA; Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China.
| | - Zhiyong Long
- Department of Physical Medicine and Rehabilitation, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wang Xiang
- The First People's Hospital of Changde City, Changde, China
| | - Shuman Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Yaru Zheng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Yexing Yan
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China; Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
5
|
Jin WS, Yin LX, Sun HQ, Zhao Z, Yan XF. Mesenchymal Stem Cells Injection Is More Effective Than Hyaluronic Acid Injection in the Treatment of Knee Osteoarthritis With Similar Safety: A Systematic Review and Meta-Analysis. Arthroscopy 2024:S0749-8063(24)00555-3. [PMID: 39154667 DOI: 10.1016/j.arthro.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024]
Abstract
PURPOSE To evaluate the efficacy and safety of intra-articular injection of mesenchymal stem cells (MSCs) versus hyaluronic acid (HA) in the treatment of knee osteoarthritis (KOA). METHODS Eligible randomized controlled trials (RCTs) were identified through a search of PubMed, Embase, the Cochrane Library, Web of Science, SinoMed, and CNKI databases from inception to March 2024. For meta-analysis, data on clinical outcomes were measured using visual analog scale (VAS) and the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), and data on cartilage repair were measured using the Whole-Organ Magnetic Resonance Imaging Score (WORMS); data on safety were evaluated by the incidence of adverse events. Two researchers independently read the included literature, extracted data and evaluated the quality, used the Cochrane risk bias assessment tool for bias risk assessment, and used RevMan5.3 software for meta-analysis. RESULTS Ten RCTs involving 818 patients with KOA ranging from I to Ⅲ on the Kellgren-Lawrence grading scale were included in this meta-analysis. Meta-analysis results showed that at 12 months, the WOMAC total score (mean difference [MD] = -10.22, 95% confidence interval [CI]: -14.86 to -5.59, P < .0001, Z = 4.32), VAS score (MD = -1.31, 95% CI: -1.90 to -0.73, P < .0001, Z = 4.40); and WORMS score (MD = -26.01, 95% CI: -31.88 to -20.14, P < .001, Z = 8.69) of the MSCs group all decreased significantly (P < .05) compared with the HA control group and reached the minimal clinically important differences. Furthermore, there was no significant difference in the incidence of adverse events (relative risk = 1.54, 95% CI: 0.85-2.79, P = .16, I2 = 0) between the 2 groups (P > .05). CONCLUSIONS Compared with HA, intra-articular injection of MSCs therapy appears to alleviate joint pain effectively, improving clinical function of KOA patients. These benefits are observed to last for at least 12 months without an increase in adverse events. Due to limited, varied, and lacking minimal clinically important differences results in existing literature, further research is needed. LEVEL OF EVIDENCE Level I, meta-analysis of Level I studies.
Collapse
Affiliation(s)
- Wen-Shu Jin
- Department of Hospital-Acquired Infection Control, The Second Affiliated Hospital of Shandong First Medical University, Taian City, Shandong Province, China
| | - Lu-Xu Yin
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan City, Shandong Province, China
| | - Hua-Qiang Sun
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan City, Shandong Province, China
| | - Zhang Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan City, Shandong Province, China
| | - Xin-Feng Yan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan City, Shandong Province, China.
| |
Collapse
|
6
|
V S, R A, Haran H, I SK, James S. Enhanced Bone Marrow Aspirate Concentrate (BMAC) Preparation Strategy in the Management of Chondromalacia Patella: A Case Report. Cureus 2024; 16:e59321. [PMID: 38817498 PMCID: PMC11137323 DOI: 10.7759/cureus.59321] [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: 04/09/2024] [Accepted: 04/29/2024] [Indexed: 06/01/2024] Open
Abstract
Chondromalacia patellae (CMP) is a widespread cause of patellofemoral pain syndrome (PFPS), which manifests as anterior knee pain and functional limitations. Current treatments frequently fail to give long-term relief, necessitating the exploration of new therapeutic techniques. Recent research has demonstrated the efficacy of Bone Marrow Aspirate Concentrate (BMAC) therapy, which utilizes the regeneration characteristics of mesenchymal stem cells (MSCs) and growth factors. We present the case of a 36-year-old male patient with Grade III CMP who was resistant to conservative treatment but was successfully treated with BMAC therapy. Detailed methods for BMAC preparation, such as double centrifugation and growth factor analysis, are presented. At six and 12 weeks after therapy, the patient showed significant improvements in pain and functional results, as well as enhanced levels of growth factors and CD34+ cells in the BMAC. This study provides insights into the regeneration potential of BMAC therapy and highlights its promising role in managing chondral abnormalities. Larger clinical trials and standardization of BMAC preparation procedures are necessary for establishing its effectiveness and consistency as a standard treatment approach for CMP.
Collapse
Affiliation(s)
- Soundharya V
- Transfusion Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Arthi R
- Transfusion Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Hari Haran
- Transfusion Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Suresh Kumar I
- Transfusion Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Sahayaraj James
- Transfusion Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| |
Collapse
|
7
|
Sunwoong P, Rim YA, Sohn Y, Nam Y, Ju JH. Exploration of Efficient HLA Haplotypes by Comparing the Proportion of Applicable Populations. Cell Transplant 2024; 33:9636897241283539. [PMID: 39437338 PMCID: PMC11504281 DOI: 10.1177/09636897241283539] [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: 05/29/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 10/25/2024] Open
Abstract
With the aging population, the incidence of degenerative diseases such as dementia and arthritis is on the rise. To combat these diseases, cell therapies using induced pluripotent stem cells (iPSCs) are being developed worldwide. However, challenges such as high development costs and immune compatibility persist. Thus, methods such as generating patient-specific iPSCs or genetically edited iPSCs with deleted immune-related genes are being researched. Applying these approaches is limited due to high cost and safety concerns of gene editing. Therefore, we focused on an alternative method using human leukocyte antigen (HLA)-homozygous cell lines, which could overcome immune rejection issues economically. We investigated diseases that could potentially be treated with cell therapy and identified which HLA-homozygous cell lines could be most effectively used for the efficient production of therapeutic cell lines. The results of the study showed that cell therapy could be applied to a wide range of diseases, and expanding the population that can benefit from HLA-homozygous iPSC lines could help popularize these treatment methods. We highlight the necessity of a global HLA-homozygous iPSC bank.
Collapse
Affiliation(s)
- Paeng Sunwoong
- School of Applied Biology, Seoul National University, Seoul, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Yoojun Nam
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
- Yipscell Inc., Seoul, Korea
| | - Ji Hyeon Ju
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Yipscell Inc., Seoul, Korea
| |
Collapse
|
8
|
Razak HRBA, Corona K, Totlis T, Chan LYT, Salreta JF, Sleiman O, Vasso M, Baums MH. Mesenchymal stem cell implantation provides short-term clinical improvement and satisfactory cartilage restoration in patients with knee osteoarthritis but the evidence is limited: a systematic review performed by the early-osteoarthritis group of ESSKA-European knee associates section. Knee Surg Sports Traumatol Arthrosc 2023; 31:5306-5318. [PMID: 37737920 PMCID: PMC10719133 DOI: 10.1007/s00167-023-07575-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE Implantation of mesenchymal stem cells (MSCs) is a potential cell-based modality for cartilage repair. Currently, its clinical use largely surrounds focal cartilage defect repair and intra-articular injections in knee osteoarthritis. The MSCs' implantation efficacy as a treatment option for osteoarthritis remains contentious. This systematic review aims to evaluate studies that focused on MSCs implantation in patients with knee OA to provide a summary of this treatment option outcomes. METHODS A systematic search was performed in PubMed (Medline), Scopus, Cinahl, and the Cochrane Library. Original studies investigating outcomes of MSCs implantations in patients with knee OA were included. Data on clinical outcomes using subjective scores, radiological outcomes, and second-look arthroscopy gradings were extracted. RESULTS Nine studies were included in this review. In all included studies, clinical outcome scores revealed significantly improved functionality and better postoperative pain scores at 2-3 years follow-up. Improved cartilage volume and quality at the lesion site was observed in five studies that included a postoperative magnetic resonance imaging assessment and studies that performed second-look arthroscopy. No major complications or tumorigenesis occurred. Outcomes were consistent in both single MSCs implantation and concurrent HTO with MSCs implantation in cases with excessive varus deformity. CONCLUSION According to the available literature, MSCs implantation in patients with mild to moderate knee osteoarthritis is safe and provides short-term clinical improvement and satisfactory cartilage restoration, either as a standalone procedure or combined with HTO in cases with axial deformity. However, the evidence is limited due to the high heterogeneity among studies and the insufficient number of studies including a control group and mid-term outcomes. LEVEL OF EVIDENCE IV.
Collapse
Affiliation(s)
| | - Katia Corona
- Orthopedics and Traumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS-Sacred Heart Catholic University, Rome, Italy
| | - Trifon Totlis
- Thessaloniki Minimally Invasive Surgery (The-MIS) Orthopaedic Centre, St. Luke's Hospital, Thessaloniki, Greece.
- Department of Anatomy and Surgical Anatomy, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Li Yi Tammy Chan
- Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117597, Singapore
| | - Jose Filipe Salreta
- Orthopaedic and Traumatology Department, Hospital Garcia de Orta, Almada, Portugal
| | - Obeida Sleiman
- Department of Orthopedics, Trauma Surgery and Sports Traumatology, Catholic Clinical Center Ruhr North (KKRN), Dorsten, Germany
| | - Michele Vasso
- Department of Medicine and Health Sciences, University of Molise, Via Francesco De Sanctis, 86100, Campobasso, Italy
| | - Mike H Baums
- Department of Orthopedics, Trauma Surgery and Sports Traumatology, Catholic Clinical Center Ruhr North (KKRN), Dorsten, Germany
| |
Collapse
|
9
|
Qiao X, Yan L, Feng Y, Li X, Zhang K, Lv Z, Xu C, Zhao S, Liu F, Yang X, Tian Z. Efficacy and safety of corticosteroids, hyaluronic acid, and PRP and combination therapy for knee osteoarthritis: a systematic review and network meta-analysis. BMC Musculoskelet Disord 2023; 24:926. [PMID: 38037038 PMCID: PMC10687893 DOI: 10.1186/s12891-023-06925-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/26/2023] [Indexed: 12/02/2023] Open
Abstract
OBJECTIVE There are many injectable treatments for knee osteoarthritis with different characteristics and effects, the aim is to understand which one can lead to better and safer results. METHODS The PRISMA principles were followed when doing the literature search. Web of Science databases, Embase, the Cochrane Library, PubMed, and the Wanfang database were searched to identified randomized controlled trials that assessed the efficacy of corticosteroids (CSC), platelet-rich plasma (PRP), hyaluronic acid (HA), and combination therapy in treating KOA. Risk of bias was assessed using the relevant Cochrane tools (version 1.0). The outcome measure included the visual analog scale (VAS) score, the Western Ontario and McMaster Universities Osteoarthritis (WOMAC) score, and treatment-related adverse events. The network meta-analysis was performed using STATA17 software and a Bayesian stratified random effects model. RESULTS Network meta-analysis using the Bayesian random-effects model revealed 35 studies with 3104 participants. PRP showed the best WOMAC score at a 3-month follow-up, followed by PRP + HA, HA, placebo, and CSC; PRP + HA scored the highest VAS, followed by PRP, CSC, HA, and placebo. PRP, CSC, HA, and placebo had the highest WOMAC scores six months following treatment; PRP + HA showed the best VAS scores. PRP showed the best WOMAC score at 12 months, followed by PRP + HA, HA, placebo, and CSC; The best VAS score was obtained with PRP, followed by PRP + HA, HA, and CSC. No therapy demonstrated a rise in adverse events linked to the treatment in terms of safety. CONCLUSIONS The current study found that PRP and PRP + HA were the most successful in improving function and alleviating pain after 3, 6, and 12 months of follow-up. CSC, HA, PRP, and combination therapy did not result in an increase in the incidence of treatment-related side events as compared to placebo.
Collapse
Affiliation(s)
- Xiaochen Qiao
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
- Department of Orthopedics, JinZhong Hospital Affiliated to Shanxi Medical University, 689 Huitong South Road, Jinzhong, Shanxi, 030600, PR China
| | - Lei Yan
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
| | - Yi Feng
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
| | - Xiaoyan Li
- Xihua Yang Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, 030013, PR China
| | - Kun Zhang
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
| | - Zhi Lv
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
| | - Chaojian Xu
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China
| | - Sen Zhao
- Taiyuan Hand Surgery Hospital, Taiyuan, Shanxi, 030001, PR China
| | - Fengrui Liu
- Orthopedics Department, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Xihua Yang
- Xihua Yang Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, 030013, PR China.
| | - Zhi Tian
- Second Clinical Medical College, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China.
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, PR China.
| |
Collapse
|
10
|
Dias de Oliveira FB, Antonioli E, Dias OFM, de Souza JG, Agarwal S, Chudzinski-Tavassi AM, Ferretti M. Comparative Effects of Intra-Articular versus Intravenous Mesenchymal Stromal Cells Therapy in a Rat Model of Osteoarthritis by Destabilization of Medial Meniscus. Int J Mol Sci 2023; 24:15543. [PMID: 37958526 PMCID: PMC10649289 DOI: 10.3390/ijms242115543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/15/2023] Open
Abstract
Transplanted mesenchymal stromal cells (MSCs) exhibit a robust anti-inflammatory and homing capacity in response to high inflammatory signals, as observed in studies focused on rheumatic diseases that target articular cartilage (AC) health. However, AC degradation in osteoarthritis (OA) does not necessarily coincide with a highly inflammatory joint profile. Often, by the time patients seek medical attention, they already have damaged AC. In this study, we examined the therapeutic potential of a single bone marrow MSC transplant (2 × 106 cells/kgbw) through two different routes: intra-articular (MSCs-IAt) and intravenous (MSCs-IVt) in a preclinical model of low-grade inflammatory OA with an established AC degeneration. OA was induced through the destabilization of the medial meniscus (DMM) in female Wistar Kyoto rats. The animals received MSCs 9 weeks after surgery and were euthanized 4 and 12 weeks post-transplant. In vivo and ex vivo tracking of MSCs were analyzed via bioluminescence and imaging flow cytometry, respectively. Cytokine/chemokine modulation in serum and synovial fluid was measured using a multiplex panel. AC degeneration was quantified through histology, and hindlimb muscle balance was assessed with precision weighing. To our knowledge, we are the first group to show the in vivo (8 h) and ex vivo (12 h) homing of cells to the DMM-OA joint following MSCs-IVt. In the case of MSCs-IAt, the detection of cellular bioluminescence at the knee joint persisted for up to 1 week. Intriguingly, intra-articular saline injection (placebo-IAt) resulted in a worse prognosis of OA when compared to a non-invasive control (placebo-IVt) without joint injection. The systemic cytokines/chemokines profile exhibited a time-dependent variation between transplant routes, displaying a transient anti-inflammatory systemic response for both MSCs-IVt and MSCs-IAt. A single injection of MSCs, whether administered via the intra-articular or intravenous route, performed 9 weeks after DMM surgery, did not effectively inhibit AC degeneration when compared to a non-invasive control.
Collapse
Affiliation(s)
| | - Eliane Antonioli
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.B.D.d.O.)
| | | | - Jean Gabriel de Souza
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA;
- CENTD Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo 05503-900, Brazil
| | - Sudha Agarwal
- Division of Rheumatology and Immunology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH 43210, USA
| | - Ana Marisa Chudzinski-Tavassi
- CENTD Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo 05503-900, Brazil
- Laboratório de Desenvolvimento e Inovação, Butantan Institute, São Paulo 05503-900, Brazil
| | - Mario Ferretti
- Hospital Israelita Albert Einstein, São Paulo 05652-900, Brazil; (F.B.D.d.O.)
- Departamento de Ortopedia e Traumatologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| |
Collapse
|
11
|
Sadri B, Hassanzadeh M, Bagherifard A, Mohammadi J, Alikhani M, Moeinabadi-Bidgoli K, Madani H, Diaz-Solano D, Karimi S, Mehrazmay M, Mohammadpour M, Vosough M. Cartilage regeneration and inflammation modulation in knee osteoarthritis following injection of allogeneic adipose-derived mesenchymal stromal cells: a phase II, triple-blinded, placebo controlled, randomized trial. Stem Cell Res Ther 2023; 14:162. [PMID: 37316949 DOI: 10.1186/s13287-023-03359-8] [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: 11/19/2022] [Accepted: 04/28/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Intra-articular injection of mesenchymal stromal cells (MSCs) with immunomodulatory features and their paracrine secretion of regenerative factors proposed a noninvasive therapeutic modality for cartilage regeneration in knee osteoarthritis (KOA). METHODS Total number of 40 patients with KOA enrolled in two groups. Twenty patients received intra-articular injection of 100 × 106 allogeneic adipose-derived mesenchymal stromal cells (AD-MSCs), and 20 patients as control group received placebo (normal saline). Questionnaire-based measurements, certain serum biomarkers, and some cell surface markers were evaluated for 1 year. Magnetic resonance imaging (MRI) before and 1 year after injection was performed to measure possible changes in the articular cartilage. RESULTS Forty patients allocated including 4 men (10%) and 36 women (90%) with average age of 56.1 ± 7.2 years in control group and 52.8 ± 7.5 years in AD-MSCs group. Four patients (two patients from AD-MSCs group and two patients from the control group) excluded during the study. Clinical outcome measures showed improvement in AD-MSCs group. Hyaluronic acid and cartilage oligomeric matrix protein levels in blood serum decreased significantly in patients who received AD-MSCs (P < 0.05). Although IL-10 level significantly increased after 1 week (P < 0.05), the serum level of inflammatory markers dramatically decreased after 3 months (P < 0.001). Expressions of CD3, CD4, and CD8 have a decreasing trend during 6-month follow-up (P < 0.05), (P < 0.001), and (P < 0.001), respectively. However, the number of CD25+ cells increased remarkably in the treatment group 3 months after intervention (P < 0.005). MRI findings showed a slight increase in the thickness of tibial and femoral articular cartilages in AD-MSCs group. The changes were significant in the medial posterior and medial anterior areas of the tibia with P < 0.01 and P < 0.05, respectively. CONCLUSION Inter-articular injection of AD-MSCs in patients with KOA is safe. Laboratory data, MRI findings, and clinical examination of patients at different time points showed notable articular cartilage regeneration and significant improvement in the treatment group. TRIAL REGISTRATION Iranian registry of clinical trials (IRCT, https://en.irct.ir/trial/46 ), IRCT20080728001031N23. Registered 24 April 2018.
Collapse
Affiliation(s)
- Bahareh Sadri
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Hassanzadeh
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Bagherifard
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Javad Mohammadi
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mehdi Alikhani
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Kasra Moeinabadi-Bidgoli
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hoda Madani
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Dylana Diaz-Solano
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, 1020-A, Caracas, Venezuela
| | - Shahedeh Karimi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Mehdi Mohammadpour
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
12
|
Vallejo R, Benyamin R, Orduña-Valls J, Vallejo A, Thomas SM, Cedeño DL. A randomized controlled study of the long-term efficacy of cooled and monopolar radiofrequency ablation for the treatment of chronic pain related to knee osteoarthritis. INTERVENTIONAL PAIN MEDICINE 2023; 2:100249. [PMID: 39238667 PMCID: PMC11372914 DOI: 10.1016/j.inpm.2023.100249] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 09/07/2024]
Abstract
Background Chronic knee pain due to osteoarthritis (OA) is expected to become more prevalent. Although conventional therapies may provide relief they are not long-lasting. Persistent pain may lead to total knee replacement, which is not free of adverse outcomes. Monopolar and cooled radiofrequency ablation (RFA) of genicular nerves is an effective option. However, either method may provide distinctive results depending on expected lesion size, a key aspect considering the anatomical variability of knee innervations. This prospective, double-blind, randomized controlled trial evaluated the efficacy and durability of knee RFA using a cooled probe or a monopolar probe of comparable diameter. Methods This investigator-initiated, post-market, double-blinded, prospective, randomized controlled trial was approved by the Western IRB. 79 subjects with chronic knee pain due to knee OA were enrolled in multiple locations of a single center. 75 subjects were randomized (1:1) into RFA treatment with either a 4 mm/17G cooled active tip (CRFA) or a 10 mm/16G monopolar active tip (MRFA) using conventional procedures. Primary endpoint was change in knee pain level (100 mm VAS score) from baseline at 24-week post-treatment. Other endpoints include change in functionality, global perceived effect, and frequency of adverse events. Evaluation spanned to 52-week post-treatment. Significance of results (p < 0.05) was calculated using standard statistical analyses. Results Both CRFA and MRFA provided significant reduction (41 mm and 39 mm, respectively) of chronic knee pain at 24-week. At the 52-week visit, reduction in pain level was sustained for CRFA (42 mm) but seems to decrease for MRFA (31 mm). Improvements in functionality were also significant and sustained with both treatments, although tend to decrease with MRFA at 52-week. Most patients also perceived a very good/good effect of treatments along the duration of the study. Conclusion RFA of knee genicular nerves for the treatment of OA chronic pain is effective for 52 weeks post-ablation when using a CRFA (4 mm/17G active tip) or MRFA (10 mm/16G active tip). The benefits of CRFA seems to be better sustained beyond 24 weeks than the ones of MRFA, although no significant differences were observed at 52 weeks.
Collapse
Affiliation(s)
- Ricardo Vallejo
- Millennium Pain Center, 2406 E. Empire St., Bloomington, IL, 61704, USA
- Lumbrera Research, 33 Derby Way, Bloomington, IL, 61704, USA
| | - Ramsin Benyamin
- Millennium Pain Center, 2406 E. Empire St., Bloomington, IL, 61704, USA
| | - Jorge Orduña-Valls
- Hospital Quirón Salud Valencia, Av. de Blasco Ibáñez, 14, 46010, Valencia, Spain
- Hospital Clínico Universitario de Valencia, Av. de Blasco Ibáñez, 17, 46010, Valencia, Spain
| | - Alejandro Vallejo
- Millennium Pain Center, 2406 E. Empire St., Bloomington, IL, 61704, USA
- Midwestern University, 555 31st Street, Downers Grove, IL, 60515, USA
| | - Samuel M Thomas
- Millennium Pain Center, 2406 E. Empire St., Bloomington, IL, 61704, USA
- College of Osteopathic Medicine, Munroe, 3200 Grand Ave, Des Moines, IA, 50312, USA
| | - David L Cedeño
- Millennium Pain Center, 2406 E. Empire St., Bloomington, IL, 61704, USA
- Lumbrera Research, 33 Derby Way, Bloomington, IL, 61704, USA
| |
Collapse
|
13
|
Thoene M, Bejer-Olenska E, Wojtkiewicz J. The Current State of Osteoarthritis Treatment Options Using Stem Cells for Regenerative Therapy: A Review. Int J Mol Sci 2023; 24:ijms24108925. [PMID: 37240271 DOI: 10.3390/ijms24108925] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/27/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Articular cartilage has very low metabolic activity. While minor injuries may be spontaneously repaired within the joint by chondrocytes, there is very little chance of a severely impaired joint regenerating itself when damaged. Therefore, any significant joint injury has little chance of spontaneously healing without some type of therapy. This article is a review that will examine the causes of osteoarthritis, both acute and chronic, and how it may be treated using traditional methods as well as with the latest stem cell technology. The latest regenerative therapy is discussed, including the use and potential risks of mesenchymal stem cells for tissue regeneration and implantation. Applications are then discussed for the treatment of OA in humans after using canine animal models. Since the most successful research models of OA were dogs, the first applications for treatment were veterinary. However, the treatment options have now advanced to the point where patients suffering from osteoarthritis may be treated with this technology. A survey of the literature was performed in order to determine the current state of stem cell technology being used in the treatment of osteoarthritis. Then, the stem cell technology was compared with traditional treatment options.
Collapse
Affiliation(s)
- Michael Thoene
- Department of Medical Biology, School of Public Health, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Ewa Bejer-Olenska
- Department of Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| |
Collapse
|
14
|
Meng S, Tang C, Deng M, Yuan J, Fan Y, Gao S, Feng Y, Yang J, Chen C. Tropoelastin-Pretreated Exosomes from Adipose-Derived Stem Cells Improve the Synthesis of Cartilage Matrix and Alleviate Osteoarthritis. J Funct Biomater 2023; 14:jfb14040203. [PMID: 37103293 PMCID: PMC10143921 DOI: 10.3390/jfb14040203] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 04/28/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have recently been widely used to treat osteoarthritis (OA). Our prior research shows that tropoelastin (TE) increases MSC activity and protects knee cartilage from OA-related degradation. The underlying mechanism might be that TE regulates the paracrine of MSCs. Exosomes (Exos), the paracrine secretion of MSCs, have been found to protect chondrocytes, reduce inflammation, and preserve the cartilage matrix. In this study, we used Exos derived from TE-pretreated adipose-derived stem cells (ADSCs) (TE-ExoADSCs) as an injection medium, and compared it with Exos derived from unpretreated ADSCs (ExoADSCs). We found that TE-ExoADSCs could effectively enhance the matrix synthesis of chondrocytes in vitro. Moreover, TE pretreatment increased the ability of ADSCs to secrete Exos. In addition, compared with ExoADSCs, TE-ExoADSCs exhibited therapeutic effects in the anterior cruciate ligament transection (ACLT)-induced OA model. Further, we observed that TE altered the microRNA expression in ExoADSCs and identified one differentially upregulated microRNA: miR-451-5p. In conclusion, TE-ExoADSCs helped maintain the chondrocyte phenotype in vitro, and promoted cartilage repair in vivo. These therapeutic effects might be related with the altered expression of miR-451-5p in the ExoADSCs. Thus, the intra-articular delivery of Exos derived from ADSCs with TE pretreatment could be a new approach to treat OA.
Collapse
Affiliation(s)
- Shuo Meng
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Cong Tang
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Muhai Deng
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Jie Yuan
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Yanli Fan
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Shasha Gao
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| | - Yong Feng
- Department of Orthopaedic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing 400014, China
| | - Junjun Yang
- Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Chongqing University, Ministry of Education, Chongqing 400044, China
| | - Cheng Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
15
|
CD44 mediates hyaluronan to promote the differentiation of human amniotic mesenchymal stem cells into chondrocytes. Biotechnol Lett 2023; 45:411-422. [PMID: 36680638 DOI: 10.1007/s10529-022-03322-2] [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: 06/09/2022] [Revised: 11/02/2022] [Accepted: 11/14/2022] [Indexed: 01/22/2023]
Abstract
OBJECTIVES CD44 is the major receptor for hyaluronan (HA), but its effect on HA-induced differentiation of human amnion mesenchymal stem cells into chondrocytes is unclear. This study aimed to investigate the effects and mechanisms of CD44 in HA-induced chondrogenesis. METHODS Immunocytochemistry and toluidine blue staining were used to assess the secretion of type II collagen and aggrecan, respectively. qRT-PCR and western blotting were performed to evaluate the expression of key genes and proteins. RESULTS The expression of aggrecan and type II collagen was downregulated after using the anti-CD44 antibody (A3D8). The transcriptional levels of chondrocytes‑associated genes SRY‑box transcription factor 9, aggrecan, and collagen type II alpha 1 chain were also decreased. Thus, CD44 may mediate HA-induced differentiation of hAMSCs into chondrocytes. Further investigation indicated that expression of phosphorylated (p)‑Erk1/2 and p‑Smad2 decreased following CD44 inhibition. The changes in the expression of p-Erk1/2 and p-Smad2 were consistent after using the ERK1/2 inhibitor (U0126) and agonist (EGF), respectively. After administering the p-Smad2 inhibitor, the expression levels of p-ERK1/2 and p-Smad2 appeared downregulated. The results showed crosstalk between Erk1/2 and Smad2. Moreover, inhibition of p-Erk1/2 and p-Smad2 significantly reduced the accumulation of aggrecan and type II collagen. CONCLUSION These data indicate that CD44 mediates HA-induced differentiation of hAMSCs into chondrocytes by regulating Erk1/2 and Smad2 signaling.
Collapse
|
16
|
Dilogo IH, Lubis AMT, Perwida NG, Sani SA, Rasyidah RA, Hartanto BR. The Efficacy of Intra-articular Umbilical Cord-Mesenchymal Stem Cell Injection for Knee Osteoarthritis: a Systematic Review. CURRENT STEM CELL REPORTS 2023. [DOI: 10.1007/s40778-023-00223-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
17
|
Madrazo-Ibarra A, Barve R, Carroll KM, Proner R, Topar C, Ibarra C, Coleman SH, Vad V. Carboplasty, a Simple Tibial Marrow Technique for Knee Osteoarthritis: A Placebo-Controlled Randomized Trial. Orthop J Sports Med 2022; 10:23259671221143743. [PMID: 36582935 PMCID: PMC9793044 DOI: 10.1177/23259671221143743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/26/2022] [Indexed: 12/24/2022] Open
Abstract
Background Carboplasty is a new minimally invasive technique for knee osteoarthritis (OA) that consists of injecting tibial marrow aspirate into the bone-cartilage interface as well as intra-articularly. Purpose To compare the clinical and imaging outcomes, as well as the safety, of carboplasty for symptomatic knee OA in a placebo-controlled trial. Study Design Randomized controlled trial; Level of evidence, 1. Methods The authors conducted a randomized controlled trial to compare carboplasty with placebo for the treatment of symptomatic knee OA. Patients who had failed medical treatment and had bone edema on magnetic resonance imaging (MRI) were randomized in a 1:1 ratio to carboplasty or placebo. The primary outcome of the study was the Numeric Pain Rating Scale (NPRS) for the knee at 1 year (scores range from 0 to 10, with a higher score indicating worse pain). Secondary outcomes were the Knee injury and Osteoarthritis Outcome Score (KOOS), treatment responder rate (based on achieving the minimal clinically important difference of the NPRS), MRI bone edema reduction, and treatment safety. Results In total, 50 patients (25 carboplasty vs 25 placebo) were enrolled and followed up with for an average of 18 months (range, 14-24 months). The average NPRS at baseline decreased from 7.1 ± 0.9 to 2.9 ± 2.1 (P < .001) at 1 year in the carboplasty group and from 7.7 ± 0.9 to 4.9 ± 2.2 (P < .001) in the placebo group. On average, patients after carboplasty improved 60% from their initial NPRS, and patients after placebo improved 37% (P = .003). Patients had a statistically significantly greater improvement from baseline in all KOOS subscales in the carboplasty group compared with the placebo group (P < .001). The responder rates were 96% for carboplasty and 76% for placebo (P = .098). Bone edema was reduced in 72% of patients in the carboplasty group and 44% of patients in the placebo group (P = .045). Neither group had adverse events related to treatment. Conclusion Carboplasty resulted in greater pain reduction, a significantly greater improvement in all KOOS subscales, and a similar safety profile compared with placebo in patients with symptomatic knee OA and bone edema. Registration ISRCTN69838191 (ISRCT Registry).
Collapse
Affiliation(s)
| | | | | | - Robert Proner
- Hospital for Special Surgery, New York, New York, USA
| | | | | | | | - Vijay Vad
- Hospital for Special Surgery, New York, New York, USA.,Vijay Vad, MD, Hospital for Special Surgery, 519 East 72nd
Street, Suite 203, New York, NY 10021, USA (
)
| |
Collapse
|
18
|
Islam MS, Ebrahimi-Barough S, Al Mahtab M, Shirian S, Aghayan HR, Arjmand B, Allahverdi A, Ranjbar FE, Sadeg AB, Ai J. Encapsulation of rat bone marrow-derived mesenchymal stem cells (rBMMSCs) in collagen type I containing platelet-rich plasma for osteoarthritis treatment in rat model. Prog Biomater 2022; 11:385-396. [PMID: 36271317 DOI: 10.1007/s40204-022-00200-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022] Open
Abstract
Osteoarthritis (OA) is the most common form of degenerative joint disease, affecting more than 25% of the adults despite its prevalence in the elderly population. Most of the current therapeutic modalities aim at symptomatic treatment which lingers the disease progression. In recent years, regenerative medicine such as stem cell transplantation and tissue engineering has been suggested as a potential curative intervention for OA. The objective of this current study was to assess the safety and efficacy of an injectable tissue-engineered construct composed of rat bone marrow mesenchymal stem cells (rBMMSCs), platelet-rich plasma (PRP), and collagen type I in rat model of OA. To produce collagen type I, PRP and rBMMSCs, male Wistar rats were ethically euthanized. After isolation, culture, expansion and characterization of rBMMSCs, tissue-engineered construct was formed by a combination of appropriate amount of collagen type I, PRP and rBMMSCs. In vitro studies were conducted to evaluate the effect of PRP on chondrogenic differentiation capacity of encapsulated cells. In the following, the tissue-engineered construct was injected in knee joints of rat models of OA (24 rats in 4 groups: OA, OA + MSC, OA + collagen + MSC + PRP, OA + MSC + collagen). After 6 weeks, the animals were euthanized and knee joint histopathology examinations of knee joint samples were performed to evaluate the effect of each treatment on OA. Tissue-engineered construct was successfully manufactured and in vitro assays demonstrated the relevant chondrogenic genes and proteins expression were higher in PRP group than that of others. Histopathological findings of the knee joint samples showed favorable regenerative effect of rBMMSCs + PRP + collagen group compared to others. We introduced an injectable tissue-engineered product composed of rBMMSCs + PRP + collagen with potential regenerative effect on cartilage that has been damaged by OA.
Collapse
Affiliation(s)
- Md Shahidul Islam
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mamun Al Mahtab
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Dhaka, Bangladesh
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shefa Neuroscience Research Center, Khatam-Alanbia Hospital, Tehran, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Allahverdi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Esmaeili Ranjbar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Amin Bigham Sadeg
- Department of Clinical Sciences, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shiraz Molecular Pathology Research Center, Dr Daneshbod Path Lab, Shiraz, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Dias IE, Viegas CA, Requicha JF, Saavedra MJ, Azevedo JM, Carvalho PP, Dias IR. Mesenchymal Stem Cell Studies in the Goat Model for Biomedical Research-A Review of the Scientific Literature. BIOLOGY 2022; 11:1276. [PMID: 36138755 PMCID: PMC9495984 DOI: 10.3390/biology11091276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells, defined by their ability to self-renew, while maintaining the capacity to differentiate into different cellular lineages, presumably from their own germinal layer. MSCs therapy is based on its anti-inflammatory, immunomodulatory, and regenerative potential. Firstly, they can differentiate into the target cell type, allowing them to regenerate the damaged area. Secondly, they have a great immunomodulatory capacity through paracrine effects (by secreting several cytokines and growth factors to adjacent cells) and by cell-to-cell contact, leading to vascularization, cellular proliferation in wounded tissues, and reducing inflammation. Currently, MSCs are being widely investigated for numerous tissue engineering and regenerative medicine applications. Appropriate animal models are crucial for the development and evaluation of regenerative medicine-based treatments and eventual treatments for debilitating diseases with the hope of application in upcoming human clinical trials. Here, we summarize the latest research focused on studying the biological and therapeutic potential of MSCs in the goat model, namely in the fields of orthopedics, dermatology, ophthalmology, dentistry, pneumology, cardiology, and urology fields.
Collapse
Affiliation(s)
- Inês E. Dias
- CITAB—Centre for the Research and Technology of Agro-Environmental and Biological Sciences, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, 5000-801 Vila Real, Portugal
| | - Carlos A. Viegas
- CITAB—Centre for the Research and Technology of Agro-Environmental and Biological Sciences, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, 5000-801 Vila Real, Portugal
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences (ECAV), UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
- CECAV—Centre for Animal Sciences and Veterinary Studies, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
- AL4AnimalS—Associate Laboratory for Animal and Veterinary Sciences, 1300-477 Lisboa, Portugal
| | - João F. Requicha
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences (ECAV), UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
- CECAV—Centre for Animal Sciences and Veterinary Studies, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
- AL4AnimalS—Associate Laboratory for Animal and Veterinary Sciences, 1300-477 Lisboa, Portugal
| | - Maria J. Saavedra
- CITAB—Centre for the Research and Technology of Agro-Environmental and Biological Sciences, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, 5000-801 Vila Real, Portugal
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences (ECAV), UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Jorge M. Azevedo
- CECAV—Centre for Animal Sciences and Veterinary Studies, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
- AL4AnimalS—Associate Laboratory for Animal and Veterinary Sciences, 1300-477 Lisboa, Portugal
- Department of Animal Science, ECAV, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Pedro P. Carvalho
- CIVG—Vasco da Gama Research Center, University School Vasco da Gama (EUVG), Av. José R. Sousa Fernandes, Campus Universitário, Lordemão, 3020-210 Coimbra, Portugal
- Vetherapy—Research and Development in Biotechnology, 3020-210 Coimbra, Portugal
| | - Isabel R. Dias
- CITAB—Centre for the Research and Technology of Agro-Environmental and Biological Sciences, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, 5000-801 Vila Real, Portugal
- Department of Veterinary Sciences, School of Agricultural and Veterinary Sciences (ECAV), UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
- CECAV—Centre for Animal Sciences and Veterinary Studies, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
- AL4AnimalS—Associate Laboratory for Animal and Veterinary Sciences, 1300-477 Lisboa, Portugal
| |
Collapse
|
20
|
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
|
21
|
Fernandes TL, Bueno DF, Shimomura K, Shao Z, Gomoll AH. Editorial: Tissue Engineering and Cell Therapy for Cartilage Restoration. Front Cell Dev Biol 2022; 10:947588. [PMID: 35846365 PMCID: PMC9284499 DOI: 10.3389/fcell.2022.947588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/06/2022] [Indexed: 12/04/2022] Open
Affiliation(s)
- Tiago Lazzaretti Fernandes
- Sports Medicine Division, Institute of Orthopedics and Traumatology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Hospital Sírio-Libanês, São Paulo, Brazil
- *Correspondence: Tiago Lazzaretti Fernandes,
| | | | - Kazunori Shimomura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Zhenxing Shao
- Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Andreas H. Gomoll
- Department of Orthopaedic Surgery, Hospital for Special Surgery, New York, NY, United States
| |
Collapse
|
22
|
Application Effect of Different Concentrations of Platelet-Rich Plasma Combined with Quadriceps Training on Cartilage Repair of Knee Osteoarthritis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7878064. [PMID: 35111289 PMCID: PMC8801772 DOI: 10.1155/2022/7878064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 12/20/2022]
Abstract
We investigated the application effect of different concentrations of platelet-rich plasma (PRP) combined with quadriceps training on cartilage repair of knee osteoarthritis. Data of 37 patients with knee osteoarthritis (KOA) treated in our hospital (November 2019–February 2021) were retrospectively analyzed and the patients were divided into low concentration group (LCG) (n = 12), medium concentration group (MCG) (n = 12), and high concentration group (HCG) (n = 13) according to the order of admission. All patients received quadriceps training. Three groups above received knee injection of PRP, and the platelet concentrations were 1000–1400 × 109/L, 1400–1800 × 109/L, and 1800–2100 × 109/L, respectively. Articular cartilage thickness of the medial and lateral femur, knee joint function scores, inflammatory factor levels, and matrix metalloproteinases (MMPs) levels were compared. After treatment, compared with the MCG and HCG, articular cartilage thickness of the medial and lateral femur of the diseased side in the LCG was obviously lower (P < 0.05). At 2 months after treatment (T3), compared with the HCG, articular cartilage thickness of the medial and lateral femur of the diseased side in the MCG was obviously higher (P < 0.05), without remarkable difference in articular cartilage thickness of the medial and lateral femur of the healthy side among three groups (P > 0.05). After treatment, compared with the LCG, knee joint function scores of the MCG and HCG were obviously better (P < 0.001). Compared with the HCG, the knee function score at T3 in the MCG was obviously better (P < 0.001). After treatment, compared with the LCG, inflammatory factor levels and levels of MMPs in the MCG and HCG were obviously lower (P < 0.05). Compared with the HCG, inflammatory factor levels and levels of MMPs at T3 in the MCG were obviously lower (P < 0.05). PRP combined with quadriceps training can accelerate cartilage repair of patients with KOA and reduce inflammatory factor levels and levels of MMPs, but the treatment effect of PRP depends on platelet concentration, with the best range of 1400–1800 × 109/L. Too high or too low platelet concentrations will affect recovery of knee function.
Collapse
|
23
|
Veronesi F, Fini M, Martini L, Berardinelli P, Russo V, Filardo G, Di Matteo B, Marcacci M, Kon E. In Vivo Model of Osteoarthritis to Compare Allogenic Amniotic Epithelial Stem Cells and Autologous Adipose Derived Cells. BIOLOGY 2022; 11:biology11050681. [PMID: 35625409 PMCID: PMC9138403 DOI: 10.3390/biology11050681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 12/20/2022]
Abstract
Simple Summary An early resolution of osteoarthritis (OA), through minimally invasive orthobiological solutions, would be important to enable a return to daily and sport activities, and delay prosthesis solutions. No study has yet evaluated amniotic epithelial stem cells (AECs) in OA. They could be considered a valid alternative to adipose derived cells, expanded or concentrated, because they differentiate into three lineages and express mesenchymal and embryonic markers, without a tumorigenic phenotype. The innovative aspects of this study are the comparison of three injective orthobiological treatments, the in vivo use of AECs in OA, and the evaluation of structural and inflammatory fronts of OA for up to six months. Abstract The challenge of osteoarthritis (OA) is to find a minimally invasive orthobiological therapy to contrast OA progression, on inflammatory and structural fronts. The aim of the present study is to compare the effects of an intra-articular injection of three orthobiological treatments, autologous culture expanded adipose-derived mesenchymal stromal cells (ADSCs), autologous stromal vascular fraction (SVF) and allogenic culture expanded amniotic epithelial stem cells (AECs), in an animal model of OA. OA was induced in 24 sheep by bilateral lateral meniscectomy and, at 3 and 6 months post-treatment, the results were analyzed with macroscopy, histology, histomorphometry, and biochemistry. All the three treatments showed better results than control (injection of NaCl), but SVF and AECs showed superiority over ADSCs, because they induced higher cartilage regeneration and lower inflammation. SVF showed better results than AECs at 3 and 6 months. To conclude, SVF seems to be more favorable than the other biological options, because it is easily obtained and rapidly used after harvesting, with good healing potential. AECs cause no discomfort and could be also considered for the treatment of OA joints.
Collapse
Affiliation(s)
- Francesca Veronesi
- Complex Structure of Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136 Bologna, Italy; (F.V.); (M.F.)
| | - Milena Fini
- Complex Structure of Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136 Bologna, Italy; (F.V.); (M.F.)
| | - Lucia Martini
- Complex Structure of Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136 Bologna, Italy; (F.V.); (M.F.)
- Correspondence: ; Tel.: +39-0516366557
| | - Paolo Berardinelli
- Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.)
| | - Valentina Russo
- Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy; (P.B.); (V.R.)
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, Via Di Barbiano 1/10, 40136 Bologna, Italy;
| | - Berardo Di Matteo
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy; (B.D.M.); (M.M.); (E.K.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini n. 4, 20090 Milan, Italy
| | - Maurilio Marcacci
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy; (B.D.M.); (M.M.); (E.K.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini n. 4, 20090 Milan, Italy
| | - Elizaveta Kon
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy; (B.D.M.); (M.M.); (E.K.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini n. 4, 20090 Milan, Italy
| |
Collapse
|
24
|
Effects of polyacrylamide hydrogel used in the treatment of osteoarthritis on mesenchymal stem cells and human osteoblasts. JOURNAL OF SURGERY AND MEDICINE 2022. [DOI: 10.28982/josam.1006577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
25
|
Insight in Hypoxia-Mimetic Agents as Potential Tools for Mesenchymal Stem Cell Priming in Regenerative Medicine. Stem Cells Int 2022; 2022:8775591. [PMID: 35378955 PMCID: PMC8976669 DOI: 10.1155/2022/8775591] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Hypoxia-mimetic agents are new potential tools in MSC priming instead of hypoxia incubators or chambers. Several pharmaceutical/chemical hypoxia-mimetic agents can be used to induce hypoxia in the tissues: deferoxamine (DFO), dimethyloxaloylglycine (DMOG), 2,4-dinitrophenol (DNP), cobalt chloride (CoCl2), and isoflurane (ISO). Hypoxia-mimetic agents can increase cell proliferation, preserve or enhance differentiation potential, increase migration potential, and induce neovascularization in a concentration- and stem cell source-dependent manner. Moreover, hypoxia-mimetic agents may increase HIF-1α, changing the metabolism and enhancing glycolysis like hypoxia. So, there is clear evidence that treatment with hypoxia-mimetic agents is beneficial in regenerative medicine, preserving stem cell capacities. These agents are not studied so wildly as hypoxia but, considering the low cost and ease of use, are believed to find application as pretreatment of many diseases such as ischemic heart disease and myocardial fibrosis and promote cardiac and cartilage regeneration. The knowledge of MSC priming is critical in evaluating safety procedures and use in clinics. In this review, similarities and differences between hypoxia and hypoxia-mimetic agents in terms of their therapeutic efficiency are considered in detail. The advantages, challenges, and future perspectives in MSC priming with hypoxia mimetic agents are also discussed.
Collapse
|
26
|
McKinney JM, Pucha KA, Doan TN, Wang L, Weinstock LD, Tignor BT, Fowle KL, Levit RD, Wood LB, Willett NJ. Sodium alginate microencapsulation of human mesenchymal stromal cells modulates paracrine signaling response and enhances efficacy for treatment of established osteoarthritis. Acta Biomater 2022; 141:315-332. [PMID: 34979327 DOI: 10.1016/j.actbio.2021.12.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 01/15/2023]
Abstract
Mesenchymal stromal cells (MSCs) have shown promise as osteoarthritis (OA) treatments; however, effective translation has been limited by high variability and heterogeneity of MSCs, suboptimal delivery strategies, and poor understanding of critical quality and potency attributes. Furthermore, most pre-clinical studies of MSC therapeutics for OA have focused on delaying OA development and not on treating established OA, which brings added clinical relevance. Thus, the objective of the current study was to assess the effects of sodium alginate microencapsulation on human MSC (hMSC) secretion of immunomodulatory cytokines in an OA microenvironment and therapeutic efficacy in treating established OA. A Medial Meniscal Transection (MMT) pre-clinical model of OA was implemented. Three weeks post-surgery, after OA was established, intra-articular injections of encapsulated hMSCs or nonencapsulated hMSCs were administered. Six weeks post-surgery, microstructural changes in the knee joint were quantified using microCT. Encapsulated hMSCs reduced articular cartilage degeneration and subchondral bone remodeling. A multiplexed immunoassay panel was used to profile the in vitro secretome of hMSCs in response to IL-1β. Nonencapsulated hMSCs showed an indiscriminate increase in all cytokines in response to IL-1β while encapsulated hMSCs showed a targeted secretory response with increased expression of pro-inflammatory (IL-1β, IL-6, IL-7, IL-8), anti-inflammatory (IL-1RA), and chemotactic (G-CSF, MDC, IP10) cytokines. These data show that sodium alginate microencapsulation can modulate hMSC paracrine signaling and enhance the therapeutic efficacy of the hMSCs in treating established OA. This cytokine profile provides a foundation for the identification of key factors affecting the overall potency of hMSC therapeutics for OA. STATEMENT OF SIGNIFICANCE: While there has been considerable interest in material based MSC encapsulation for treatment of OA, there are critical gaps in our translational understanding of these biomaterial-based technologies for OA. More specifically, previous studies have several important limitations: (1) they have been largely focused on preventing OA development, which limits their translational utility and (2) little prior work has been done to delineate potential routes/mechanisms by which material encapsulation alters MSC therapeutic action. In our manuscript, we aimed to fill these gaps in knowledge by testing the hypotheses that: (1) hMSC encapsulation can attenuate established disease progression, which is a more clinically relevant scenario and (2) hMSC encapsulation significantly changes the secreted paracrine factors from hMSCs.
Collapse
Affiliation(s)
- Jay M McKinney
- Research Division, VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA; Department of Orthopaedics, Emory University, 49 Jesse Hill Jr Dr SE, Atlanta, GA 30303, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Krishna A Pucha
- Research Division, VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA
| | - Thanh N Doan
- Research Division, VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA; Department of Orthopaedics, Emory University, 49 Jesse Hill Jr Dr SE, Atlanta, GA 30303, USA
| | - Lanfang Wang
- Department of Medicine, Division of Cardiology, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Laura D Weinstock
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Benjamin T Tignor
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Kelsey L Fowle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Rebecca D Levit
- Department of Medicine, Division of Cardiology, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA 30332, USA.
| | - Nick J Willett
- Research Division, VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA; Department of Orthopaedics, Emory University, 49 Jesse Hill Jr Dr SE, Atlanta, GA 30303, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; Phil and Penny Knight Campus for Accelerating Scientific Impact, 6231 University of Oregon, Eugene, Oregon, USA.
| |
Collapse
|
27
|
Wang G, Xing D, Liu W, Zhu Y, Liu H, Yan L, Fan K, Liu P, Yu B, Li JJ, Wang B. Preclinical studies and clinical trials on mesenchymal stem cell therapy for knee osteoarthritis: A systematic review on models and cell doses. Int J Rheum Dis 2022; 25:532-562. [PMID: 35244339 DOI: 10.1111/1756-185x.14306] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/30/2022] [Accepted: 02/07/2022] [Indexed: 12/15/2022]
Abstract
AIM To provide a systematic analysis of the study design in knee osteoarthritis (OA) preclinical studies, focusing on the characteristics of animal models and cell doses, and to compare these to the characteristics of clinical trials using mesenchymal stem cells (MSCs) for the treatment of knee OA. METHOD A systematic and comprehensive search was conducted using the PubMed, Web of Science, Ovid, and Embase electronic databases for research papers published in 2009-2020 on testing MSC treatment in OA animal models. The PubMed database and ClinicalTrials.gov website were used to search for published studies reporting clinical trials of MSC therapy for knee OA. RESULTS In total, 9234 articles and two additional records were retrieved, of which 120 studies comprising preclinical and clinical studies were included for analysis. Among the preclinical studies, rats were the most commonly used species for modeling knee OA, and anterior cruciate ligament transection was the most commonly used method for inducing OA. There was a correlation between the cell dose and body weight of the animal. In clinical trials, there was large variation in the dose of MSCs used to treat knee OA, ranging from 1 × 106 to 200 × 106 cells with an average of 37.91 × 106 cells. CONCLUSION Mesenchymal stem cells have shown great potential in improving pain relief and tissue protection in both preclinical and clinical studies of knee OA. Further high-quality preclinical and clinical studies are needed to explore the dose effectiveness relationship of MSC therapy and to translate the findings from preclinical studies to humans.
Collapse
Affiliation(s)
- Guishan Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China.,Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Beijing, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, China
| | - Yuanyuan Zhu
- Department of Pharmacy, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Haifeng Liu
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Lei Yan
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Kenan Fan
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Peidong Liu
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Baofeng Yu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Jiao Jiao Li
- Faculty of Engineering and IT, School of Biomedical Engineering, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Bin Wang
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China.,Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
28
|
Wang B, Liu W, Li JJ, Chai S, Xing D, Yu H, Zhang Y, Yan W, Xu Z, Zhao B, Du Y, Jiang Q. A low dose cell therapy system for treating osteoarthritis: In vivo study and in vitro mechanistic investigations. Bioact Mater 2022; 7:478-490. [PMID: 34466747 PMCID: PMC8379370 DOI: 10.1016/j.bioactmat.2021.05.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be effective in alleviating the progression of osteoarthritis (OA). However, low MSC retention and survival at the injection site frequently require high doses of cells and/or repeated injections, which are not economically viable and create additional risks of complications. In this study, we produced MSC-laden microcarriers in spinner flask culture as cell delivery vehicles. These microcarriers containing a low initial dose of MSCs administered through a single injection in a rat anterior cruciate ligament (ACL) transection model of OA achieved similar reparative effects as repeated high doses of MSCs, as evaluated through imaging and histological analyses. Mechanistic investigations were conducted using a co-culture model involving human primary chondrocytes grown in monolayer, together with MSCs grown either within 3D constructs or as a monolayer. Co-culture supernatants subjected to secretome analysis showed significant decrease of inflammatory factors in the 3D group. RNA-seq of co-cultured MSCs and chondrocytes using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed processes relating to early chondrogenesis and increased extracellular matrix interactions in MSCs of the 3D group, as well as phenotypic maintenance in the co-cultured chondrocytes. The cell delivery platform we investigated may be effective in reducing the cell dose and injection frequency required for therapeutic applications.
Collapse
Affiliation(s)
- Bin Wang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Department of Orthopaedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 10081, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Senlin Chai
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Hongsheng Yu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenjin Yan
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Zhihong Xu
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Bin Zhao
- Department of Orthopaedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| |
Collapse
|
29
|
Cell-based regenerative joint therapy: a hot topic. Knee Surg Sports Traumatol Arthrosc 2022; 30:1129-1131. [PMID: 35362721 PMCID: PMC9007778 DOI: 10.1007/s00167-022-06959-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
30
|
Lee JS, Shim DW, Kang KY, Chae DS, Lee WS. Method Categorization of Stem Cell Therapy for Degenerative Osteoarthritis of the Knee: A Review. Int J Mol Sci 2021; 22:ijms222413323. [PMID: 34948119 PMCID: PMC8704290 DOI: 10.3390/ijms222413323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 01/02/2023] Open
Abstract
Current clinical applications of mesenchymal stem cell therapy for osteoarthritis lack consistency because there are no established criteria for clinical processes. We aimed to systematically organize stem cell treatment methods by reviewing the literature. The treatment methods used in 27 clinical trials were examined and reviewed. The clinical processes were separated into seven categories: cell donor, cell source, cell preparation, delivery methods, lesion preparation, concomitant procedures, and evaluation. Stem cell donors were sub-classified as autologous and allogeneic, and stem cell sources included bone marrow, adipose tissue, peripheral blood, synovium, placenta, and umbilical cord. Mesenchymal stem cells can be prepared by the expansion or isolation process and attached directly to cartilage defects using matrices or injected into joints under arthroscopic observation. The lesion preparation category can be divided into three subcategories: chondroplasty, microfracture, and subchondral drilling. The concomitant procedure category describes adjuvant surgery, such as high tibial osteotomy. Classification codes were assigned for each subcategory to provide a useful and convenient method for organizing documents associated with stem cell treatment. This classification system will help researchers choose more unified treatment methods, which will facilitate the efficient comparison and verification of future clinical outcomes of stem cell therapy for osteoarthritis.
Collapse
Affiliation(s)
- Jae Sun Lee
- Stem Cell Therapy Center, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Korea;
| | - Dong Woo Shim
- Department of Orthopedic Surgery, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Korea;
| | - Kyung-Yil Kang
- Department of Medicine, Catholic Kwandong Graduate School, Gangneung-si 25601, Korea;
| | - Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Korea;
- Correspondence: (D.-S.C.); (W.-S.L.); Tel.: +82-32-290-3878 (D.-S.C.); +82-2-2019-3410 (W.-S.L.); Fax: +82-32-290-3879 (D.-S.C.); +82-2-573-5393 (W.-S.L.)
| | - Woo-Suk Lee
- Department of Orthopedic Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06276, Korea
- Correspondence: (D.-S.C.); (W.-S.L.); Tel.: +82-32-290-3878 (D.-S.C.); +82-2-2019-3410 (W.-S.L.); Fax: +82-32-290-3879 (D.-S.C.); +82-2-573-5393 (W.-S.L.)
| |
Collapse
|
31
|
Ding W, Xu YQ, Zhang Y, Li AX, Qiu X, Wen HJ, Tan HB. Efficacy and Safety of Intra-Articular Cell-Based Therapy for Osteoarthritis: Systematic Review and Network Meta-Analysis. Cartilage 2021; 13:104S-115S. [PMID: 32693632 PMCID: PMC8808819 DOI: 10.1177/1947603520942947] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a chronic joint disease characterized by degeneration of articular cartilage and secondary osteogenesis. Cell-based agents, such as mesenchymal stem cells, have turned into the most extensively explored new therapeutic agents for OA. However, evidence-based research is still lacking. METHODS We searched public databases up to February 2020 and only included randomized controlled trials. The outcomes included the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), the Knee Injury and Osteoarthritis Outcome Score (KOOS), the visual analogue scale (VAS) score, and serious adverse events (SAEs). A network meta-analysis was also performed in this work. RESULTS We included 13 studies in the meta-analysis. The effect size showed that cell-based therapy did not significantly reduce the WOMAC score at the 6-month follow-up (standard mean difference [SMD] -3.6; 95% confidence interval [CI] -0.90 to 0.18; P = 0.1928). However, cell-based therapy significantly improved the KOOS at the 12-month follow-up (SMD 0.68; 95% CI 0.07-1.30; P = 0.0288) and relieved pain (SMD -1.05; 95% CI -1.46 to -0.64; P < 0.0001). The findings also indicated that high-dosage adipose-derived mesenchymal stem cells (ADMSCs) may be more advantageous in terms of long-term effects. CONCLUSIONS Cell-based therapy had a better effect on KOOS improvement and pain relief without safety concerns. However, cell-based therapy did not show a benefit in terms of the WOMAC. Allogeneic cells might have advantages compared to controls in the WOMAC and KOOS scores. The long-term effect of high-dose ADMSC treatment for OA is worthy of further study.
Collapse
Affiliation(s)
- Wei Ding
- Medical College, Yunnan University of
Business Management, Kunming, Yunnan, People’s Repuplic of China
| | - Yong-qing Xu
- Department of Orthopaedic, The 920th
Hospital of Joint Logistics Support Force, Kunming, Yunnan, People’s Republic of
China
| | - Ying Zhang
- Department of Orthopaedic, The 920th
Hospital of Joint Logistics Support Force, Kunming, Yunnan, People’s Republic of
China
| | - An-xu Li
- Department of Orthopaedic, The 920th
Hospital of Joint Logistics Support Force, Kunming, Yunnan, People’s Republic of
China
| | - Xiong Qiu
- Department of Orthopaedic, The 920th
Hospital of Joint Logistics Support Force, Kunming, Yunnan, People’s Republic of
China
| | - Hong-jie Wen
- Department of Orthopedic Surgery, The
Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, Peoples’
Republic of China
| | - Hong-bo Tan
- Department of Orthopaedic, The 920th
Hospital of Joint Logistics Support Force, Kunming, Yunnan, People’s Republic of
China,Hong-bo Tan, Department of Orthopaedic, The
920th Hospital of Joint Logistics Support Force, NO. 212, Daguan Road, Xishan
District, Kunming, Yunnan 650020, People’s Republic of China.
| |
Collapse
|
32
|
Pagani S, Veronesi F, Giavaresi G, Filardo G, Papio T, Romandini I, Fini M. Autologous Protein Solution Effect on Chondrogenic Differentiation of Mesenchymal Stem Cells from Adipose Tissue and Bone Marrow in an Osteoarthritic Environment. Cartilage 2021; 13:225S-237S. [PMID: 33583216 PMCID: PMC8804741 DOI: 10.1177/1947603521993217] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is an inflammatory and degenerative disease, and the numerous treatments currently used are not fully effective. Mesenchymal stem cells (MSCs) and platelet-rich plasma (PRP) are proposed for OA treatment as biologic therapies. The aim of the study was to observe the role of autologous protein solution (APS), a type of PRP, on chondrogenic differentiation of 2 types of MSCs, from bone marrow (BMSCs) and adipose tissue (ADSCs), in an in vitro osteoarthritic microenvironment. DESIGN Inflammatory culture conditions, mimicking OA, were obtained by adding interleukin-1β (IL-1β) and tumor necrosis factor α (TNFα), or synovial fluid from patient osteoarthritic knees (OSF), to the culture medium. MSCs were then treated with APS. RESULTS After 1 month of culture, both cell types formed mature micromasses, partially altered in the presence of IL-1β and TNFα but quite preserved with OSF. Inflammatory conditions hindered differentiation in terms of gene expression, not counterbalanced by APS. APS triggered type I collagen deposition and above all contributed to decrease the expression of metalloproteinases in the most aggressive conditions (IL-1β and TNFα in the culture medium). ADSCs originated micromasses more mature and less prone toward osteogenic lineage than BMSCs, thus showing to better adapt in an aggressive environment than BMSC. CONCLUSIONS APS seems to act better on inflammation front and, between cell types, ADSCs respond better to the inflammatory microenvironment of OA and to the treatment with APS than BMSCs.
Collapse
Affiliation(s)
- Stefania Pagani
- Complex Structure of Surgical Sciences
and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Veronesi
- Complex Structure of Surgical Sciences
and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy,Francesca Veronesi, Complex Structure of
Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di
Barbiano 1/10, Bologna, 40136, Italy.
| | - Gianluca Giavaresi
- Complex Structure of Surgical Sciences
and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giuseppe Filardo
- Applied and Translational Research
Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Tiziana Papio
- Applied and Translational Research
Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Iacopo Romandini
- 2nd Orthopaedic and Traumatologic
Clinic, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Milena Fini
- Complex Structure of Surgical Sciences
and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
33
|
Bar JK, Lis-Nawara A, Grelewski PG. Dental Pulp Stem Cell-Derived Secretome and Its Regenerative Potential. Int J Mol Sci 2021; 22:ijms222112018. [PMID: 34769446 PMCID: PMC8584775 DOI: 10.3390/ijms222112018] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/11/2022] Open
Abstract
The therapeutic potential of the dental pulp stem (DSC) cell-derived secretome, consisting of various biomolecules, is undergoing intense research. Despite promising in vitro and in vivo studies, most DSC secretome-based therapies have not been implemented in human medicine because the paracrine effect of the bioactive factors secreted by human dental pulp stem cells (hDPSCs) and human exfoliated deciduous teeth (SHEDs) is not completely understood. In this review, we outline the current data on the hDPSC- and SHED-derived secretome as a potential candidate in the regeneration of bone, cartilage, and nerve tissue. Published reports demonstrate that the dental MSC-derived secretome/conditional medium may be effective in treating neurodegenerative diseases, neural injuries, cartilage defects, and repairing bone by regulating neuroprotective, anti-inflammatory, antiapoptotic, and angiogenic processes through secretome paracrine mechanisms. Dental MSC-secretomes, similarly to the bone marrow MSC-secretome activate molecular and cellular mechanisms, which determine the effectiveness of cell-free therapy. Many reports emphasize that dental MSC-derived secretomes have potential application in tissue-regenerating therapy due to their multidirectional paracrine effect observed in the therapy of many different injured tissues.
Collapse
|
34
|
Hsu GCY, Cherief M, Sono T, Wang Y, Negri S, Xu J, Peault B, James AW. Divergent effects of distinct perivascular cell subsets for intra-articular cell therapy in posttraumatic osteoarthritis. J Orthop Res 2021; 39:2388-2397. [PMID: 33512030 PMCID: PMC8319216 DOI: 10.1002/jor.24997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/30/2020] [Accepted: 01/24/2021] [Indexed: 02/04/2023]
Abstract
Intra-articular injection of mesenchymal stem cells has shown benefit for the treatment of osteoarthritis (OA). However, mesenchymal stem/stromal cells at the origin of these clinical results are heterogenous cell populations with limited cellular characterization. Here, two transgenic reporter mice were used to examine the differential effects of two precisely defined perivascular cell populations (Pdgfrα+ and Pdgfrβ+ cells) from white adipose tissue for alleviation of OA. Perivascular mesenchymal cells were isolated from transgenic Pdgfrα-and Pdgfrβ-CreERT2 reporter animals and delivered as a one-time intra-articular dose to C57BL/6J mice after destabilization of the medial meniscus (DMM). Both Pdgfrα+ and Pdgfrβ+ cell preparations improved metrics of cartilage degradation and reduced markers of chondrocyte hypertrophy. While some similarities in cell distribution were identified within the synovial and perivascular spaces, injected Pdgfrα+ cells remained in the superficial layers of articular cartilage, while Pdgfrβ+ cells were more widely dispersed. Pdgfrβ+ cell therapy prevented subchondral sclerosis induced by DMM, while Pdgfrα+ cell therapy had no effect. In summary, while both cell therapies showed beneficial effects in the DMM model, important differences in cell incorporation, persistence, and subchondral sclerosis were identified.
Collapse
Affiliation(s)
- Ginny Ching-Yun Hsu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States;,Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095;,Center For Cardiovascular Science and Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| |
Collapse
|
35
|
H19 Overexpression Improved Efficacy of Mesenchymal Stem Cells in Ulcerative Colitis by Modulating the miR-141/ICAM-1 and miR-139/CXCR4 Axes. DISEASE MARKERS 2021; 2021:7107705. [PMID: 34630738 PMCID: PMC8494579 DOI: 10.1155/2021/7107705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/30/2021] [Accepted: 09/02/2021] [Indexed: 12/30/2022]
Abstract
Overexpression of C-X-C motif chemokine receptor 4 (CXCR4) and intercellular cell adhesion molecule-1 (ICAM-1) may promote homing of mesenchymal stem cells (MSC). In this study, we treated ulcerative colitis animals with MSC preconditioned with or without H19 and compared the therapeutic effect of MSC and MSC-H19. We evaluated the regulatory relationship of H19 vs. miR-141/miR-139 and miR-141/miR-139 vs. ICAM-1/CXCR4. We established an ulcerative colitis mouse model to assess the effect of MSC and MSC-H19. H19 was found to bind to miR-141 and miR-139. The activity of H19 was strongly decreased in cells c-transfected with miR-141/miR-139 and WT H19. ICAM-1 was confirmed to be targeted by miR-141 and CXCR4 was targeted by miR-139. The H19 expression showed a negative regulatory relationship with the miR-141 and miR-139 expression but a positive regulatory relationship with the ICAM-1 and CXCR4 expression. In summary, the overexpression of H19 in MSC downregulated miR-139 and miR-141, thus increasing the activity of their targets ICAM-1 and CXCR4, respectively, to exhibit therapeutic effects in ulcerative colitis.
Collapse
|
36
|
Mahmoudian A, Lohmander LS, Mobasheri A, Englund M, Luyten FP. Early-stage symptomatic osteoarthritis of the knee - time for action. Nat Rev Rheumatol 2021; 17:621-632. [PMID: 34465902 DOI: 10.1038/s41584-021-00673-4] [Citation(s) in RCA: 153] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) remains the most challenging arthritic disorder, with a high burden of disease and no available disease-modifying treatments. Symptomatic early-stage OA of the knee (the focus of this Review) urgently needs to be identified and defined, as efficient early-stage case finding and diagnosis in primary care would enable health-care providers to proactively and substantially reduce the burden of disease through proper management including structured education, exercise and weight management (when needed) and addressing lifestyle-related risk factors for disease progression. Efforts to define patient populations with symptomatic early-stage knee OA on the basis of validated classification criteria are ongoing. Such criteria, as well as the identification of molecular and imaging biomarkers of disease risk and/or progression, would enable well-designed clinical studies, facilitate interventional trials, and aid the discovery and validation of cellular and molecular targets for novel therapies. Treatment strategies, relevant outcomes and ethical issues also need to be considered in the context of the cost-effective management of symptomatic early-stage knee OA. To move forwards, a multidisciplinary and sustained international effort involving all major stakeholders is required.
Collapse
Affiliation(s)
- Armaghan Mahmoudian
- Department of Development & Regeneration, KU Leuven, Leuven, Belgium.,Department of Clinical Sciences Lund, Orthopaedics, Lund University, Lund, Sweden
| | - L Stefan Lohmander
- Department of Clinical Sciences Lund, Orthopaedics, Lund University, Lund, Sweden
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Martin Englund
- Department of Clinical Sciences Lund, Orthopaedics, Lund University, Lund, Sweden
| | - Frank P Luyten
- Department of Development & Regeneration, KU Leuven, Leuven, Belgium.
| |
Collapse
|
37
|
Vahedi P, Moghaddamshahabi R, Webster TJ, Calikoglu Koyuncu AC, Ahmadian E, Khan WS, Jimale Mohamed A, Eftekhari A. The Use of Infrapatellar Fat Pad-Derived Mesenchymal Stem Cells in Articular Cartilage Regeneration: A Review. Int J Mol Sci 2021; 22:ijms22179215. [PMID: 34502123 PMCID: PMC8431575 DOI: 10.3390/ijms22179215] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cartilage is frequently damaged with a limited capacity for repair. Current treatment strategies are insufficient as they form fibrocartilage as opposed to hyaline cartilage, and do not prevent the progression of degenerative changes. There is increasing interest in the use of autologous mesenchymal stem cells (MSC) for tissue regeneration. MSCs that are used to treat articular cartilage defects must not only present a robust cartilaginous production capacity, but they also must not cause morbidity at the harvest site. In addition, they should be easy to isolate from the tissue and expand in culture without terminal differentiation. The source of MSCs is one of the most important factors that may affect treatment. The infrapatellar fat pad (IPFP) acts as an important reservoir for MSC and is located in the anterior compartment of the knee joint in the extra-synovial area. The IPFP is a rich source of MSCs, and in this review, we discuss studies that demonstrate that these cells have shown many advantages over other tissues in terms of ease of isolation, expansion, and chondrogenic differentiation. Future studies in articular cartilage repair strategies and suitable extraction as well as cell culture methods will extend the therapeutical application of IPFP-derived MSCs into additional orthopedic fields, such as osteoarthritis. This review provides the latest research concerning the use of IPFP-derived MSCs in the treatment of articular cartilage damage, providing critical information for the field to grow.
Collapse
Affiliation(s)
- Parviz Vahedi
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran;
| | - Rana Moghaddamshahabi
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta 99628, North Cyprus, Turkey;
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA;
| | - Ayse Ceren Calikoglu Koyuncu
- Materials and Metallurgical Engineering Department, Faculty of Technology, Marmara University, Istanbul 34722, Turkey;
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz 51666-15731, Iran;
| | - Wasim S. Khan
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
- Correspondence: (W.S.K.); (A.E.)
| | - Ali Jimale Mohamed
- Department of Pharmacology, Faculty of Medicine, Somali National University, Mogadishu 801, Somalia;
| | - Aziz Eftekhari
- Department of Toxicology and Pharmacology, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran
- Department of Synthesis and Characterization of Polymers, Polymer Institute, Slovak Academy of Sciences (SAS), Dúbravská cesta, 9, 845 41 Bratislava, Slovakia
- Correspondence: (W.S.K.); (A.E.)
| |
Collapse
|
38
|
Human Umbilical Cord Mesenchymal Stem Cells in Combination with Hyaluronic Acid Ameliorate the Progression of Knee Osteoarthritis. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11146650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The aim of this study is to evaluate the feasibility and usefulness of the human umbilical cord mesenchymal stem cells (hUC-MSCs) and hyaluronan acid (HA) combination to attenuate osteoarthritis progression in the knee while simultaneously providing some insights on the mitigation mechanism. In vitro, the effect of hUC-MSCs with HA treatment on chondrocyte cell viability and the cytokine profile were analyzed. Additionally, the antioxidation capability of hUC-MSCs-CM (conditioned medium) with HA towards H2O2-induced chondrocyte cell damage was evaluated. The HA addition increased the hUC-MSC antioxidation capability and cytokine secretion, such as Dickkopf-related protein 1 (DKK-1) and hepatocyte growth factor (HGF), while no adverse effect on the cell viability was observed. In vivo, the intra-articular injection of hUC-MSCs with HA to a mono-iodoacetate (MIA)-induced knee osteoarthritis (KOA) rat model was performed and investigated. Attenuation of the KOA progression in the MIA-damaged rat model was seen best in hUC-MSCs with a HA combination compared to the vehicle control or each individual element. Combining hUC-MSCs and HA resulted in a synergistic effect, such as increasing the cell therapeutic capability while incurring no observable adverse effects. Therefore, this combinatorial therapy is feasible and has promising potential to ameliorate KOA progression.
Collapse
|
39
|
Aitong W, Leisheng Z, Hao Y. Visualized analyses of investigations upon mesenchymal stem/stromal cell-based cytotherapy and underlying mechanisms for COVID-19 associated ARDS. Curr Stem Cell Res Ther 2021; 17:2-12. [PMID: 34254927 DOI: 10.2174/1574888x16666210712212421] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 11/22/2022]
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) triggered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a widespread pandemic globally and seriously threatened the public health. Patients with COVID-19 infection, and in particular, those with severe pneumonia-associated acute respiratory distress syndrome (ARDS) manifested rapid disease progression and the resultant high mortality and morbidity. Advances in fundamental and clinical studies have suggested the feasibility of mesenchymal stem/stromal cell (MSC)-based therapy as an inspiring alternative for ARDS administration. However, the systematic characteristics of the MSC-based cytotherapy and underlying mechanism for COVID-19 associated ARDS by bibliometric analyses are still unknowable. Herein, we took advantage of visual analyses to reveal the overview of ARDS-associated updates, core authors and focused issues, as well as to summarize the comprehensive knowledge of the keywords, authors, institutions with the aid of indicated software. Meanwhile, we have provided a brief overview on the molecular mechanisms and discussed the safety and efficacy of MSC-based therapy for ARDS on the basis of clinical trials.
Collapse
Affiliation(s)
- Wang Aitong
- National Engineering Research Center of Cell Products, AmCellGene Engineering Co., Ltd, Tianjin 300457, China
| | - Zhang Leisheng
- Institute of Stem Cells, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd., Tianjin, 301700, China
| | - Yu Hao
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
40
|
Proteomic Analysis Reveals Commonly Secreted Proteins of Mesenchymal Stem Cells Derived from Bone Marrow, Adipose Tissue, and Synovial Membrane to Show Potential for Cartilage Regeneration in Knee Osteoarthritis. Stem Cells Int 2021; 2021:6694299. [PMID: 34306096 PMCID: PMC8264516 DOI: 10.1155/2021/6694299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/28/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022] Open
Abstract
Paracrine factors secreted by mesenchymal stem cells (MSCs) reportedly modulate inflammation and reparative processes in damaged tissues and have been explored for knee osteoarthritis (OA) therapy. Although various studies have reported the effects of paracrine factors in knee OA, it is not yet clear which paracrine factors directly affect the regeneration of damaged cartilage and which are secreted under various knee OA conditions. In this study, we cultured MSCs derived from three types of tissues and treated each type with IL-1β and TNF-α or not to obtain conditioned medium. Each conditioned medium was used to analyse the paracrine factors related to cartilage regeneration using liquid chromatography-tandem mass spectrometry. Bone marrow-, adipose tissue-, and synovial membrane-MSCs (all-MSCs) exhibited expression of 93 proteins under normal conditions and 105 proteins under inflammatory conditions. It was confirmed that the types of secreted proteins differed depending on the environmental conditions, and the proteins were validated using ELISA. The results of Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis using a list of proteins secreted by all-MSCs under each condition confirmed that the secreted proteins were closely related to cartilage repair under inflammatory conditions. Protein-protein interaction networks were confirmed to change depending on environmental differences and were found to enhance the secretion of paracrine factors related to cartilage regeneration under inflammatory conditions. In conclusion, our results demonstrated that compared with knee OA conditions, the differential expression proteins may contribute to the regeneration of damaged cartilage. In addition, the detailed information on commonly secreted proteins by all-MSCs provides a comprehensive basis for understanding the potential of paracrine factors to influence tissue repair and regeneration in knee OA.
Collapse
|
41
|
Ragni E, Colombini A, Viganò M, Libonati F, Perucca Orfei C, Zagra L, de Girolamo L. Cartilage Protective and Immunomodulatory Features of Osteoarthritis Synovial Fluid-Treated Adipose-Derived Mesenchymal Stem Cells Secreted Factors and Extracellular Vesicles-Embedded miRNAs. Cells 2021; 10:cells10051072. [PMID: 33946524 PMCID: PMC8147187 DOI: 10.3390/cells10051072] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/20/2022] Open
Abstract
Intra-articular administration of adipose-derived mesenchymal stem cells (ASCs), either in vitro expanded or within adipose tissue-based products obtained at point-of-care, has gained popularity as innovative regenerative medicine approach for osteoarthritis (OA) treatment. ASCs can stimulate tissue repair and immunomodulation through paracrine factors, both soluble and extracellular vesicles (EV) embedded, collectively defining the secretome. Interaction with the degenerative/inflamed environment is a crucial factor in understanding the finely tuned molecular message but, to date, the majority of reports have described ASC-secretome features in resting conditions or under chemical stimuli far from the in vivo environment of degenerated OA joints. In this report, the secretory profile of ASCs treated with native synovial fluid from OA patients was evaluated, sifting 200 soluble factors and 754 EV-embedded miRNAs. Fifty-eight factors and 223 EV-miRNAs were identified, and discussed in the frame of cartilage and immune cell homeostasis. Bioinformatics gave a molecular basis for M2 macrophage polarization, T cell proliferation inhibition and T reg expansion enhancement, as well as cartilage protection, further confirmed in an in vitro model of OA chondrocytes. Moreover, a strong influence on immune cell chemotaxis emerged. In conclusion, obtained molecular data support the regenerative and immunomodulatory properties of ASCs when interacting with osteoarthritic joint environment.
Collapse
Affiliation(s)
- Enrico Ragni
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, I-20161 Milano, Italy; (E.R.); (A.C.); (M.V.); (F.L.); (C.P.O.)
| | - Alessandra Colombini
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, I-20161 Milano, Italy; (E.R.); (A.C.); (M.V.); (F.L.); (C.P.O.)
| | - Marco Viganò
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, I-20161 Milano, Italy; (E.R.); (A.C.); (M.V.); (F.L.); (C.P.O.)
| | - Francesca Libonati
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, I-20161 Milano, Italy; (E.R.); (A.C.); (M.V.); (F.L.); (C.P.O.)
| | - Carlotta Perucca Orfei
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, I-20161 Milano, Italy; (E.R.); (A.C.); (M.V.); (F.L.); (C.P.O.)
| | - Luigi Zagra
- Hip Department, IRCCS Istituto Ortopedico Galeazzi, I-20161 Milano, Italy;
| | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, I-20161 Milano, Italy; (E.R.); (A.C.); (M.V.); (F.L.); (C.P.O.)
- Correspondence: ; Tel.: +39-02-6621-4067
| |
Collapse
|
42
|
Alessio N, Stellavato A, Aprile D, Cimini D, Vassallo V, Di Bernardo G, Galderisi U, Schiraldi C. Timely Supplementation of Hydrogels Containing Sulfated or Unsulfated Chondroitin and Hyaluronic Acid Affects Mesenchymal Stromal Cells Commitment Toward Chondrogenic Differentiation. Front Cell Dev Biol 2021; 9:641529. [PMID: 33912558 PMCID: PMC8072340 DOI: 10.3389/fcell.2021.641529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are currently used for cartilage cell therapy because of their well proven capacity to differentiate in chondrocytes. The advantage of MSC-based therapy is the possibility of producing a high number of chondrocytes for implants. The transplant procedure, however, has some limitations, since MSCs may produce non-functional chondrocytes. This limit has been challenged by cultivating MSC in media with hydrogels containing hyaluronic acid (HA), extractive chondroitin sulfate (CS), or bio-fermentative unsulphated chondroitin (BC) alone or in combination. Nevertheless, a clear study of the effect of glycosaminoglycans (GAGs) on chondrocyte differentiation is still lacking, especially for the newly obtained unsulfated chondroitin of biotechnological origin. Are these GAGs playing a role in the commitment of stem cells to chondrocyte progenitors and in the differentiation of progenitors to mature chondrocytes? Alternatively, do they have a role only in one of these biological processes? We evaluated the role of HA, CS, and - above all - BC in cell commitment and chondrocyte differentiation of MSCs by supplementing these GAGs in different phases of in vitro cultivation. Our data provided evidence that a combination of HA and CS or of HA and BC supplemented during the terminal in vitro differentiation and not during cell commitment of MSCs improved chondrocytes differentiation without the presence of fibrosis (reduced expression of Type I collagen). This result suggests that a careful evaluation of extracellular cues for chondrocyte differentiation is fundamental to obtaining a proper maturation process.
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Antonietta Stellavato
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Domenico Aprile
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Donatella Cimini
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Valentina Vassallo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA, United States
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA, United States.,Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | - Chiara Schiraldi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| |
Collapse
|
43
|
Motejunas MW, Bonneval L, Carter C, Reed D, Ehrhardt K. Biologic Therapy in Chronic Pain Management: a Review of the Clinical Data and Future Investigations. Curr Pain Headache Rep 2021; 25:30. [PMID: 33761016 DOI: 10.1007/s11916-021-00947-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE With the aging population, it is clear that the demand for future chronic pain treatment modalities is at an all-time high. One of the newest treatment modalities that is gaining popularity with both practitioners and patients alike is that of regenerative medicine and the use of stem cells to treat chronic painful conditions. This article aims to distill the most recent, available data from both laboratory research and clinical trials to better illuminate the potentials for these therapies in the treatment of chronic pain. RECENT FINDINGS There are numerous investigations underway using mesenchymal stem cells (MSCs) to treat painful, largely degenerative conditions. A large majority of these investigations focus on osteoarthritis of the knee and have demonstrated significantly improved pain scores. Some of these investigations have demonstrated significantly increased articular cartilage and meniscus growth as well as improved function. These studies have been smaller (n, 18) and need to be corroborated on a macrolevel. Platelet-rich plasma (PRP)-based therapies have been most extensively studied in the treatment of knee osteoarthritis. Multiple prospective and randomized trials and meta-analyses have afforded level I evidence in support of PRP's safety and efficacy in chronic knee pain demonstrating both decreased pain (via VAS) and increased functional status (via WOMAC and IKDC). There have been randomized controlled trials examining PRP therapies in treatment degenerative disc disease (intradiscal treatment), facet arthropathy (intra-facet injections), and sacroiliitis (SIJ) which have all yielded similar positive results. Each RTC demonstrated decreased pain scores and increased function but lacks the scale to derive concrete guidelines. Newer investigations are underway examining modified PRP formulas with increased fibrin (PRF) or various growth factors (PRGF) and have shown positive outcomes with respect to osteoarthritic conditions in small trials. Animal trials are underway further investigating these therapies as well as specific gene modulation therapies. This review of the most recent investigations into the application and uses of biologic stem cell-derived treatments for chronic painful conditions should act to illustrate the growing, favorable data for these types of modalities both with respect to pain control and functional improvement.
Collapse
Affiliation(s)
| | | | | | | | - Ken Ehrhardt
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Recent Updates of Diagnosis, Pathophysiology, and Treatment on Osteoarthritis of the Knee. Int J Mol Sci 2021; 22:ijms22052619. [PMID: 33807695 PMCID: PMC7961389 DOI: 10.3390/ijms22052619] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative and chronic joint disease characterized by clinical symptoms and distortion of joint tissues. It primarily damages joint cartilage, causing pain, swelling, and stiffness around the joint. It is the major cause of disability and pain. The prevalence of OA is expected to increase gradually with the aging population and increasing prevalence of obesity. Many potential therapeutic advances have been made in recent years due to the improved understanding of the underlying mechanisms, diagnosis, and management of OA. Embryonic stem cells and induced pluripotent stem cells differentiate into chondrocytes or mesenchymal stem cells (MSCs) and can be used as a source of injectable treatments in the OA joint cavity. MSCs are known to be the most studied cell therapy products in cell-based OA therapy owing to their ability to differentiate into chondrocytes and their immunomodulatory properties. They have the potential to improve cartilage recovery and ultimately restore healthy joints. However, despite currently available therapies and advances in research, unfulfilled medical needs persist for OA treatment. In this review, we focused on the contents of non-cellular and cellular therapies for OA, and briefly summarized the results of clinical trials for cell-based OA therapy to lay a solid application basis for clinical research.
Collapse
|
45
|
Primorac D, Molnar V, Matišić V, Hudetz D, Jeleč Ž, Rod E, Čukelj F, Vidović D, Vrdoljak T, Dobričić B, Antičević D, Smolić M, Miškulin M, Ćaćić D, Borić I. Comprehensive Review of Knee Osteoarthritis Pharmacological Treatment and the Latest Professional Societies' Guidelines. Pharmaceuticals (Basel) 2021; 14:ph14030205. [PMID: 33801304 PMCID: PMC8001498 DOI: 10.3390/ph14030205] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis is the most common musculoskeletal progressive disease, with the knee as the most commonly affected joint in the human body. While several new medications are still under research, many symptomatic therapy options, such as analgesics (opioid and non-opioid), nonsteroid anti-inflammatory drugs, symptomatic slow-acting drugs in osteoarthritis, and preparations for topical administration, are being used, with a diverse clinical response and inconsistent conclusions across various professional societies guidelines. The concept of pharmacogenomic-guided therapy, which lies on principles of the right medication for the right patient in the right dose at the right time, can significantly increase the patient’s response to symptom relief therapy in knee osteoarthritis. Corticosteroid intra-articular injections and hyaluronic acid injections provoke numerous discussions and disagreements among different guidelines, even though they are currently used in daily clinical practice. Biological options, such as platelet-rich plasma and mesenchymal stem cell injections, have shown good results in the treatment of osteoarthritis symptoms, greatly increasing the patient’s quality of life, especially when combined with other therapeutic options. Non-inclusion of the latter therapies in the guidelines, and their inconsistent stance on numerous therapy options, requires larger and well-designed studies to examine the true effects of these therapies and update the existing guidelines.
Collapse
Affiliation(s)
- Dragan Primorac
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
- Eberly College of Science, The Pennsylvania State University, University Park, State College, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
- Medical School, University of Split, 21000 Split, Croatia;
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96450 Coburg, Germany
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Correspondence: ; Tel.: +385-98-470-710
| | - Vilim Molnar
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Vid Matišić
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
| | - Damir Hudetz
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Željko Jeleč
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
- Department of Nursing, University North, 48000 Varaždin, Croatia
| | - Eduard Rod
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
| | - Fabijan Čukelj
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
- Medical School, University of Split, 21000 Split, Croatia;
- Department of Health Studies, University of Split, 21000 Split, Croatia
- Clinic for Traumatology, University Hospital “Sisters of Mercy”, 10000 Zagreb, Croatia
| | - Dinko Vidović
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
- Clinic for Traumatology, University Hospital “Sisters of Mercy”, 10000 Zagreb, Croatia
| | - Trpimir Vrdoljak
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
- Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Borut Dobričić
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
- Department of Orthopaedics and Traumatology, University Hospital Dubrava, 10000 Zagreb, Croatia
| | - Darko Antičević
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
| | - Martina Smolić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Mladen Miškulin
- Medical School, University of Split, 21000 Split, Croatia;
- Aksis Specialty Hospital, 10000 Zagreb, Croatia
| | - Damir Ćaćić
- General Hospital Karlovac, 47000 Karlovac, Croatia;
| | - Igor Borić
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (V.M.); (D.H.); (Ž.J.); (E.R.); (F.Č.); (D.V.); (T.V.); (B.D.); (D.A.); (I.B.)
- Medical School, University of Split, 21000 Split, Croatia;
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Department of Health Studies, University of Split, 21000 Split, Croatia
| |
Collapse
|
46
|
Research Progress on Stem Cell Therapies for Articular Cartilage Regeneration. Stem Cells Int 2021; 2021:8882505. [PMID: 33628274 PMCID: PMC7895563 DOI: 10.1155/2021/8882505] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Injury of articular cartilage can cause osteoarthritis and seriously affect the physical and mental health of patients. Unfortunately, current surgical treatment techniques that are commonly used in the clinic cannot regenerate articular cartilage. Regenerative medicine involving stem cells has entered a new stage and is considered the most promising way to regenerate articular cartilage. In terms of theories on the mechanism, it was thought that stem cell-mediated articular cartilage regeneration was achieved through the directional differentiation of stem cells into chondrocytes. However, recent evidence has shown that the stem cell secretome plays an important role in biological processes such as the immune response, inflammation regulation, and drug delivery. At the same time, the stem cell secretome can effectively mediate the process of tissue regeneration. This new theory has attributed the therapeutic effect of stem cells to their paracrine effects. The application of stem cells is not limited to exogenous stem cell transplantation. Endogenous stem cell homing and in situ regeneration strategies have received extensive attention. The application of stem cell derivatives, such as conditioned media, extracellular vesicles, and extracellular matrix, is an extension of stem cell paracrine theory. On the other hand, stem cell pretreatment strategies have also shown promising therapeutic effects. This article will systematically review the latest developments in these areas, summarize challenges in articular cartilage regeneration strategies involving stem cells, and describe prospects for future development.
Collapse
|
47
|
Wang AT, Zhao M, Feng Y, Jia H, Zhang L, Yu H, Li Z, Han Z, Han Z. Multifaceted Optimization of MSC-Based Formulation upon Sodium Iodoacetate-Induced Osteoarthritis Models by Combining Advantageous HA/PG Hydrogel and Fluorescent Tracer. Stem Cells Int 2021; 2021:1-13. [DOI: 10.1155/2021/8827212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Owing to the boundedness of conventional remedies upon articular cartilage for self-rehabilitation and the incrementally senior citizens, the incidence of osteoarthritis (OA) is increasing worldwide. Empirical studies have revealed the advantageous and promising potentials of mesenchymal stem/stromal cells (MSCs) on the refractory OA, whereas the deficiency of systematic and detailed exploration of MSC-based therapy largely hampers the large-scale applications in regenerative medicine. Herein, we initially utilized the monosodium iodoacetate- (MIA-) induced OA rabbit models and investigated the therapeutic effect of human umbilical cord-derived UC-MSCs at serial dose gradients with the splendid hyaluronic acid and/or propylene glycol hydrogels (HA, HA/PG), respectively. Afterwards, we turned to a dual-luciferase reporter tracing system and evaluated the spatiotemporal distribution and metabolokinetics of bifluorescence expressing UC-MSCs (BF-MSCs) in OA rats. Of the aforementioned trials, we verified that the combination of HA/PG and middle-dose MSCs (
cells/ml) eventually manifested the optimal efficacy on OA rabbits. Furthermore, with the aid of the bioluminescence imaging (BLI) technology for dynamic in vitro and in vivo tracking, we intuitively delineated the spatiotemporal distribution and therapeutic process of BF-MSCs in OA rats, which substantially confirmed the reinforcement of HA/PG on BF-MSCs for OA treatment. Collectively, our data conformably demonstrated that the middle dose of UC-MSCs combined with HA/PG hydrogel was sufficient for optimal MSC-based formulation for blocking OA progression and promoting cartilage repair, which supplied overwhelming new references and enlightened MSC-based therapeutic strategies for cartilage defects.
Collapse
Affiliation(s)
- Ai-tong Wang
- Cell Products of National Engineering Center & National Stem Cell Engineering Research Center, Tianjin IMCELL Stem Cell and Gene Technology Co., Ltd., Tianjin, China
| | - Meng Zhao
- Cell Products of National Engineering Center & National Stem Cell Engineering Research Center, Tianjin IMCELL Stem Cell and Gene Technology Co., Ltd., Tianjin, China
| | - Ying Feng
- Cell Products of National Engineering Center & National Stem Cell Engineering Research Center, Tianjin IMCELL Stem Cell and Gene Technology Co., Ltd., Tianjin, China
| | - Honghong Jia
- Cell Products of National Engineering Center & National Stem Cell Engineering Research Center, Tianjin IMCELL Stem Cell and Gene Technology Co., Ltd., Tianjin, China
| | - Leisheng Zhang
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, China
- Precision Medicine Division, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd., Tianjin 301700, China
- State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Jiangxi Research Center of Stem Cell Engineering, Jiangxi Health-Biotech Stem Cell Technology Co., Ltd., Shangrao 334000, China
| | - Hao Yu
- Cell Products of National Engineering Center & National Stem Cell Engineering Research Center, Tianjin IMCELL Stem Cell and Gene Technology Co., Ltd., Tianjin, China
| | - Zongjin Li
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, China
| | - Zhibo Han
- Cell Products of National Engineering Center & National Stem Cell Engineering Research Center, Tianjin IMCELL Stem Cell and Gene Technology Co., Ltd., Tianjin, China
- State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zhongchao Han
- Cell Products of National Engineering Center & National Stem Cell Engineering Research Center, Tianjin IMCELL Stem Cell and Gene Technology Co., Ltd., Tianjin, China
- Precision Medicine Division, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd., Tianjin 301700, China
- State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Jiangxi Research Center of Stem Cell Engineering, Jiangxi Health-Biotech Stem Cell Technology Co., Ltd., Shangrao 334000, China
| |
Collapse
|
48
|
Laplace-Builhé B, Bahraoui S, Jorgensen C, Djouad F. From the Basis of Epimorphic Regeneration to Enhanced Regenerative Therapies. Front Cell Dev Biol 2021; 8:605120. [PMID: 33585444 PMCID: PMC7873919 DOI: 10.3389/fcell.2020.605120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/14/2020] [Indexed: 01/01/2023] Open
Abstract
Current cell-based therapies to treat degenerative diseases such as osteoarthritis (OA) fail to offer long-term beneficial effects. The therapeutic effects provided by mesenchymal stem cell (MSC) injection, characterized by reduced pain and an improved functional activity in patients with knee OA, are reported at short-term follow-up since the improved outcomes plateau or, even worse, decline several months after MSC administration. This review tackles the limitations of MSC-based therapy for degenerative diseases and highlights the lessons learned from regenerative species to comprehend the coordination of molecular and cellular events critical for complex regeneration processes. We discuss how MSC injection generates a positive cascade of events resulting in a long-lasting systemic immune regulation with limited beneficial effects on tissue regeneration while in regenerative species fine-tuned inflammation is required for progenitor cell proliferation, differentiation, and regeneration. Finally, we stress the direct or indirect involvement of neural crest derived cells (NCC) in most if not all adult regenerative models studied so far. This review underlines the regenerative potential of NCC and the limitations of MSC-based therapy to open new avenues for the treatment of degenerative diseases such as OA.
Collapse
Affiliation(s)
| | | | - Christian Jorgensen
- IRMB, Univ Montpellier, INSERM, Montpellier, France.,CHU Montpellier, Montpellier, France
| | | |
Collapse
|
49
|
Kamm JL, Riley CB, Parlane N, Gee EK, McIlwraith CW. Interactions Between Allogeneic Mesenchymal Stromal Cells and the Recipient Immune System: A Comparative Review With Relevance to Equine Outcomes. Front Vet Sci 2021; 7:617647. [PMID: 33521090 PMCID: PMC7838369 DOI: 10.3389/fvets.2020.617647] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/02/2020] [Indexed: 12/27/2022] Open
Abstract
Despite significant immunosuppressive activity, allogeneic mesenchymal stromal cells (MSCs) carry an inherent risk of immune rejection when transferred into a recipient. In naïve recipients, this immune response is initially driven by the innate immune system, an immediate reaction to the foreign cells, and later, the adaptive immune system, a delayed response that causes cell death due to recognition of specific alloantigens by host cells and antibodies. This review describes the actions of MSCs to both suppress and activate the different arms of the immune system. We then review the survival and effectiveness of the currently used allogeneic MSC treatments.
Collapse
Affiliation(s)
- J Lacy Kamm
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Christopher B Riley
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Natalie Parlane
- Hopkirk Laboratory, AgResearch, Palmerston North, New Zealand
| | - Erica K Gee
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - C Wayne McIlwraith
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Medical Institute, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
50
|
Mesenchymal Stem Cell Therapy for Osteoarthritis: Practice and Possible Promises. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1387:107-125. [DOI: 10.1007/5584_2021_695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|