1
|
Moghassemi S, Nikanfar S, Dadashzadeh A, Sousa MJ, Wan Y, Sun F, Colson A, De Windt S, Kwaspen L, Kanbar M, Sobhani K, Yang J, Vlieghe H, Li Y, Debiève F, Wyns C, Amorim CA. The revolutionary role of placental derivatives in biomedical research. Bioact Mater 2025; 49:456-485. [PMID: 40177109 PMCID: PMC11964572 DOI: 10.1016/j.bioactmat.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The human placenta is a transient yet crucial organ that plays a key role in sustaining the relationship between the maternal and fetal organisms. Despite its historical classification as "biowaste," placental tissues have garnered increasing attention since the early 1900s for their significant medical potential, particularly in wound repair and surgical application. As ethical considerations regarding human placental derivatives have largely been assuaged in many countries, they have gained significant attention due to their versatile applications in various biomedical fields, such as biomedical engineering, regenerative medicine, and pharmacology. Moreover, there is a substantial trend toward various animal product substitutions in laboratory research with human placental derivatives, reflecting a broader commitment to advancing ethical and sustainable research methodologies. This review provides a comprehensive examination of the current applications of human placental derivatives, explores the mechanisms behind their therapeutic effects, and outlines the future potential and directions of this rapidly advancing field.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Saba Nikanfar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yuting Wan
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Fengxuan Sun
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Arthur Colson
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sven De Windt
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Lena Kwaspen
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Keyvan Sobhani
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yongqian Li
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christine Wyns
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
2
|
Dhillon J, Maguire JA, Kraeutler MJ. Stromal cell-based injection therapies for the treatment of knee osteoarthritis: A systematic review of level I randomized controlled trials. OSTEOARTHRITIS AND CARTILAGE OPEN 2025; 7:100608. [PMID: 40290651 PMCID: PMC12033899 DOI: 10.1016/j.ocarto.2025.100608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Objective To systematically review randomized controlled trials (RCTs) to compare clinical outcomes of stromal cell-based injection therapies versus other non-operative treatment modalities for the treatment of knee osteoarthritis (OA). Method A systematic review was performed by searching PubMed, Cochrane Library, and EMBASE to locate RCTs, published since 2019, comparing stromal cell-based injection therapies versus other non-operative modalities for the treatment of knee OA. The search terms used were: knee AND osteoarthritis AND injection AND randomized. Results Seventeen studies (all Level I evidence) were included in this review with 972 patients undergoing treatment with stromal cell-based therapy (Intervention Group) and 651 patients in the control group (Control Group). Among the 17 studies, 7 used autologous adipose-derived mesenchymal stromal cells (MSCs) (ADMSCs), 2 studies used allogeneic ADMSCs, 4 used autologous bone marrow-derived MSCs (BMMSCs), 1 used allogeneic BMMSCs, 1 used allogeneic placental MSCs, 1 used umbilical cord-derived MSCs (UCMSCs), and 1 study used autologous ADMSCs, BMMSCs, or allogeneic UCMSCs. All but 3 studies reported significantly better clinical or radiological outcomes in the Intervention Group at final follow-up. A total of 5 and 3 studies reported adverse events occurring in the Intervention and the Control groups, respectively, but they were all self-limiting. Conclusions Patients undergoing treatment of knee OA with MSCs might be expected to experience improvements in clinical and radiological outcomes in comparison to other non-operative modalities. Additional studies with mid-to long-term outcomes are needed to better determine the efficacy and safety of MSCs for the treatment of knee OA.
Collapse
Affiliation(s)
- Jaydeep Dhillon
- Samaritan Health Services, Department of Orthopedics, Corvallis, OR 97330, USA
| | - James A. Maguire
- St. Joseph's University Medical Center, Department of Orthopaedic Surgery, Paterson, NJ 07503, USA
| | - Matthew J. Kraeutler
- Texas Tech University Health Sciences Center, Department of Orthopaedic Surgery & Rehabilitation, Lubbock, TX 79430, USA
| |
Collapse
|
3
|
Wu C, Xu H, Wu Z, Huang H, Ge Q, Xu J, Chen J, Wang P, Yuan W, Jin H, Tong P. Subchondral injection of human umbilical cord mesenchymal stem cells ameliorates knee osteoarthritis by inhibiting osteoblast apoptosis and TGF-beta activity. Stem Cell Res Ther 2025; 16:235. [PMID: 40346614 PMCID: PMC12065343 DOI: 10.1186/s13287-025-04366-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common degenerative disease caused by multiple pathological mechanisms wherein subchondral bone malfunction plays a substantial role. Recently, subchondral (SC) injection of orthobiologics has been attracting growing interest albeit the mainstream delivery method of mesenchymal stem cells (MSCs) is through intra-articular (IA). This study investigates the effect of SC injection of human umbilical cord mesenchymal stem cells (UCMSCs) on OA and its possible therapeutic mechanism compared to IA injection. METHODS Male Sprague-Dawley rats with anterior cruciate ligament transection (ACLT) received saline or UCMSC injections via SC or IA. Consecutive injections once a week for three weeks and withdrawal for another four weeks, followed by Radiographical scanning, histopathological, immunohistochemical, and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labelling (TUNEL) staining. Cell counting Kit-8 (CCK-8) assay, alkaline phosphatase (ALP), alizarin red staining (ARS), TUNEL, flow cytometry, quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were employed in TNFα-induced MC3T3-E1 cells to illustrate the exact pathogenesis mechanism. RESULTS IA and SC UCMSC injections preserved cartilage, synovium, and subchondral bone parameters like trabecular bone volume fraction (BV/TV). SC injection uniquely improved Trabecular separation (Tb.Sp) and Trabecular number (Tb.N). SC and IA injections of UCMSCs demonstrated equivalent efficacy in promoting osteoblastic bone formation and attenuating aberrant angiogenesis of subchondral bone. In addition, we demonstrated that osteoblast apoptosis and Smad2-dependent TGF-beta (TGF-β) are crucial and interactive subchondral bone pathological features in OA. In vivo and vitro studies further revealed that UCMSCs inhibited excessive TGF-β/pSmad2 signaling to regulate aberrant vascularization, osteoblast apoptosis and differentiation imbalance, ultimately maintaining osteochondral homeostasis. CONCLUSIONS The efficacy of UCMSCs for treating OA rats via SC injection was equivalent to that of IA; and even superior to IA in terms of subchondral bone phenotype via regulating apoptosis and TGF-β/pSmad2 signaling in osteoblasts, suggesting SC injection of UCMSCs as a potential and promising cell therapy for OA treatment.
Collapse
Affiliation(s)
- Congzi Wu
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
- Department of Orthopaedic Surgery, Hangzhou Red Cross Hospital (Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine), No.208 East Huancheng Road, Gongshu District, Hangzhou, Zhejiang, 310003, PR China
| | - HuiHui Xu
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China
| | - Zhen Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
- Department of Orthopaedic Surgery, Tongde Hospital of Zhejiang Province, No.234 Gucui Road, Xihu District, Hangzhou, Zhejiang,, 310012, PR China
| | - Haipeng Huang
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
| | - Qinwen Ge
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China
| | - Jianbo Xu
- The First People's Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, No.199 Shixinnan Road, Xiaoshan District, Hangzhou, Zhejiang, 311200, PR China
| | - Jiali Chen
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China
| | - Pinger Wang
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China
| | - Wenhua Yuan
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China.
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China.
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No.548 Binwen Road, Binwen District, Hangzhou, Zhejiang, 310053, PR China.
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China.
| |
Collapse
|
4
|
Zhang H, Felthaus O, Prantl L. Adipose Tissue-Derived Therapies for Osteoarthritis: Multifaceted Mechanisms and Clinical Prospects. Cells 2025; 14:669. [PMID: 40358193 PMCID: PMC12071781 DOI: 10.3390/cells14090669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease that significantly impacts quality of life and poses a growing economic burden. Adipose tissue-derived therapies, including both cell-based and cell-free products, have shown promising potential in promoting cartilage repair, modulating inflammation, and improving joint function. Recent studies and clinical trials have demonstrated their regenerative effects, highlighting their feasibility as a novel treatment approach for OA. This review summarises the therapeutic mechanisms and latest advancements in adipose tissue-derived therapies, providing insights into their clinical applications and future prospects.
Collapse
Affiliation(s)
- Hanwen Zhang
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | | | | |
Collapse
|
5
|
Holiuk Y, Birsa R, Bukreieva T, Nemtinov P, Kyryk V, Ustymenko A, Mazevych V, Sokolov M, Lobyntseva G, Shablii V. Effectiveness and safety of multiple injections of human placenta-derived MSCs for knee osteoarthritis: a nonrandomized phase I trial. BMC Musculoskelet Disord 2025; 26:418. [PMID: 40281581 PMCID: PMC12032682 DOI: 10.1186/s12891-025-08664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
OBJECTIVE This study investigates the safety and efficacy of three intra-articular (IA) injections of cryopreserved human placenta-derived mesenchymal stem cells (hP-MSCs) for knee osteoarthritis (KOA) over a 1-year follow-up period. METHODS A total of 26 patients with stage II-III KOA were enrolled in this non-randomized, open-label study. Patients received either conventional therapy with hyaluronic acid (HA) alone (Control group, n = 11) or in combination with hP-MSCs (MSC group, n = 15) via three intra-articular injections with 4-week intervals. Clinical outcomes were assessed using the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), Visual Analogue Scale (VAS), and magnetic resonance imaging (MRI) at 6 and 12 months following the first injection. Blood samples were analyzed for cytokine levels. RESULTS Three injections of hP-MSCs combined with HA were well-tolerated, with no severe adverse events observed. Significant improvements in WOMAC and VAS scores were noted in the MSC group compared to the Control group at both 6 and 12 months. MRI analysis revealed no significant differences in cartilage thickness or optical density index between the groups. Additionally, serum cytokine analysis showed a significant decrease in interleukin-2 (IL-2) levels in the MSC group, indicating an anti-inflammatory effect of hP-MSCs. However, no significant changes were observed in other cytokines. CONCLUSION This study demonstrates that three intra-articular injections of cryopreserved hP-MSCs in combination with HA are safe and effective for treating KOA, providing sustained clinical improvement at the 1-year follow-up. TRIAL REGISTRATION NCT04453111, #7/09.26.2018. Registered 02 January 2020, https://www. CLINICALTRIALS gov/study/NCT04453111 .
Collapse
Affiliation(s)
- Yevhen Holiuk
- State Institution "The Institute of Traumatology and Orthopedics by NAMS of Ukraine", 27 Bulvarno-Kudriavska Street, Kyiv, 01601, Ukraine
| | - Roman Birsa
- Department of Traumatology, Kyiv City Clinical Hospital, #6, 3 Guzara Ave, Kyiv, 03680, Ukraine
| | - Tetiana Bukreieva
- Laboratory of Biosynthesis of Nucleic Acids, Institute of Molecular Biology and Genetics of National Academy of Science of Ukraine, 150 Zabolotnogo Str, Kyiv, 03143, Ukraine
- Placenta Stem Cell Laboratory, Institute of Cell Therapy, 9 Mokra str, Cryobank, Kyiv, 03035, Ukraine
| | - Petro Nemtinov
- Institute of Cell Therapy, 9 Mokra str, Kyiv, 03035, Ukraine
| | - Vitalii Kyryk
- Cell and Tissue Technologies Department, M. D. Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine of the National Academy of Medical Sciences of Ukraine, 5 Svyatoslav Khorobrygo str, Kyiv, 03151, Ukraine
- Laboratory of Pathological Physiology and Immunology, D. F. Chebotarev Institute of Gerontology of the National Academy of Medical Sciences of Ukraine, 67 Vyshgorodska Street, Kyiv, 04114, Ukraine
| | - Alina Ustymenko
- Cell and Tissue Technologies Department, M. D. Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine of the National Academy of Medical Sciences of Ukraine, 5 Svyatoslav Khorobrygo str, Kyiv, 03151, Ukraine
- Laboratory of Pathological Physiology and Immunology, D. F. Chebotarev Institute of Gerontology of the National Academy of Medical Sciences of Ukraine, 67 Vyshgorodska Street, Kyiv, 04114, Ukraine
| | - Vadym Mazevych
- State Institution "The Institute of Traumatology and Orthopedics by NAMS of Ukraine", 27 Bulvarno-Kudriavska Street, Kyiv, 01601, Ukraine
| | - Mykola Sokolov
- Institute of Cell Therapy, 9 Mokra str, Kyiv, 03035, Ukraine
| | | | - Volodymyr Shablii
- Placenta Stem Cell Laboratory, Institute of Cell Therapy, 9 Mokra str, Cryobank, Kyiv, 03035, Ukraine.
- Department of Protein Synthesis Enzymology, Institute of Molecular Biology and Genetics of National Academy of Science of Ukraine, 150 Zabolotnogo Str, 03143, Kyiv, Ukraine.
| |
Collapse
|
6
|
Whittle SL, Johnston RV, McDonald S, Worthley D, Campbell TM, Cyril S, Bapna T, Zhang J, Buchbinder R. Stem cell injections for osteoarthritis of the knee. Cochrane Database Syst Rev 2025; 4:CD013342. [PMID: 40169165 PMCID: PMC11961299 DOI: 10.1002/14651858.cd013342.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
BACKGROUND Stem cells are specialised precursor cells that can replace aged or damaged cells and thereby maintain healthy tissue function. Stem cell therapy is increasingly used as a treatment for knee osteoarthritis, despite the lack of clarity around the mechanism by which stem cell therapy may slow down disease progression in osteoarthritis, and uncertainty regarding its benefits and harms. OBJECTIVES To assess the benefits and harms of stem cell injections for people with osteoarthritis of the knee. A secondary objective is to maintain the currency of the evidence, using a living systematic review approach. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE and Embase on 15 September 2023, unrestricted by date or language of publication. We also searched ClinicalTrials.gov and the WHO International Clinical Trials Registry Platform (ICTRP) for relevant trial protocols and ongoing trials. SELECTION CRITERIA We included randomised controlled trials (RCTs), or trials using quasi-randomised methods of participant allocation, comparing stem cell injection with placebo injection, no treatment or usual care, glucocorticoid injection, other injections, exercise, drug therapy, surgical interventions, and supplements and complementary therapies in people with knee osteoarthritis. DATA COLLECTION AND ANALYSIS Two review authors selected studies for inclusion, extracted trial characteristics and outcome data, assessed risk of bias and assessed the certainty of evidence using the GRADE approach. The primary comparison was stem cell injection compared with placebo injection. The primary time point for pain, function and quality of life was three to six months, and the end of the trial period for participant-reported success, joint structure changes and adverse event outcomes. Major outcomes were pain, function, quality of life, global assessment of success, radiographic joint progression, withdrawals due to adverse events and serious adverse events. MAIN RESULTS We found 25 randomised trials (1341 participants) comparing stem cell injections with placebo injection (eight trials), no treatment or usual care (analgesia, weight loss and exercise) (two trials), glucocorticoid injection (one trial), hyaluronic acid injection (seven trials), platelet-rich plasma injections (two trials), oral acetaminophen (paracetamol) (one trial), non-steroidal anti-inflammatory drugs plus physical therapy plus hyaluronic acid injection (one trial) and stem cell injection plus intra-articular co-intervention versus co-intervention alone (three trials) in people with osteoarthritis of the knee. Trials were predominantly small, with sample sizes ranging from 6 to 252 participants, with only two trials having more than 100 participants. The average age of participants across trials ranged from 51 to 66 years, and symptom duration varied from one to 10 years. Placebo-controlled trials were largely free from bias, while most trials without a placebo control were susceptible to performance and detection biases. Here, we limit reporting to the main comparison, stem cell injection versus placebo injection. Compared with placebo injection, stem cell injection may slightly improve pain and function up to six months after treatment. Mean pain (0 to 10 scale, 0 no pain) was 4.5 out of 10 points with placebo injection and 1.2 points better (2.5 points better to 0 points better) with stem cell injection (I2 = 80%; 7 studies, 445 participants). Mean function (0 to 100 scale, 0 best function) was 46.3 points with placebo injection and 14.2 points better (25.3 points better to 3.1 points better) with stem cell injection (I2 = 82%; 7 studies, 432 participants). We are uncertain whether stem cell injections improve quality of life or increase the number of people who report treatment success compared to placebo injection, because the certainty of the evidence was very low. Mean quality of life was 45.3 points with placebo injection and 22.8 points better (18.0 points worse to 63.7 points better) with stem cell injection (I2 = 96%; 2 studies, 288 participants) at up to six months follow-up. At the end of follow-up, 89/168 participants (530 per 1000) in the placebo injection group reported treatment success compared with 126/180 participants (683 per 1000) in the stem cell injection group (risk ratio (RR) 1.29, 95% CI 1.10 to 1.53; I2 = 0%; 4 trials, 348 participants). We downgraded the evidence to low certainty for pain and function due to indirectness (as the source, method of preparation and dose of stem cells varied across studies), and suspected publication bias (up to three larger RCTs have been conducted but withdrawn prior to reporting of results). For quality of life and treatment success, we further downgraded the evidence to very low certainty due to imprecision in addition to indirectness and suspected publication bias. We are uncertain of the potential harms associated with stem cell injection, as there were very low event rates for serious adverse events. At the end of follow-up, 5/219 participants (23 per 1000) in the placebo injection group experienced serious adverse events compared with 4/242 participants (16 per 1000) in the stem cell injection group (RR 0.72, 95% CI 0.20 to 2.64; I2 = 0%; 7 trials, 461 participants) and there were no reported withdrawals due to adverse events. We downgraded the evidence to very low certainty due to indirectness, suspected publication bias and imprecision. Radiographic progression was not assessed in any of the included studies. AUTHORS' CONCLUSIONS Compared with placebo injections and based upon low-certainty evidence, stem cell injections for people with knee osteoarthritis may slightly improve pain and function. We are uncertain of the effects of stem cell injections on quality of life or the number who report treatment success. Although the putative benefits of stem cell therapies for osteoarthritis include potential regenerative effects on damaged tissues, particularly articular cartilage, we remain uncertain of the effect of stem cell injections on structural progression in the knee (measured by radiographic appearance). There is also uncertainty regarding the safety of stem cell injections. Serious adverse events were infrequently reported, although all invasive joint procedures (including injections) carry a small risk of septic arthritis. The risk of other important harms, including potential concerns related to the use of a therapy with the theoretical capacity to promote cell growth, or to the use of allogeneic cells, remains unknown.
Collapse
Affiliation(s)
- Samuel L Whittle
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
- Rheumatology Unit, Queen Elizabeth Hospital, Woodville South, Australia
| | - Renea V Johnston
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Steve McDonald
- Cochrane Australia, School of Public Health & Preventive Medicine, Monash University, Melbourne, Australia
| | - Daniel Worthley
- Gastrointestinal Cancer Biology Group, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - T Mark Campbell
- Physical Medicine and Rehabilitation, Elisabeth Bruyère Hospital, Ottawa, Canada
| | - Sheila Cyril
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Tanay Bapna
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Jason Zhang
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Rachelle Buchbinder
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
7
|
Lin H, Zhou C, Li Q, Xie Q, Xia L, Liu L, Bao W, Xiong X, Zhang H, Zheng Z, Zhao J, Liang W. Nanotechnology-Assisted mesenchymal stem cells treatment for improved cartilage regeneration: A review of current practices. Biochem Pharmacol 2025; 237:116895. [PMID: 40154890 DOI: 10.1016/j.bcp.2025.116895] [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: 11/18/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Cartilage tissue does not promptly elicit an inflammatory response upon injury, hence constraining its capacity for healing and self-regeneration. Mesenchymal Stem Cells (MSC) therapy, enhanced by nanotechnology, offers promising advancements in cartilage repair. Injuries to cartilage often cause chronic pain, where current treatments are inadequate. As MSCs can readily differentiate into chondrocytes and secrete soluble factors, they are essential components in tissue engineering of cartilage repair. Although, like other stem cell applications, clinical applications are restricted by poor post implantation survival and differentiation. Recent studies show that nanoparticles (NPs) can further improve MSC outcomes by promoting cell adhesion, and chondrogenic differentiation allowing for sustained growth factor release. In addition, nanomaterials can improve the biological activity of MSCs, by also facilitating the composition of a conducive microenvironment for cartilage repair. In this review, the application of nanofibrous scaffolds, hydrogels and nanoscale particulate matter to improve mechanical properties in cartilage tissue engineering, are discussed. Moreover, the MSCs and nanotechnology synergistic effects present hope of overcoming the limitations of conventional treatments. Nanotechnology greatly enhances the MSC based cartilage regeneration strategies and could provide better treatment for cartilage related diseases in the future. Future research should be aimed at standardizing MSC harvesting and culturing protocols and contrasting their long-term efficacy.
Collapse
Affiliation(s)
- Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua hospital, Zhoushan 316000 Zhejiang Province, China
| | - Qingping Li
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China.
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China.
| |
Collapse
|
8
|
Safwan M, Bourgleh MS, Haider KH. Clinical experience with cryopreserved mesenchymal stem cells for cardiovascular applications: A systematic review. World J Stem Cells 2025; 17:102067. [PMID: 40160690 PMCID: PMC11947892 DOI: 10.4252/wjsc.v17.i3.102067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/17/2025] [Accepted: 02/24/2025] [Indexed: 03/21/2025] Open
Abstract
BACKGROUND As living biodrugs, mesenchymal stem cells (MSCs) have progressed to phase 3 clinical trials for cardiovascular applications. However, their limited immediate availability hampers their routine clinical use. AIM To validate our hypothesis that cryopreserved MSCs (CryoMSCs) are as safe and effective as freshly cultured MSC counterparts but carry logistical advantages. METHODS Four databases were systematically reviewed for relevant randomized controlled trials (RCTs) evaluating the safety and efficacy of CryoMSCs from various tissue sources in treating patients with heart disease. A subgroup analysis was performed based on MSC source and post-thaw cell viability to determine treatment effects across different CryoMSCs sources and viability status. Weighted mean differences (WMDs) and odds ratios were calculated to measure changes in the estimated treatment effects. All statistical analyses were performed using RevMan version 5.4.1 software. RESULTS Seven RCTs (285 patients) met the eligibility criteria for inclusion in the meta-analysis. During short-term follow-up, CryoMSCs demonstrated a significant 2.11% improvement in left ventricular ejection fraction (LVEF) [WMD (95%CI) = 2.11 (0.66-3.56), P = 0.004, I 2 = 1%], with umbilical cord-derived MSCs being the most effective cell type. However, the significant effect on LVEF was not sustained over the 12 months of follow-up. Subgroup analysis demonstrated a substantial 3.44% improvement in LVEF [WMD (95%CI) = 3.44 (1.46-5.43), P = 0.0007, I 2 = 0%] when using MSCs with post-thaw viability exceeding 80%. There was no statistically significant difference in the frequency of major cardiac adverse events observed in rehospitalization or mortality in patients treated with CryoMSCs vs the control group. CONCLUSION CryoMSCs are a promising option for heart failure patients, particularly considering the current treatment options for cardiovascular diseases. Our data suggest that CryoMSCs could be a viable alternative or complementary treatment to the current options, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Moaz Safwan
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, AlQaseem, Saudi Arabia
| | - Mariam Safwan Bourgleh
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, AlQaseem, Saudi Arabia
| | - Khawaja Husnain Haider
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, AlQaseem, Saudi Arabia.
| |
Collapse
|
9
|
Goulian AJ, Goldstein B, Saad MA. Advancements in Regenerative Therapies for Orthopedics: A Comprehensive Review of Platelet-Rich Plasma, Mesenchymal Stem Cells, Peptide Therapies, and Biomimetic Applications. J Clin Med 2025; 14:2061. [PMID: 40142869 PMCID: PMC11943164 DOI: 10.3390/jcm14062061] [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: 01/27/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Regenerative therapies have gained interest in orthopedic applications for their potential to enhance tissue regeneration, functional recovery, and pain modification. This review evaluates the clinical efficacy of platelet-rich plasma (PRP), mesenchymal stem cells (MSCs), peptide-based treatments, and biomimetic materials in orthopedic care, with a focus on pain reduction and functional outcomes. Methods: A structured literature search in PubMed (January 2009-January 2025) identified 160 studies. After applying inclusion criteria prioritizing randomized controlled trials (RCTs) and clinical trials, 59 studies were included: 20 on PRP, 20 on MSCs, 10 on peptide therapies, and 7 on biomimetics. Data extraction focused on pain reduction and functional recovery, with risk of bias assessed using the Cochrane Risk of Bias (RoB) tool and ROBINS-I tool. A random-effects meta-regression analysis was conducted to evaluate the impact of therapy type, sample size, and risk of bias on reported pain reduction outcomes. Results: Meta-regression analysis identified MSC therapy as the most effective intervention for pain reduction (β = 8.45, p < 0.05), with PRP and peptide-based therapies showing moderate improvements, and biomimetic therapies demonstrating the lowest effect. PRP provided short-term pain relief, particularly in acute injuries and tendon repair, though inconsistencies in preparation methods limited success in chronic conditions. MSC therapies demonstrated cartilage regeneration and early osteoarthritis improvement, but high costs and ethical concerns remain barriers to widespread adoption. Peptide-based therapies and biomimetic materials, including engineered scaffolds and autologous protein solutions, showed promise for infection control and wound healing, though further research is needed to optimize dosing, delivery methods, and long-term safety. Conclusions: Regenerative therapies offer significant potential in orthopedic care, with MSC therapies demonstrating the most reliable regenerative effects, PRP providing short-term symptomatic relief, and peptide-based and biomimetic treatments emerging as promising adjuncts. However, standardized protocols and large-scale clinical trials are needed to establish long-term efficacy and improve clinical translation for broader adoption.
Collapse
Affiliation(s)
- Andrew J. Goulian
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.J.G.); (B.G.)
| | - Brielle Goldstein
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.J.G.); (B.G.)
| | - Maarouf A. Saad
- Department of Orthopaedic Surgery, University of California, Sacramento, CA 95817, USA
| |
Collapse
|
10
|
Fenger Carlander AL, Jakobsen KK, Todsen T, Paaske N, Østergaard Madsen AK, Bendtsen SK, Kastrup J, Friborg J, Duch Lynggaard C, Hauge AW, Christensen R, Grønhøj C, von Buchwald C. Long-term Effectiveness and Safety of Mesenchymal Stromal Cell Therapy for Radiation-Induced Hyposalivation in Head and Neck Cancer Survivors: A Randomized Phase II Trial. Clin Cancer Res 2025; 31:824-831. [PMID: 39751638 DOI: 10.1158/1078-0432.ccr-24-2663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/30/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE The long-term effect of adipose-derived mesenchymal stromal cells (ASC) on restoring radiation-induced salivary gland hypofunction in patients with previous head and neck cancer has not been validated in larger settings. PATIENTS AND METHODS The study was a 12-month follow-up of a randomized trial, including patients with hyposalivation. Patients were randomized to receive allogeneic ASC or placebo in the submandibular glands. The primary endpoint was unstimulated whole saliva (UWS) followed by stimulated whole saliva, patient-reported outcomes (European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire, Head and Neck Module, and the xerostomia questionnaire), and safety. RESULTS Of the 120 enrolled patients, 117 (97.5%) were assessed at 12 months. Treatment with ASC did not increase UWS compared with placebo: Increase in UWS was 0.02 mL/minute [95% confidence interval (CI), 0.01-0.04] in the ASC group and 0.02 mL/minute (95% CI, 0-0.03) in the placebo group (P = 0.56). ASC reduced the symptom burden for dry mouth with -10.07 units (95% CI, -13.39 to -6.75) compared with -4.15 units (95% CI, -7.46 to -0.84) in the placebo group (P = 0.01). Compared with placebo, ASC did not improve sticky saliva (-9.27 vs. -4.55 units; P = 0.13), swallowing (-4.50 vs. 3.49 units; P = 0.5), or xerostomia (-3.12 vs. -2.74 units; P = 0.82). Treatment was safe and associated with a transient immune response. CONCLUSIONS Intraglandular ACS therapy in the submandibular glands significantly relieved subjective dry mouth symptoms. Both ASC and placebo increased UWS, but ASC did not prove superior to placebo in restoring salivary gland function, based on the salivary flow rate.
Collapse
Affiliation(s)
- Amanda-Louise Fenger Carlander
- Department of Otorhinolaryngology, Head and Neck Surgery, and Audiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Kathrine Kronberg Jakobsen
- Department of Otorhinolaryngology, Head and Neck Surgery, and Audiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Tobias Todsen
- Department of Otorhinolaryngology, Head and Neck Surgery, and Audiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Natasja Paaske
- Department of Otorhinolaryngology, Head and Neck Surgery, and Audiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Anne Kathrine Østergaard Madsen
- Department of Otorhinolaryngology, Head and Neck Surgery, and Audiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Simone Kloch Bendtsen
- Department of Otorhinolaryngology, Head and Neck Surgery, and Audiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Jens Kastrup
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Jeppe Friborg
- Department of Oncology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Charlotte Duch Lynggaard
- Department of Otorhinolaryngology, Head and Neck Surgery, and Audiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Anne Werner Hauge
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Robin Christensen
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Research, Research Unit of Rheumatology, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Christian Grønhøj
- Department of Otorhinolaryngology, Head and Neck Surgery, and Audiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Christian von Buchwald
- Department of Otorhinolaryngology, Head and Neck Surgery, and Audiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
11
|
Shi Y, Zhu H, Xu S, Zhao J, Wang Y, Pan X, Zhao B, Sun Z, Yin Y, Xu L, Wei F, He S, Hou X, Xue J. Injectable doxorubicin-loaded hyaluronic acid-based hydrogel for locoregional therapy and inhibiting metastasis of breast cancer. Colloids Surf B Biointerfaces 2025; 247:114433. [PMID: 39647423 DOI: 10.1016/j.colsurfb.2024.114433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/17/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
Therapy and metastasis pose significant challenges for breast cancer therapy. Locoregional chemotherapy presents a promising strategy to address these dilemmas. In this study, a doxorubicin-loaded injectable hydrogel based on hyaluronic acid (DOX-MCHAgel) was fabricated for locoregional chemotherapy and inhibiting the metastasis of breast cancer. The high bio-safety of cargo-free hydrogels (MCHAgel) would enhance patient compliance. The sustained DOX release behaviors from DOX-MCHAgel (over 10 days) could reduce dosing frequency and achieve long-term therapeutic effects. The potent in vivo anti-tumor activity of DOX-MCHAgel was verified by the smallest tumor volumes, the largest number of apoptotic cells, and the strongest fluorescence intensity in TUNEL sections. Notably, the injectable DOX-MCHAgel not only greatly suppressed the growth of 4T1 tumor tissues, but also effectively curbed the liver and lung metastasis in vivo. Moreover, the survival of 4T1-tumor bearing mice was extended without obvious systemic toxicity. In brief, the novel injectable hydrogel developed in this study offers a new strategy for locoregional therapy and inhibiting metastasis of breast cancer.
Collapse
Affiliation(s)
- Yongli Shi
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China.
| | - Huiqing Zhu
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Suyue Xu
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Jingya Zhao
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Yuxin Wang
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Xiaofei Pan
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Bingqian Zhao
- Basic medicine college, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Zeyu Sun
- First clinical college, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Yili Yin
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Linyin Xu
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Fengjiao Wei
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China
| | - Sisi He
- Department of Oncology, the Second Affiliated Hospital of Zunyi Medical University, e6c000, Guizhou, PR China.
| | - Xueyan Hou
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China; Pingyuan Laboratory, dec007, Xinxiang, Henan, PR China.
| | - Jintao Xue
- College of pharmacy, Xinxiang Medical University, dec00c, Xinxiang, PR China.
| |
Collapse
|
12
|
Wu W, An X, Gong W, Yang L, Liu N, Liu B, Guo B, Jiang Q, Li L. ShK-modified UCMSCs Inhibit M1-Like Macrophage Polarization and Alleviate Osteoarthritis Progression via PI3K/Akt Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406822. [PMID: 39721037 PMCID: PMC11884619 DOI: 10.1002/advs.202406822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/15/2024] [Indexed: 12/28/2024]
Abstract
The potassium channel Kv1.3 plays an important role in regulating immune cell functions in many inflammatory diseases whereas rarely in osteoarthritis (OA). Here, it is demonstrated that the Kv1.3 of macrophages is upregulated in response to LPS stimulation, as well as in human OA synovium samples than non-OA. Administration of Stichodactyla toxin (ShK), a Kv1.3 blocker, significantly inhibited cartilage degeneration and synovial inflammation in animal models of OA in vivo by inhibiting M1 macrophage polarization and reducing the production of inflammatory factors. In this study, a transgenically engineered human umbilical cord mesenchymal stem cell (UCMSC) delivery system is developed that secreted a peptide ShK, a Kv1.3 potassium blocker, into the knee articular cavity. Collectively, the results identified Kv1.3 as a potential therapeutic target for OA and demonstrated the efficacy of using ShK transgenic engineered UCMSCs as a delivery for the peptide in OA treatment.
Collapse
Affiliation(s)
- Wenshu Wu
- Division of Sports Medicine and Adult Reconstructive SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University321 Zhongshan RoadNanjingJiangsu210008P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University22 Hankou RoadNanjingJiangsu210093P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation321 Zhongshan RoadNanjingJiangsu210008P. R. China
- Institute of Medical 3D PrintingNanjing UniversityNanjing211166P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting321 Zhongshan RoadNanjing210000P. R. China
| | - Xueying An
- Division of Sports Medicine and Adult Reconstructive SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University321 Zhongshan RoadNanjingJiangsu210008P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University22 Hankou RoadNanjingJiangsu210093P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation321 Zhongshan RoadNanjingJiangsu210008P. R. China
- Institute of Medical 3D PrintingNanjing UniversityNanjing211166P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting321 Zhongshan RoadNanjing210000P. R. China
| | - Wang Gong
- Division of Sports Medicine and Adult Reconstructive SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University321 Zhongshan RoadNanjingJiangsu210008P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University22 Hankou RoadNanjingJiangsu210093P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation321 Zhongshan RoadNanjingJiangsu210008P. R. China
- Institute of Medical 3D PrintingNanjing UniversityNanjing211166P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting321 Zhongshan RoadNanjing210000P. R. China
| | - Lin Yang
- Division of Sports Medicine and Adult Reconstructive SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University321 Zhongshan RoadNanjingJiangsu210008P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University22 Hankou RoadNanjingJiangsu210093P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation321 Zhongshan RoadNanjingJiangsu210008P. R. China
- Institute of Medical 3D PrintingNanjing UniversityNanjing211166P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting321 Zhongshan RoadNanjing210000P. R. China
- Department of Sports Medicine and Adult Reconstructive SurgeryNanjing Drum Tower HospitalClinical College of Nanjing Medical UniversityNanjing210008P.R. China
| | - Na Liu
- Division of Sports Medicine and Adult Reconstructive SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University321 Zhongshan RoadNanjingJiangsu210008P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University22 Hankou RoadNanjingJiangsu210093P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation321 Zhongshan RoadNanjingJiangsu210008P. R. China
- Institute of Medical 3D PrintingNanjing UniversityNanjing211166P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting321 Zhongshan RoadNanjing210000P. R. China
| | - Bin Liu
- Division of Sports Medicine and Adult Reconstructive SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University321 Zhongshan RoadNanjingJiangsu210008P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University22 Hankou RoadNanjingJiangsu210093P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation321 Zhongshan RoadNanjingJiangsu210008P. R. China
- Institute of Medical 3D PrintingNanjing UniversityNanjing211166P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting321 Zhongshan RoadNanjing210000P. R. China
| | - Baosheng Guo
- Division of Sports Medicine and Adult Reconstructive SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University321 Zhongshan RoadNanjingJiangsu210008P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University22 Hankou RoadNanjingJiangsu210093P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation321 Zhongshan RoadNanjingJiangsu210008P. R. China
- Institute of Medical 3D PrintingNanjing UniversityNanjing211166P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting321 Zhongshan RoadNanjing210000P. R. China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University321 Zhongshan RoadNanjingJiangsu210008P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University22 Hankou RoadNanjingJiangsu210093P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation321 Zhongshan RoadNanjingJiangsu210008P. R. China
- Institute of Medical 3D PrintingNanjing UniversityNanjing211166P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting321 Zhongshan RoadNanjing210000P. R. China
- Department of Sports Medicine and Adult Reconstructive SurgeryNanjing Drum Tower HospitalClinical College of Nanjing Medical UniversityNanjing210008P.R. China
| | - Lan Li
- Division of Sports Medicine and Adult Reconstructive SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University321 Zhongshan RoadNanjingJiangsu210008P. R. China
- State Key Laboratory of Pharmaceutical Biotechnology Nanjing University22 Hankou RoadNanjingJiangsu210093P. R. China
- Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation321 Zhongshan RoadNanjingJiangsu210008P. R. China
- Institute of Medical 3D PrintingNanjing UniversityNanjing211166P. R. China
- Jiangsu Engineering Research Center for 3D Bioprinting321 Zhongshan RoadNanjing210000P. R. China
| |
Collapse
|
13
|
Ishak-Samrin M, Naina-Mohamed I, Zulfarina MS, Abdul Wahid SF, Mohd Don AF, Mohamad N, Ramlan MKR, Badrul AHMY. Treatment of Knee Osteoarthritis and Chondral Injury with Umbilical Cord/Wharton's Jelly-Derived Mesenchymal Stem Cells: A Systematic Review of Safety and Efficacy. J Funct Biomater 2025; 16:84. [PMID: 40137363 PMCID: PMC11942840 DOI: 10.3390/jfb16030084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/17/2024] [Accepted: 08/20/2024] [Indexed: 03/27/2025] Open
Abstract
Knee osteoarthritis (KOA) is a major cause of pain and disability worldwide, with no current treatment modality able to prevent the progressive destruction of articular cartilage. Mesenchymal stem cells (MSCs) have attracted interest in treating KOA and cartilage injury due to their self-renewal and multidirectional differentiation potential, as well as local bioactive factors with immunomodulatory and regenerative potential. This study aimed to evaluate the currently available studies using the intra-articular injection of Wharton's jelly MSCs for KOA and cartilage injury. We analyzed all clinical trials published from inception to 31 December 2023. Six studies met the inclusion criteria, for a total of 97 patients and 134 knees. The follow-up period ranged from 3 to 48 months. There were no serious adverse effects noted. There was significant improvement in functional outcomes in the form of VAS, WOMAC, KOOS, and IKDC parameters, though radiological outcomes demonstrated mixed results. In conclusion, patients with KOA treated with intra-articular injections of Wharton's jelly MSCs showed improvements in functional outcomes with no severe adverse effects. Multiple injections of Wharton's jelly MSCs showed better outcomes compared to single-injection techniques. Wharton's jelly MSCs may have potential as a cornerstone in the future treatment of KOA.
Collapse
Affiliation(s)
- Mohd Ishak-Samrin
- Department of Orthopaedic and Traumatology, Faculty of Medicine, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.I.-S.)
| | - Isa Naina-Mohamed
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mohamed S. Zulfarina
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - S. Fadilah Abdul Wahid
- Cell Therapy Center, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Ahmad Farihan Mohd Don
- Department of Orthopaedic and Traumatology, Faculty of Medicine, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.I.-S.)
| | - Norlelawati Mohamad
- Department of Orthopaedic and Traumatology, Faculty of Medicine, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.I.-S.)
| | - Muhamad Karbela Reza Ramlan
- Department of Orthopaedic and Traumatology, Faculty of Medicine, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.I.-S.)
| | - Akmal Hisham Md Yusoff Badrul
- Department of Orthopaedic and Traumatology, Faculty of Medicine, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.I.-S.)
| |
Collapse
|
14
|
Bensa A, Bianco Prevot L, Moraca G, Sangiorgio A, Boffa A, Filardo G. Corticosteroids, hyaluronic acid, platelet-rich plasma, and cell-based therapies for knee osteoarthritis - literature trends are shifting in the injectable treatments' evidence: a systematic review and expert opinion. Expert Opin Biol Ther 2025; 25:309-318. [PMID: 40028854 DOI: 10.1080/14712598.2025.2465833] [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: 11/26/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION The aim of this systematic review was to quantify the data available on corticosteroids (CS), hyaluronic acid, (HA), platelet-rich plasma (PRP), and cell-based therapies for knee osteoarthritis (OA) treatment. METHODS A literature search was conducted on PubMed, Cochrane, and Web of Science according to the PRISMA guidelines. Inclusion criteria: clinical studies of any level of evidence, written in English, evaluating the intra-articular use of CS, HA, PRP, or cell-based therapies for knee OA treatment. RESULTS The initial search identified 17,415 records. A total of 766 studies from 1959 were included. Of these, 401 were randomized controlled trials (RCTs), 110 comparative studies, and 255 case series, for a total of 75,834 patients. (11,245 treated with CS, 40,862 with HA, 16,174 with PRP, and 7,553 with cell-based therapies). The number of placebo-controlled RCTs remains limited and a negligible percentage of studies investigated possible disease-modifying effects of these treatments for knee OA. CONCLUSIONS The evidence on injective knee OA treatments is increasing at different speeds with a more rapidly growing literature focusing on orthobiologics. Currently, HA has the largest evidence, followed by PRP that recently surpassed the number of studies evaluating CS. Cell-based therapies are also growing rapidly, although the number of studies is still lower. The rapid literature shift toward orthobiologics urges an update in societies' guidelines to align with the new body of evidence on knee OA treatments. PROTOCOL REGISTRATION www.crd.york.ac.uk/prosperoi dentifier is CRD42024592972.
Collapse
Affiliation(s)
- Alessandro Bensa
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Luca Bianco Prevot
- Residency Program in Orthopedics and Traumatology, University of Milan, Milan, Italy
- Dipartimento di Ortopedia e Traumatologia, IRCCS Ospedale Galeazzi - S. Ambrogio, Milan, Italy
| | - Giacomo Moraca
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
| | - Alessandro Sangiorgio
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
| | - Angelo Boffa
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Giuseppe Filardo
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
15
|
Quan J, Liu Q, Li P, Yang Z, Zhang Y, Zhao F, Zhu G. Mesenchymal stem cell exosome therapy: current research status in the treatment of neurodegenerative diseases and the possibility of reversing normal brain aging. Stem Cell Res Ther 2025; 16:76. [PMID: 39985030 PMCID: PMC11846194 DOI: 10.1186/s13287-025-04160-5] [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: 11/04/2024] [Accepted: 01/21/2025] [Indexed: 02/23/2025] Open
Abstract
With the exacerbation of the aging population trend, a series of neurodegenerative diseases caused by brain aging have become increasingly common, significantly impacting the daily lives of the elderly and imposing heavier burdens on nations and societies. Brain aging is a complex process involving multiple mechanisms, including oxidative stress, apoptosis of damaged neuronal cells, chronic inflammation, and mitochondrial dysfunction, and research into new therapeutic strategies to delay brain aging has gradually become a research focus in recent years. Mesenchymal stem cells (MSCs) have been widely used in cell therapy due to their functions such as antioxidative stress, anti-inflammation, and tissue regeneration. However, accompanying safety issues such as immune rejection, tumor development, and pulmonary embolism cannot be avoided. Studies have shown that using exosome derived from mesenchymal stem cells (MSC-Exo) for the treatment of neurodegenerative diseases is a safe and effective method. It not only has the therapeutic effects of stem cells but also avoids the risks associated with cell therapy. Therefore, exploring new therapeutic strategies to delay normal brain aging from the mechanism of MSC-Exo in the treatment of neurodegenerative diseases is feasible. This review summarizes the characteristics of MSC-Exo and their clinical progress in the treatment of neurodegenerative diseases, aiming to explore the possibility and potential mechanisms of MSC-Exo in reversing brain aging.
Collapse
Affiliation(s)
- Jinglan Quan
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Qing Liu
- Department of Library, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Pinghui Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Zhiyu Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Yaohui Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Fuxing Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China
| | - Gaohong Zhu
- Department of Nuclear Medicine, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming, Yunnan, 650032, China.
| |
Collapse
|
16
|
Li X, Jian X, Yan Z, Liu H, Zhang L. Multiple Intra-Articular Injections of Adipose-Derived Mesenchymal Stem Cells for Canine Osteoarthritis Treatment. Cells 2025; 14:323. [PMID: 40072052 PMCID: PMC11899304 DOI: 10.3390/cells14050323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
Osteoarthritis (OA) is one of the most common degenerative diseases in dogs and humans, which can lead to articular cartilage deterioration, chronic pain, and decreased quality of life. The anti-inflammatory, anti-fibrotic, analgesic, and cartilage regeneration properties of mesenchymal stem cell (MSC) therapy provide a new direction for the treatment development of OA in the future. Currently, MSC therapy lacks confirmed ideal sources, dosages, formulations, and specific characteristics. In this study, we evaluated the efficacy of multiple canine adipose-derived mesenchymal stem cell (ADSC) injections on anti-inflammation and joint cartilage damage in a canine OA model. Considering animal ethics, we simulated the effects of inflammation and cartilage repair during treatment through a mouse OA model. In the mouse OA model, through the detection of cartilage repair and inflammation-related key factors via histology and molecular biology, it was found that MSC therapy has a certain repair effect on cartilage, but the anti-inflammatory effect is time-dependent. In the canine OA model, we verified the feasibility of multiple injections of ADSCs. Compared with the control group, the cartilage repair effect of the treatment group was obvious, and the inflammatory factors decreased, showing an obvious therapeutic effect. This study demonstrates that multiple intra-articular injections of canine ADSCs could be effective in treating OA symptoms.
Collapse
Affiliation(s)
| | | | | | | | - Lisheng Zhang
- College of Veterinary Medicine/Bio-Medical Center, Huazhong Agricultural University, Wuhan 430070, China; (X.L.); (X.J.); (Z.Y.); (H.L.)
| |
Collapse
|
17
|
Aabling RR, Rusan M, Møller AMJ, Munk-Pedersen N, Holm C, Elmengaard B, Pedersen M, Møller BK. A Narrative Review on Manufacturing Methods Employed in the Production of Mesenchymal Stromal Cells for Knee Osteoarthritis Therapy. Biomedicines 2025; 13:509. [PMID: 40002922 PMCID: PMC11853043 DOI: 10.3390/biomedicines13020509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Knee osteoarthritis (OA) is a chronic, progressive, inflammatory, and degenerative whole-joint disease. Early-stage OA treatments typically include physiotherapy, weight-loss, pain relief medications, and intra-articular knee injections, such as corticosteroids, hyaluronic acid, or platelet-rich plasma. These treatments primarily provide symptomatic relief rather than reversing or halting disease progression. Recently, mesenchymal stromal cell (MSC) injections have garnered attention due to their immunomodulatory and regenerative capacities. MSCs, which can be derived from sources such as bone marrow, umbilical cord, or adipose tissue, and can be allogeneic or autologous, have demonstrated promising results in both animal models and several human studies. However, different protocols have been employed, presenting challenges for comparing outcomes. In this review, we address these variable settings, evaluate current practices, and identify key factors critical in optimizing MSC-based therapies by critically reviewing clinical trials of ex vivo expanded MSC therapies for OA undertaken between 2008 and 2023. Specific attention was given to two key aspects: (1) the cell culture process employed in manufacturing of autologous or allogeneic MSC products, and (2) the post-culture methods employed in storage, reconstitution and administration of the MSCs. Our findings suggest that standardizing MSC production for clinical applications remains a significant challenge, primarily due to variations in tissue sources, harvesting techniques, and manufacturing protocols, and due to broad discrepancies in reporting. Thus, we propose a set of minimal reporting criteria to guide future clinical trials. A common reporting guideline is a critical step towards a more standardized MSC production across different laboratories and clinical settings, thereby enhancing reproducibility and advancing the field of regenerative medicine for knee OA, as well as other disease settings.
Collapse
Affiliation(s)
- Rasmus Roost Aabling
- Comparative Medicine Lab, SDCA-Steno Diabetes Center Aarhus, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99 and 11, DK-8200 Aarhus, Denmark
| | - Maria Rusan
- Department of Molecular Medicine, Aarhus University Hospital, Brendstrupgårdsvej 21A, DK-8200 Aarhus, Denmark;
- Department of Clinical Pharmacology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark;
| | - Anaïs Marie Julie Møller
- Center for Gene and Cellular Therapy, Department of Clinical Immunology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark;
| | - Naija Munk-Pedersen
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark; (N.M.-P.); (M.P.)
| | - Carsten Holm
- Department of Orthopedic Surgery, Elective Surgery Centre, Silkeborg Regional Hospital, Falkevej 1G, DK-8600 Silkeborg, Denmark; (C.H.); (B.E.)
| | - Brian Elmengaard
- Department of Orthopedic Surgery, Elective Surgery Centre, Silkeborg Regional Hospital, Falkevej 1G, DK-8600 Silkeborg, Denmark; (C.H.); (B.E.)
| | - Michael Pedersen
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark; (N.M.-P.); (M.P.)
| | - Bjarne Kuno Møller
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark;
- Center for Gene and Cellular Therapy, Department of Clinical Immunology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus, Denmark;
| |
Collapse
|
18
|
Pakdaman Kolour SS, Nematollahi S, Dehbozorgi M, Fattahi F, Movahed F, Esfandiari N, Kahrizi MS, Ghavamikia N, Hajiagha BS. Extracecellulr vesicles (EVs) microRNAs (miRNAs) derived from mesenchymal stem cells (MSCs) in osteoarthritis (OA); detailed role in pathogenesis and possible therapeutics. Heliyon 2025; 11:e42258. [PMID: 40007782 PMCID: PMC11850152 DOI: 10.1016/j.heliyon.2025.e42258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
The primary cause of pain and disability in the world is osteoarthritis (OA), a common joint disease characterized by the primary pathological alteration in articular cartilage deterioration. The general outcome of treatment is not acceptable despite current interventions. Therefore, joint replacement surgery is frequently needed by patients with severe OA. Mesenchymal stem cells (MSCs) have become a practical treatment choice for preclinical and clinical OA palliation in recent years, mainly due to their unique immunomodulatory attributes. Further, attractive candidates for cell-free therapy for OA are MSC-derived extracecellulr vesicles (EVs) that convey bioactive molecules of the original cells, such as microRNAs. These EVs have been shown to significantly influence the regulation of various physiological activities of cells in the joint cavity. Dysregulated miRNAs upregulate the synthesis of enzymes that degrade cartilage, downregulate the expression of components in the cartilage matrix, promote the production of proinflammatory cytokines, induce programmed cell death in chondrocytes, inhibit the process of autophagy in chondrocytes, and participate in pathways related to pain. MiRNAs are also found in extracellular membranous vesicles (EVs), such as exosomes, and play a role in intercellular communication in osteoarthritic joints. Thus, the biosynthesis, chemical makeup, and mechanism of action of miRNAs-enriched EVs in OA are all thoroughly covered in this review. We additionally discussed how miRNA-enriched MSC-EVs might be used therapeutically to change intercellular interaction in OA.
Collapse
Affiliation(s)
| | - Saeide Nematollahi
- Department of Radiology, Kerman University of Paramedical Sciences, Kerman, Iran
| | | | | | - Fatemeh Movahed
- Department of Gynecology, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Nima Ghavamikia
- Cardiovascular Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Salmanian Hajiagha
- Department of Cellular and Molecular Biology, Faculty of Basic Science, East Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
19
|
Zeng L, Liu C, Wu Y, Liu S, Zheng Y, Hao W, Wang D, Sun L. Efficacy and safety of mesenchymal stromal cell transplantation in the treatment of autoimmune and rheumatic immune diseases: a systematic review and meta-analysis of randomized controlled trials. Stem Cell Res Ther 2025; 16:65. [PMID: 39934871 PMCID: PMC11817852 DOI: 10.1186/s13287-025-04184-x] [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: 12/04/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
OBJECTIVE This study aims to assess the effectiveness and safety of mesenchymal stem cell (MSC) transplantation in the treatment of autoimmune and rheumatic immune diseases through randomized controlled trials (RCTs). METHODS Two researchers conducted a comprehensive search of Chinese and English databases from their inception until Dec. 2023. The literature screening and data extraction were then performed. Statistical analysis was carried out using RevMan 5.4 software. RESULTS A total of 42 relevant RCTs, involving 2,183 participants, were ultimately included in this study. These RCTs encompassed four types of rheumatic immune and bone diseases, namely rheumatoid arthritis (RA), osteoarthritis (OA), spondyloarthritis, systemic sclerosis arthritis, systemic lupus erythematosus (SLE), inflammatory bowel disease, multiple sclerosis, primary Sjögren's syndrome (PSS). The systematic review indicates that MSC transplantation may improve spondyloarthritis, RA, PSS. The meta-analysis reveals that MSC transplantation significantly improved symptoms in patients with OA [VAS (visual analogue scale): bone marrow: SMD = - 0.95, 95% CI - 1.55 to - 0.36, P = 0.002; umbilical cord: SMD = - 1.25, 95% CI - 2.04 to - 0.46, P = 0.002; adipose tissue: SMD = -1.26, 95% CI -1.99 to - 0.52, P = 0.0009)], SLE [Systemic lupus erythematosus disease activity index (SLEDAI): SMD = - 2.32, 95% CI - 3.59 to - 1.06, P = 0.0003], inflammatory bowel disease [clinical efficacy: RR = 2.02, 95% CI 1.53 to 2.67, P < 0.00001]. However, MSC transplantation may not improve the symptoms of multiple sclerosis and systemic sclerosis (Ssc). Importantly, MSC transplantation did not increase the incidence of adverse events (OA: RR = 1.23, 95% CI 0.93 to 1.65, P = 0.15; SLE: RR = 0.83, 95% CI 0.28 to 2.51, P = 0.76; Inflammatory bowel disease: RR = 0.99, 95% CI 0.81 to 1.22, P = 0.96; Multiple sclerosis: RR = 1.12, 95% CI 0.81 to 1.53, P = 0.50), supporting its safety profile across the included studies. These findings suggest that MSC transplantation holds promise for several rheumatic and autoimmune diseases while highlighting areas where further research is warranted. CONCLUSION MSC transplantation may have the potential to treat autoimmune and rheumatic immune diseases. Moreover. MSC transplantation appears to be relatively safe and could be considered as a viable alternative treatment option for autoimmune and rheumatic immune diseases.
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.
| | - Chang 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
| | - Yang Wu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 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
| | - Wensa Hao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dandan Wang
- 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.
| | - 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
|
20
|
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 2025; 41:318-332. [PMID: 39154667 DOI: 10.1016/j.arthro.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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 In terms of efficacy, the clinical effects of intra-articular injection therapy using MSCs for KOA are superior to those of HA, and the cartilage repair effect of MSCs is also markedly better than that of HA. Although the clinical effects varied across time periods, the functional score reached the minimum clinically significant difference at both 6 and 12 months. In terms of safety, adverse reactions mainly manifest as joint pain, swelling, and joint effusion. Both intra-articular injections of MSCs and HA did not result in severe adverse reactions, indicating that MSCs and HA have similar safety profiles. 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
|
21
|
Pico OA, Espinoza F, Cádiz MI, Sossa CL, Becerra-Bayona SM, Salgado MCC, Rodríguez JER, Cárdenas OFV, Cure JMQ, Khoury M, Arango-Rodríguez ML. Efficacy of a single dose of cryopreserved human umbilical cord mesenchymal stromal cells for the treatment of knee osteoarthritis:a randomized, controlled, double-blind pilot study. Cytotherapy 2025; 27:188-200. [PMID: 39503681 DOI: 10.1016/j.jcyt.2024.09.005] [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: 06/26/2024] [Revised: 09/14/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Knee osteoarthritis (OA) is the most prevalent degenerative musculoskeletal disorder, which is particularly common in older population. While conventional treatments have limited effectiveness, the development of more effective therapeutic strategies is necessary to address this primary source of pain and disability. Umbilical cord mesenchymal stromal cells (UC-MSCs) offer a promising therapeutic approach for treating knee OA. AIM This randomized, prospective, double-blind and controlled pilot study was carried out to evaluate and compare the safety and therapeutic efficacy of a single intra-articular injection of a standardized product CellistemOA (5 × 106 ± 5 × 105 UC-MSCs), vs. triamcinolone (a synthetic corticosteroid) (10 mg/mL) in thirty patients with symptomatic knee OA (Kellgren-Lawrence grade II or III). METHODS The outcomes included changes in Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores based on a Likert scale, numerical rating score (NRS) for pain, Magnetic Resonance Imaging (MRI), and quality of life (SF-36 questionnaire), from baseline and throughout 12-months of follow-up. RESULTS Patients treated with CellistemOA showed significant improvement in WOMAC score (including the three subscale scores (pain, stiffness and function), NRS in pain, and SF-36 profile from baseline to 12 months (p < 0.05) compared to the triamcinolone group, and no severe adverse events were reported. There were no significant differences in MRI WORMS scores between the two groups. However, patients who received the cellular treatment experienced a significant improvement in their SF-36 profile (p < 0.05). CONCLUSIONS This pilot study revealed that a single dose of CellistemOA is safe and superior to the active comparator in knee OA at 1-year of follow-up, making it a compelling therapeutic alternative to treat symptomatic OA patients.
Collapse
Affiliation(s)
- Omar Amado Pico
- Fundación Oftalmológica de Santander - FOSCAL, Floridablanca, Colombia; Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | - Francisco Espinoza
- Cells for Cells & Consorcio Regenero, Santiago, Chile; Program for Translational Research in Cell Therapy, Universidad de los Andes, Santiago, Chile; Department of Rheumatology, Universidad de los Andes, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María Ignacia Cádiz
- Cells for Cells & Consorcio Regenero, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Laboratory of Nano-Regenerative Medicine, Universidad de los Andes, Santiago, Chile
| | - Claudia L Sossa
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia; Programa para el Tratamiento y Estudio de Enfermedades Hematológicas y Oncológicas de Santander (PROTEHOS), Floridablanca, Colombia
| | - Silvia M Becerra-Bayona
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | - María C Canencio Salgado
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | | | | | | | - Maroun Khoury
- Cells for Cells & Consorcio Regenero, Santiago, Chile; Program for Translational Research in Cell Therapy, Universidad de los Andes, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Laboratory of Nano-Regenerative Medicine, Universidad de los Andes, Santiago, Chile
| | - Martha L Arango-Rodríguez
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia; Banco Multitejidos y Centro de Terapias Avanzadas, Clínica FOSCAL Internacional, Floridablanca, Colombia.
| |
Collapse
|
22
|
Lei J, Chen X, Xie H, Dai Y, Chen Z, Xu L. Therapeutic efficacy of intra-articular injection of human adipose-derived mesenchymal stem cells in a sheep model of knee osteoarthritis. Stem Cell Res Ther 2025; 16:24. [PMID: 39849597 PMCID: PMC11755983 DOI: 10.1186/s13287-025-04143-6] [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: 11/06/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Mesenchymal stem cells have great potential for repairing articular cartilage and treating knee osteoarthritis (KOA). Nonetheless, little is known about the efficacy of human adipose-derived mesenchymal stem cells (haMSCs) for KOA in large animal models. METHODS This study evaluated the therapeutic efficacy of haMSCs in knee articular cartilage repair in a sheep model of KOA. haMSCs were isolated, cultured, and characterized. KOA was surgically induced by anterior cruciate ligament transection and medial meniscectomy, followed by intra-articular injection of saline (negative control group) or haMSCs (haMSC group) into the right knee joint at 6 and 9 weeks after surgery. Sheep were sacrificed 21 weeks after surgery, and samples (whole knee joints, femoral condyles, and tibias) were collected, processed, and analyzed. Changes in knee articular cartilage were assessed by magnetic resonance imaging, micro-computed tomography, macroscopic analysis, histology, and immunohistochemistry. RESULTS KOA caused the degeneration of the medial femoral condyle in the sheep model of KOA. Conversely, haMSCs repaired chondral defects and increased the thickness of knee articular cartilage. CONCLUSIONS These data suggest that the intra-articular injection of haMSCs can effectively repair articular cartilage defects in the knee.
Collapse
Affiliation(s)
- Jigang Lei
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Xingyi Chen
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Haohao Xie
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Yuhao Dai
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Zhongjin Chen
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China
| | - Liang Xu
- Cellular Biopharma (Shanghai) Co., Ltd, Building 3, No.85, Faladi Road, Pudong New Area, Shanghai, 200233, China.
| |
Collapse
|
23
|
Figueroa-Valdés AI, Luz-Crawford P, Herrera-Luna Y, Georges-Calderón N, García C, Tobar HE, Araya MJ, Matas J, Donoso-Meneses D, de la Fuente C, Cuenca J, Parra E, Lillo F, Varela C, Cádiz MI, Vernal R, Ortloff A, Nardocci G, Castañeda V, Adasme-Vidal C, Kunze-Küllmer M, Hidalgo Y, Espinoza F, Khoury M, Alcayaga-Miranda F. Clinical-grade extracellular vesicles derived from umbilical cord mesenchymal stromal cells: preclinical development and first-in-human intra-articular validation as therapeutics for knee osteoarthritis. J Nanobiotechnology 2025; 23:13. [PMID: 39806427 PMCID: PMC11730155 DOI: 10.1186/s12951-024-03088-x] [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: 06/25/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Osteoarthritis (OA) is a joint disease characterized by articular cartilage degradation. Persistent low-grade inflammation defines OA pathogenesis, with crucial involvement of pro-inflammatory M1-like macrophages. While mesenchymal stromal cells (MSC) and their small extracellular vesicles (sEV) hold promise for OA treatment, achieving consistent clinical-grade sEV products remains a significant challenge. This study aims to develop fully characterized, reproducible, clinical-grade batches of sEV derived from umbilical cord (UC)-MSC for the treatment of OA while assessing its efficacy and safety. Initially, a standardized, research-grade manufacturing protocol was established to ensure consistent sEV production. UC-MSC-sEV characterization under non-cGMP conditions showed consistent miRNA and protein profiles, suggesting their potential for standardized manufacturing. In vitro studies evaluated the efficacy, safety, and potency of sEV; animal studies confirmed their effectiveness and safety. In vitro, UC-MSC-sEV polarized macrophages to an anti-inflammatory M2b-like phenotype, through STAT1 modulation, indicating their potential to create an anti-inflammatory environment in the affected joints. In silico studies confirmed sEV's immunosuppressive signature through miRNA and proteome analysis. In an OA mouse model, sEV injected intra-articularly (IA) induced hyaline cartilage regeneration, validated by histological and μCT analyses. The unique detection of sEV signals within the knee joint over time highlights its safety profile by confirming the retention of sEV in the joint. The product development of UC-MSC-sEV involved refining, standardizing, and validating processes in compliance with GMP standards. The initial assessment of the safety of the clinical-grade product via IA administration in a first-in-human study showed no adverse effects after a 12 month follow-up period. These results support the progress of this sEV-based therapy in an early-phase clinical trial, the details of which are presented and discussed in this work. This study provides data on using UC-MSC-sEV as local therapy for OA, highlighting their regenerative and anti-inflammatory properties and safety in preclinical and a proof-of-principle clinical application.
Collapse
Affiliation(s)
- Aliosha I Figueroa-Valdés
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Nicolás Georges-Calderón
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Hugo E Tobar
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - José Matas
- Centro de Terapia Celular, Clínica Universidad de los Andes, Santiago, Chile
- Departmento de Cirugía Ortopédica, Clínica Universidad de los Andes, Santiago, Chile
| | - Darío Donoso-Meneses
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | | | - Jimena Cuenca
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Eliseo Parra
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Fernando Lillo
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Cristóbal Varela
- Departmento de Radiología, Clínica Universidad de los Andes, Santiago, Chile
| | - María Ignacia Cádiz
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Rolando Vernal
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Gino Nardocci
- Laboratorio de Biología Molecular y Bioinformática, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Verónica Castañeda
- Laboratorio de Biología Molecular y Bioinformática, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Catalina Adasme-Vidal
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Maximiliano Kunze-Küllmer
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- Cells for Cells, Santiago, Chile
- EVast Bio, Miami, FL, USA
| | - Yessia Hidalgo
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Francisco Espinoza
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Centro de Terapia Celular, Clínica Universidad de los Andes, Santiago, Chile
- Departmento de Reumatología, Clínica Universidad de los Andes, Santiago, Chile
| | - Maroun Khoury
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile.
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cells for Cells, Santiago, Chile.
- EVast Bio, Miami, FL, USA.
| | - Francisca Alcayaga-Miranda
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile.
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cells for Cells, Santiago, Chile.
| |
Collapse
|
24
|
Woolley K, Milan N, Master Z, Feeley BT. Evaluation of Spin in Clinical Trials of Mesenchymal Stromal Cells for the Treatment of Knee Osteoarthritis: A Systematic Review. Am J Sports Med 2025:3635465241274155. [PMID: 39772944 DOI: 10.1177/03635465241274155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
BACKGROUND The regenerative potential of mesenchymal stromal cells (MSCs) has sparked interest in their use for knee osteoarthritis. Concurrently, there have been investigations on how data in scientific journals are reported and how they may influence readers' interpretations, or "spin bias." These studies are at risk for bias, given the limited number of patients and inconsistent blinding or controls. The risk of spin bias also complicates the interpretation, as results may be presented in a way that favors a particular outcome. PURPOSE To quantify and characterize spin bias in clinical trials of MSCs for knee osteoarthritis. STUDY DESIGN Systematic review. METHODS PubMed and Embase searches were conducted using the terms "mesenchymal stem cells" or "MSCs" and "knee arthritis" or "osteoarthritis" and "therapy" or "treatment" or "regeneration." Overall, 2 independent reviewers classified spin as high, moderate, or low and as 1 of 3 types: (1) emphasizing statistically significant results, (2) interpreting nonsignificant results as treatment effectiveness, and (3) claiming treatment benefits despite nonsignificant results. Journals were categorized as orthopaedic or nonorthopaedic. Descriptive statistics, the chi-square test, and the Fisher exact test were used to analyze the data, with alpha set at P < .05. RESULTS Among the 54 studies, spin was found in 80.0% of articles, with 14.5% having high, 25.5% moderate, and 40.0% low levels of spin. Type 1 was found in 54.5% of articles, type 2 in 18.2%, and type 3 in 29.1%. Spin was less frequently observed in the Methods section of articles compared with the abstract (17.52; P = .003). Reports on adipose-derived MSCs were associated with a higher frequency and level of spin compared with reports on MSCs from other sources (18.92; P = .026). There was no difference in the frequency of spin between orthopaedic and nonorthopaedic journals (0.48; P = .49) and no association with the impact factor (5.34; P = .07). There was no association between spin and financial disclosures (0.02; P = .577). CONCLUSION Spin bias was present in most MSC-related trials for knee osteoarthritis, with a higher frequency among those that utilized adipose-derived MSCs. Understanding the prevalence and strategies of spin can mitigate any potential misinterpretations of study outcomes.
Collapse
Affiliation(s)
- Katherine Woolley
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Nesa Milan
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Zubin Master
- Department of Social Sciences and Health Policy, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Maya Angelou Center for Health Equity, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Brian T Feeley
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
25
|
Vega-Letter AM, García-Guerrero C, Yantén-Fuentes L, Pradenas C, Herrera-Luna Y, Lara-Barba E, Bustamante-Barrientos FA, Rojas M, Araya MJ, Jeraldo N, Aros C, Troncoso F, Poblete D, Court A, Ortloff A, Barraza J, Velarde F, Farkas C, Carril C, Luque-Campos N, Almarza G, Barahona M, Matas J, Cereceda L, Lorca R, Toledo J, Oyarce K, Vernal R, Caicedo A, Del Campo A, Hidalgo Y, Elizondo-Vega R, Djouad F, Khoury M, Figueroa FE, Luz-Crawford P. Safety and efficacy of mesenchymal stromal cells mitochondria transplantation as a cell-free therapy for osteoarthritis. J Transl Med 2025; 23:26. [PMID: 39773289 PMCID: PMC11706173 DOI: 10.1186/s12967-024-05945-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE The inflammatory responses from synovial fibroblasts and macrophages and the mitochondrial dysfunction in chondrocytes lead to oxidative stress, disrupt extracellular matrix (ECM) homeostasis, and accelerate the deterioration process of articular cartilage in osteoarthritis (OA). In recent years, it has been proposed that mesenchymal stromal cells (MSC) transfer their functional mitochondria to damaged cells in response to cellular stress, becoming one of the mechanisms underpinning their therapeutic effects. Therefore, we hypothesize that a novel cell-free treatment for OA could involve direct mitochondria transplantation, restoring both cellular and mitochondrial homeostasis. METHODS Mitochondria were isolated from Umbilical Cord (UC)-MSC (Mito-MSC) and characterized based on their morphology, phenotype, functions, and their ability to be internalized by different articular cells. Furthermore, the transcriptional changes following mitochondrial uptake by chondrocytes were evaluated using an Affymetrix analysis, Lastly, the dose dependence therapeutic efficacy, biodistribution and immunogenicity of Mito-MSC were assessed in vivo, through an intra-articular injection in male C57BL6 mice in a collagenase-induced OA (CIOA) model. RESULTS Our findings demonstrate the functional integrity of Mito-MSC and their ability to be efficiently transferred into chondrocytes, synovial macrophages, and synovial fibroblasts. Moreover, the transcriptomic analysis showed the upregulation of genes involved in stress such as DNA reparative machinery and inflammatory antiviral responses. Finally, Mito-MSC transplantation yielded significant reductions in joint mineralization, a hallmark of OA progression, as well as improvements in OA-related histological signs, with the lower dose exhibiting better therapeutic efficacy. Furthermore, Mito-MSC was detected within the knee joint for up to 24 h post-injection without eliciting an inflammatory response in CIOA mice. CONCLUSION Collectively, our results reveal that mitochondria derived from MSC are transferred to key articular cells and are retained in the joint without generating an inflammatory immune response mitigating articular cartilage degradation in OA, probably through a restorative effect triggered by the stress antiviral response within OA chondrocytes.
Collapse
Affiliation(s)
- Ana Maria Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Cynthia García-Guerrero
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Liliana Yantén-Fuentes
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Carolina Pradenas
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Yeimi Herrera-Luna
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Eliana Lara-Barba
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Masyelly Rojas
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - María Jesús Araya
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Nicole Jeraldo
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Red de Equipamiento Científico Avanzado-REDECA, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Constanza Aros
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Francisca Troncoso
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Daniela Poblete
- Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Angela Court
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Jose Barraza
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Francesca Velarde
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Carlos Farkas
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Claudio Carril
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Chile
| | - Noymar Luque-Campos
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Gonzalo Almarza
- Laboratorio de Fisiología y Bioenergetica Celular, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maximiliano Barahona
- Departamento de Ortopedia y Traumatología, Hospital Clinico Universidad de Chile, Independencia, Chile
| | - Jose Matas
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
| | - Lucas Cereceda
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Rocío Lorca
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Jorge Toledo
- Red de Equipamiento Científico Avanzado-REDECA, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Karina Oyarce
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Chile
| | - Rolando Vernal
- Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Andrés Caicedo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud e Instituto de Investigaciones en Biomedicina iBioMed, Escuela de Medicina, Quito, Ecuador-Mito-Act Research Consortium, Quito, Ecuador
| | - Andrea Del Campo
- Laboratorio de Fisiología y Bioenergetica Celular, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yessia Hidalgo
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Farida Djouad
- IRMB, Université de Montpellier, INSERM, Montpellier, France
- CHU Montpellier, Montpellier, France
| | - Maroun Khoury
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile.
| | - Fernando E Figueroa
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile.
| | - Patricia Luz-Crawford
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
| |
Collapse
|
26
|
Huang L, Guo Z, Yang X, Zhang Y, Liang Y, Chen X, Qiu X, Chen X. Advancements in GelMA bioactive hydrogels: Strategies for infection control and bone tissue regeneration. Theranostics 2025; 15:460-493. [PMID: 39744697 PMCID: PMC11671377 DOI: 10.7150/thno.103725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/04/2024] [Indexed: 01/11/2025] Open
Abstract
Infectious bone defects present a significant clinical challenge, characterized by infection, inflammation, and subsequent bone tissue destruction. Traditional treatments, including antibiotic therapy, surgical debridement, and bone grafting, often fail to address these defects effectively. However, recent advancements in biomaterials research have introduced innovative solutions for managing infectious bone defects. GelMA, a three-dimensional network of hydrophilic polymers that can absorb and retain substantial amounts of water, has attracted considerable attention in the fields of materials science and biomedical engineering. Its distinctive properties, such as biocompatibility, responsiveness to stimuli, and customisable mechanical characteristics make GelMA an exemplary scaffold material for bone tissue engineering. This review aims to thoroughly explore the current literature on antibacterial and osteogenic strategies using GelMA hydrogels for the restoration of infected bones. It discusses their fabrication methods, biocompatibility, antibacterial effectiveness, and bioactivity. We conclude by discussing the existing challenges and future research directions in this field, with the hope of inspiring further innovations in the synthesis, modification, and application of GelMA-based hydrogels for infection control and bone tissue regeneration.
Collapse
Affiliation(s)
- Lei Huang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Ziyao Guo
- SCP 11A of the International Department, Guangzhou Experimental Foreign Language School, Guangzhou, China
| | - Xiaoxia Yang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yinchun Zhang
- Department of Periodontology, Shaoxing Stomatological Hospital, Shaoxing, Zhejiang, China
| | - Yiyun Liang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiaxue Chen
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiaoling Qiu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xuan Chen
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Lin J, Huang J, Jiao Z, Nian M, Li C, Dai Y, Jia S, Zhang X. Mesenchymal stem cells for osteoarthritis: Recent advances in related cell therapy. Bioeng Transl Med 2025; 10:e10701. [PMID: 39801757 PMCID: PMC11711223 DOI: 10.1002/btm2.10701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/20/2024] [Accepted: 07/04/2024] [Indexed: 01/06/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease that affects the entire joint and has been a huge burden on the health care system worldwide. Although traditional therapy and targeted cartilage cell therapy have made significant progress in the treatment of OA and cartilage regeneration, there are still many problems. Mesenchymal stem cells from various tissues are the most studied cell type and have been used in preclinical and clinical studies of OA, because they are more widely available, have a greater capacity for in vitro expansion, and have anti-inflammatory and immunomodulatory properties compared to autologous chondrocytes. This article will systematically review the latest developments in these areas. It may provide new insights for improving OA and cartilage regeneration.
Collapse
Affiliation(s)
- Jianjing Lin
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
| | - Jingtao Huang
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Zilu Jiao
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
| | - Mengyuan Nian
- Cardre Health Care DepartmentPeking University Shenzhen HospitalShenzhenChina
| | - Canfeng Li
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
| | - Yali Dai
- Cardre Health Care DepartmentPeking University Shenzhen HospitalShenzhenChina
| | - Shicheng Jia
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Xintao Zhang
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
| |
Collapse
|
28
|
Im GI. Clinical updates in mesenchymal stromal cell therapy for osteoarthritis treatment. Expert Opin Biol Ther 2025; 25:187-195. [PMID: 39710894 DOI: 10.1080/14712598.2024.2446612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/21/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) is a common chronic musculoskeletal disease with heterogeneous clinical manifestations and variable responses to different treatments. Unfortunately, there is no effective disease modifying therapy at present that can alter the natural course of the disease. Cell therapy based on mesenchymal stromal cells (MSCs) may offer an attractive therapeutic option for OA with their multiple modes of action, particularly immune-regulatory and regenerative capacities. AREAS COVERED In this narrative review, updates on mode of action based on patient's data, factors that can influence the efficacy of MSC treatment, current status in clinical application of MSCs as seen from randomized, controlled OA trials are introduced as well as the author's perspectives in the future of MSCs as OA therapeutics. EXPERT OPINION Symptomatic relief is not sufficient to justify the high cost associated with culture-expanded stem cells. Its advantages and efficacy over simple and low risk/cost modalities should be seriously reevaluated. Also, as the short-term strategy, efforts should be made to lower the cost of MSC therapy. In the future, multiomics technology may help to predict that subgroup of patients who will favorably respond to stem cell treatment, which would enhance the cost effectiveness and therapeutic benefit of MSC therapy.
Collapse
Affiliation(s)
- Gun-Il Im
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| |
Collapse
|
29
|
Zhang A, Li Q, Chen Z. Therapeutic Efficacy and Promise of Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles in Aging and Age-Related Disorders. Int J Mol Sci 2024; 26:225. [PMID: 39796081 PMCID: PMC11719504 DOI: 10.3390/ijms26010225] [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: 12/04/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
The global issue of aging populations has become increasingly prominent, thus the research and development for anti-aging therapies to assure longevity as well as to ameliorate age-related complications is put high on the agenda. The young humoral milieu has been substantiated to impart youthful characteristics to aged cells or organs. Extracellular vesicles (EVs) are a heterogeneous group of cell-derived membrane-limited structures that serve as couriers of proteins and genetic material to regulate intercellular communication. Of note, EVs appeared to be an indispensable component of young blood in prolonging lifespans, and circulating EVs have been indicated to mediate the beneficial effect of a young milieu on aging. Human umbilical cord mesenchymal stem cell-derived EVs (HUCMSC-EVs), isolated from the youngest adult stem cell source, are speculated to reproduce the function of circulating EVs in young blood and partially revitalize numerous organs in old animals. Robust evidence has suggested HUCMSC-EVs as muti-target therapeutic agents in combating aging and alleviating age-related degenerative disorders. Here, we provide a comprehensive overview of the anti-aging effects of HUCMSC-EVs in brain, heart, vasculature, kidney, muscle, bone, and other organs. Furthermore, we critically discuss the current investigation on engineering strategies of HUCMSC-EVs, intending to unveil their full potential in the field of anti-aging research.
Collapse
Affiliation(s)
- Anyuan Zhang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Qiubai Li
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhichao Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| |
Collapse
|
30
|
Malige A, Gates C, Cook JL. Mesenchymal stem cells in orthopaedics: A systematic review of applications to practice. J Orthop 2024; 58:1-9. [PMID: 39035449 PMCID: PMC11254590 DOI: 10.1016/j.jor.2024.06.026] [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: 05/12/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024] Open
Abstract
Background Mesenchymal stem cells (MSCs) have alluring interest for clinical use in orthopaedics based on their therapeutic potential through directed pluripotent differentiation. While many studies and reviews have discussed the importance of this approach, few have reduced it to practice using reproducible criteria. This study was designed to systematically review and synthesize current evidence regarding clinical use of clearly defined MSCs in orthopaedics. Methods Studies of any level of evidence and sample size, regardless of MSC source, orthopaedic pathology, and patient population, were reviewed. In vitro and animal studies, and articles written in a language other than English, were excluded. Studies were then screened for final inclusion based on documented MSC verification using testing of the therapeutic cellular population for at least one of the following phenotypic markers: CD 73, CD 90, and CD 105. In addition, therapeutic cellular populations could not have higher percentages of CD34, CD45, CD14, HLA-DR, CD11b, or CD19 markers compared to the aforementioned markers. From each studies' results, sample size, procedural methods, radiographic outcomes, clinical outcomes, patient-report outcomes (PROs), and adverse events were tabulated. Results Overall, 43 studies were included. Twenty-three studies (53.5 %) derived their MSCs from iliac crest bone marrow while 12 (27.9 %) studied adipose-derived MSCs. Included studies explored MSC use in Osteoarthritis, Cartilage Defects, Osteonecrosis, Bone Defects and Nonunions, Spine, and Other. MSC use in all pathologies led to improvement of studied radiographic, clinical, and patient-reported outcomes. Conclusions Mesenchymal stem cells have proven to have successful and safe uses in multiple orthopaedic applications, including treating chondral defects, osteoarthritis, and osteonecrosis. A stringent and reproducible process for evaluating obtained human stem cells using CD markers for clinical use is necessary to both evaluate previous studies and continue to evaluate for future uses. Level of evidence Level V.
Collapse
Affiliation(s)
- Ajith Malige
- Kerlan Jobe Orthopedic Clinic, 6801 Park Terrace, Suite 500, Los Angeles, CA, 90045, USA
| | - Carson Gates
- University of Missouri Department of Orthopaedic Surgery, Columbia, MO, USA
| | - James L. Cook
- University of Missouri Department of Orthopaedic Surgery, Columbia, MO, USA
| |
Collapse
|
31
|
Xiao KZ, Liao G, Huang GY, Huang YL, Gu RH. Efficacy of serum-free cultured human umbilical cord mesenchymal stem cells in the treatment of knee osteoarthritis in mice. World J Stem Cells 2024; 16:944-955. [DOI: 10.4252/wjsc.v16.i11.944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND We investigated the efficacy of intra-articular injection of human umbilical cord mesenchymal stem cells (hUC-MSCs) for the treatment of osteoarthritis (OA) progression in the knee joint. Although many experimental studies of hUC-MSCs have been published, these studies have mainly used fetal bovine serum-containing cultures of hUC-MSCs; serum-free cultures generally avoid the shortcomings of serum-containing cultures and are not subject to ethical limitations, have a wide range of prospects for clinical application, and provide a basis or animal experimentation for clinical experiments.
AIM To study the therapeutic effects of serum-free hUC-MSCs (N-hUCMSCs) in a mouse model of knee OA.
METHODS Fifty-five male C57BL/6 mice were randomly divided into six groups: The blank control group, model control group, serum-containing hUC-MSCs (S-hUCMSC) group, N-hUCMSC group and hyaluronic acid (HA) group. After 9 weeks of modeling, the serum levels of interleukin (IL)-1β and IL-1 were determined. Hematoxylin-eosin staining was used to observe the cartilage tissue, and the Mankin score was determined. Immunohistochemistry and western blotting were used to determine the expression of collagen type II, matrix metalloproteinase (MMP)-1 and MMP-13.
RESULTS The Mankin score and serum IL-1 and IL-1β and cartilage tissue MMP-1 and MMP-13 expression were significantly greater in the experimental group than in the blank control group (P < 0.05). Collagen II expression in the experimental group was significantly lower than that in the blank control group (P < 0.05). The Mankin score and serum IL-1 and IL-1β and cartilage tissue MMP-1 and MMP-13 levels the experimental group were lower than those in the model control group (P < 0.05). Collagen II expression in the experimental group was significantly greater than that in the model control group (P < 0.05).
CONCLUSION N-hUCMSC treatment significantly alleviate the pathological damage caused by OA. The treatment effects of the S- hUCMSC group and HA group were similar.
Collapse
Affiliation(s)
- Kai-Zhen Xiao
- Department of Orthopedic Surgery, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning 530022, Guangxi Zhuang Autonomous Region, China
- Graduate School of Guangxi Medical University, Guangxi Medical University, Nanning 530022, Guangxi Zhuang Autonomous Region, China
| | - Gui Liao
- Department of Orthopedic Surgery, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning 530022, Guangxi Zhuang Autonomous Region, China
| | - Guang-Yu Huang
- Department of Orthopedic Surgery, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning 530022, Guangxi Zhuang Autonomous Region, China
- Graduate School of Guangxi Medical University, Guangxi Medical University, Nanning 530022, Guangxi Zhuang Autonomous Region, China
| | - Yun-Long Huang
- Department of Orthopedic Surgery, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning 530022, Guangxi Zhuang Autonomous Region, China
- Graduate School of Guangxi Medical University, Guangxi Medical University, Nanning 530022, Guangxi Zhuang Autonomous Region, China
| | - Rong-He Gu
- Department of Orthopedic Surgery, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning 530022, Guangxi Zhuang Autonomous Region, China
- Department of Guangxi Key Laboratory of Intelligent Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning 530022, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
32
|
Liu L, Hao X, Zhang J, Li S, Han S, Qian P, Zhang Y, Yu H, Kang Y, Yin Y, Zhang W, Chen J, Yu Y, Jiang H, Chai J, Yin H, Chai W. The wound healing of deep partial-thickness burn in Bama miniature pigs is accelerated by a higher dose of hUCMSCs. Stem Cell Res Ther 2024; 15:437. [PMID: 39563365 PMCID: PMC11575178 DOI: 10.1186/s13287-024-04063-x] [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: 06/13/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Deep partial-thickness burns have a significant impact on both the physical and mental health of patients. Our previous study demonstrated human Umbilical Cord Mesenchymal stem cells (hUCMSCs) could enhance the healing of severe burns in small animal burn models, such as rats. Furthermore, our team has developed a deep partial-thickness burn model in Bama miniature pigs, which can be utilized for assessing drug efficacy in preclinical trials for wound healing. Therefore, this study further determine the optimal dosage of hUCMSCs in future clinical practice by comparing the efficacy of low-to-high doses of hUCMSCs on deep partial-thickness burn wounds in Bama miniature pigs. MATERIALS AND METHODS The male Bama miniature pigs (N = 8, weight: 23-28 kg and length: 71-75 cm) were used to establish deep partial-thickness burn models, which used a continuous pressure of 1 kg and contact times of 35 s by the invented electronic burn instrument at 100℃ to prepare 10 round burn wounds with diameter of 5 cm according to our previous report. And then, 0 × 10^7, 1 × 10^7, 2 × 10^7, 5 × 10^7 and 1 × 10^8 doses of hUCMSCs were respectively injected into burn wounds of their corresponding groups. After treatment for 7, 14 and 21 days, the burned wound tissues were obtained for histological evaluation, including HE staining for histopathological changes, immunohistochemistry for neutrophil (MPO+) infiltration and microvessel (CD31+) quantity, as well as Masson staining for collagen deposition. The levels of inflammatory factors TNF-α, IL-1β, IL-10 and angiogenesis factors angiopoietin-2 (Ang-2), vascular endothelial growth factor (VEGF), as well as collagen type-I/type-III of the wound tissues were quantified by ELISA. RESULTS All of doses hUCMSCs can significantly increase wound healing rate and shorten healing time of the deep partial-thickness burn pigs in a dose-dependent manner. Furthermore, all of doses hUCMSCs can significantly promote epithelialization and decreased inflammatory reaction of wound, including infiltration of inflammatory cells and levels inflammatory factors. Meanwhile, the amounts of microvessel were increased in all of doses hUCMSCs group than those in the burn group. Furthermore, the collagen structure was disordered and partially necrotized, and ratios of collagen type-I and type-III were significantly decreased in burn group (4:1 in normal skin tissue), and those of all hUCMSCs groups were significantly improved in a dose-dependent manner. In a word, 1 × 10^8 dose of hUCMSCs could regenerate the deep partial-thickness burn wounds most efficaciously compared to other dosages groups and the burn group. CONCLUSION This regenerative cell therapy study using hUCMSCs demonstrates the best efficacy toward a high dose, that is dose of 1 × 10^8 of hUCMSCs was used as a reference therapeutic dose for treating 20 cm2 deep partial-thickness burns wound in future clinical practice.
Collapse
Affiliation(s)
- Lingying Liu
- Department of Nutrition, The Fourth Medical Center Affiliated to PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100037, China.
- Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China.
- Hebei North University, Zhangjiakou, Hebei, 075000, China.
| | - Xingxia Hao
- Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China
| | - Jing Zhang
- Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China
| | - Shaozeng Li
- Department of Clinical Laboratory, The Fourth Medical Center Affiliated to PLA General Hospital, Beijing, 100037, China
| | - Shaofang Han
- Department of Nutrition, The Fourth Medical Center Affiliated to PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100037, China
| | - Peipei Qian
- Department of Nutrition, The Fourth Medical Center Affiliated to PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100037, China
| | - Yong Zhang
- Department of Nutrition, The Fourth Medical Center Affiliated to PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100037, China.
| | - Huaqing Yu
- Department of Nutrition, The Fourth Medical Center Affiliated to PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100037, China
| | - Yuxin Kang
- Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Yue Yin
- Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China
| | - Weiouwen Zhang
- Department of Nutrition, The Fourth Medical Center Affiliated to PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100037, China
| | - Jianmei Chen
- Department of Health Medicine, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100037, China
| | - Yang Yu
- Department of Traditional Chinese Medical Science, The Sixth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Hua Jiang
- Department of Endocrinology, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jiake Chai
- Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of PLA General Hospital, Beijing, 100037, China
| | - Huinan Yin
- Senior Department of Burns and Plastic Surgery, The Fourth Medical Center of PLA General Hospital, Beijing, 100037, China
| | - Wei Chai
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100037, China
| |
Collapse
|
33
|
Liu T, Ran C, Zhao D, Yang F, Guo Q, Yang J, Zhang X. Mesenchymal stem cells and their exosomes mitigate osteoarthritis by restoring the balance between proinflammatory Teffs and Tregs. FRONTIERS IN AGING 2024; 5:1509014. [PMID: 39629263 PMCID: PMC11611854 DOI: 10.3389/fragi.2024.1509014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Osteoarthritis (OA) is a degenerative joint disease caused by chronic inflammation that damages articular cartilage. In addition to the wear and tear of joints, aberrant remodelling driven by a significant presence of inflammatory mediators within the joint is one of the key mechanisms in the pathogenesis of OA. Among these factors, hyperactivation of Teffs subsets plays a crucial role in promoting this pathological process. The immune imbalance between proinflammatory CD4+ effector T cells (proinflammatory Teffs) and Tregs could be a crucial factor in the pathogenesis of OA. Therefore, correcting the imbalance of Tregs/proinflammatory Teffs may slow or inhibit the occurrence and development of OA, which could be a potential target for the treatment of OA. Mesenchymal stem cells (MSCs) possess anti-inflammatory and immunomodulatory properties, regulating both adaptive and innate immunity through mechanisms involving soluble factors such as IDO, PGE2, and TGF-β, as well as cell-to-cell contact and exosomes. Correcting the imbalance between Tregs and proinflammatory Teffs may be one of the mechanisms of MSCs in the treatment of OA. Therefore, this review aims to summarize the relationship between OA and the immune imbalance between Tregs and proinflammatory Teffs, the immunoregulatory role of Tregs in OA, and the role of MSCs and their exosomes in correcting the imbalance between Tregs and proinflammatory Teffs.
Collapse
Affiliation(s)
- Tianhao Liu
- Zhongshan Clinical College, Dalian University, Dalian, Liaoning, China
| | - Chunxiao Ran
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Dewei Zhao
- Zhongshan Clinical College, Dalian University, Dalian, Liaoning, China
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Fan Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Qiang Guo
- Zhongshan Clinical College, Dalian University, Dalian, Liaoning, China
| | - Jiahui Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Xiuzhi Zhang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
34
|
Semitela A, Marques PAAP, Completo A. Strategies to engineer articular cartilage with biomimetic zonal features: a review. Biomater Sci 2024; 12:5961-6005. [PMID: 39463257 DOI: 10.1039/d4bm00579a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Articular cartilage (AC) is a highly specialized tissue with restricted ability for self-regeneration, given its avascular and acellular nature. Although a considerable number of surgical treatments is available for the repair, reconstruction, and regeneration of AC defects, most of them do not prioritize the development of engineered cartilage with zonal stratification derived from biomimetic biochemical, biomechanical and topographic cues. In the absence of these zonal elements, engineered cartilage will exhibit increased susceptibility to failure and will neither be able to withstand the mechanical loading to which AC is subjected nor will it integrate well with the surrounding tissue. In this regard, new breakthroughs in the development of hierarchical stratified engineered cartilage are highly sought after. Initially, this review provides a comprehensive analysis of the composition and zonal organization of AC, aiming to enhance our understanding of the significance of the structure of AC for its function. Next, we direct our attention towards the existing in vitro and in vivo studies that introduce zonal elements in engineered cartilage to elicit appropriate AC regeneration by employing tissue engineering strategies. Finally, the advantages, challenges, and future perspectives of these approaches are presented.
Collapse
Affiliation(s)
- Angela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
35
|
Xiao Z, Wang X, Li C, Luo L, Li W. Effects of the umbilical cord mesenchymal stem cells in the treatment of knee osteoarthritis: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e40490. [PMID: 39560593 PMCID: PMC11575993 DOI: 10.1097/md.0000000000040490] [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: 05/18/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND This study aimed to evaluate the effects of umbilical cord mesenchymal stem cells (UC-MSCs) in the treatment of knee osteoarthritis. METHODS PubMed, Web of Science, Cochrane Library, Embase, Chinese National Knowledge Infrastructure and Wanfang databases were searched from inception to March 31, 2024. RevMan 5.3 was used to conduct meta-analyses of the final included studies. RESULTS Three randomized controlled studies were conducted. Western Ontario and McMaster Universities Osteoarthritis Index was reduced in the UC-MSCs group compared that in to the control group (mean difference: -25.85; 95% confidence interval: -41.50, -10.20; P = .001). Knee Lysholm Score was improved in the UC-MSCs group compared with the control group (mean difference: 18.33; 95% confidence interval: 12.89, 23.77; P < .00001). Egger test showed P = .583, indicating no publication bias. Sensitivity analysis indicated that the results were stable. CONCLUSION Intra-articular injection of UC-MSCs improved function and reduced pain in patients with knee osteoarthritis. However, the number of included studies was small and more studies are needed to confirm this.
Collapse
Affiliation(s)
- Zhijian Xiao
- Hangzhou Fuyang Hospital of TCM Orthopedics and Traumtology, Hangzhou, China
| | - Xinying Wang
- Hangzhou Tongjuntang Second TCM Clinic, Hangzhou, China
| | - Cheng Li
- Hangzhou Fuyang Hospital of TCM Orthopedics and Traumtology, Hangzhou, China
| | - Lihua Luo
- Hangzhou Tongyuderen TCM Clinic, Hangzhou, China
| | - Wei Li
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
36
|
Bolandnazar NS, Raeissadat SA, Haghighatkhah H, Rayegani SM, Oshnari RS, Keshel SH, Zahraei M, Aalipour K, Babaee M, Zamani A, Rad ZB, Soleimani M, Sefat F. Safety and efficacy of placental mesenchymal stromal cells-derived extracellular vesicles in knee osteoarthritis: a randomized, triple-blind, placebo-controlled clinical trial. BMC Musculoskelet Disord 2024; 25:856. [PMID: 39465400 PMCID: PMC11514941 DOI: 10.1186/s12891-024-07979-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Knee osteoarthritis causes pain and disability in many people worldwide, for which no definitive treatment has yet been proposed. In this study, we investigated the safety and efficacy of placental mesenchymal stromal cells-derived extracellular vesicles in patients with knee osteoarthritis. METHODS This triple-blind, randomized clinical trial included patients suffering from bilateral knee osteoarthritis with grade 2 or 3. The knees of each patient were randomized to intervention and control. For the intervention knee, 5 cc of placental mesenchymal stromal cells-derived extracellular vesicles were injected, and for the control knee, 5 cc of normal saline was injected. The patients' symptoms were evaluated before the intervention and 2 and 6 months after the intervention using VAS, WOMAC questionnaire, and Lequesne index. MRI was performed before the intervention and 6 months after the intervention to evaluate retropatellar and tibiofemoral cartilage volume, medial and lateral meniscal disintegrity, ACL injury, and effusion-synovitis. RESULTS 62 knees (31 patients) were enrolled in this study. There were 31 knees as intervention and 31 knees as control. Finally, the data of 58 knees (29 patients) were analyzed, including 28 women and 1 man. The mean age of the patients was 55.38 ± 6.07 years. No statistically significant difference was detected between the two groups in clinical outcomes (including VAS, WOMAC, and Lequesne scores) before treatment and 2 and 6 months after treatment. Also, no statistically significant difference was detected between the two groups in MRI findings before treatment and 6 months after treatment. No systemic complications or severe local reactions occurred in the patients. CONCLUSION A single intra-articular injection of placental mesenchymal stromal cells-derived extracellular vesicles (5 cc, 7 × 109 particles/cc) is safe, but does not improve clinical symptoms or MRI findings in knee osteoarthritis beyond placebo. The protocol of this study was approved on 11 May 2022 with registration number IRCT20210423051054N1.
Collapse
Affiliation(s)
- Najmeh Sadat Bolandnazar
- Physical Medicine and Rehabilitation Research Center, Medical and Educational Center of Taleghani Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ahmad Raeissadat
- Physical Medicine and Rehabilitation Department, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Haghighatkhah
- Radiology Department, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mansoor Rayegani
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasa Salmani Oshnari
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeed Heidari Keshel
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Zahraei
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kianmehr Aalipour
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Babaee
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Zamani
- Radiology Department, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford, UK
- Interdisciplinary Research Center in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford, UK
| |
Collapse
|
37
|
Muniz TDTP, Rossi MC, de Vasconcelos Machado VM, Alves ALG. Mesenchymal Stem Cells and Tissue Bioengineering Applications in Sheep as Ideal Model. Stem Cells Int 2024; 2024:5176251. [PMID: 39465229 PMCID: PMC11511598 DOI: 10.1155/2024/5176251] [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: 01/25/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
The most common technologies in tissue engineering include growth factor therapies; metal implants, such as titanium; 3D bioprinting; nanoimprinting for ceramic/polymer scaffolds; and cell therapies, such as mesenchymal stem cells (MSCs). Cell therapy is a promising alternative to organ grafts and transplants in the treatment of numerous musculoskeletal diseases. MSCs have increasingly been used in generative medicine due to their specialized self-renewal, immunomodulation, multiplication, and differentiation properties. To further expand the potential of these cells in tissue repair, significant efforts are currently dedicated to the production of biomaterials with desirable short- and long-term biophysical properties that can aid the differentiation and expansion of MSCs. Biomaterials support MSC differentiation by modulating their characteristics, such as composition, mechanical properties, porosity, and topography. This review aimed to describe recent MSC approaches, including those associated with biomaterials, from experimental, clinical, and preclinical studies with sheep models.
Collapse
Affiliation(s)
- Talita D'Paula Tavares Pereira Muniz
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| | - Mariana Correa Rossi
- Materials Engineering Department (DEMa), São Carlos Federal University (UFSCar), 13.565-905, São Carlos, Sao Paulo, Brazil
| | - Vânia Maria de Vasconcelos Machado
- Department of Veterinary Surgery and Animal Reproduction, Imaging Diagnostic Sector, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| | - Ana Liz Garcia Alves
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| |
Collapse
|
38
|
Deng Z, Zeng X, Lin B, Chen L, Wu J, Zheng J, Ma Y, Lyu FJ, Zheng Q. Human umbilical cord mesenchymal stem cells on treating osteoarthritis in a rabbit model: Injection strategies. Heliyon 2024; 10:e38384. [PMID: 39430502 PMCID: PMC11489144 DOI: 10.1016/j.heliyon.2024.e38384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/22/2024] Open
Abstract
Human umbilical cord mesenchymal stem cells (UCMSCs) are a novel stem-cell source to treat osteoarthritis (OA). Here we investigated the therapeutic effects of UCMSCs injection strategies on knee OA in a rabbit model. Thirty OA rabbits randomly received normal saline, a single dose of 1 × 106 UCMSCs, or three injections of 1 × 106 UCMSCs at 2, 4, 6 weeks. Articular cartilages were collected after 8 weeks. Macroscopic and histological assessments indicated that intra-articular injection of UCMSCs, both single and multiple injection, significantly reduced the formation of periarticular osteophytes and articular cartilage degeneration when compared with the control. Furthermore, both UCMSCs injections increased the expression of chondrogenic markers in the articular cartilage, and reduced the levels of TNF-α and IL-6 in synovium. Micro-CT showed significant reduction of sub-chondral bone degeneration and osteophytes in the multiple-injection group compared to the control and single-injection group. Taken together, intra-articular injection of UCMSCs for OA treatment is safe and effective. Single and multiple injection of UCMSCs had comparable reparative effect on cartilage lesions, while multiple injection of UCMSCs further exerted effect on enhancing subchondral bone volume.
Collapse
Affiliation(s)
- Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaoli Zeng
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Bofu Lin
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lixuan Chen
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Jiwei Wu
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Jie Zheng
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Feng-Juan Lyu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
Irwin RM, Thomas MA, Fahey MJ, Mayán MD, Smyth JW, Delco ML. Connexin 43 regulates intercellular mitochondrial transfer from human mesenchymal stromal cells to chondrocytes. Stem Cell Res Ther 2024; 15:359. [PMID: 39390589 PMCID: PMC11468299 DOI: 10.1186/s13287-024-03932-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND The phenomenon of intercellular mitochondrial transfer from mesenchymal stromal cells (MSCs) has shown promise for improving tissue healing after injury and has potential for treating degenerative diseases like osteoarthritis (OA). Recently MSC to chondrocyte mitochondrial transfer has been documented, but the mechanism of transfer is unknown. Full-length connexin 43 (Cx43, encoded by GJA1) and the truncated, internally translated isoform GJA1-20k have been implicated in mitochondrial transfer between highly oxidative cells, but have not been explored in orthopaedic tissues. Here, our goal was to investigate the role of Cx43 in MSC to chondrocyte mitochondrial transfer. In this study, we tested the hypotheses that (a) mitochondrial transfer from MSCs to chondrocytes is increased when chondrocytes are under oxidative stress and (b) MSC Cx43 expression mediates mitochondrial transfer to chondrocytes. METHODS Oxidative stress was induced in immortalized human chondrocytes using tert-Butyl hydroperoxide (t-BHP) and cells were evaluated for mitochondrial membrane depolarization and reactive oxygen species (ROS) production. Human bone-marrow derived MSCs were transduced for mitochondrial fluorescence using lentiviral vectors. MSC Cx43 expression was knocked down using siRNA or overexpressed (GJA1 + and GJA1-20k+) using lentiviral transduction. Chondrocytes and MSCs were co-cultured for 24 h in direct contact or separated using transwells. Mitochondrial transfer was quantified using flow cytometry. Co-cultures were fixed and stained for actin and Cx43 to visualize cell-cell interactions during transfer. RESULTS Mitochondrial transfer was significantly higher in t-BHP-stressed chondrocytes. Contact co-cultures had significantly higher mitochondrial transfer compared to transwell co-cultures. Confocal images showed direct cell contacts between MSCs and chondrocytes where Cx43 staining was enriched at the terminal ends of actin cellular extensions containing mitochondria in MSCs. MSC Cx43 expression was associated with the magnitude of mitochondrial transfer to chondrocytes; knocking down Cx43 significantly decreased transfer while Cx43 overexpression significantly increased transfer. Interestingly, GJA1-20k expression was highly correlated with incidence of mitochondrial transfer from MSCs to chondrocytes. CONCLUSIONS Overexpression of GJA1-20k in MSCs increases mitochondrial transfer to chondrocytes, highlighting GJA1-20k as a potential target for promoting mitochondrial transfer from MSCs as a regenerative therapy for cartilage tissue repair in OA.
Collapse
Affiliation(s)
- Rebecca M Irwin
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Matthew A Thomas
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Megan J Fahey
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - James W Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA, 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Michelle L Delco
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
40
|
Court AC, Vega-Letter AM, Parra-Crisóstomo E, Velarde F, García C, Ortloff A, Vernal R, Pradenas C, Luz-Crawford P, Khoury M, Figueroa FE. Mitochondrial transfer balances cell redox, energy and metabolic homeostasis in the osteoarthritic chondrocyte preserving cartilage integrity. Theranostics 2024; 14:6471-6486. [PMID: 39479450 PMCID: PMC11519804 DOI: 10.7150/thno.96723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/22/2024] [Indexed: 11/02/2024] Open
Abstract
Osteoarthrosis (OA) is a leading cause of disability and early mortality, with no disease modifying treatment. Mitochondrial (MT) dysfunction and changes in energy metabolism, leading to oxidative stress and apoptosis, are main drivers of disease. In reaction to stress, mesenchymal stromal/stem cells (MSCs) donate their MT to damaged tissues. Methods: To evaluate the capacity of clinically validated MSCs to spontaneously transfer their MT to human OA chondrocytes (OA-Ch), primary cultured Ch isolated from the articular cartilage of OA patients were co-cultured with MT-labeled MSCs. MT transfer (MitoT) was evidenced by flow cytometry and confocal microscopy of MitoTracker-stained and YFP-tagged MT protein. MT persistence and metabolic analysis on target cells were assessed by direct transfer of MSC-derived MT to OA-Chs (Mitoception), through SNP-qPCR analysis, ATP measurements and Seahorse technology. The effects of MitoT on MT dynamics, oxidative stress and cell viability were gauged by western blot of fusion/fission proteins, confocal image analysis, ROS levels, Annexin V/7AAD and TUNEL assays. Intra-articular injection of MSC-derived MT was tested in a collagenase-induced murine model of OA. Results: Dose-dependent cell-to-cell MitoT from MSCs to cultured OA-Chs was detected starting at 4 hours of co-culture, with increasing MT-fluorescence levels at higher MSC:Ch ratios. PCR analysis confirmed the presence of exogenous MSC-MT within MitoT+ OA-Chs up to 9 days post Mitoception. MitoT from MSCs to OA-Ch restores energetic status, with a higher ATP production and metabolic OXPHOS/Glycolisis ratio. Significant changes in the expression of MT network regulators, increased MFN2 and decreased p-DRP1, reveal that MitoT promotes MT fusion restoring the MT dynamics in the OA-Ch. Additionally, MitoT increases SOD2 transcripts, protein, and activity levels, and reduces ROS levels, confering resistance to oxidative stress and enhancing resistance to apoptosis. Intra-articular injection of MSC-derived MT improves histologic scores and bone density of the affected joints in the OA mouse model, demonstrating a protective effect of MT transplantation on cartilage degradation. Conclusion: The Mitochondria transfer of MSC-derived MT induced reversal of the metabolic dysfunction by restoring the energetic status and mitochondrial dynamics in the OA chondrocyte, while conferring resistance to oxidative stress and apoptosis. Intra-articular injection of MT improved the disease in collagenase-induced OA mouse model. The restoration of the cellular homeostasis and the preclinical benefit of the intra-articular MT treatment offer a new approach for the treatment of OA.
Collapse
Affiliation(s)
- Angela C. Court
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ana María Vega-Letter
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliseo Parra-Crisóstomo
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Francesca Velarde
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Rolando Vernal
- Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Carolina Pradenas
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Patricia Luz-Crawford
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Maroun Khoury
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Fernando E. Figueroa
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Laboratory Cell and Molecular Immunology, CIIB, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
41
|
Gong X, Jiao Y, Hu H, Zhang R, Jia W, Zhao J, Liu Z, Xin Y, Han W. A prospective randomized controlled study of multi-intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure and reduced ejection fraction (PRIME-HFrEF) trial: Rationale and design. Contemp Clin Trials Commun 2024; 41:101350. [PMID: 39246626 PMCID: PMC11377133 DOI: 10.1016/j.conctc.2024.101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/12/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024] Open
Abstract
Background and objective The use of mesenchymal stem cells for heart failure treatment has gained increasing interest. However, most studies have relied on a single injection approach, with no research yet confirming the effects of multiple administrations. The present trial aims to investigate the safety and efficacy of multi-intravenous infusion of umbilical cord-mesenchymal stem cells (UC-MSCs) in patients with heart failure and reduced ejection fraction (HFrEF). Methods The PRIME-HFrEF trial is a single-center, prospective, randomized, triple-blinded, placebo-controlled trial of multi-intravenous infusion of UC-MSCs in HFrEF patients. A total of 40 patients meeting the inclusion criteria for HFrEF were enrolled and randomized 1:1 to the MSC group or the placebo group. Patients enrolled will receive intravenous injections of either UC-MSCs or placebo every 6 weeks for three times. Both groups will be followed up for 12 months. The primary safety endpoint is the incidence of serious adverse events. The primary efficacy endpoint is a change in left ventricular ejection fraction (LVEF) measured by left ventricular opacification (LVO) with contrast echocardiography and magnetic resonance imaging (MRI) at 12 months. The secondary endpoints include a composite of the incidence of death and re-hospitalization caused by heart failure at the 12th month, serum NT-proBNP, growth stimulation expressed gene 2 (ST2), and a change of right ventricular structure and function. Conclusions The PRIME-HFrEF study is designed to shed new light on multiple UC-MSC administration regimens for heart failure treatment.
Collapse
Affiliation(s)
- Xin Gong
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuheng Jiao
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hao Hu
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Rongzhen Zhang
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wenwen Jia
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Yuanfeng Xin
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wei Han
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| |
Collapse
|
42
|
Chang HH, Liou YS, Sun DS. Unraveling the interplay between inflammation and stem cell mobilization or homing: Implications for tissue repair and therapeutics. Tzu Chi Med J 2024; 36:349-359. [PMID: 39421490 PMCID: PMC11483098 DOI: 10.4103/tcmj.tcmj_100_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 04/29/2024] [Accepted: 06/14/2024] [Indexed: 10/19/2024] Open
Abstract
Inflammation and stem cell mobilization or homing play pivotal roles in tissue repair and regeneration. This review explores their intricate interplay, elucidating their collaborative role in maintaining tissue homeostasis and responding to injury or disease. While examining the fundamentals of stem cells, we detail the mechanisms underlying inflammation, including immune cell recruitment and inflammatory mediator release, highlighting their self-renewal and differentiation capabilities. Central to our exploration is the modulation of hematopoietic stem cell behavior by inflammatory cues, driving their mobilization from the bone marrow niche into circulation. Key cytokines, chemokines, growth factors, and autophagy, an intracellular catabolic mechanism involved in this process, are discussed alongside their clinical relevance. Furthermore, mesenchymal stem cell homing in response to inflammation contributes to tissue repair processes. In addition, we discuss stem cell resilience in the face of inflammatory challenges. Moreover, we examine the reciprocal influence of stem cells on the inflammatory milieu, shaping immune responses and tissue repair. We underscore the potential of targeting inflammation-induced stem cell mobilization for regenerative therapies through extensive literature analysis and clinical insights. By unraveling the complex interplay between inflammation and stem cells, this review advances our understanding of tissue repair mechanisms and offers promising avenues for clinical translation in regenerative medicine.
Collapse
Affiliation(s)
- Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Yu-Shan Liou
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
43
|
Yin L, Ye M, Qiao Y, Huang W, Xu X, Xu S, Oh S. Unlocking the full potential of mesenchymal stromal cell therapy for osteoarthritis through machine learning-based in silico trials. Cytotherapy 2024; 26:1252-1263. [PMID: 38904585 DOI: 10.1016/j.jcyt.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024]
Abstract
Despite the potential of mesenchymal stromal cells (MSCs) in osteoarthritis (OA) treatment, the challenge lies in addressing their therapeutic inconsistency. Clinical trials revealed significantly varied therapeutic outcomes among patients receiving the same allogenic MSCs but different treatment regimens. Therefore, optimizing personalized treatment strategies is crucial to fully unlock MSCs' potential and enhance therapeutic consistency. We employed the XGBoost algorithm to train a self-collected database comprising 37 published clinical reports to create a model capable of predicting the probability of effective pain relief and Western Ontario and McMaster Universities (WOMAC) index improvement in OA patients undergoing MSC therapy. Leveraging this model, extensive in silico simulations were conducted to identify optimal personalized treatment strategies and ideal patient profiles. Our in silico trials predicted that the individually optimized MSC treatment strategies would substantially increase patients' chances of recovery compared to the strategies used in reported clinical trials, thereby potentially benefiting 78.1%, 47.8%, 94.4% and 36.4% of the patients with ineffective short-term pain relief, short-term WOMAC index improvement, long-term pain relief and long-term WOMAC index improvement, respectively. We further recommended guidelines on MSC number, concentration, and the patients' appropriate physical (body mass index, age, etc.) and disease states (Kellgren-Lawrence grade, etc.) for OA treatment. Additionally, we revealed the superior efficacy of MSC in providing short-term pain relief compared to platelet-rich plasma therapy for most OA patients. This study represents the pioneering effort to enhance the efficacy and consistency of MSC therapy through machine learning applied to clinical data. The in silico trial approach holds immense potential for diverse clinical applications.
Collapse
Affiliation(s)
- Lu Yin
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang, Jiangxi, China; Jiangxi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, China; Agency for Science Technology and Research, Bioprocessing Technology Institute, Singapore, Singapore.
| | - Meiwu Ye
- Bio-totem Pte. Ltd., Guangzhou (Nanhai) Biomedical Industrial Park, Foshan, Guangdong, China
| | - Yang Qiao
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang, Jiangxi, China; Jiangxi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, China
| | - Weilu Huang
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang, Jiangxi, China; Jiangxi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, China
| | - Xinping Xu
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang, Jiangxi, China; Jiangxi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, China
| | - Shuoyu Xu
- Bio-totem Pte. Ltd., Guangzhou (Nanhai) Biomedical Industrial Park, Foshan, Guangdong, China.
| | - Steve Oh
- Agency for Science Technology and Research, Bioprocessing Technology Institute, Singapore, Singapore; CellVec Pte. Ltd., Singapore, Singapore.
| |
Collapse
|
44
|
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
|
45
|
Court AC, Parra-Crisóstomo E, Castro-Córdova P, Abdo L, Aragão EAA, Lorca R, Figueroa FE, Bonamino MH, Khoury M. Survival advantage of native and engineered T cells is acquired by mitochondrial transfer from mesenchymal stem cells. J Transl Med 2024; 22:868. [PMID: 39334383 PMCID: PMC11429672 DOI: 10.1186/s12967-024-05627-4] [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: 02/12/2024] [Accepted: 08/14/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Apoptosis, a form of programmed cell death, is critical for the development and homeostasis of the immune system. Chimeric antigen receptor T (CAR-T) cell therapy, approved for hematologic cancers, retains several limitations and challenges associated with ex vivo manipulation, including CAR T-cell susceptibility to apoptosis. Therefore, strategies to improve T-cell survival and persistence are required. Mesenchymal stem/stromal cells (MSCs) exhibit immunoregulatory and tissue-restoring potential. We have previously shown that the transfer of umbilical cord MSC (UC-MSC)-derived mitochondrial (MitoT) prompts the genetic reprogramming of CD3+ T cells towards a Treg cell lineage. The potency of T cells plays an important role in effective immunotherapy, underscoring the need for improving their metabolic fitness. In the present work, we evaluate the effect of MitoT on apoptotis of native T lymphocytes and engineered CAR-T cells. METHODS We used a cell-free approach using artificial MitoT (Mitoception) of UC-MSC derived MT to peripheral blood mononuclear cells (PBMCs) followed by RNA-seq analysis of CD3+ MitoTpos and MitoTneg sorted cells. Target cell apoptosis was induced with Staurosporine (STS), and cell viability was evaluated with Annexin V/7AAD and TUNEL assays. Changes in apoptotic regulators were assessed by flow cytometry, western blot, and qRT-PCR. The effect of MitoT on 19BBz CAR T-cell apoptosis in response to electroporation with a non-viral transposon-based vector was assessed with Annexin V/7AAD. RESULTS Gene expression related to apoptosis, cell death and/or responses to different stimuli was modified in CD3+ T cells after Mitoception. CD3+MitoTpos cells were resistant to STS-induced apoptosis compared to MitoTneg cells, showing a decreased percentage in apoptotic T cells as well as in TUNEL+ cells. Additionally, MitoT prevented the STS-induced collapse of the mitochondrial membrane potential (MMP) levels, decreased caspase-3 cleavage, increased BCL2 transcript levels and BCL-2-related BARD1 expression in FACS-sorted CD3+ T cells. Furthermore, UC-MSC-derived MitoT reduced both early and late apoptosis in CAR-T cells following electroporation, and exhibited an increasing trend in cytotoxic activity levels. CONCLUSIONS Artificial MitoT prevents STS-induced apoptosis of human CD3+ T cells by interfering with the caspase pathway. Furthermore, we observed that MitoT confers protection to apoptosis induced by electroporation in MitoTpos CAR T-engineered cells, potentially improving their metabolic fitness and resistance to environmental stress. These results widen the physiological perspective of organelle-based therapies in immune conditions while offering potential avenues to enhance CAR-T treatment outcomes where their viability is compromised.
Collapse
Affiliation(s)
- Angela C Court
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
| | - Eliseo Parra-Crisóstomo
- Cell for Cells, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
| | - Pablo Castro-Córdova
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
| | - Luiza Abdo
- Cell and Gene Therapy Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | | | - Rocío Lorca
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
| | - Fernando E Figueroa
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile
- Consorcio Regenero and R-MATIS, Chilean Consortium for Regenerative Medicine, and Manufacture of Advanced Therapies for Innovative Science, Santiago, Chile
| | - Martín Hernán Bonamino
- Cell and Gene Therapy Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Vice-Presidency of Research and Biological Collections (VPPCB), Oswaldo Cruz Foundation, (FIOCRUZ), Rio de Janeiro, Brazil
| | - Maroun Khoury
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cell for Cells, Santiago, Chile.
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Av. La Plaza 2501, Las Condes, Santiago, Chile.
- Consorcio Regenero and R-MATIS, Chilean Consortium for Regenerative Medicine, and Manufacture of Advanced Therapies for Innovative Science, Santiago, Chile.
| |
Collapse
|
46
|
Shi X, Chen H, Yang H, Xue S, Li Y, Fang X, Ding C, Zhu Z. Aptamer-Modified Tetrahedral Framework Nucleic Acid Synergized with TGF-β3 to Promote Cartilage Protection in Osteoarthritis by Enhancing Chondrogenic Differentiation of MSCs. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50484-50496. [PMID: 39282962 DOI: 10.1021/acsami.4c12159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Characterized by progressive and irreversible degeneration of the articular cartilage (AC), osteoarthritis (OA) is the most common chronic joint disease, and there is no cure for OA at present. Recent studies suggest that enhancing the recruitment of endogenous mesenchymal stem cells (MSCs) to damaged cartilage is a promising therapeutic strategy for cartilage repair. Tetrahedral framework nucleic acid (tFNA) is a novel DNA nanomaterial and has shown great potential in the field of biomedical science. Transforming growth factor-beta 3 (TGF-β3), a vital member of the highly conserved TGF-β superfamily, is considered to induce chondrogenesis. A 66-base DNA aptamer named HM69 is reported to identify and recruit MSCs. In this study, aptamer HM69-modified tFNAs were successfully self-assembled and used to load TGF-β3 when the disulfide bonds combined. We confirmed the successful synthesis of the final composition, HM69-tFNA@TGF-β3 (HTT), by PAGE, dynamic light scattering, and atomic force microscopy. The results of in vitro experiments showed that HTT effectively induced MSC proliferation, migration, and chondrogenic differentiation. In addition, HTT-treated MSCs were shown to protect the OA chondrocytes. In DMM mice, the injection of HTT improved the therapeutic outcome of mouse pain symptoms and AC degeneration. In conclusion, this study innovatively used the disulfide bonds combined with TGF-β3 and tFNA, and an additional sequence HM69 was loaded on tFNA for the better-targeted recruitment of MSCs. HTT demonstrated its role in promoting the chondrogenesis of MSCs and cartilage protection, indicating that it might be promising for OA therapy.
Collapse
Affiliation(s)
- Xiaorui Shi
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haowei Chen
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Hao Yang
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Song Xue
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yang Li
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaofeng Fang
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Changhai Ding
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Department of Rheumatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart 7000, Australia
| | - Zhaohua Zhu
- Clinical Research Centre, Orthopedic Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling Institute, University of Sydney, Sydney 2065, Australia
| |
Collapse
|
47
|
D'Souza RS, Her YF, Hussain N, Karri J, Schatman ME, Calodney AK, Lam C, Buchheit T, Boettcher BJ, Chang Chien GC, Pritzlaff SG, Centeno C, Shapiro SA, Klasova J, Grider JS, Hubbard R, Ege E, Johnson S, Epstein MH, Kubrova E, Ramadan ME, Moreira AM, Vardhan S, Eshraghi Y, Javed S, Abdullah NM, Christo PJ, Diwan S, Hassett LC, Sayed D, Deer TR. Evidence-Based Clinical Practice Guidelines on Regenerative Medicine Treatment for Chronic Pain: A Consensus Report from a Multispecialty Working Group. J Pain Res 2024; 17:2951-3001. [PMID: 39282657 PMCID: PMC11402349 DOI: 10.2147/jpr.s480559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Purpose Injectable biologics have not only been described and developed to treat dermal wounds, cardiovascular disease, and cancer, but have also been reported to treat chronic pain conditions. Despite emerging evidence supporting regenerative medicine therapy for pain, many aspects remain controversial. Methods The American Society of Pain and Neuroscience (ASPN) identified the educational need for an evidence-based guideline on regenerative medicine therapy for chronic pain. The executive board nominated experts spanning multiple specialties including anesthesiology, physical medicine and rehabilitation, and sports medicine based on expertise, publications, research, and clinical practice. A steering committee selected preliminary questions, which were reviewed and refined. Evidence was appraised using the United States Preventive Services Task Force (USPSTF) criteria for evidence level and degree of recommendation. Using a modified Delphi approach, consensus points were distributed to all collaborators and each collaborator voted on each point. If collaborators provided a decision of "disagree" or "abstain", they were invited to provide a rationale in a non-blinded fashion to the committee chair, who incorporated the respective comments and distributed revised versions to the committee until consensus was achieved. Results Sixteen questions were selected for guideline development. Questions that were addressed included type of injectable biologics and mechanism, evidence in treating chronic pain indications (eg, tendinopathy, muscular pathology, osteoarthritis, intervertebral disc disease, neuropathic pain), role in surgical augmentation, dosing, comparative efficacy between injectable biologics, peri-procedural practices to optimize therapeutic response and quality of injectate, federal regulations, and complications with mitigating strategies. Conclusion In well-selected individuals with certain chronic pain indications, use of injectable biologics may provide superior analgesia, functionality, and/or quality of life compared to conventional medical management or placebo. Future high-quality randomized clinical trials are warranted with implementation of minimum reporting standards, standardization of preparation protocols, investigation of dose-response associations, and comparative analysis between different injectable biologics.
Collapse
Affiliation(s)
- Ryan S D'Souza
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yeng F Her
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nasir Hussain
- Department of Anesthesiology, The Ohio State Wexner Medical Center, Columbus, OH, USA
| | - Jay Karri
- Departments of Orthopedic Surgery and Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael E Schatman
- Department of Anesthesiology, Perioperative Care, & Pain Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Christopher Lam
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Thomas Buchheit
- Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Brennan J Boettcher
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | | | - Scott G Pritzlaff
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Sacramento, CA, USA
| | | | - Shane A Shapiro
- Department of Orthopedic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Johana Klasova
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jay S Grider
- Department of Anesthesiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Ryan Hubbard
- Department of Sports Medicine, Anderson Orthopedic Clinic, Arlington, VA, USA
| | - Eliana Ege
- Department of Physical Medicine & Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Shelby Johnson
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Max H Epstein
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Eva Kubrova
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Mohamed Ehab Ramadan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Alexandra Michelle Moreira
- Department of Physical Medicine & Rehabilitation, University of Miami/Jackson Memorial Hospital, Miami, FL, USA
| | - Swarnima Vardhan
- Department of Internal Medicine, Yale New Haven Health - Bridgeport Hospital, Bridgeport, CT, USA
| | - Yashar Eshraghi
- Department of Anesthesiology & Critical Care Medicine, Ochsner Health System, New Orleans, LA, USA
| | - Saba Javed
- Department of Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Newaj M Abdullah
- Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Paul J Christo
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Sudhir Diwan
- Department of Pain Medicine, Advanced Spine on Park Avenue, New York City, NY, USA
| | | | - Dawood Sayed
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Timothy R Deer
- Department of Anesthesiology and Pain Medicine, West Virginia University School of Medicine, Charleston, WV, USA
| |
Collapse
|
48
|
Tabet CG, Pacheco RL, Martimbianco ALC, Riera R, Hernandez AJ, Bueno DF, Fernandes TL. Advanced therapy with mesenchymal stromal cells for knee osteoarthritis: Systematic review and meta-analysis of randomized controlled trials. J Orthop Translat 2024; 48:176-189. [PMID: 39360004 PMCID: PMC11445595 DOI: 10.1016/j.jot.2024.07.012] [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: 03/06/2024] [Revised: 07/10/2024] [Accepted: 07/29/2024] [Indexed: 10/04/2024] Open
Abstract
Background Advanced cell therapies emerged as promising candidates for treatment of knee articular diseases, but robust evidence regarding their clinical applicability is still lacking. Objective To assess the efficacy and safety of advanced mesenchymal stromal cells (MSC) therapy for knee osteoarthritis (OA) and chondral lesions. Methods Systematic review of randomized controlled trials conducted in accordance with Cochrane Handbook and reported following PRISMA checklist. GRADE approach was used for assessing the evidence certainty. Results 25 randomized controlled trials that enrolled 1048 participants were included. Meta-analyses data showed that, compared to viscosupplementation (VS), advanced MSC therapy resulted in a 1.91 lower pain VAS score (95 % CI -3.23 to -0.59; p < 0.00001) for the treatment of knee OA after 12 months. Compared to placebo, the difference was 0.99 lower pain VAS points (95 % CI -1.94 to -0.03; p = 0.76). According to the GRADE approach, the evidence was very uncertain for both comparisons. By excluding studies with high risk of bias, there was a similar size of effect (VAS MD -1.54, 95 % CI -2.09 to -0.98; p = 0.70) with improved (moderate) certainty of evidence, suggesting that MSC therapy probably reduces pain slightly better than VS. Regarding serious adverse events, there was no difference from advanced MSC therapy to placebo or to VS, with very uncertain evidence. Conclusion Advanced MSC therapy resulted in lower pain compared to placebo or VS for the treatment of knee OA after 12 months, with no difference in adverse events. However, the evidence was considered uncertain. The Translational Potential of this Article Currently, there is a lack of studies with good methodological structure aiming to evaluate the real clinical impact of advanced cell therapy for knee OA. The present study was well structured and conducted, with Risk of Bias, GRADE certainty assessment and sensitivity analysis. It explores the translational aspect of the benefits and safety of MSC compared with placebo and gold-standard therapy to give practitioners and researchers support to expand this therapy in their practice. PROSPERO registration number CRD42020158173. Access at https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=158173.
Collapse
Affiliation(s)
- Caio Gomes Tabet
- Sports Medicine Division, Instituto de Ortopedia e Traumatologia da Faculdade de Medicina do Hospital das Clínicas da Universidade de São Paulo (USP), São Paulo, Brazil
- Hospital Sírio-Libanês, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Rafael Leite Pacheco
- Centre of Health Technology Assessment, Hospital Sírio-Libanês, São Paulo, Brazil
- Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, Brazil
| | - Ana Luiza Cabrera Martimbianco
- Centre of Health Technology Assessment, Hospital Sírio-Libanês, São Paulo, Brazil
- Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, Brazil
- Postgraduate Program of Health and Environment, Universidade Metropolitana de Santos, Santos, Brazil
| | - Rachel Riera
- Centre of Health Technology Assessment, Hospital Sírio-Libanês, São Paulo, Brazil
- Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, Brazil
| | - Arnaldo José Hernandez
- Sports Medicine Division, Instituto de Ortopedia e Traumatologia da Faculdade de Medicina do Hospital das Clínicas da Universidade de São Paulo (USP), São Paulo, Brazil
- Hospital Sírio-Libanês, São Paulo, Brazil
| | | | - Tiago Lazzaretti Fernandes
- Sports Medicine Division, Instituto de Ortopedia e Traumatologia da Faculdade de Medicina do Hospital das Clínicas da Universidade de São Paulo (USP), São Paulo, Brazil
- Hospital Sírio-Libanês, São Paulo, Brazil
| |
Collapse
|
49
|
Huang K, Liu X, Qin H, Li Y, Zhu J, Yin B, Zheng Q, Zuo C, Cao H, Tong Z, Sun Z. FGF18 encoding circular mRNA-LNP based on glycerolipid engineering of mesenchymal stem cells for efficient amelioration of osteoarthritis. Biomater Sci 2024; 12:4427-4439. [PMID: 39037353 DOI: 10.1039/d4bm00668b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Mesenchymal stem cells (MSCs) exhibit substantial potential for osteoarthritis (OA) therapy through cartilage regeneration, yet the realization of optimal therapeutic outcomes is hampered by their limited intrinsic reparative capacities. Herein, MSCs are engineered with circular mRNA (cmRNA) encoding fibroblast growth factor 18 (FGF18) encapsulated within lipid nanoparticles (LNP) derived from a glycerolipid to facilitate OA healing. A proprietary biodegradable and ionizable glycerolipid, TG6A, with branched tails and five ester bonds, forms LNP exhibiting above 9-fold and 41-fold higher EGFP protein expression in MSCs than commercial LNP from DLin-MC3-DMA and ALC-0315, respectively. The introduction of FGF18 not only augmented the proliferative capacity of MSCs but also upregulated the expression of chondrogenic genes and glycosaminoglycan (GAG) content. Additionally, FGF18 enhanced the production of proteoglycans and type II collagen in chondrocyte pellet cultures in a three-dimensional culture. In an OA rat model, transplantation with FGF18-engineered MSCs remarkably preserved cartilage integrity and facilitated functional repair of cartilage lesions, as evidenced by thicker cartilage layers, reduced histopathological scores, maintenance of zone structure, and incremental type II collagen and extracellular matrix (ECM) deposition. Taken together, our findings suggest that TG6A-based LNP loading with cmRNA for engineering MSCs present an innovative strategy to overcome the current limitations in OA treatment.
Collapse
Affiliation(s)
- Ke Huang
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, 215000, China
| | - Xiaoyun Liu
- Jiangsu Purecell Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Haitang Qin
- Jiangsu Purecell Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Yingwen Li
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Jiafeng Zhu
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Bo Yin
- National University of Singapore (Suzhou) Research Institute, Suzhou, 215123, China.
| | - Qijun Zheng
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, 215000, China
| | - Chijian Zuo
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Hui Cao
- Jiangsu Purecell Biopharma Technology Co., Ltd, Suzhou 215125, China.
| | - Zhenbo Tong
- Southeast University-Monash University Joint Research Institute, Suzhou 215125, China
| | - Zhenhua Sun
- Suzhou CureMed Biopharma Technology Co., Ltd, Suzhou 215125, China.
| |
Collapse
|
50
|
Soufan S, Al Khoury J, Hamdan Z, Rida MA. Intra-articular interventions in osteoarthritis: Navigating the landscape of hyaluronic acid, mesenchymal stem cells, and platelet-rich plasma. World J Orthop 2024; 15:704-712. [PMID: 39165875 PMCID: PMC11331319 DOI: 10.5312/wjo.v15.i8.704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/13/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024] Open
Abstract
Osteoarthritis (OA) poses a substantial burden on patients, leading to pain, functional decline, and reduced quality of life. While conventional treatments focus on symptom management, disease-modifying interventions are yet to be established. This review explores the efficacy of intra-articular interventions, particularly hyaluronic acid (HA), mesenchymal stem cells (MSCs), and platelet-rich plasma (PRP), in the context of OA management. HA injections, with diverse formulations like Hylan G-F20, sodium hyaluronate, and hyaluronan, present varying outcomes, necessitating a nuanced understanding of their effectiveness and timing. MSC therapy, derived from adipose tissue, umbilical cord, or bone marrow, shows promising results in clinical improvement, with adipose-derived MSCs demonstrating efficacy in maintaining benefits over 6 mo. Conversely, bone-marrow-derived MSCs show limited effectiveness, highlighting the need for source-specific considerations. PRP has emerged as a superior option for long-term pain reduction and quality of life improvement, with leukocyte-poor formulations and a critical platelet count of 10 billion demonstrating optimal results. This comprehensive analysis underscores the potential of intra-articular interventions in OA management, emphasizing the need for personalized and evidence-based approaches to enhance treatment efficacy and patient outcomes.
Collapse
Affiliation(s)
- Somar Soufan
- Department of Medicine, University of Balamand, Beirut 00000, Lebanon
| | - Joenne Al Khoury
- Department of Medicine, University of Balamand, Beirut 00000, Lebanon
| | - Zahi Hamdan
- Department of Medicine, University of Balamand, Beirut 00000, Lebanon
| | - Mohamad Ali Rida
- Department of Internal Medicine, Clemenceau Medical Center, Beirut 12345, Lebanon
| |
Collapse
|