1
|
Trapana J, Weinerman J, Lee D, Sedani A, Constantinescu D, Best TM, Hornicek FJ, Hare JM. Cell-based therapy in the treatment of musculoskeletal diseases. Stem Cells Transl Med 2024; 13:959-978. [PMID: 39226104 PMCID: PMC11465182 DOI: 10.1093/stcltm/szae049] [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: 03/13/2023] [Accepted: 05/21/2024] [Indexed: 09/04/2024] Open
Abstract
A limited number of tissues can spontaneously regenerate following injury, and even fewer can regenerate to a state comparable to mature, healthy adult tissue. Mesenchymal stem cells (MSCs) were first described in the 1960s-1970s by Friedenstein et al as a small population of bone marrow cells with osteogenic potential and abilities to differentiate into chondrocytes. In 1991, Arnold Caplan coined the term "mesenchymal cells" after identifying these cells as a theoretical precursor to bone, cartilage, tendon, ligament, marrow stroma, adipocyte, dermis, muscle, and connective tissues. MSCs are derived from periosteum, fat, and muscle. Another attractive property of MSCs is their immunoregulatory and regenerative properties, which result from crosstalk with their microenvironment and components of the innate immune system. Collectively, these properties make MSCs potentially attractive for various therapeutic purposes. MSCs offer potential in sports medicine, aiding in muscle recovery, meniscal tears, and tendon and ligament injuries. In joint disease, MSCs have the potential for chondrogenesis and reversing the effects of osteoarthritis. MSCs have also demonstrated potential application to the treatment of degenerative disc disease of the cervical, thoracic, and lumbar spine.
Collapse
Affiliation(s)
- Justin Trapana
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Jonathan Weinerman
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Danny Lee
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Anil Sedani
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - David Constantinescu
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
| | - Thomas M Best
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Francis J Hornicek
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, United States
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, United States
| |
Collapse
|
2
|
Johnbosco C, Karbaat L, Korthagen NM, Warmink K, Koerselman M, Coeleveld K, Becker M, van Loo B, Zoetebier B, Both S, Weinans H, Karperien M, Leijten J. Microencapsulated stem cells reduce cartilage damage in a material dependent manner following minimally invasive intra-articular injection in an OA rat model. Mater Today Bio 2023; 22:100791. [PMID: 37731960 PMCID: PMC10507156 DOI: 10.1016/j.mtbio.2023.100791] [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: 06/13/2023] [Revised: 08/05/2023] [Accepted: 09/04/2023] [Indexed: 09/22/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative disease of the joints for which no curative treatment exists. Intra-articular injection of stem cells is explored as a regenerative approach, but rapid clearance of cells from the injection site limits the therapeutic outcome. Microencapsulation of mesenchymal stem cells (MSCs) can extend the retention time of MSCs, but the outcomes of the few studies currently performed are conflicting. We hypothesize that the composition of the micromaterial's shell plays a deciding factor in the treatment outcome of intra-articular MSC injection. To this end, we microencapsulate MSCs using droplet microfluidic generators in flow-focus mode using various polymers and polymer concentrations. We demonstrate that polymer composition and concentration potently alter the metabolic activity as well as the secretome of MSCs. Moreover, while microencapsulation consistently prolongs the retention time of MSC injected in rat joints, distinct biodistribution within the joint is demonstrated for the various microgel formulations. Furthermore, intra-articular injections of pristine and microencapsulated MSC in OA rat joints show a strong material-dependent effect on the reduction of cartilage degradation and matrix loss. Collectively, this study highlights that micromaterial composition and concentration are key deciding factors for the therapeutic outcome of intra-articular injections of microencapsulated stem cells to treat degenerative joint diseases.
Collapse
Affiliation(s)
- Castro Johnbosco
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Lisanne Karbaat
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Nicoline M. Korthagen
- Faculty of Veterinary Sciences Department of equine sciences, University of Utrecht, the Netherlands
- Department of Orthopaedics, University Medical Centre Utrecht, the Netherlands
| | - Kelly Warmink
- Department of Orthopaedics, University Medical Centre Utrecht, the Netherlands
| | - Michelle Koerselman
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Katja Coeleveld
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, the Netherlands
| | - Malin Becker
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Bas van Loo
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Bram Zoetebier
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Sanne Both
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Harrie Weinans
- Department of Orthopaedics, University Medical Centre Utrecht, the Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Jeroen Leijten
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| |
Collapse
|
3
|
Xiao K, Liu C, Wang H, Hou F, Shi Y, Qian ZR, Zhang H, Deng DYB, Xie L. Umbilical cord mesenchymal stem cells overexpressing CXCR7 facilitate treatment of ARDS-associated pulmonary fibrosis via inhibition of Notch/Jag1 mediated by the Wnt/β-catenin pathway. Biomed Pharmacother 2023; 165:115124. [PMID: 37454589 DOI: 10.1016/j.biopha.2023.115124] [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: 05/12/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023] Open
Abstract
The therapeutic efficacy of umbilical cord mesenchymal stem cells (UCMSCs) in acute respiratory distress syndrome (ARDS) is mainly limited by the efficiency of homing of UCMSCs toward tissue damage. C-X-C chemokine receptor type 7 (CXCR7), which is involved in the mobilization of UCMSCs, is only expressed on the surface of a small proportion of UCMSCs. This study examined whether overexpression of CXCR7 in UCMSCs (UCMSCsOE-CXCR7) could improve their homing efficiency, and therefore, improve their effectiveness in fibrosis repair at the site of lung injury caused by ARDS. A lentiviral vector expressing CXCR7 was built and then transfect into UCMSCs. The impacts of CXCR7 expression of the proliferationand homing of UCMSCs were examined in a lipopolysaccharide-induced ARDS mouse model. The potential role and underlying mechanism of CXCR7 were examined by performing scratch assays, transwell assays, and immunoassays. The therapeutic dose and treatment time of UCMSCsOE-CXCR7 were directly proportional to their therapeutic effect on lung injury. In addition, overexpression of CXCR7 increased SDF-1-induced proliferation and migration of lung epithelial cells (Base-2b cells), and upregulation of CXCR7 inhibited α-SMA expression, suggesting that CXCR7 may have a role in alleviating pulmonary fibrosis caused by ARDS. Overexpression of CXCR7 in UCMSCs may improve their therapeutic effect of acute lung injury mouse, The mechanism of fibrosis repair by CXCR7 is inhibition of Jag1 via suppression of the Wnt/β-catenin pathway under the chemotaxis of SDF-1.
Collapse
Affiliation(s)
- Kun Xiao
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Chang Liu
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100853, China; School of medicine Nankai university, Tianjin 300071, China
| | - Heming Wang
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresource, Hainan University, Haikou 570228, China
| | - Fei Hou
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Yinghan Shi
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhi Rong Qian
- Department of Scientific Research Center, The Seventh Affiliated Hospital of Sun YatSen University, Shenzhen 518106, China; Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Hao Zhang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - David Y B Deng
- Department of Scientific Research Center, The Seventh Affiliated Hospital of Sun YatSen University, Shenzhen 518106, China.
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
4
|
Liu M, Cheng L, Li X, Wang H, Wang M, Gan L. Resveratrol Reverses Myogenic Induction Suppression Caused by High Glucose Through Activating the SIRT1/AKT/FOXO1 Pathway. Nat Prod Commun 2023. [DOI: 10.1177/1934578x231159722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
Background Differentiated bone marrow mesenchymal stem cells (BMSCs) may be a therapeutic strategy to treat sarcopenia caused by high glucose. The effects of resveratrol in the myogenic induction of BMSCs under high glucose are unknown. We evaluated the effects and possible mechanisms of high glucose and resveratrol on myogenic induction of rat BMSCs. Methods Primary rat BMSCs were isolated and purified from Sprague-Dawley rats aged between 3 and 4 weeks. Rat BMSCs were differentiated into myogenic cells using conditioned medium and treated with glucose and/or resveratrol along with EX527 (a specific silent information regulator 1 [SIRT1] inhibitor). The expressions of MyoD1 and Myogenin were measured. The reactive oxygen species (ROS) level, superoxide dismutase (SOD) activity, and the expressions of FOXO1 and p-AKT/AKT during myogenic induction were also examined. Results High glucose decreased cell viability, cell proliferation, and SOD activity, increased intracellular ROS levels, and inhibited the AKT/FOXO1. Resveratrol reversed myogenic induction suppression caused by high glucose, partly through restoring cell proliferation and viability, reducing peroxidative damage, and activating the AKT/FOXO1 pathway; this effect was eliminated by EX527. Conclusion Our results indicate that resveratrol promoted myogenic induction and partially reversed the suppression of myogenic induction caused by high glucose through activating the SIRT1/AKT/FOXO1 pathway.
Collapse
Affiliation(s)
- Meiling Liu
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Luyang Cheng
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xianglu Li
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongzhi Wang
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Manfeng Wang
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lu Gan
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
5
|
A Novel Approach for Fluoroscopic Guided Intra-articular Hip Injections: Technique Description and Case Series. Am J Phys Med Rehabil 2023; 102:e15-e17. [PMID: 36166654 DOI: 10.1097/phm.0000000000002112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
ABSTRACT Fluoroscopic guided intra-articular hip injections generally utilize a standard anterior-posterior view. However, this approach can expose patients to inadvertent femoral nerve or vessel infiltration owing to the proximity of the neurovascular bundle to the joint space. This case-series study describes a novel technique using fluoroscopic ipsilateral oblique angulation and caudal tilt of the image intensifier. With this view, the clinician can advance the needle in a lateral to medial trajectory to obtain intra-articular access and minimize the risk of complications. This method was performed in five patients with refractory chronic hip osteoarthritis, which resulted in notable pain improvements and no reported adverse events. The suggested technique could provide a safer alternative to the anterior-posterior imaging technique for intra-articular hip injections by avoiding the femoral neurovascular bundle, limiting needle repositioning, and offering a satisfactory postprocedural analgesic effect.
Collapse
|
6
|
Everts PA, Panero AJ. Basic Science of Autologous Orthobiologics. Phys Med Rehabil Clin N Am 2023; 34:25-47. [DOI: 10.1016/j.pmr.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
7
|
De Becker A, Heestermans R, De Brouwer W, Bockstaele K, Maes K, Van Riet I. Genetic profiling of human bone marrow mesenchymal stromal cells after in vitro expansion in clinical grade human platelet lysate. Front Bioeng Biotechnol 2022; 10:1008271. [DOI: 10.3389/fbioe.2022.1008271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are non-hematopoietic cells that have a broad therapeutic potential. To obtain sufficient cells for clinical application, they must be expanded ex vivo. In the initial expansion protocols described, fetal calf serum (FCS) was used as the reference growth supplement, but more recently different groups started to replace FCS with platelet lysate (PL). We investigated in this study the impact of the culture supplement on gene expression of MSCs. Human bone marrow derived MSCs were expanded in vitro in FCS and PL supplemented medium. We found that MSCs expanded in PL-containing medium (PL-MSCs) express typical MSC immunomorphological features and can migrate, as their counterparts expanded in FCS-containing medium, through a layer of endothelial cells in vitro. Additionally, they show an increased proliferation rate compared to MSCs expanded in FCS medium (FCS-MSCs). RNA sequencing performed for MSCs cultured in both types of expansion medium revealed a large impact of the choice of growth supplement on gene expression: 1974 genes were at least twofold up- or downregulated. We focused on impact of genes involved in apoptosis and senescence. Our data showed that PL-MSCs express more anti-apoptotic genes and FCS-MSCs more pro-apoptotic genes. FCS-MSCs showed upregulation of senescence-related genes after four passages whereas this was rarer in PL-MSCs at the same timepoint. Since PL-MSCs show higher proliferation rates and anti-apoptotic gene expression, they might acquire features that predispose them to malignant transformation. We screened 10 MSC samples expanded in PL-based medium for the presence of tumor-associated genetic variants using a 165 gene panel and detected only 21 different genetic variants. According to our analysis, none of these were established pathogenic mutations. Our data show that differences in culture conditions such as growth supplement have a significant impact on the gene expression profile of MSCs and favor the use of PL over FCS for expansion of MSCs.
Collapse
|
8
|
Peláez P, Damiá E, Torres-Torrillas M, Chicharro D, Cuervo B, Miguel L, del Romero A, Carrillo JM, Sopena JJ, Rubio M. Cell and Cell Free Therapies in Osteoarthritis. Biomedicines 2021; 9:1726. [PMID: 34829953 PMCID: PMC8615373 DOI: 10.3390/biomedicines9111726] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is the most common articular disease in adults and has a current prevalence of 12% in the population over 65 years old. This chronic disease causes damage to articular cartilage and synovial joints, causing pain and leading to a negative impact on patients' function, decreasing quality of life. There are many limitations regarding OA conventional therapies-pharmacological therapy can cause gastrointestinal, renal, and cardiac adverse effects, and some of them could even be a threat to life. On the other hand, surgical options, such as microfracture, have been used for the last 20 years, but hyaline cartilage has a limited regeneration capacity. In recent years, the interest in new therapies, such as cell-based and cell-free therapies, has been considerably increasing. The purpose of this review is to describe and compare bioregenerative therapies' efficacy for OA, with particular emphasis on the use of mesenchymal stem cells (MSCs) and platelet-rich plasma (PRP). In OA, these therapies might be an alternative and less invasive treatment than surgery, and a more effective option than conventional therapies.
Collapse
Affiliation(s)
- Pau Peláez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Elena Damiá
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Belén Cuervo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Laura Miguel
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Ayla del Romero
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Jose Maria Carrillo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Joaquín J. Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Mónica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain; (P.P.); (M.T.-T.); (D.C.); (B.C.); (L.M.); (A.d.R.); (J.M.C.); (J.J.S.); (M.R.)
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| |
Collapse
|
9
|
Rodas G, Soler-Rich R, Rius-Tarruella J, Alomar X, Balius R, Orozco L, Masci L, Maffulli N. Effect of Autologous Expanded Bone Marrow Mesenchymal Stem Cells or Leukocyte-Poor Platelet-Rich Plasma in Chronic Patellar Tendinopathy (With Gap >3 mm): Preliminary Outcomes After 6 Months of a Double-Blind, Randomized, Prospective Study. Am J Sports Med 2021; 49:1492-1504. [PMID: 33783227 DOI: 10.1177/0363546521998725] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Patellar tendinopathy is common. The success of traditional management, including isometric or eccentric exercises combined with shockwave therapy and even surgery, is limited. Therefore, it is important to determine whether biological treatments such as ultrasound-guided intratendinous and peritendinous injections of autologous expanded bone marrow mesenchymal stem cells (BM-MSCs) or leukocyte-poor platelet-rich plasma (Lp-PRP) improve clinical outcomes in athletic patients with patellar tendinopathy. STUDY DESIGN Randomized controlled trial; Level of evidence, 2. METHODS A prospective, double-blinded, randomized, 2-arm parallel group, active controlled, phase 1/2 single-center clinical study was performed in patients who had proximal patellar tendinopathy with a lesion >3 mm. A total of 20 participants (age 18-48 years) with pain for >4 months (mean, 23.6 months) and unresponsive to nonoperative treatments were randomized into 2 groups. Of these, 10 participants were treated with BM-MSC (20 × 106 cells) and 10 with Lp-PRP. Both groups performed the same postintervention rehabilitation protocol. Outcomes included the Victorian Institute of Sport Assessment for pain (VISA-P), self-reported tendon pain during activity (visual analog scale [VAS]), muscle function by dynamometry, tendon thickness and intratendinous vascularity by ultrasonographic imaging and Doppler signal, ultrasound tissue characterization (UTC) echo type changes, and magnetic resonance imaging (MRI) T2-weighted mapping changes. Participants were followed longitudinally for 6 months. RESULTS The average VAS scores improved in both groups at all time points, and there was a significant reduction in pain during sporting activities (P < .05). In both groups, the average mean VISA-P scores at 6 months were significantly increased compared with baseline (66 BM-MSC group and 72.90 Lp-PRP group), with no significant differences in VAS or VISA-P scores between the groups. There were statistically significant greater improvements in tendon structure on 2-dimensional ultrasound and UTC in the BM-MSC group compared with the Lp-PRP group at 6 months. Similarly, the BM-MSC group demonstrated significant evidence of restoration of tendon structure on MRI compared with the Lp-PRP group at 6 months. Only the participants in the BM-MSC group showed evidence of normalization of tendon structure, with statistically significant differences between the groups on T2-weighted, fat-saturated sagittal and coronal scans and hypersignal in T2-weighted on spin-echo T2-weighted coronal MRI scan. Both treatments were safe, and no significant adverse events were reported in either group. CONCLUSION Treatment with BM-MSC or Lp-PRP in combination with rehabilitation in chronic patellar tendinopathy is effective in reducing pain and improving activity levels in active participants. Participants who received BM-MSC treatment demonstrated greater improvement in tendon structure compared with those who received Lp-PRP. REGISTRATION 2016-001262-28 (EudraCT identifier); NCT03454737 (ClinicalTrials.gov identifier).
Collapse
Affiliation(s)
- Gil Rodas
- Medical Department FC Barcelona, Barcelona, Spain.,Sports Medicine Unit, Clínic Hospital and Sant Joan de Déu Hospital, Barcelona, Spain
| | - Robert Soler-Rich
- Institut de Teràpia Regenerativa Tissular, Centro Médico Teknon, Barcelona, Spain
| | - Joan Rius-Tarruella
- Institut de Teràpia Regenerativa Tissular, Centro Médico Teknon, Barcelona, Spain
| | - Xavier Alomar
- Diagnóstico por la Imagen, Clínica Creu Blanca, Barcelona, Spain
| | - Ramon Balius
- Consell Català de l'Esport, Generalitat de Catalunya, Barcelona, Spain
| | - Lluís Orozco
- Institut de Teràpia Regenerativa Tissular, Centro Médico Teknon, Barcelona, Spain
| | - Lorenzo Masci
- Institute of Sports Exercise and Health (ISEH), London, UK
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, University of Salerno School of Medicine, Surgery and Dentistry, Salerno, Italy.,Centre for Sports and Exercise Medicine, Queen Mary University of London, London, UK.,School of Pharmacy and Bioengineering, Keele University School of Medicine, Staffordshire, UK
| |
Collapse
|
10
|
Abstract
Orthobiologics are exciting tools providing promising results for difficult orthopedic conditions. In the elbow there is high-level evidence for their use in lateral epicondylopathy and encouraging evidence for other elbow pathologies. This article provides an in-depth review of the current literature for the use of orthobiologics in elbow injuries.
Collapse
Affiliation(s)
- Cleo D Stafford
- Department of Orthopaedics and Rehabilitation Medicine, Emory University School of Medicine, 49 Jessie Hill Junior Drive Southeast 3rd Floor, Atlanta, GA 30303, USA. https://twitter.com/CleoStaffordMD
| | - Ricardo E Colberg
- Andrews Sports Medicine and Orthopedic Center, American Sports Medicine Institute, 805 St Vincent's Drive Suite 100, Birmingham, AL 35205, USA.
| | - Hunter Garrett
- American Sports Medicine Institute, 805 St Vincent's Drive Suite 100, Birmingham, AL 35205, USA
| |
Collapse
|
11
|
Liu M, Li X, Zhou C, Wang M, Wang H, Ding H, Cheng L, Gan L, Wu X, Du Z. Thioredoxin mitigates H 2 O 2 -induced inhibition of myogenic differentiation of rat bone marrow mesenchymal stem cells by enhancing AKT activation. FEBS Open Bio 2020; 10:835-846. [PMID: 32160414 PMCID: PMC7193161 DOI: 10.1002/2211-5463.12835] [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: 09/12/2019] [Revised: 02/06/2020] [Accepted: 03/06/2020] [Indexed: 11/20/2022] Open
Abstract
Thioredoxin (Trx) is a hydrogen acceptor of ribonucleotide reductase and a regulator of some enzymes and receptors. It has been previously shown that significantly elevated levels of Trx expression are associated with the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), but it is not clear how Trx regulates the effects of hydrogen peroxide (H2O2) on myogenic differentiation of BMSCs. Here, we report that rat BMSCs treated with a high dose (150 µm) of H2O2 exhibited a significant reduction in viability, cell cycling, and superoxide dismutase and glutathione peroxidase levels, and an increase in reactive oxygen species and malondialdehyde levels, which was accompanied by reductions in protein kinase B activation and forkhead Box O1, myogenic differentiation 1 and myogenin expression during myogenic differentiation. Furthermore, treatment with recombinant human Trx significantly mitigated the effects of H2O2 on the myogenic differentiation of BMSCs, and this was abrogated by cotreatment with wortmannin [a specific phosphatidylinositol 3‐kinase inhibitor]. In summary, our results suggest that treatment with recombinant human Trx mitigates H2O2‐induced oxidative stress and may promote myogenic differentiation of rat BMSCs by enhancing phosphatidylinositol 3‐kinase/protein kinase B/forkhead Box O1 signaling.
Collapse
Affiliation(s)
- Meiling Liu
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, China
| | - Xianglu Li
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, China
| | - Changlin Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, China
| | - Manfeng Wang
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, China
| | - Hongzhi Wang
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, China
| | - Haifeng Ding
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, China
| | - Luyang Cheng
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, China
| | - Lu Gan
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, China
| | - Xiaowei Wu
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, China
| | - Zhimin Du
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University, China.,State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Harbin, China
| |
Collapse
|
12
|
McInnis KC, Chen ET, Finnoff JT, Roh EY, Borg Stein J. Orthobiologics for the Hip Region: A Narrative Review. PM R 2020; 12:1045-1054. [PMID: 31953917 DOI: 10.1002/pmrj.12327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022]
Abstract
Management of hip region disorders is challenging. Orthobiologic treatments including platelet rich plasma (PRP), mesenchymal stem cells, and amniotic injectables have gained popularity as promising treatments despite a lack of robust evidence for their effectiveness. We review rationale and current evidence for orthobiologics for three common hip region conditions: hip osteoarthritis, gluteal tendinopathy, and proximal hamstring tendinopathy. Overall, the current state of evidence is extremely limited for orthobiologic treatments and is predominantly relevant to PRP injections. There is currently a lack of data to support the use of mesenchymal stem cells or amniotic injectables in these conditions of the hip.
Collapse
Affiliation(s)
- Kelly C McInnis
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA
| | - Eric T Chen
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA
| | - Jonathan T Finnoff
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Eugene Y Roh
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, Stanford University, Redwood City, CA
| | - Joanne Borg Stein
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA
| |
Collapse
|
13
|
Wang W, Xin J, Chen W, Jing L, Zhang P. Icariin alleviates hypoxia-induced damage in MC3T3-E1 cells by downregulating TALNEC2. Biotechnol Appl Biochem 2019; 67:1000-1010. [PMID: 31845407 DOI: 10.1002/bab.1874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/11/2019] [Indexed: 12/27/2022]
Abstract
Osteonecrosis is a harmful musculoskeletal disease. We aim to detect the effects of icariin (ICA) in MC3T3-E1 cell. MC3T3-E1 cell was pretreated with ICA and was subjected to hypoxia stimuli. The tumor-associated long noncoding RNA expressed on chromosome 2 (TALNEC2) overexpression or silencing vectors (pTALNEC2 or si-TALNEC2) was utilized for MC3T3-E1 cell transfection. Viability and apoptosis rate were individually tested by cell counting kit-8 and Annexin V-fluorescein isothiocyanate/propidium iodide kit untied with flow cytometry. The alkaline phosphatase activity (ALP) activity was tested through ALP assay. The quantitative reverse transcription PCR or Western blot was performed for elements detection at the RNA or protein level. Hypoxia treatment induced viability inhibition and CyclinD1 reduction, but elevation of p53 and p16. It also promoted apoptosis by increasing apoptotic cells, Bax, and cleaved-poly ADP-ribose polymerase but decreasing Bcl-2. Also, hypoxia stimuli restrained ALP activity, and osteopontin, osteocalcin, and Runt-related transcription factor 2 expression. Those effects caused by hypoxia stimuli were all reversed by ICA. TALNEC2 was downregulated by ICA, whose impacts were subsequently abolished by pTALNEC2. Silencing TALNEC2 displayed similar effects with ICA. But the apoptosis was not affected by si-TALNEC2. ICA blocked ste20-related proline/alanine-rich kinase/c-Jun N-terminal kinase (SPAK/JNK) but triggered phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway in MC3T3-E1 cell by suppressing TALNEC2. ICA relieved hypoxia-stimulated damage by restraining TALNEC2 through blocking SPAK/JNK and triggering PI3K/AKT/mTOR in the MC3T3-E1 cell.
Collapse
Affiliation(s)
- Weiguo Wang
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Jian Xin
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Wenming Chen
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Lizhong Jing
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Peng Zhang
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| |
Collapse
|
14
|
Torres-Torrillas M, Rubio M, Damia E, Cuervo B, Del Romero A, Peláez P, Chicharro D, Miguel L, Sopena JJ. Adipose-Derived Mesenchymal Stem Cells: A Promising Tool in the Treatment of Musculoskeletal Diseases. Int J Mol Sci 2019; 20:ijms20123105. [PMID: 31242644 PMCID: PMC6627452 DOI: 10.3390/ijms20123105] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 02/08/2023] Open
Abstract
Chronic musculoskeletal (MSK) pain is one of the most common medical complaints worldwide and musculoskeletal injuries have an enormous social and economical impact. Current pharmacological and surgical treatments aim to relief pain and restore function; however, unsatiscactory outcomes are commonly reported. In order to find an accurate treatment to such pathologies, over the last years, there has been a significantly increasing interest in cellular therapies, such as adipose-derived mesenchymal stem cells (AMSCs). These cells represent a relatively new strategy in regenerative medicine, with many potential applications, especially regarding MSK disorders, and preclinical and clinical studies have demonstrated their efficacy in muscle, tendon, bone and cartilage regeneration. Nevertheless, several worries about their safety and side effects at long-term remain unsolved. This article aims to review the current state of AMSCs therapy in the treatment of several MSK diseases and their clinical applications in veterinary and human medicine.
Collapse
Affiliation(s)
- Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Monica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Elena Damia
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Belen Cuervo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Ayla Del Romero
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Pau Peláez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Laura Miguel
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Joaquin J Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| |
Collapse
|
15
|
Everts PA, Malanga GA, Paul RV, Rothenberg JB, Stephens N, Mautner KR. Assessing clinical implications and perspectives of the pathophysiological effects of erythrocytes and plasma free hemoglobin in autologous biologics for use in musculoskeletal regenerative medicine therapies. A review. Regen Ther 2019; 11:56-64. [PMID: 31193111 PMCID: PMC6517793 DOI: 10.1016/j.reth.2019.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/19/2019] [Accepted: 03/28/2019] [Indexed: 02/07/2023] Open
Abstract
Autologous biologics, defined as platelet-rich plasma (PRP) and bone marrow aspirate concentrate (BMC), are cell-based therapy treatment options in regenerative medicine practices, and have been increasingly used in orthopedics, sports medicine, and spinal disorders. These biological products are produced at point-of-care; thereby, avoiding expensive and cumbersome culturing and expansion techniques. Numerous commercial PRP and BMC systems are available but reports and knowledge of bio-cellular formulations produced by these systems are limited. This limited information hinders evaluating clinical and research outcomes and thus making conclusions about their biological effectiveness. Some of their important cellular and protein properties have not been characterized, which is critical for understanding the mechanisms of actions involved in tissue regenerative processes. The presence and role of red blood cells (RBCs) in any biologic has not been addressed extensively. Furthermore, some of the pathophysiological effects and phenomena related to RBCs have not been studied. A lack of a complete understanding of all of the biological components and their functional consequences hampers the development of clinical standards for any biological preparation. This paper aims to review the clinical implications and pathophysiological effects of RBCs in PRP and BMC; emphasizes hemolysis, eryptosis, and the release of macrophage inhibitory factor; and explains several effects on the microenvironment, such as inflammation, oxidative stress, vasoconstriction, and impaired cell metabolism. Different biological formulations optimize disease specific regenerative treatment protocols. Disintegrated RBC's release harmful components to regenerative therapy treatment vials. The effectiveness of MSC injection depends on the quality of the bone marrow aspiration procedure. PRP and BMC should contain minimal to no erythrocytes.
Collapse
Key Words
- BM-MSCs, bone marrow-mesenchymal cells
- BMA, bone marrow aspiration
- BMC, bone marrow concentrate
- Bone marrow mesenchymal cells
- Eryptosis
- HSCs, hematopoietic stem cells
- Hb, hemoglobin
- Hp, haptoglobin
- Hx, hemopexin
- Inflammation
- MIF, Macrophage migration inhibitory factor
- MNCs, mononucleated cells
- Macrophage migration inhibitor factor
- NO, nitric oxide
- OA, osteoarthritis
- Oxidative stress
- PAF, platelet activating factor
- PFH, plasma free hemoglobin
- PRP, platelet-rich plasma
- PS, phosphatidylserine
- Plasma free hemoglobin
- Platelet-rich plasma
- RBC, red blood cell
- ROS, reactive oxygen species
Collapse
Affiliation(s)
- Peter A. Everts
- Gulf Coast Biologics, Scientific and Research Department, Fort Myers, FL, USA
- Corresponding author. Gulf Coast Biologics, 6900 Daniels Pkwy, Suite #29-282, Fort Myers, FL 33912, USA.
| | - Gerard A. Malanga
- New Jersey Regenerative Institute LLC, Cedar Knolls, NJ, USA
- Department of Physical Medicine and Rehabilitation, Rutgers University, New Jersey Medical School, Newark, NJ, USA
| | - Rowan V. Paul
- California Pacific Orthopedics, San Francisco, CA, USA
- California Pacific Medical Center, San Francisco, CA, USA
- Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Joshua B. Rothenberg
- Boca Raton Regional Hospital, Regenerative Medicine and Orthopedics Biologic Department, Boca Raton, FL, USA
- BocaCare Orthopedics, Boca Raton, FL, USA
| | | | - Kenneth R. Mautner
- Emory University, Department of Physical Medicine & Rehabilitation, Atlanta GA, USA
- Emory University, Department of Orthopedics, Atlanta GA, USA
| |
Collapse
|
16
|
Liu X, Zheng L, Zhou Y, Chen Y, Chen P, Xiao W. BMSC Transplantation Aggravates Inflammation, Oxidative Stress, and Fibrosis and Impairs Skeletal Muscle Regeneration. Front Physiol 2019; 10:87. [PMID: 30814953 PMCID: PMC6382023 DOI: 10.3389/fphys.2019.00087] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 01/24/2019] [Indexed: 12/26/2022] Open
Abstract
Skeletal muscle contusion is one of the most common muscle injuries in sports medicine and traumatology. Bone marrow mesenchymal stem cell (BMSC) transplantation has been proposed as a promising strategy to promote skeletal muscle regeneration. However, the roles and underlying mechanisms of BMSCs in the regulation of skeletal muscle regeneration are still not completely clear. Here, we investigated the role of BMSC transplantation after muscle contusion. BMSCs were immediately transplanted into gastrocnemius muscles (GMs) following direct contusion. Comprehensive morphological and genetic analyses were performed after BMSC transplantation. BMSC transplantation exacerbated muscle fibrosis and inflammation, as evidenced by increased leukocyte and macrophage infiltration, increased inflammatory cytokines and chemokines, and increased matrix metalloproteinases. BMSC transplantation also increased muscle oxidative stress. Overall, BMSC transplantation aggravated inflammation, oxidative stress and fibrosis and impaired skeletal muscle regeneration. These results, shed new light on the role of BMSCs in regenerative medicine and indicate that caution is needed in the application of BMSCs for muscle injury.
Collapse
Affiliation(s)
- Xiaoguang Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lifang Zheng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yongzhan Zhou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yingjie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Weihua Xiao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
17
|
Guerra E, Fabbri D, Cavallo M, Marinelli A, Rotini R. Treatment of Capitellar Osteochondritis Dissecans With a Novel Regenerative Technique: Case Report of 3 Patients After 4 Years. Orthop J Sports Med 2018; 6:2325967118795831. [PMID: 30228993 PMCID: PMC6137555 DOI: 10.1177/2325967118795831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
| | - Daniele Fabbri
- IRCCS Galeazzi Orthopaedic Institute, San Siro Clinical Institute, Milan, Italy
| | | | | | | |
Collapse
|
18
|
Hong B, Lee S, Shin N, Ko Y, Kim D, Lee J, Lee W. Bone regeneration with umbilical cord blood mesenchymal stem cells in femoral defects of ovariectomized rats. Osteoporos Sarcopenia 2018; 4:95-101. [PMID: 30775550 PMCID: PMC6362973 DOI: 10.1016/j.afos.2018.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/04/2018] [Accepted: 08/27/2018] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Current treatments for osteoporosis were prevention of progression, yet it has been questionable in the stimulation of bone growth. The mesenchymal stem cells (MSCs) treatment for osteoporosis aims to induce differentiation of bone progenitor cells into bone-forming osteoblasts. We investigate whether human umbilical cord blood (hUCB)-MSCs transplantation may induce bone regeneration for osteoporotic rat model induced by ovariectomy. METHODS The ovariectomized (OVX) group (n = 10) and OVX-MSCs group (n = 10) underwent bilateral ovariectomy to induce osteoporosis, while the Sham group (n = 10) underwent sham operation at aged 12 weeks. After a femoral defect was made at 9 months, Sham group and OVX group were injected with Hartmann solution, while the OVX-MSCs group was injected with Hartmann solution containing 1 × 107 hUCB-MSCs. The volume of regenerated bone was evaluated using micro-computed tomography at 4 and 8 weeks postoperation. RESULTS At 4- and 8-week postoperation, the OVX group (5.0% ± 1.5%; 6.1% ± 0.7%) had a significantly lower regenerated bone volume than the Sham group (8.6% ± 1.3%; 12.0% ± 1.8%, P < 0.01), respectively. However, there was no significant difference between the OVX-MSCs and Sham groups. The OVX-MSCs group resulted in about 53% and 65% significantly higher new bone formation than the OVX group (7.7% ± 1.9%; 10.0% ± 2.9%, P < 0.05). CONCLUSIONS hUCB-MSCs in bone defects may enhance bone regeneration in osteoporotic rat model similar to nonosteoporotic bone regeneration. hUCB-MSCs may be a promising alternative stem cell therapy for osteoporosis.
Collapse
Affiliation(s)
- Boohwi Hong
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Sunyeul Lee
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - Nara Shin
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Korea
| | - Youngkwon Ko
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, Korea
| | - DongWoon Kim
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jun Lee
- Department of Orofascial Surgery, Wonkwang Dental Hospital, Daejeon, Korea
| | - Wonhyung Lee
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, Korea
| |
Collapse
|
19
|
Damia E, Chicharro D, Lopez S, Cuervo B, Rubio M, Sopena JJ, Vilar JM, Carrillo JM. Adipose-Derived Mesenchymal Stem Cells: Are They a Good Therapeutic Strategy for Osteoarthritis? Int J Mol Sci 2018; 19:ijms19071926. [PMID: 29966351 PMCID: PMC6073660 DOI: 10.3390/ijms19071926] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/21/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is a major cause of disability in elderly population around the world. More than one-third of people over 65 years old shows either clinical or radiological evidence of OA. There is no effective treatment for this degenerative disease, due to the limited capacity for spontaneous cartilage regeneration. Regarding the use of regenerative therapies, it has been reported that one option to restore degenerated cartilage are adipose-derived mesenchymal stem cells (ASCs). The purpose of this review is to describe and compare the efficacy of ASCs versus other therapies in OA. Methods: Recent studies have shown that ASCs exert paracrine effects protecting against degenerative changes in chondrocytes. According to the above, we have carried out a review of the literature using a combination of osteoarthritis, stem cells, and regenerative therapies as keywords. Results: Conventional pharmacological therapies for OA treatment are considered before the surgical option, however, they do not stop the progression of the disease. Moreover, total joint replacement is not recommended for patients under 55 years, and high tibia osteotomy (HTO) is a viable solution to address lower limb malalignment with concomitant OA, but some complications have been described. In recent years, the use of mesenchymal stem cells (MSCs) as a treatment strategy for OA is increasing considerably, thanks to their capacity to improve symptoms together with joint functionality and, therefore, the patients’ quality of life. Conclusions: ASC therapy has a positive effect on patients with OA, although there is limited evidence and little long-term follow-up.
Collapse
Affiliation(s)
- Elena Damia
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
| | - Sergio Lopez
- Department of Animal Pathology. Instituto Universitario de Investigaciones Biomédicas y Sanitarias. University of Las Palmas de Gran Canaria, 35416 Las Palmas de Gran Canaria, Spain.
| | - Belen Cuervo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
| | - Monica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
| | - Joaquin J Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
| | - Jose Manuel Vilar
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
- Department of Animal Pathology. Instituto Universitario de Investigaciones Biomédicas y Sanitarias. University of Las Palmas de Gran Canaria, 35416 Las Palmas de Gran Canaria, Spain.
| | - Jose Maria Carrillo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- Garcia Cugat Foundation CEU UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain.
| |
Collapse
|
20
|
Mesenchymal stem cells and porous β-tricalcium phosphate composites prepared through stem cell screen-enrich-combine(-biomaterials) circulating system for the repair of critical size bone defects in goat tibia. Stem Cell Res Ther 2018; 9:157. [PMID: 29895312 PMCID: PMC5998551 DOI: 10.1186/s13287-018-0906-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/07/2018] [Accepted: 05/15/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Efficacious bone substitute is essential for the treatment of a critical size bone defect. Currently, the bone substitutes commonly used in clinical practice lack osteogenic capacity and the therapeutic efficacy is not ideal. Herein, a novel stem cell screen-enrich-combine(-biomaterials) circulating system (SECCS) was introduced to provide the substitutes with osteogenic ability. The stem cell screening, enrichment, and recombination with substitutes could be integrated during the surgical operation. Using SECCS, the bioactive mesenchymal stem cells (MSCs) and porous β-tricalcium phosphate (β-TCP) composites (MSCs/β-TCP) were rapidly prepared for critical size bone defect repair and validated in goat models of critical size tibia defects. METHODS Twelve goats with right hind limb tibia defects of 30 mm were randomly divided into two groups: six (the experimental group) were treated with MSCs/β-TCP prepared by SECCS and the other six goats (the control group) were treated with pure porous β-TCP. The repair effect was assessed by x-ray, computed tomography (CT), micro-CT, histology and histomorphology 6 months after the operation. In addition, the enrichment efficacy of MSCs and the characteristics of the MSCs/β-TCP prepared by SECCS were evaluated. RESULTS The SECCS could compound about 81.3 ± 3.0% of the MSCs in bone marrow to the porous β-TCP without affecting the cell viability. The average number of MSCs for retransplantation was 27,655.0 ± 5011.6 for each goat from the experimental group. In vitro, satisfactory biocompatibility of the MSCs/β-TCP was performed, with the MSCs spreading adequately, proliferating actively, and retaining the osteogenic potential. In vivo, the defect repair by MSCs/β-TCP with good medullary cavity recanalization and cortical remodeling was significantly superior to that of pure porous β-TCP. CONCLUSIONS The MSCs/β-TCP prepared through SECCS demonstrated significant therapeutic efficacy in goat models of critical size bone defect. This provides a novel therapeutic strategy for critical size bone defects caused by severe injury, infection, and bone tumor resection with a high profile of safety, effectiveness, simplicity, and ease.
Collapse
|
21
|
Combined use of bone marrow-derived mesenchymal stromal cells (BM-MSCs) and platelet rich plasma (PRP) stimulates proliferation and differentiation of myoblasts in vitro: new therapeutic perspectives for skeletal muscle repair/regeneration. Cell Tissue Res 2018; 372:549-570. [PMID: 29404727 DOI: 10.1007/s00441-018-2792-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 01/06/2018] [Indexed: 10/18/2022]
Abstract
Satellite cell-mediated skeletal muscle repair/regeneration is compromised in cases of extended damage. Bone marrow mesenchymal stromal cells (BM-MSCs) hold promise for muscle healing but some criticisms hamper their clinical application, including the need to avoid animal serum contamination for expansion and the scarce survival after transplant. In this context, platelet-rich plasma (PRP) could offer advantages. Here, we compare the effects of PRP or standard culture media on C2C12 myoblast, satellite cell and BM-MSC viability, survival, proliferation and myogenic differentiation and evaluate PRP/BM-MSC combination effects in promoting myogenic differentiation. PRP induced an increase of mitochondrial activity and Ki67 expression comparable or even greater than that elicited by standard media and promoted AKT signaling activation in myoblasts and BM-MSCs and Notch-1 pathway activation in BM-MSCs. It stimulated MyoD, myogenin, α-sarcomeric actin and MMP-2 expression in myoblasts and satellite cell activation. Notably, PRP/BM-MSC combination was more effective than PRP alone. We found that BM-MSCs influenced myoblast responses through a paracrine activation of AKT signaling, contributing to shed light on BM-MSC action mechanisms. Our results suggest that PRP represents a good serum substitute for BM-MSC manipulation in vitro and could be beneficial towards transplanted cells in vivo. Moreover, it might influence muscle resident progenitors' fate, thus favoring the endogenous repair/regeneration mechanisms. Finally, within the limitations of an in vitro experimentation, this study provides an experimental background for considering the PRP/BM-MSC combination as a potential therapeutic tool for skeletal muscle damage, combining the beneficial effects of BM-MSCs and PRP on muscle tissue, while potentiating BM-MSC functionality.
Collapse
|
22
|
Liu L, Chen JX, Zhang XW, Sun Q, Yang L, Liu A, Hu S, Guo F, Liu S, Huang Y, Yang Y, Qiu HB. Chemokine receptor 7 overexpression promotes mesenchymal stem cell migration and proliferation via secreting Chemokine ligand 12. Sci Rep 2018; 8:204. [PMID: 29317710 PMCID: PMC5760632 DOI: 10.1038/s41598-017-18509-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 12/13/2017] [Indexed: 01/06/2023] Open
Abstract
Great interest has been shown in mesenchymal stem cell (MSC) therapy in a wide variety of clinical domains. However, the therapeutic efficiency depends on the proliferation and migration of MSCs. Chemokine receptors are involved in regulating the proliferation and migration to the specific organs of MSCs in different microenvironments. CXC receptor seven (CXCR7), a newly discovered Chemokine ligand 12 (CXCL12) receptor, has organ specificity for tumour migration. We hypothesized that CXCR7 expression affects proliferation and migration of MSCs. In present study, we constructed long-term and stable mMSCs lines overexpressing and suppressing CXCR7 modifications with lentiviral vectors. The transduction efficiencies, mRNA and protein expression of CXCR7 were significantly regulated. CXCR7 gene overexpression promoted mMSCs proliferation and migration, whereas suppressing CXCR7 had the opposite effect. Additional CXCL12 improved the vertical migration of mMSCs. The overexpression of CXCR7 increased the MSC-secreted CXCL12, VCAM-1, CD44 and MMP2 levels, which contributed to the improvement of mMSC proliferation and migration. Therefore, overexpressing CXCR7 improved the proliferation and migration of mMSCs, which may be attributable to the CXCL12 secreted by MSCs, leading to a positive feedback loop for CXCL12/CXCR7 axis. Our results may provide a potential method for improving the treatment effectiveness of mMSCs by overexpressing CXCR7.
Collapse
Affiliation(s)
- Ling Liu
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jian-Xiao Chen
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xi-Wen Zhang
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Qin Sun
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Lan Yang
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Airan Liu
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Shuling Hu
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Fengmei Guo
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Songqiao Liu
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yingzi Huang
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yi Yang
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Hai-Bo Qiu
- Department of Critical Care Medicine, Nanjing Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
23
|
Emerging Concepts in Treating Cartilage, Osteochondral Defects, and Osteoarthritis of the Knee and Ankle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:25-62. [PMID: 29736568 DOI: 10.1007/978-3-319-76735-2_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The management and treatment of cartilage lesions, osteochondral defects, and osteoarthritis remain a challenge in orthopedics. Moreover, these entities have different behaviors in different joints, such as the knee and the ankle, which have inherent differences in function, biology, and biomechanics. There has been a huge development on the conservative treatment (new technologies including orthobiologics) as well as on the surgical approach. Some surgical development upraises from technical improvements including advanced arthroscopic techniques but also from increased knowledge arriving from basic science research and tissue engineering and regenerative medicine approaches. This work addresses the state of the art concerning basic science comparing the knee and ankle as well as current options for treatment. Furthermore, the most promising research developments promising new options for the future are discussed.
Collapse
|
24
|
Implications of anticoagulants and gender on cell counts and growth factor concentration in platelet-rich plasma and platelet-rich gel supernatants from rabbits. Vet Comp Orthop Traumatol 2017; 29:115-24. [DOI: 10.3415/vcot-15-01-0011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 10/18/2015] [Indexed: 11/17/2022]
Abstract
SummaryObjectives: Our objectives were as follows: 1) to validate a protocol for producing rabbit platelet-rich plasma (PRP); 2) to determine the influence of two anticoagulants, sodium citrate and acid-citrate-dextrose solution A, and gender on cell count in PRP and growth factor concentration in pure platelet-rich gel supernatants; 3) to correlate the variables evaluated.Methods: Whole blood from 18 New Zealand rabbits (9 males and 9 females) was obtained with sodium citrate and acid- citrate-dextrose solution A for processing PRP fractions (A and B), which were evaluated for haematology. The PRP fractions were either activated with calcium gluconate or lysated with a detergent. The concentrations of transforming growth factor beta 1 and platelet-derived growth factor BB were assayed by ELISA.Results: The sodium citrate PRP-B had significantly higher counts of platelets in comparison to PRP-A and whole blood obtained with the same anticoagulant and the homologous acid-citrate-dextrose solution A PRP fraction. The sodium citrate PRP-A had a significantly higher count of leukocytes compared to the homologous acid-citrate-dextrose solution A fraction. All the PRP fractions had a significant leuko-reduction when compared to whole blood. The sodium citrate PRP-A fraction from female rabbits had significantly lower platelet counts and significantly higher leukocyte counts than the same acid-citrate-dextrose solution A fraction. Growth factor concentration was not affected by the type of anticoagulant or gender.Clinical significance: The type of anticoagulant and gender affected the cell counts in PRP, but they did not influence the growth factor concentration. More complete rabbit PRP studies should be performed before evaluating this type of substance in models of disease.
Collapse
|
25
|
A novel cytotherapy device for rapid screening, enriching and combining mesenchymal stem cells into a biomaterial for promoting bone regeneration. Sci Rep 2017; 7:15463. [PMID: 29133959 PMCID: PMC5684202 DOI: 10.1038/s41598-017-15451-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 10/24/2017] [Indexed: 01/26/2023] Open
Abstract
Bone defects are a common challenge in clinic, usually warranting bone grafts. However, current strategies to obtain effective graft materials have many drawbacks. Mesenchymal stem cell (MSC)-based therapy is a promising alternative. We designed an innovative appliance named the stem cell screen-enrich-combine(-biomaterials) circulating system (SECCS). In this study, 42 patients who required bone graft underwent SECCS-based treatment. Their bone marrow samples and beta-tricalcium phosphate (β-TCP) granules were processed in the SECCS for 10-15 minutes, to produce MSC/β-TCP composites. These composites were grafted back into bone defect sites. The results showed 85.53% ± 7.95% autologous MSCs were successfully screened, enriched, and seeded on the β-TCP scaffolds synchronously. The cell viability remained unchanged after SECCS processing. Clinically, all patients obtained satisfactory bone healing. Thus, without in vitro culture, the SECCS can produce bioactive MSC/β-TCP composites for bone regeneration during surgery. The SECCS represents a convenient, rapid, low-cost, and safe method for bone regeneration.
Collapse
|
26
|
Wehling P, Evans C, Wehling J, Maixner W. Effectiveness of intra-articular therapies in osteoarthritis: a literature review. Ther Adv Musculoskelet Dis 2017; 9:183-196. [PMID: 28835778 DOI: 10.1177/1759720x17712695] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/25/2017] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis is a painful, chronic disease with widespread burden on patients, communities, health and social care systems. Conservative therapies, such as nonpharmacological interventions, systemic drug treatment and intra-articular therapies are used before resorting to surgery; nonetheless, disease control often remains inadequate. Recent advances in osteoarthritis management have aimed to provide greater variety of treatment options. Here, we summarize a targeted literature review evaluating efficacy and safety of intra-articular therapies for osteoarthritis. Injections of intra-articular therapies directly into the joint avoid conventional barriers to joint entry, increase bioavailability and lower systemic toxicity. Intra-articular corticosteroids and hyaluronic acid are established United States Food and Drug Administration (US FDA)/European Medicines Agency (EMA)-approved treatments; however, concerns exist regarding effect duration, safety, effectiveness across populations and heterogeneity. Newer therapies, such as autologous blood products and mesenchymal stem cells, are in development. Benefits of autologous blood products (e.g. platelet-rich plasma, autologous conditioned serum) include an expected improved safety profile and direct targeting of osteoarthritis-related pathophysiology. Autologous conditioned serum is cell-free and manufactured by a standardized process, whereas platelet-rich plasma composition and characteristics can vary. Currently, only limited efficacy comparisons between these biological treatments can be drawn; long-term clinical and safety studies are needed to increase the efficacy evidence base and earn consideration in treatment frameworks.
Collapse
Affiliation(s)
- Peter Wehling
- Orthogen AG, Ernst-Schneider-Platz 1, 40212 Düsseldorf, Germany
| | - Christopher Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN, USA
| | - Jana Wehling
- Center for Molecular Orthopaedics and Regenerative Medicine, Stadttor 1, Düsseldorf, Germany
| | - William Maixner
- Center for Translational Pain Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
27
|
Qazi TH, Mooney DJ, Duda GN, Geissler S. Biomaterials that promote cell-cell interactions enhance the paracrine function of MSCs. Biomaterials 2017. [PMID: 28644976 DOI: 10.1016/j.biomaterials.2017.06.019] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cells (MSCs) secrete paracrine factors that play crucial roles during tissue regeneration. Whether this paracrine function is influenced by the properties of biomaterials in general, and those used for cell delivery in particular, largely remains unexplored. Here, we investigated if three-dimensional culture in distinct microenvironments - nanoporous hydrogels (mean pore size ∼5 nm) and macroporous scaffolds (mean pore size ∼120 μm) - affects the secretion pattern of MSCs, and consequently leads to differential paracrine effects on target progenitor cells such as myoblasts. We report that compared to MSCs encapsulated in hydrogels, scaffold seeded MSCs show an enhanced secretion profile and exert beneficial paracrine effects on various myoblast functions including migration and proliferation. Additionally, we show that the heightened paracrine effects of scaffold seeded cells can in part be attributed to N-cadherin mediated cell-cell interactions during culture. In hydrogels, this physical interaction between cells is prevented by the encapsulating matrix. Functionally blocking N-cadherin negatively affected the secretion profile and paracrine effects of MSCs on myoblasts, with stronger effects observed for scaffold seeded compared to hydrogel encapsulated cells. Together, these findings demonstrate that the therapeutic potency of MSCs can be enhanced by biomaterials that promote cell-cell interactions.
Collapse
Affiliation(s)
- Taimoor H Qazi
- Julius Wolff Institute, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies & Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford St., Cambridge, MA 02138, USA
| | - Georg N Duda
- Julius Wolff Institute, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies & Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sven Geissler
- Julius Wolff Institute, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies & Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
28
|
In Vitro Biocompatibility Assessment and In Vivo Behavior of a New Osteoconductive βTCP Bone Substitute. IMPLANT DENT 2017; 25:456-63. [PMID: 27455428 DOI: 10.1097/id.0000000000000442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Beta-tricalcium phosphate (βTCP) granules (OsproLife) exhibit a pure crystalline phase and a rough microporous surface for promoting cell adhesion and microsized intragranule porosity for improving wettability and resorption necessary for bone regeneration. OsproLife is a fully resorbable, space-maintaining, and osteoconductive synthetic material for the filling of bone defects. To asses OsproLife properties, a similar synthetic biomaterial, already on the market, has been chosen as reference: Cerasorb has the same chemical composition, but different crystal structure, surface morphology, and granule size. The aim of this study is to compare the properties of OsproLife and Cerasorb. METHODS Chemical purity, composition and physical properties, in vitro cytotoxicity, and in vivo bone performance in a rabbit model were analyzed. βTCP OsproLife granules (test) were compared with Cerasorb (control). Histological and μCT analyses were performed at 6, 12, and 56 weeks after implantation. RESULTS βTCP OsproLife and Cerasorb granules result to be both biocompatible and characterized by the same osteoconductivity and resorption rate. CONCLUSION βTCP OsproLife granules are a promising bone substitute for dental and orthopedic applications.
Collapse
|
29
|
Bertone AL, Reisbig NA, Kilborne AH, Kaido M, Salmanzadeh N, Lovasz R, Sizemore JL, Scheuermann L, Kopp RJ, Zekas LJ, Brokken MT. Equine Dental Pulp Connective Tissue Particles Reduced Lameness in Horses in a Controlled Clinical Trial. Front Vet Sci 2017; 4:31. [PMID: 28344975 PMCID: PMC5344919 DOI: 10.3389/fvets.2017.00031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 02/20/2017] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE To assess if injection of allogeneic dental pulp tissue particles would improve lameness in horses with naturally occurring osteoarthritis (OA) or soft tissue (ST) injury. DESIGN Prospective, randomized, blinded, and controlled clinical trial and client survey assessment. ANIMALS Forty lame client-owned horses. PROCEDURES Sterile dental pulp, recovered from otherwise healthy foals that perish during dystocia, was processed under good manufacturing processing to produce mechanically manipulated, unexpanded pulp tissue particles containing viable cells surrounded in extracellular matrix. Forty lame client-owned horses with confirmed OA (n = 20), or ST injury (desmitis or tendonitis) received a 2 mL intra-articular (n = 20 OA) or intra-lesional (n = 20) injection of control transport vehicle (n = 20) or 10 × 106 dental pulp tissue particles (n = 20). Acclimatized horses had baseline measurements performed and were then injected on day 0. Horses were treadmill exercised for 2 weeks, evaluated by clinical parameters, lameness score, edema (score and circumference), pain on flexion (OA) or pressure (ST), and clients' scores for pain and discomfort before and through 45 days after pulp injection. Twenty horses were available for >2.5-year follow-up. RESULTS Pulp-treated horses showed decrease in lameness compared to baseline (P < 0.009) or placebo controls (P < 0.013) for at least 2 weeks. Client assessments of comfort were improved between before and 45 days after pulp injection (P < 0.001). Clinical improvement with ST injury was significantly greater than OA (P < 0.001). At >2.5-year follow-up, at least 10 horses were in work. CONCLUSION AND CLINICAL RELEVANCE Dental pulp tissue particles can be considered as a treatment option for equine lameness due to OA, desmitis, or tendonitis.
Collapse
Affiliation(s)
- Alicia L. Bertone
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Nathalie A. Reisbig
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Allison H. Kilborne
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Mari Kaido
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Navid Salmanzadeh
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Rebecca Lovasz
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Joy L. Sizemore
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Logan Scheuermann
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Rosalind J. Kopp
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Lisa J. Zekas
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Matthew T. Brokken
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
30
|
Parsha K, Mir O, Satani N, Yang B, Guerrero W, Mei Z, Cai C, Chen PR, Gee A, Hanley PJ, Aronowski J, Savitz SI. Mesenchymal stromal cell secretomes are modulated by suspension time, delivery vehicle, passage through catheter, and exposure to adjuvants. Cytotherapy 2017; 19:36-46. [DOI: 10.1016/j.jcyt.2016.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 10/06/2016] [Accepted: 10/17/2016] [Indexed: 01/09/2023]
|
31
|
Hofer HR, Tuan RS. Secreted trophic factors of mesenchymal stem cells support neurovascular and musculoskeletal therapies. Stem Cell Res Ther 2016; 7:131. [PMID: 27612948 PMCID: PMC5016979 DOI: 10.1186/s13287-016-0394-0] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adult mesenchymal stem cells (MSCs) represent a subject of intense experimental and biomedical interest. Recently, trophic activities of MSCs have become the topic of a number of revealing studies that span both basic and clinical fields. In this review, we focus on recent investigations that have elucidated trophic mechanisms and shed light on MSC clinical efficacy relevant to musculoskeletal applications. Innate differences due to MSC sourcing may play a role in the clinical utility of isolated MSCs. Pain management, osteochondral, nerve, or blood vessel support by MSCs derived from both autologous and allogeneic sources have been examined. Recent mechanistic insights into the trophic activities of these cells point to ultimate regulation by nitric oxide, nuclear factor-kB, and indoleamine, among other signaling pathways. Classic growth factors and cytokines-such as VEGF, CNTF, GDNF, TGF-β, interleukins (IL-1β, IL-6, and IL-8), and C-C ligands (CCL-2, CCL-5, and CCL-23)-serve as paracrine control molecules secreted or packaged into extracellular vesicles, or exosomes, by MSCs. Recent studies have also implicated signaling by microRNAs contained in MSC-derived exosomes. The response of target cells is further regulated by their microenvironment, involving the extracellular matrix, which may be modified by MSC-produced matrix metalloproteinases (MMPs) and tissue inhibitor of MMPs. Trophic activities of MSCs, either resident or introduced exogenously, are thus intricately controlled, and may be further fine-tuned via implant material modifications. MSCs are actively being investigated for the repair and regeneration of both osteochondral and other musculoskeletal tissues, such as tendon/ligament and meniscus. Future rational and effective MSC-based musculoskeletal therapies will benefit from better mechanistic understanding of MSC trophic activities, for example using analytical "-omics" profiling approaches.
Collapse
Affiliation(s)
- Heidi R Hofer
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
32
|
De Becker A, Riet IV. Homing and migration of mesenchymal stromal cells: How to improve the efficacy of cell therapy? World J Stem Cells 2016; 8:73-87. [PMID: 27022438 PMCID: PMC4807311 DOI: 10.4252/wjsc.v8.i3.73] [Citation(s) in RCA: 334] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 12/24/2015] [Accepted: 01/29/2016] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are currently being investigated for use in a wide variety of clinical applications. For most of these applications, systemic delivery of the cells is preferred. However, this requires the homing and migration of MSCs to a target tissue. Although MSC homing has been described, this process does not appear to be highly efficacious because only a few cells reach the target tissue and remain there after systemic administration. This has been ascribed to low expression levels of homing molecules, the loss of expression of such molecules during expansion, and the heterogeneity of MSCs in cultures and MSC culture protocols. To overcome these limitations, different methods to improve the homing capacity of MSCs have been examined. Here, we review the current understanding of MSC homing, with a particular focus on homing to bone marrow. In addition, we summarize the strategies that have been developed to improve this process. A better understanding of MSC biology, MSC migration and homing mechanisms will allow us to prepare MSCs with optimal homing capacities. The efficacy of therapeutic applications is dependent on efficient delivery of the cells and can, therefore, only benefit from better insights into the homing mechanisms.
Collapse
|
33
|
Australasian College of Sports Physicians-Position Statement: The Place of Mesenchymal Stem/Stromal Cell Therapies in Sport and Exercise Medicine. Clin J Sport Med 2016; 26:87-95. [PMID: 26784119 DOI: 10.1097/jsm.0000000000000298] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
34
|
Augusto LMM, Aguiar DP, Bonfim DC, Dos Santos Cavalcanti A, Casado PL, Duarte MEL. Mesenchymal stromal cells from bone marrow treated with bovine tendon extract acquire the phenotype of mature tenocytes. Rev Bras Ortop 2016; 51:70-4. [PMID: 26962503 PMCID: PMC4767843 DOI: 10.1016/j.rboe.2015.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 02/03/2015] [Indexed: 11/16/2022] Open
Abstract
Objective This study evaluated in vitro differentiation of mesenchymal stromal cells isolated from bone marrow, in tenocytes after treatment with bovine tendon extract. Methods Bovine tendons were used for preparation of the extract and were stored at −80 °C. Mesenchymal stromal cells from the bone marrow of three donors were used for cytotoxicity tests by means of MTT and cell differentiation by means of qPCR. Results The data showed that mesenchymal stromal cells from bone marrow treated for up to 21 days in the presence of bovine tendon extract diluted at diminishing concentrations (1:10, 1:50 and 1:250) promoted activation of biglycan, collagen type I and fibromodulin expression. Conclusion Our results show that bovine tendon extract is capable of promoting differentiation of bone marrow stromal cells in tenocytes.
Collapse
|
35
|
Augusto LMM, Aguiar DP, Bonfim DC, Cavalcanti ADS, Casado PL, Duarte MEL. Células mesenquimais do estroma da medula óssea tratadas com extrato de tendão bovino adquirem o fenótipo de tenócitos maduros. Rev Bras Ortop 2016. [DOI: 10.1016/j.rbo.2015.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
36
|
Osborne H, Anderson L, Burt P, Young M, Gerrard D. Australasian College of Sports Physicians—position statement: the place of mesenchymal stem/stromal cell therapies in sport and exercise medicine. Br J Sports Med 2015; 50:1237-1244. [DOI: 10.1136/bjsports-2015-095711] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2015] [Indexed: 12/18/2022]
|
37
|
Development of mRuby2-Transfected C3H10T1/2 Fibroblasts for Musculoskeletal Tissue Engineering. PLoS One 2015; 10:e0139054. [PMID: 26407291 PMCID: PMC4583363 DOI: 10.1371/journal.pone.0139054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/07/2015] [Indexed: 11/19/2022] Open
Abstract
Mouse C3H10T1/2 fibroblasts are multipotent, mesenchymal stem cell (MSC)-like progenitor cells that are widely used in musculoskeletal research. In this study, we have established a clonal population of C3H10T1/2 cells stably-transfected with mRuby2, an orange-red fluorescence reporter gene. Flow cytometry analysis and fluorescence imaging confirmed successful transfection of these cells. Cell counting studies showed that untransfected C3H10T1/2 cells and mRuby2-transfected C3H10T1/2 cells proliferated at similar rates. Adipogenic differentiation experiments demonstrated that untransfected C3H10T1/2 cells and mRuby2-transfected C3H10T1/2 cells stained positive for Oil Red O and showed increased expression of adipogenic genes including adiponectin and lipoprotein lipase. Chondrogenic differentiation experiments demonstrated that untransfected C3H10T1/2 cells and mRuby2-transfected C3H10T1/2 cells stained positive for Alcian Blue and showed increased expression of chondrogenic genes including aggrecan. Osteogenic differentiation experiments demonstrated that untransfected C3H10T1/2 cells and mRuby2-transfected C3H10T1/2 cells stained positive for alkaline phosphatase (ALP) as well as Alizarin Red and showed increased expression of osteogenic genes including alp, ocn and osf-1. When seeded on calcium phosphate-based ceramic scaffolds, mRuby2-transfected C3H10T1/2 cells maintained even fluorescence labeling and osteogenic differentiation. In summary, mRuby2-transfected C3H10T1/2 cells exhibit mRuby2 fluorescence and showed little-to-no difference in terms of cell proliferation and differentiation as untransfected C3H10T1/2 cells. These cells will be available from American Type Culture Collection (ATCC; CRL-3268™) and may be a valuable tool for preclinical studies.
Collapse
|
38
|
Centeno CJ, Al-Sayegh H, Bashir J, Goodyear S, Freeman MD. A dose response analysis of a specific bone marrow concentrate treatment protocol for knee osteoarthritis. BMC Musculoskelet Disord 2015; 16:258. [PMID: 26385099 PMCID: PMC4575428 DOI: 10.1186/s12891-015-0714-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/08/2015] [Indexed: 12/14/2022] Open
Abstract
Background Prior studies describing the treatment of symptomatic knee osteoarthritis with injections of bone marrow concentrate have provided encouraging results. The relationship between the cellular dose contained within the bone marrow concentrate and efficacy of the treatment, however, is unclear. In the present study we describe clinical outcomes for symptomatic knee osteoarthritis in relation to higher and lower cell concentrations contained within a bone marrow concentrate treatment protocol. Methods Data from an ongoing patient registry was culled to identify 373 patients that received bone marrow concentrate injections for the treatment of 424 osteoarthritic knee joints. The clinical scales for these patients were assessed at baseline and then tracked post-procedure at 1, 3, 6 and 12 months, and annually thereafter. Tracked outcomes included the numeric pain scale; a lower extremity functional questionnaire; an International Knee Documentation Committee scale; and a subjective improvement rating scale. Using pain and functional outcome measures, a receiver operating characteristic analysis was used to define an optimal clinical outcome threshold at which bone marrow nucleated cell count could be divided into either a lower or higher cell count group within a treatment protocol. Results The lower and higher cell count groups were defined using a threshold of 4 × 108 cells. There were 224 and 185 knee joints treated in the lower (≤4 × 108) and higher (>4 × 108) cell count groups respectively. Most joints were diagnosed with early stage knee osteoarthritis. Both the lower and higher cell count groups demonstrated significant positive results with the treatment for all of the pain and functional metrics. The higher cell count group reported lower post treatment numeric pain scale values, in comparison with the lower cell count group (1.6 vs. 3.2; P < 0.001). No significant differences were detected for the other metrics, however. Conclusions Improved function and reduced pain was observed in patients treated with a bone marrow concentrate protocol regardless of cellular dose; however, patients receiving a higher concentration of cells reported a better pain outcome in comparison with the lower dose group. These preliminary findings suggest that cell dose may be an important factor governing clinical outcomes in autologous bone marrow concentrate treatment of knee osteoarthritis. Further studies using a larger patient population may help elucidate these findings.
Collapse
Affiliation(s)
| | - Hasan Al-Sayegh
- Centeno-Schultz Clinic, 403 Summit Blvd Suite 201, Broomfield, CO, 80021, USA.
| | - Jamil Bashir
- Centeno-Schultz Clinic, 403 Summit Blvd Suite 201, Broomfield, CO, 80021, USA
| | | | | |
Collapse
|
39
|
The Addition of Platelet-Rich Plasma to Scaffolds Used for Cartilage Repair: A Review of Human and Animal Studies. Arthroscopy 2015; 31:1607-25. [PMID: 25823672 DOI: 10.1016/j.arthro.2015.01.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/15/2015] [Accepted: 01/22/2015] [Indexed: 02/02/2023]
Abstract
PURPOSE To review the available literature on studies focusing on platelet-rich plasma (PRP)-enhanced scaffolds for cartilage lesion repair in animals and to analyze the clinical outcomes of similar biologically augmented cartilage regeneration techniques in humans. METHODS We conducted a literature search and subsequent review investigating the potential of PRP to enhance articular cartilage repair using scaffolds or bioengineered implants. RESULTS Of the 14 animal model studies reviewed, 10 reported positive effects with PRP whereas only 2 showed negative overall effects. The remaining 2 studies reported no significant differences, or neutral results, with the use of PRP. With the addition of PRP, the gross appearance and histologic analysis of repair cartilage were improved or no difference was seen compared with control (11 of 12 studies that looked at this). Human studies of the knee or talar dome showed improvements in clinical assessment scores as soon as 6 months after surgery. There was great variability in the method of PRP preparation, choice of scaffold, and cell source between studies. CONCLUSIONS PRP-augmented scaffolds have been shown to be beneficial in the articular cartilage repair process in animals and humans based on macroscopic, histologic, and biochemical analysis and based on clinical outcome scores, respectively. Comparison between studies is difficult because there is great variability in PRP preparation and administration. LEVEL OF EVIDENCE Level IV, systematic review of Level III and IV studies.
Collapse
|
40
|
Gianakos AL, Ross KA, Hannon CP, Duke GL, Prado MP, Kennedy JG. Functional Outcomes of Tibialis Posterior Tendoscopy With Comparison to Magnetic Resonance Imaging. Foot Ankle Int 2015; 36:812-9. [PMID: 25759276 DOI: 10.1177/1071100715576485] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The purpose of the current study was to report functional outcomes of tendoscopy for treatment of tibialis posterior tendon pathology as well as compare its diagnostic capability with magnetic resonance imaging (MRI). METHODS Clinical records and MRI of 12 patients who underwent tendoscopy of the tibialis posterior tendon (TPT) were retrospectively reviewed. Mean follow-up was 31 months (range, 26-43 months). Preoperative MRI findings were compared with tendoscopic findings to assess the diagnostic agreement between each modality. Functional outcomes were assessed using the Foot and Ankle Outcome Score (FAOS) and Short Form-12 (SF-12) General Health Questionnaire pre- and postoperatively. Mean patient age was 43 years (range, 17-63 years). Mean duration of preoperative symptoms was 15.5 months (range, 3-36 months). RESULTS Pathologies addressed via tendoscopy included tenosynovitis, tendinosis, stenosis, tendon subluxation, and partial thickness tear (via mini-arthrotomy). Preoperative MRI findings were in agreement with tendoscopic findings in 8 of 12 cases (67%). Tendoscopy diagnosed and allowed access for treating pathology that was missed on MRI in the remaining four cases. The FAOS improved from a mean preoperative score of 58 (range, 36-78) to a mean postoperative score of 81 (range, 44-98) (P < .01). The SF-12 score improved from a mean preoperative score of 34 (range, 13-51) to a mean postoperative score of 51 (range, 21-76) (P = .01). CONCLUSIONS Although MRI is considered an effective imaging technique for tendon pathology, tendoscopy may be a more sensitive diagnostic tool. Tendoscopy was an effective minimally invasive tool to diagnose and treat tibialis posterior tendon pathology resulting in functional improvements in the short-term for early stage TPT dysfunction. Further studies comparing tendoscopy with traditional open approaches are warranted. LEVEL OF EVIDENCE Level IV case series.
Collapse
Affiliation(s)
| | - Keir A Ross
- Hospital for Special Surgery, New York, NY, USA
| | | | | | | | | |
Collapse
|
41
|
Sun HB, Schaniel C, Leong DJ, Wang JHC. Biology and mechano-response of tendon cells: Progress overview and perspectives. J Orthop Res 2015; 33:785-92. [PMID: 25728946 PMCID: PMC4422159 DOI: 10.1002/jor.22885] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/24/2015] [Indexed: 02/04/2023]
Abstract
In this review, we summarize the group discussions on Cell Biology & Mechanics from the 2014 ORS/ISMMS New Frontiers in Tendon Research Conference. The major discussion topics included: (1) the biology of tendon stem/progenitor cells (TSPCs) and the potential of stem cell-based tendon therapy using TSPCs and other types of stem cells, namely, embryonic and/or induced pluripotent stem cells (iPSCs), (2) the biological concept and potential impact of cellular senescence on tendon aging, tendon injury repair and the development of degenerative disease, and (3) the effects of tendon cells' mechano-response on tendon cell fate and metabolism. For each topic, a brief overview is presented which summarizes the major points discussed by the group participants. The focus of the discussions ranged from current research progress, challenges and opportunities, to future directions on these topics. In the preparation of this manuscript, authors consulted relevant references as a part of their efforts to present an accurate view on the topics discussed.
Collapse
Affiliation(s)
- Hui B. Sun
- Department of Orthopaedic Surgery, Albert Einstein College of Medicine, Bronx, NY
,Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY
,Corresponding Author: 1300 Morris Park Avenue, Golding 101 Bronx, NY 10461 USA Tel: (718) 430-4291 Fax: (718) 430-3259
| | - Christoph Schaniel
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY
,Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY
,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Daniel J. Leong
- Department of Orthopaedic Surgery, Albert Einstein College of Medicine, Bronx, NY
,Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY
| | - James H-C. Wang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
42
|
Ro H, Park J, Yang K, Kim J, Yim HG, Jung G, Lee H, Cho SW, Hwang NS. Osteogenic priming of mesenchymal stem cells by chondrocyte-conditioned factors and mineralized matrix. Cell Tissue Res 2015; 362:115-26. [DOI: 10.1007/s00441-015-2195-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 04/11/2015] [Indexed: 12/13/2022]
|
43
|
Sepúlveda F, Baerga L, Micheo W. The role of physiatry in regenerative medicine: the past, the present, and future challenges. PM R 2015; 7:S76-S80. [PMID: 25864663 DOI: 10.1016/j.pmrj.2015.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/30/2014] [Accepted: 01/09/2015] [Indexed: 01/13/2023]
Abstract
Historically, the foundation of physical medicine and rehabilitation training has provided the capabilities to optimize nonoperative treatments of a variety of musculoskeletal conditions, including acute and chronic muscle, tendon, ligament, and cartilage disorders. Such treatments include the use of nonsteroidal anti-inflammatory drugs (NSAIDs), therapeutic modalities (eg, thermal and manual therapies), and corticosteroid injections in conjunction with specific rehabilitation exercises. Although NSAIDs, modalities, and corticosteroids may be helpful for short-term pain reduction and early recovery of function, they do not typically reverse the structural changes associated with degenerative conditions and may contribute to worse long-term outcomes by potentially interfering with tissue healing. Regenerative interventions, including platelet-rich plasma and mesenchymal stem cells, recently have been used to treat refractory painful conditions such as chronic tendinopathies because of the potential of these interventions to facilitate tissue healing. The future development of these regenerative techniques will require a variety of conditions to be met, including determining the most appropriate procedures based on the disease being treated; establishing the optimal preparations of these regenerative techniques; and providing clinicians, patients, and regulatory agencies with high-quality evidence demonstrating the safety, effectiveness, and long-term results of these treatments. Clarification of current regulatory uncertainty, improved access for all patients, proper training for clinicians who incorporate these techniques into their practice, and determination of the most appropriate postinjection protocols will allow physical medicine and rehabilitation specialists to play a unique role in the long-term management of patients with musculoskeletal and sports injuries. This article will also address the role physiatrists should have in the inevitable growth of regenerative medicine applications.
Collapse
Affiliation(s)
- Fernando Sepúlveda
- Department of Physical Medicine, Rehabilitation & Sports Medicine, Sports Medicine Fellowship Program, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Luis Baerga
- Department of Physical Medicine, Rehabilitation & Sports Medicine, Sports Medicine Fellowship Program, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - William Micheo
- Department of Physical Medicine, Rehabilitation & Sports Medicine, Sports Medicine Fellowship Program, University of Puerto Rico School of Medicine, San Juan, Puerto Rico; Department of Physical Medicine, Rehabilitation & Sports Medicine, Sports Medicine Fellowship Program, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| |
Collapse
|
44
|
Lewallen EA, Riester SM, Bonin CA, Kremers HM, Dudakovic A, Kakar S, Cohen RC, Westendorf JJ, Lewallen DG, van Wijnen AJ. Biological strategies for improved osseointegration and osteoinduction of porous metal orthopedic implants. TISSUE ENGINEERING PART B-REVIEWS 2014; 21:218-30. [PMID: 25348836 DOI: 10.1089/ten.teb.2014.0333] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The biological interface between an orthopedic implant and the surrounding host tissue may have a dramatic effect upon clinical outcome. Desired effects include bony ingrowth (osseointegration), stimulation of osteogenesis (osteoinduction), increased vascularization, and improved mechanical stability. Implant loosening, fibrous encapsulation, corrosion, infection, and inflammation, as well as physical mismatch may have deleterious clinical effects. This is particularly true of implants used in the reconstruction of load-bearing synovial joints such as the knee, hip, and the shoulder. The surfaces of orthopedic implants have evolved from solid-smooth to roughened-coarse and most recently, to porous in an effort to create a three-dimensional architecture for bone apposition and osseointegration. Total joint surgeries are increasingly performed in younger individuals with a longer life expectancy, and therefore, the postimplantation lifespan of devices must increase commensurately. This review discusses advancements in biomaterials science and cell-based therapies that may further improve orthopedic success rates. We focus on material and biological properties of orthopedic implants fabricated from porous metal and highlight some relevant developments in stem-cell research. We posit that the ideal primary and revision orthopedic load-bearing metal implants are highly porous and may be chemically modified to induce stem cell growth and osteogenic differentiation, while minimizing inflammation and infection. We conclude that integration of new biological, chemical, and mechanical methods is likely to yield more effective strategies to control and modify the implant-bone interface and thereby improve long-term clinical outcomes.
Collapse
|