151
|
Roato I, Alotto D, Belisario DC, Casarin S, Fumagalli M, Cambieri I, Piana R, Stella M, Ferracini R, Castagnoli C. Adipose Derived-Mesenchymal Stem Cells Viability and Differentiating Features for Orthopaedic Reparative Applications: Banking of Adipose Tissue. Stem Cells Int 2016; 2016:4968724. [PMID: 28018432 PMCID: PMC5153503 DOI: 10.1155/2016/4968724] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/24/2016] [Accepted: 11/02/2016] [Indexed: 01/05/2023] Open
Abstract
Osteoarthritis is characterized by loss of articular cartilage also due to reduced chondrogenic activity of mesenchymal stem cells (MSCs) from patients. Adipose tissue is an attractive source of MSCs (ATD-MSCs), representing an effective tool for reparative medicine, particularly for treatment of osteoarthritis, due to their chondrogenic and osteogenic differentiation capability. The treatment of symptomatic knee arthritis with ATD-MSCs proved effective with a single infusion, but multiple infusions could be also more efficacious. Here we studied some crucial aspects of adipose tissue banking procedures, evaluating ATD-MSCs viability, and differentiation capability after cryopreservation, to guarantee the quality of the tissue for multiple infusions. We reported that the presence of local anesthetic during lipoaspiration negatively affects cell viability of cryopreserved adipose tissue and cell growth of ATD-MSCs in culture. We observed that DMSO guarantees a faster growth of ATD-MSCs in culture than trehalose. At last, ATD-MSCs derived from fresh and cryopreserved samples at -80°C and -196°C showed viability and differentiation ability comparable to fresh samples. These data indicate that cryopreservation of adipose tissue at -80°C and -196°C is equivalent and preserves the content of ATD-MSCs in Stromal Vascular Fraction (SVF), guaranteeing the differentiation ability of ATD-MSCs.
Collapse
Affiliation(s)
- Ilaria Roato
- CeRMS, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Daniela Alotto
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | | | - Stefania Casarin
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Mara Fumagalli
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Irene Cambieri
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Raimondo Piana
- Department of Orthopaedic Oncology, CTO Hospital, Torino, Italy
| | - Maurizio Stella
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | | | - Carlotta Castagnoli
- Skin Bank, Department of General and Specialized Surgery, A.O.U. Città della Salute e della Scienza, Torino, Italy
| |
Collapse
|
152
|
Fekrazad R, Eslaminejad MB, Shayan AM, Kalhori KA, Abbas FM, Taghiyar L, Sepehr Pedram M, Ghuchani MS. Effects of Photobiomodulation and Mesenchymal Stem Cells on Articular Cartilage Defects in a Rabbit Model. Photomed Laser Surg 2016; 34:543-549. [DOI: 10.1089/pho.2015.4028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Reza Fekrazad
- Department of Periodontology, Dental Faculty, AJA University of Medical Sciences, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Arman M. Shayan
- Department of Orthodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Fatemeh Mashhadi Abbas
- Department of Oral & Maxillofacial Pathology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mir Sepehr Pedram
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mostafa Sadeghi Ghuchani
- Department of Orthodontics, Gorgan Faculty of Dentistry, Golestan University of Medical Sciences, Golestan, Iran
| |
Collapse
|
153
|
Upregulation of miR-98 Inhibits Apoptosis in Cartilage Cells in Osteoarthritis. Genet Test Mol Biomarkers 2016; 20:645-653. [DOI: 10.1089/gtmb.2016.0011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
154
|
Ondrésik M, Azevedo Maia FR, da Silva Morais A, Gertrudes AC, Dias Bacelar AH, Correia C, Gonçalves C, Radhouani H, Amandi Sousa R, Oliveira JM, Reis RL. Management of knee osteoarthritis. Current status and future trends. Biotechnol Bioeng 2016; 114:717-739. [DOI: 10.1002/bit.26182] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 07/13/2016] [Accepted: 09/09/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Marta Ondrésik
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
| | - Fatima R. Azevedo Maia
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
| | - Alain da Silva Morais
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Ana C. Gertrudes
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Ana H. Dias Bacelar
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Cristina Correia
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Cristiana Gonçalves
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Hajer Radhouani
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Rui Amandi Sousa
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Joaquim M. Oliveira
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
| | - Rui L. Reis
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
| |
Collapse
|
155
|
Bielli A, Scioli MG, Gentile P, Cervelli V, Orlandi A. Adipose-derived stem cells in cartilage regeneration: current perspectives. Regen Med 2016; 11:693-703. [PMID: 27599358 DOI: 10.2217/rme-2016-0077] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Repair of cartilage injuries represents a musculoskeletal medicine criticism because of the poor ability to self-renewal of adult cartilage. Therefore, research focuses on developing new regenerative strategies combining chondrocytes or stem cells, scaffolds and growth factors. Because of the low proliferation capability of explanted chondrocytes, new chondrogenesis models, employing human adipose-derived stem cells (ASCs), have been investigated. ASCs are readily accessible with no morbidity and display the capability to differentiate into several cell lineages, including the spontaneous chondrogenic differentiation when entrapped in collagen gel scaffolds. Recent studies also defined some biomolecular mechanisms involved in ASC chondrogenesis in vitro, and their regenerative properties in bioengineered scaffolds and in the presence of growth factors. However, further investigations are required to validate these exciting preclinical results for the application of bioenginereed ASCs in the clinical practice.
Collapse
Affiliation(s)
- Alessandra Bielli
- Anatomic Pathology, Department of Biomedicine & Prevention, Tor Vergata University of Rome, Italy
| | - Maria Giovanna Scioli
- Anatomic Pathology, Department of Biomedicine & Prevention, Tor Vergata University of Rome, Italy
| | - Pietro Gentile
- Plastic Surgery, Department of Biomedicine & Prevention, Tor Vergata University of Rome, Italy
| | - Valerio Cervelli
- Plastic Surgery, Department of Biomedicine & Prevention, Tor Vergata University of Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine & Prevention, Tor Vergata University of Rome, Italy
| |
Collapse
|
156
|
Gugjoo MB, Amarpal, Sharma GT, Aithal HP, Kinjavdekar P. Cartilage tissue engineering: Role of mesenchymal stem cells along with growth factors & scaffolds. Indian J Med Res 2016; 144:339-347. [PMID: 28139532 PMCID: PMC5320839 DOI: 10.4103/0971-5916.198724] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Indexed: 01/13/2023] Open
Abstract
Articular cartilage injury poses a major challenge for both the patient and orthopaedician. Articular cartilage defects once formed do not regenerate spontaneously, rather replaced by fibrocartilage which is weaker in mechanical competence than the normal hyaline cartilage. Mesenchymal stem cells (MSCs) along with different growth factors and scaffolds are currently incorporated in tissue engineering to overcome the deficiencies associated with currently available surgical methods and to facilitate cartilage healing. MSCs, being readily available with a potential to differentiate into chondrocytes which are enhanced by the application of different growth factors, are considered for effective repair of articular cartilage after injury. However, therapeutic application of MSCs and growth factors for cartilage repair remains in its infancy, with no comparative clinical study to that of the other surgical techniques. The present review covers the role of MSCs, growth factors and scaffolds for the repair of articular cartilage injury.
Collapse
Affiliation(s)
- M. B. Gugjoo
- Division of Surgery, Modular Laboratory Building, Indian Veterinary Research Institute, Izatnagar, India
- Clinical Veterinary Services Complex, Faculty of Veterinary Sciences & Animal Husbandry, Shuhama, Sher-e-Kashmir University of Agricultural Sciences & Technology, Srinagar, India
| | - Amarpal
- Division of Surgery, Modular Laboratory Building, Indian Veterinary Research Institute, Izatnagar, India
| | - G. T. Sharma
- Division of Physiology & Climatology, Indian Veterinary Research Institute, Izatnagar, India
| | - H. P. Aithal
- Division of Surgery, Modular Laboratory Building, Indian Veterinary Research Institute, Izatnagar, India
| | - P. Kinjavdekar
- Division of Surgery, Modular Laboratory Building, Indian Veterinary Research Institute, Izatnagar, India
| |
Collapse
|
157
|
Nguyen PD, Tran TD, Nguyen HT, Vu HT, Le PT, Phan NL, Vu NB, Phan NK, Van Pham P. Comparative Clinical Observation of Arthroscopic Microfracture in the Presence and Absence of a Stromal Vascular Fraction Injection for Osteoarthritis. Stem Cells Transl Med 2016; 6:187-195. [PMID: 28170179 PMCID: PMC5442736 DOI: 10.5966/sctm.2016-0023] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 07/28/2016] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative cartilage disease that is characterized by a local inflammatory reaction. Consequently, many studies have been performed to identify suitable prevention and treatment interventions. In recent years, both arthroscopic microfracture (AM) and stem cell therapy have been used clinically to treat OA. This study aimed to evaluate the clinical effects of AM in the presence and absence of a stromal vascular fraction (SVF) injection in the management of patients with OA. Thirty patients with grade 2 or 3 (Lawrence scale) OA of the knee participated in this study. Placebo group patients (n = 15) received AM alone; treatment group patients (n = 15) received AM and an adipose tissue‐derived SVF injection. The SVF was suspended in platelet‐rich plasma (PRP) before injection into the joint. Patient groups were monitored and scored with the Western Ontario and McMaster Universities Arthritis Index (WOMAC), Lysholm, Visual Analog Pain Scale (VAS), and modified Outerbridge classifications before treatment and at 6, 12, and 18 months post‐treatment. Bone marrow edema was also assessed at these time points. Patients were evaluated for knee activity (joint motion amplitude) and adverse effects relating to surgery and stem cell injection. Treatment efficacy was significantly different between placebo and treatment groups. All treatment group patients had significantly reduced pain and WOMAC scores, and increased Lysholm and VAS scores compared with the placebo group. These findings suggest that the SVF/PRP injection efficiently improved OA for 18 months after treatment. This study will be continuously monitored for additional 24 months. Stem Cells Translational Medicine2017;6:187–195
Collapse
Affiliation(s)
| | | | | | | | | | - Nhan Lu‐Chinh Phan
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Ngoc Bich Vu
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Ngoc Kim Phan
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Phuc Van Pham
- Laboratory of Stem Cell Research and Application, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| |
Collapse
|
158
|
Phull AR, Eo SH, Abbas Q, Ahmed M, Kim SJ. Applications of Chondrocyte-Based Cartilage Engineering: An Overview. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1879837. [PMID: 27631002 PMCID: PMC5007317 DOI: 10.1155/2016/1879837] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 06/24/2016] [Accepted: 06/26/2016] [Indexed: 12/31/2022]
Abstract
Chondrocytes are the exclusive cells residing in cartilage and maintain the functionality of cartilage tissue. Series of biocomponents such as different growth factors, cytokines, and transcriptional factors regulate the mesenchymal stem cells (MSCs) differentiation to chondrocytes. The number of chondrocytes and dedifferentiation are the key limitations in subsequent clinical application of the chondrocytes. Different culture methods are being developed to overcome such issues. Using tissue engineering and cell based approaches, chondrocytes offer prominent therapeutic option specifically in orthopedics for cartilage repair and to treat ailments such as tracheal defects, facial reconstruction, and urinary incontinence. Matrix-assisted autologous chondrocyte transplantation/implantation is an improved version of traditional autologous chondrocyte transplantation (ACT) method. An increasing number of studies show the clinical significance of this technique for the chondral lesions treatment. Literature survey was carried out to address clinical and functional findings by using various ACT procedures. The current study was conducted to study the pharmacological significance and biomedical application of chondrocytes. Furthermore, it is inferred from the present study that long term follow-up studies are required to evaluate the potential of these methods and specific positive outcomes.
Collapse
Affiliation(s)
- Abdul-Rehman Phull
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongjudaehakro 56, Gongju 32588, Republic of Korea
| | - Seong-Hui Eo
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongjudaehakro 56, Gongju 32588, Republic of Korea
| | - Qamar Abbas
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongjudaehakro 56, Gongju 32588, Republic of Korea
| | - Madiha Ahmed
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Song Ja Kim
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongjudaehakro 56, Gongju 32588, Republic of Korea
| |
Collapse
|
159
|
Burke J, Hunter M, Kolhe R, Isales C, Hamrick M, Fulzele S. Therapeutic potential of mesenchymal stem cell based therapy for osteoarthritis. Clin Transl Med 2016; 5:27. [PMID: 27510262 PMCID: PMC4980326 DOI: 10.1186/s40169-016-0112-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/28/2016] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease affecting articular cartilage in joints, and it is a leading cause of disability in the United States. Current pharmacological treatment strategies are ineffective to prevent the OA progression; however, cellular therapies have the potential to regenerate the lost cartilage, combat cartilage degeneration, provide pain relief, and improve patient mobility. One of the most promising sources of cellular regenerative medicine is from mesenchymal stem cells (MSCs). MSCs can be isolated from adipose tissue, bone marrow, synovial tissue, and other sources. The aim of this review is to compile recent advancement in cellular based therapy more specifically in relation to MSCs in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- John Burke
- Department of Orthopedics, Georgia Regents University, Augusta, GA, USA
| | - Monte Hunter
- Department of Orthopedics, Georgia Regents University, Augusta, GA, USA
| | - Ravindra Kolhe
- Department of Pathology, Georgia Regents University, Augusta, GA, USA
| | - Carlos Isales
- Department of Orthopedics, Georgia Regents University, Augusta, GA, USA.,Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA
| | - Mark Hamrick
- Department of Cell Biology and Anatomy, Georgia Regents University, Augusta, GA, USA.,Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Orthopedics, Georgia Regents University, Augusta, GA, USA. .,Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA. .,Department of Orthopedics Surgery, Augusta University, Augusta, GA, 30904, USA.
| |
Collapse
|
160
|
Maguire T, Davis M, Marrero-Berrios I, Zhu C, Gaughan C, Weinberg J, Manchikalapati D, SchianodiCola J, Kamath H, Schloss R, Yarmush J. Control Release Anesthetics to Enable an Integrated Anesthetic-mesenchymal Stromal Cell Therapeutic. INTERNATIONAL JOURNAL OF ANESTHESIOLOGY & PAIN MEDICINE 2016; 2:3. [PMID: 31106286 PMCID: PMC6519947 DOI: 10.21767/2471-982x.100012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
While general anesthetics control pain via consciousness regulation, local anesthetics (LAs) act by decreasing sensation in the localized area of administration by blocking nerve transmission to pain centers. Perioperative intra-articular administration of LAs is a commonly employed practice in orthopedic procedures to minimize patient surgical and post-surgical pain and discomfort. LAs are also co-administered with cellular mesenchymal stromal cell (MSC) therapies for a variety of tissue regenerative and inflammatory applications including osteoarthritis (OA) treatment; however, LAs can affect MSC viability and function. Therefore, finding an improved method to co-administer LAs with cells has become critically important. We have developed a sustained release LA delivery model that could enable the co-administration of LAs and MSCs. Encapsulation of liposomes within an alginate matrix leads to sustained release of bupivacaine as compared to bupivacaine-containing liposomes alone. Furthermore, drug release is maintained for a minimum of 4 days and the alginate-liposome capsules mitigated the adverse effects of bupivacaine on MSC viability.
Collapse
Affiliation(s)
- T Maguire
- Rutgers Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- BeauRidge Pharmaceuticals, LLC, New York, USA
| | - M Davis
- Rutgers Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - I Marrero-Berrios
- Rutgers Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - C Zhu
- Rutgers Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - C Gaughan
- BeauRidge Pharmaceuticals, LLC, New York, USA
| | - J Weinberg
- Department of Anesthesiology, New York Methodist Hospital, Brooklyn, New York, USA
| | - D Manchikalapati
- Department of Anesthesiology, New York Methodist Hospital, Brooklyn, New York, USA
| | - J SchianodiCola
- Department of Anesthesiology, New York Methodist Hospital, Brooklyn, New York, USA
| | - H Kamath
- Department of Anesthesiology, New York Methodist Hospital, Brooklyn, New York, USA
| | - R Schloss
- Rutgers Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - J Yarmush
- Department of Anesthesiology, New York Methodist Hospital, Brooklyn, New York, USA
| |
Collapse
|
161
|
Soler R, Orozco L, Munar A, Huguet M, López R, Vives J, Coll R, Codinach M, Garcia-Lopez J. Final results of a phase I-II trial using ex vivo expanded autologous Mesenchymal Stromal Cells for the treatment of osteoarthritis of the knee confirming safety and suggesting cartilage regeneration. Knee 2016; 23:647-54. [PMID: 26783191 DOI: 10.1016/j.knee.2015.08.013] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 08/11/2015] [Accepted: 08/13/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND Cellular therapies have shown encouraging results in the treatment of chronic osteoarthritis (OA). Herein, we present the final results of a phase I-II clinical trial assessing the feasibility, safety and efficacy of ex vivo expanded autologous bone marrow Mesenchymal Stromal Cells (MSC, XCEL-M-ALPHA), infused intra-articularly, in patients with knee OA. METHODS Fifteen patients (median age=52years) with grade II(9) or III(6) gonarthrosis (Kellgren & Lawrence classification) and chronic pain were treated with an intra-articular infusion of 40.9×10(6)±0.4×10(6) MSCin a phase I-II prospective, open-label, single-dose, single-arm clinical trial. Endpoints were safety and tolerability. Efficacy was measured by the Visual Analogue Scale for pain, algofunctional Health Assessment Questionnaire, Quality of Life (QoL) SF-36 questionnaire, Lequesne functional index and WOMAC score. Cartilage integrity was assessed by Magnetic Resonance Imaging and quantitative T2-mapping at 0, 6 and 12months. RESULTS The cell-based product was well tolerated with few reported Adverse Events (mild arthralgia and low back pain). There was a relevant decrease in the intensity of pain since day 8 after the infusion, that was maintained after 12months. The SF-36 QoL test showed improvement of parameters including bodily pain, role physical and physical functioning at month 12. The health assessment questionnaire revealed a significant decrease of incapacity. Moreover, T2 mapping showed signs of cartilage regeneration in all patients at 12months post-treatment. CONCLUSIONS Single intra-articular infusion of XCEL-M-ALPHA is a safe and well-tolerated cell-based product, associated with a long-lasting amelioration of pain, improvement of QoL (up to four years), and signs of cartilage repair.
Collapse
Affiliation(s)
- Robert Soler
- Institut de Teràpia Regenerativa Tissular (ITRT), Hospital Quirón Teknon, Carrer de Vilana, 12, 08022 Barcelona, Spain
| | - Lluis Orozco
- Institut de Teràpia Regenerativa Tissular (ITRT), Hospital Quirón Teknon, Carrer de Vilana, 12, 08022 Barcelona, Spain
| | - Ana Munar
- Institut de Teràpia Regenerativa Tissular (ITRT), Hospital Quirón Teknon, Carrer de Vilana, 12, 08022 Barcelona, Spain
| | - Marina Huguet
- Department of Magnetic Resonance Imaging, CETIR Clínica del Pilar, Carrer de Balmes, 271, 08006 Barcelona, Spain
| | - Ramon López
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Joaquim Vives
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Ruth Coll
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Margarita Codinach
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Joan Garcia-Lopez
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain; Chair of Transfusion Medicine and Cellular and Tissue Therapies, Universitat Autònoma de Barcelona, Campus UAB, Cerdanyola del Vallès, 08193 Bellaterra, Spain.
| |
Collapse
|
162
|
Mamidi MK, Das AK, Zakaria Z, Bhonde R. Mesenchymal stromal cells for cartilage repair in osteoarthritis. Osteoarthritis Cartilage 2016; 24:1307-16. [PMID: 26973328 DOI: 10.1016/j.joca.2016.03.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 02/09/2016] [Accepted: 03/03/2016] [Indexed: 02/08/2023]
Abstract
Treatment for articular cartilage damage is quite challenging as it shows limited repair and regeneration following injury. Non-operative and classical surgical techniques are inefficient in restoring normal anatomy and function of cartilage in osteoarthritis (OA). Thus, investigating new and effective strategies for OA are necessary to establish feasible therapeutic solutions. The emergence of the new discipline of regenerative medicine, having cell-based therapy as its primary focus, may enable us to achieve repair and restore the damaged articular cartilage. This review describes progress and development of employing mesenchymal stromal cell (MSC)-based therapy as a promising alternative for OA treatment. The objective of this review is to first, discuss how in vitro MSC chondrogenic differentiation mimics in vivo embryonic cartilage development, secondly, to describe various chondrogenic differentiation strategies followed by pre-clinical and clinical studies demonstrating their feasibility and efficacy. However, several challenges need to be tackled before this research can be translated to the clinics. In particular, better understanding of the post-transplanted cell behaviour and learning to enhance their potency in the disease microenvironment is essential. Final objective is to underscore the importance of isolation, storage, cell shipment, route of administration, optimum dosage and control batch to batch variations to realise the full potential of MSCs in OA clinical trials.
Collapse
Affiliation(s)
- M K Mamidi
- School of Regenerative Medicine, Manipal University, Bangalore 560065, India
| | - A K Das
- Department of Surgery, Taylor's University School of Medicine, Sungai Buloh Hospital, Selangor, Malaysia
| | - Z Zakaria
- Hematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588 Kuala Lumpur, Malaysia
| | - R Bhonde
- School of Regenerative Medicine, Manipal University, Bangalore 560065, India.
| |
Collapse
|
163
|
Malanga G, Abdelshahed D, Jayaram P. Orthobiologic Interventions Using Ultrasound Guidance. Phys Med Rehabil Clin N Am 2016; 27:717-31. [DOI: 10.1016/j.pmr.2016.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
164
|
Rodriguez-Merchan EC. Intra-articular injections of mesenchymal stem cells (MSCs) as a treatment for hemophilic arthropathy. Expert Rev Hematol 2016; 9:737-41. [DOI: 10.1080/17474086.2016.1203780] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
165
|
No significant effects of Poly(I:C) on human umbilical cord-derived mesenchymal stem cells in the treatment of B6.MRL-Fas(lpr) mice. Curr Res Transl Med 2016; 64:55-60. [PMID: 27316386 DOI: 10.1016/j.retram.2016.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/11/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Our study aimed to compare the curative effect and immunoregulation between MSCs activated by Poly(I:C) for 24hours and unactivated MSCs on lupus mice. MATERIALS AND METHODS MSCs were pretreated by Poly(I:C) at 50μg/mL for 24h. B6.MRL-Fas(lpr) mice were divided into UC-MSC treated group, FLS treated group, Poly(I:C) preconditioned MSC treated group (P-MSC) and untreated group randomly. All treated mice were infused with 1×10(6) MSCs or FLSs at the 24th week and were sacrificed 4 weeks later. The spleen weight, serum immunoglobulin G (IgG) levels, serum anti-double stranded DNA (anti-dsDNA) antibody levels, immune cell subsets, renal lesions and IgG deposition in the kidney were evaluated. The effects of two kinds of MSCs on the proliferation and apoptosis of CD4+ T cells were detected by flow cytometry. The TLR3 expression at protein level in MSCs was assessed with and without Poly(I:C) treatment. The expression of immunoregulatory factors were detected by qRT-PCR in different dose and duration of Poly(I:C). RESULT Poly(I:C) preconditioned MSCs had similar therapeutic effects in lupus mice compared with untreated MSCs in vivo. Furthermore, Poly(I:C) treated MSCs and untreated MSCs had comparable inhibitory effects on proliferation of T cells, and Poly(I:C) could enhance the expression of TLR3 at protein and mRNA level. Poly(I:C) could partly alter the mRNA levels of immunoregulatory factors, such as hepatocyte growth factor, transforming growth factor β1, vascular endothelial growth factor, but did not have significant changes in cyclooxygenase 2, interleukin 6, tumor necrosis factor α, indoleamine 2,3-dioxygenase, interferon γ and chemokine (C-C motif) ligand 2. CONCLUSION Our study did not find that Poly(I:C) treatment could enhance the therapeutic effect of MSCs in lupus mice in vivo.
Collapse
|
166
|
Freitag J, Bates D, Boyd R, Shah K, Barnard A, Huguenin L, Tenen A. Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy - a review. BMC Musculoskelet Disord 2016; 17:230. [PMID: 27229856 PMCID: PMC4880954 DOI: 10.1186/s12891-016-1085-9] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/17/2016] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis is a leading cause of pain and disability across the world. With an aging population its prevalence is likely to further increase. Current accepted medical treatment strategies are aimed at symptom control rather than disease modification. Surgical options including joint replacement are not without possible significant complications. A growing interest in the area of regenerative medicine, led by an improved understanding of the role of mesenchymal stem cells in tissue homeostasis and repair, has seen recent focused efforts to explore the potential of stem cell therapies in the active management of symptomatic osteoarthritis. Encouragingly, results of pre-clinical and clinical trials have provided initial evidence of efficacy and indicated safety in the therapeutic use of mesenchymal stem cell therapies for the treatment of knee osteoarthritis. This paper explores the pathogenesis of osteoarthritis and how mesenchymal stem cells may play a role in future management strategies of this disabling condition.
Collapse
Affiliation(s)
- Julien Freitag
- Melbourne Stem Cell Centre, Level 2, 116-118 Thames St, Box Hill North, VIC, 3128, Australia.
| | - Dan Bates
- Melbourne Stem Cell Centre, Level 2, 116-118 Thames St, Box Hill North, VIC, 3128, Australia
| | | | - Kiran Shah
- Magellan Stem Cells, Melbourne, Australia
| | | | - Leesa Huguenin
- Melbourne Stem Cell Centre, Level 2, 116-118 Thames St, Box Hill North, VIC, 3128, Australia
| | - Abi Tenen
- Monash University, Melbourne, Australia
| |
Collapse
|
167
|
Infrapatellar Fat Pad: An Alternative Source of Adipose-Derived Mesenchymal Stem Cells. ARTHRITIS 2016; 2016:4019873. [PMID: 27239342 PMCID: PMC4861778 DOI: 10.1155/2016/4019873] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/07/2016] [Accepted: 02/14/2016] [Indexed: 12/12/2022]
Abstract
Introduction. The Infrapatellar fat pad (IPFP) represents an emerging alternative source of adipose-derived mesenchymal stem cells (ASCs). We compared the characteristics and differentiation capacity of ASCs isolated from IPFP and SC. Materials and Methods. ASCs were harvested from either IPFP or SC. IPFPs were collected from patients undergoing total knee arthroplasty (TKA), whereas subcutaneous tissues were collected from patients undergoing lipoaspiration. Immunophenotypes of surface antigens were evaluated. Their ability to form colony-forming units (CFUs) and their differentiation potential were determined. The ASCs karyotype was evaluated. Results. There was no difference in the number of CFUs and size of CFUs between IPFP and SC sources. ASCs isolated from both sources had a normal karyotype. The mesenchymal stem cells (MSCs) markers on flow cytometry was equivalent. IPFP-ASCs demonstrated significantly higher expression of SOX-9 and RUNX-2 over ASCs isolated from SC (6.19 ± 5.56-, 0.47 ± 0.62-fold; p value = 0.047, and 17.33 ± 10.80-, 1.56 ± 1.31-fold; p value = 0.030, resp.). Discussion and Conclusion. CFU assay of IPFP-ASCs and SC-ASCs harvested by lipoaspiration technique was equivalent. The expression of key chondrogenic and osteogenic genes was increased in cells isolated from IPFP. IPFP should be considered a high quality alternative source of ASCs.
Collapse
|
168
|
Saeed H, Ahsan M, Saleem Z, Iqtedar M, Islam M, Danish Z, Khan AM. Mesenchymal stem cells (MSCs) as skeletal therapeutics - an update. J Biomed Sci 2016; 23:41. [PMID: 27084089 PMCID: PMC4833928 DOI: 10.1186/s12929-016-0254-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 04/03/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells hold the promise to treat not only several congenital and acquired bone degenerative diseases but also to repair and regenerate morbid bone tissues. Utilizing MSCs, several lines of evidences advocate promising clinical outcomes in skeletal diseases and skeletal tissue repair/regeneration. In this context, both, autologous and allogeneic cell transfer options have been utilized. Studies suggest that MSCs are transplanted either alone by mixing with autogenous plasma/serum or by loading onto repair/induction supportive resorb-able scaffolds. Thus, this review is aimed at highlighting a wide range of pertinent clinical therapeutic options of MSCs in the treatment of skeletal diseases and skeletal tissue regeneration. Additionally, in skeletal disease and regenerative sections, only the early and more recent preclinical evidences are discussed followed by all the pertinent clinical studies. Moreover, germane post transplant therapeutic mechanisms afforded by MSCs have also been conversed. Nonetheless, assertive use of MSCs in the clinic for skeletal disorders and repair is far from a mature therapeutic option, therefore, posed challenges and future directions are also discussed. Importantly, for uniformity at all instances, term MSCs is used throughout the review.
Collapse
Affiliation(s)
- Hamid Saeed
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan.
| | - Muhammad Ahsan
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan
| | - Zikria Saleem
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan
| | - Mehwish Iqtedar
- Department of Bio-technology, Lahore College for Women University, Lahore, Pakistan
| | - Muhammad Islam
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan
| | - Zeeshan Danish
- Section of Clinical Pharmacy, University College of Pharmacy, University of the Punjab, Allama Iqbal Campus, 54000, Lahore, Pakistan
| | - Asif Manzoor Khan
- Department of Biochemistry and Molecular Biology, University of the Southern Denmark, 5230, Odense, Denmark
| |
Collapse
|
169
|
Filardo G, Perdisa F, Roffi A, Marcacci M, Kon E. Stem cells in articular cartilage regeneration. J Orthop Surg Res 2016; 11:42. [PMID: 27072345 PMCID: PMC4830073 DOI: 10.1186/s13018-016-0378-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/29/2016] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising option to treat articular defects and early osteoarthritis (OA) stages. However, both their potential and limitations for a clinical use remain controversial. Thus, the aim of this systematic review was to examine MSCs treatment strategies in clinical settings, in order to summarize the current evidence of their efficacy for the treatment of cartilage lesions and OA.Among the 60 selected studies, 7 were randomized, 13 comparative, 31 case series, and 9 case reports; 26 studies reported the results after injective administration, whereas 33 used surgical implantation. One study compared the two different modalities. With regard to the cell source, 20 studies concerned BMSCs, 17 ADSCs, 16 BMC, 5 PBSCs, 1 SDSCs, and 1 compared BMC versus PBSCs. Overall, despite the increasing literature on this topic, the evidence is still limited, in particular for high-level studies. On the other hand, the available studies allow to draw some indications. First, no major adverse events related to the treatment or to the cell harvest have been reported. Second, a clinical benefit of using MSCs therapies has been reported in most of the studies, regardless of cell source, indication, or administration method. This effectiveness has been reflected by clinical improvements and also positive MRI and macroscopic findings, whereas histologic features gave more controversial results among different studies. Third, young age, lower BMI, smaller lesion size for focal lesions, and earlier stages of OA joints have been shown to correlate with better outcomes, even though the available data strength does not allow to define clear cutoff values. Finally, definite trends can be observed with regard to the delivery method: currently cultured cells are mostly being administered by i.a. injection, while one-step surgical implantation is preferred for cell concentrates. In conclusion, while promising results have been shown, the potential of these treatments should be confirmed by reliable clinical data through double-blind, controlled, prospective and multicenter studies with longer follow-up, and specific studies should be designed to identify the best cell sources, manipulation, and delivery techniques, as well as pathology and disease phase indications.
Collapse
Affiliation(s)
- Giuseppe Filardo
- />II Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Francesco Perdisa
- />II Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Alice Roffi
- />Nanobiotechnology Laboratory, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Maurilio Marcacci
- />II Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
- />Nanobiotechnology Laboratory, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Elizaveta Kon
- />II Orthopaedic and Traumatologic Clinic, Rizzoli Orthopaedic Institute, Bologna, Italy
- />Nanobiotechnology Laboratory, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| |
Collapse
|
170
|
Zhang W, Ouyang H, Dass CR, Xu J. Current research on pharmacologic and regenerative therapies for osteoarthritis. Bone Res 2016; 4:15040. [PMID: 26962464 PMCID: PMC4772471 DOI: 10.1038/boneres.2015.40] [Citation(s) in RCA: 310] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 12/05/2015] [Accepted: 12/06/2015] [Indexed: 01/05/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disorder commonly encountered in clinical practice, and is the leading cause of disability in elderly people. Due to the poor self-healing capacity of articular cartilage and lack of specific diagnostic biomarkers, OA is a challenging disease with limited treatment options. Traditional pharmacologic therapies such as acetaminophen, non-steroidal anti-inflammatory drugs, and opioids are effective in relieving pain but are incapable of reversing cartilage damage and are frequently associated with adverse events. Current research focuses on the development of new OA drugs (such as sprifermin/recombinant human fibroblast growth factor-18, tanezumab/monoclonal antibody against β-nerve growth factor), which aims for more effectiveness and less incidence of adverse effects than the traditional ones. Furthermore, regenerative therapies (such as autologous chondrocyte implantation (ACI), new generation of matrix-induced ACI, cell-free scaffolds, induced pluripotent stem cells (iPS cells or iPSCs), and endogenous cell homing) are also emerging as promising alternatives as they have potential to enhance cartilage repair, and ultimately restore healthy tissue. However, despite currently available therapies and research advances, there remain unmet medical needs in the treatment of OA. This review highlights current research progress on pharmacologic and regenerative therapies for OA including key advances and potential limitations.
Collapse
Affiliation(s)
- Wei Zhang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China
- School of Pathology and Laboratory Medicine, The University of Western Australia, Perth WA 6009, Australia
| | - Hongwei Ouyang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Crispin R Dass
- School of Pharmacy, Building 306, Curtin University, Bentley, Perth WA 6102, Australia
| | - Jiake Xu
- School of Pathology and Laboratory Medicine, The University of Western Australia, Perth WA 6009, Australia
| |
Collapse
|
171
|
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]
|
172
|
Reissis D, Tang QO, Cooper NC, Carasco CF, Gamie Z, Mantalaris A, Tsiridis E. Current clinical evidence for the use of mesenchymal stem cells in articular cartilage repair. Expert Opin Biol Ther 2016; 16:535-57. [PMID: 26798997 DOI: 10.1517/14712598.2016.1145651] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Articular cartilage is renowned for its poor intrinsic capacity for repair. Current treatments for osteoarthritis are limited in their ability to reliably restore the native articular cartilage structure and function. Mesenchymal stem cells (MSCs) present an attractive treatment option for articular cartilage repair, with a recent expansion of clinical trials investigating their use in patients. AREAS COVERED This paper provides a current overview of the clinical evidence on the use of MSCs in articular cartilage repair. EXPERT OPINION The article demonstrates robust clinical evidence that MSCs have significant potential for the regeneration of hyaline articular cartilage in patients. The majority of clinical trials to date have yielded significantly positive results with minimal adverse effects. However the clinical research is still in its infancy. The optimum MSC source, cell concentrations, implantation technique, scaffold, growth factors and rehabilitation protocol for clinical use are yet to be identified. A larger number of randomised control trials are required to objectively compare the clinical efficacy and long-term safety of the various techniques. As the clinical research continues to evolve and address these challenges, it is likely that MSCs may become integrated into routine clinical practice in the near future.
Collapse
Affiliation(s)
- Dimitris Reissis
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Quen Oak Tang
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Nina Catherine Cooper
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Clare Francesca Carasco
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Zakareya Gamie
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Athanasios Mantalaris
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK
| | - Eleftherios Tsiridis
- a Department of Chemical Engineering, Biological Systems Engineering Laboratory (BSEL) , Imperial College London , London , UK.,b Academic Orthopaedic Unit , Aristotle University Medical School , Thessaloniki , Greece
| |
Collapse
|
173
|
Fodor PB, Paulseth SG. Adipose Derived Stromal Cell (ADSC) Injections for Pain Management of Osteoarthritis in the Human Knee Joint. Aesthet Surg J 2016; 36:229-36. [PMID: 26238455 DOI: 10.1093/asj/sjv135] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND This safety and feasibility study used autologous adipose-derived stromal vascular cells (the stromal vascular fraction [SVF] of adipose tissue), to treat 8 osteoarthritic knees in 6 patients of grade I to III (K-L scale) with initial pain of 4 or greater on a 10-point Visual Analog Scale (VAS). OBJECTIVES The primary objective of the study was evaluation of the safety of intra-articular injection of SVF. The secondary objective was to assess initial feasibility for reduction of pain in osteoarthritic knees. METHODS Adipose-derived SVF cells were obtained through enzymatic disaggregation of lipoaspirate, resuspension in 3 mL of Lactated Ringer's Solution, and injection directly into the intra-articular space of the knee, with a mean of 14.1 million viable, nucleated SVF cells per knee. Metrics included monitoring of adverse events and preoperative to postoperative changes in the Western Ontario and McMaster Universities Arthritis Index (WOMAC), the VAS pain scale, range of motion (ROM), timed up-and-go (TUG), and MRI. RESULTS No infections, acute pain flares, or other adverse events were reported. At 3-months postoperative, there was a statistically significant improvement in WOMAC and VAS scores (P < .02 and P < .001, respectively), which was maintained at 1 year. Physical therapy measurements for ROM and TUG both improved from preoperative to 3-months postoperative. Standard MRI assessment from preoperative to 3-months postoperative showed no detectable structural differences. All patients attained full activity with decreased knee pain. CONCLUSIONS Autologous SVF was shown to be safe and to present a new potential therapy for reduction of pain for osteoarthritis of the knee. LEVEL OF EVIDENCE 4: Therapeutic.
Collapse
Affiliation(s)
- Peter B Fodor
- Dr Fodor is an Associate Clinical Professor of Plastic Surgery, UCLA Medical Center, Los Angeles, California. Dr Paulseth is an Adjunct Instructor of Clinical Physical Therapy, Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, California
| | - Stephen G Paulseth
- Dr Fodor is an Associate Clinical Professor of Plastic Surgery, UCLA Medical Center, Los Angeles, California. Dr Paulseth is an Adjunct Instructor of Clinical Physical Therapy, Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, California
| |
Collapse
|
174
|
Onishi K, Jones DL, Riester SM, Lewallen EA, Lewallen DG, Sellon JL, Dietz AB, Qu W, van Wijnen AJ, Smith J. Human Adipose-Derived Mesenchymal Stromal/Stem Cells Remain Viable and Metabolically Active Following Needle Passage. PM R 2016; 8:844-54. [PMID: 26826615 DOI: 10.1016/j.pmrj.2016.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/10/2016] [Accepted: 01/20/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To assess the biological effects of passage through clinically relevant needles on the viability and metabolic activity of culture-expanded, human adipose tissue-derived mesenchymal stromal/stem cells (AMSCs). DESIGN Prospective observational pilot study. SETTING Academic medical center. PARTICIPANTS Patient-derived clinical-grade culture expanded AMSCs. INTERVENTIONS AMSCs were passed through syringes without a needle attached (control), with an 18-gauge (25.4-mm) needle attached and with a 30-gauge (19-mm) needle attached at a constant injection flow rate and constant cell concentrations. Each injection condition was completed in triplicate. MAIN OUTCOME MEASURES Cell number and viability, proliferative capacity, metabolic activity, and acute gene expression as measured by cell counts, mitochondrial activity, and quantitative real time reverse-transcription polymerase chain reaction on day 0 (immediately), day 1, and day 4 after injection. RESULTS AMSC viability was not significantly affected by injection, and cells proliferated normally regardless of study group. Postinjection, AMSCs robustly expressed both proliferation markers and extracellular matrix proteins. Stress-response mRNAs were markedly but transiently increased independently of needle size within the first day in culture postinjection. CONCLUSIONS Human, culture-expanded AMSCs maintain their viability, proliferative capacity, and metabolic function following passage through needles as small as 30-gauge at constant flow rates of 4 mL/min, despite an early, nonspecific stress/cytoprotective response. These initial findings suggest that culture-expanded AMSCs should tolerate the injection process during most cell-based therapeutic interventions.
Collapse
Affiliation(s)
- Kentaro Onishi
- Department of Physical Medicine & Rehabilitation, Mayo Clinic Sports Medicine Center, Mayo Clinic, Rochester, MN(∗)
| | - Dakota L Jones
- Department of Biomedical Engineering and Physiology, Mayo Graduate School, Mayo Clinic, Rochester, MN; Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(†)
| | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(‡)
| | - Eric A Lewallen
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(§)
| | - David G Lewallen
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(‖)
| | - Jacob L Sellon
- Department of Physical Medicine & Rehabilitation, Mayo Clinic Sports Medicine Center, Mayo Clinic, Rochester, MN(¶)
| | - Allan B Dietz
- Department of Biochemistry & Molecular Biology, Mayo Graduate School, Mayo Clinic, Rochester, MN; Department of Laboratory Medicine & Pathology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(#)
| | - Wenchun Qu
- Department of Physical Medicine & Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN; Department of Anesthesiology Division of Pain Medicine, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(∗∗)
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Medical Sciences Building, Rm S3-69, Mayo Clinic, 200 1st St, SW, Rochester, MN 55905; Department of Biomedical Engineering and Physiology, Mayo Graduate School, Mayo Clinic, Rochester, MN; Department of Biochemistry & Molecular Biology, Mayo Graduate School, Mayo Clinic, Rochester, MN(††).
| | - Jay Smith
- Department of Physical Medicine & Rehabilitation, W14, Mayo Building, Mayo Clinic, 200 1st St, SW, Rochester, MN 55905; Department of Radiology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN; Department of Anatomy, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(‡‡).
| |
Collapse
|
175
|
Gao F, Chiu SM, Motan DAL, Zhang Z, Chen L, Ji HL, Tse HF, Fu QL, Lian Q. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis 2016; 7:e2062. [PMID: 26794657 PMCID: PMC4816164 DOI: 10.1038/cddis.2015.327] [Citation(s) in RCA: 772] [Impact Index Per Article: 96.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/13/2015] [Accepted: 09/25/2015] [Indexed: 12/11/2022]
Abstract
The unique immunomodulatory properties of mesenchymal stem cells (MSCs) make them an invaluable cell type for the repair of tissue/ organ damage caused by chronic inflammation or autoimmune disorders. Although they hold great promise in the treatment of immune disorders such as graft versus host disease (GvHD) and allergic disorders, there remain many challenges to overcome before their widespread clinical application. An understanding of the biological properties of MSCs will clarify the mechanisms of MSC-based transplantation for immunomodulation. In this review, we summarize the preclinical and clinical studies of MSCs from different adult tissues, discuss the current hurdles to their use and propose the future development of pluripotent stem cell-derived MSCs as an approach to immunomodulation therapy.
Collapse
Affiliation(s)
- F Gao
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - S M Chiu
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - D A L Motan
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Z Zhang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - L Chen
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - H-L Ji
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| | - H-F Tse
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Q-L Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Q Lian
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.,Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
176
|
Afizah H, Hui JHP. Mesenchymal stem cell therapy for osteoarthritis. J Clin Orthop Trauma 2016; 7:177-82. [PMID: 27489413 PMCID: PMC4949411 DOI: 10.1016/j.jcot.2016.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 02/08/2023] Open
Abstract
The versatility of mesenchymal stem cells (MSCs) as a treatment modality has landed it another repair target: osteoarthritis, a crippling cartilage disease that frequently afflicts the aged population. Through many studies, this newly discovered method has been shown to significantly alleviate the pain experienced by osteoarthritic patients. Notwithstanding the effectiveness of MSCs in this regard, varying degrees of success rates have also been reported, which is probably attributable to the different approaches adopted in harnessing MSCs' therapeutic value. Accordingly, it is pertinent to understand the contributory factors like MSC type, dosage, size of osteoarthritic lesion, MSC carrier, and mode of infusion, which would be briefly discussed in this review.
Collapse
Affiliation(s)
- Hassan Afizah
- Department of Orthopaedic Surgery, National University Health System, Yong Loo Lin School of Medicine, Singapore,Tissue Engineering Programme, Department of Orthopaedic Surgery, Life Sciences Institute, National University of Singapore, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, National University Health System, Yong Loo Lin School of Medicine, Singapore,Tissue Engineering Programme, Department of Orthopaedic Surgery, Life Sciences Institute, National University of Singapore, Singapore,Corresponding author at: Department of Orthopaedic Surgery, National University of Singapore, NUHS Tower Block, Level 11, IE Kent Ridge Road, Singapore 119228, Singapore. Tel.: +65 67724321.Department of Orthopaedic Surgery, National University of SingaporeNUHS Tower Block, Level 11, IE Kent Ridge RoadSingapore119228Singapore
| |
Collapse
|
177
|
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]
|
178
|
Kasir R, Vernekar VN, Laurencin CT. Regenerative Engineering of Cartilage Using Adipose-Derived Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2015; 1:42-49. [PMID: 26998511 PMCID: PMC4795960 DOI: 10.1007/s40883-015-0005-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/19/2015] [Indexed: 12/13/2022]
Abstract
Injury to the articular cartilage occurs commonly in the general population and undergoes minimal spontaneous healing. Traditional methods of cartilage repair provide no long-term cure and are significant causes of morbidity. For this reason, stem cell therapies have recently been investigated for their ability to regenerate cartilage, and the results have been promising. Since the discovery that adipose tissue is a major source of mesenchymal stem cells in 2001, scientists have been studying the use of adipose-derived stem cells (ASCs) for the treatment of various disorders including lesions of the articular cartilage. ASCs hold several advantages over autologous chondrocytes for cartilage repair, including but not limited to their anti-inflammatory effects, their multi-lineage differentiation potential, and their ability to form new cartilage in a defect. Whereas several investigations have been made in in vitro and animal models, there have been surprisingly little clinical studies on the intra-articular use of adipose-derived stem cells, despite their first isolation about a decade and a half ago. The few studies that have been conducted are encouraging. With approval for various stem cell therapies on the horizon, this review seeks to update the clinician and the researcher on the current state-of-the-art use of adipose-derived stem cells for the treatment of cartilage disorders and the regenerative engineering of cartilaginous tissue.
Collapse
Affiliation(s)
- Rafid Kasir
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Varadraj N. Vernekar
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Cato T. Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
179
|
Effect of the direct injection of bone marrow mesenchymal stem cells in hyaluronic acid and bone marrow stimulation to treat chondral defects in the canine model. Regen Ther 2015; 2:42-48. [PMID: 31245458 PMCID: PMC6581783 DOI: 10.1016/j.reth.2015.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/19/2015] [Accepted: 10/29/2015] [Indexed: 01/22/2023] Open
Abstract
Introduction The purpose of this study was to assess the direct injection of bone marrow-derived mesenchymal stem cells (BMSCs) suspended in hyaluronic acid (HA) combined with drilling as a treatment for chondral defects in a canine model. Methods Tibial bone marrow was aspirated, and BMSCs were isolated and cultured. One 8.0-mm diameter chondral defect was created in the femoral groove, and nine 0.9-mm diameter holes were drilled into the defect. BMSCs (2.14 × 107 cells) suspended in HA were injected into the defect. HA alone was injected into a similar defect on the contralateral knee as a control. Animals were sacrificed at 3 and 6 months. Results Although the percentage of coverage assessed macroscopically was significantly better at 6 months than at 3 months in both the BMSC (p = 0.02) and control (p = 0.001) groups, there were no significant differences in the International Cartilage Repair Society grades. The Wakitani histological score was significantly better at 6 months than at 3 months in the BMSC and control groups. While the control defects were mostly filled with fibrocartilage, several of the defects in the BMSC group contained hyaline-like cartilage. The mean Wakitani scores of the BMSC group improved from 7.0 ± 1.0 at 3 months to 4.6 ± 0.9 at 6 months, and those of the control group improved from 9.4 ± 1.2 to 6.0 ± 0.6. The BMSC group showed significantly better regeneration than the control group at 3 months (p = 0.04), but the difference at 6 months was not significant (p = 0.06). Conclusions The direct injection of BMSCs in HA combined with drilling enhanced cartilage regeneration.
Collapse
|
180
|
Delling U, Brehm W, Ludewig E, Winter K, Jülke H. Longitudinal Evaluation of Effects of Intra-Articular Mesenchymal Stromal Cell Administration for the Treatment of Osteoarthritis in an Ovine Model. Cell Transplant 2015; 24:2391-407. [DOI: 10.3727/096368915x686193] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In this study, the therapeutic effect of intra-articularly injected autologous mesenchymal stromal cells (MSCs) was evaluated in an ovine osteoarthritis (OA) model using consecutive magnetic resonance imaging (MRI), radiography, and macroscopic and histologic postmortem examination. In 12 sheep, OA was induced by bilateral, lateral meniscectomy. After 6 weeks, 20 × 106 bone marrow-derived MSCs (50% MSCs were superparamagnetic iron oxide particle labeled) were injected intra-articularly into one knee joint. The contralateral knee served as negative control. MR images were acquired before OA induction, immediately before and after MSC injection, and 1, 4, 8, and 12 weeks thereafter using a 0.5T unit and a T2* gradient echo sequence. Radiographs were obtained before OA induction, at MSC injection, and 12 weeks thereafter. The MRI scoring system included articular cartilage, bone, joint capsule, and synovial fluid evaluation. The radiographic scoring system included the joint space and bone. Postmortem evaluation entailed macroscopic and histologic assessment. Longitudinal MRI revealed a significant deceleration of OA progression in MSC-treated joints. However, at the conclusion of the study, there was no significant difference in the degree of OA detected by MRI, radiography, and postmortem evaluation between the treatment and control group. The degree of OA on MRI varied among the 12 animals at the time of injection, but there was no difference between the left and right limb. In conclusion, intra-articular MSCs decreased OA progression. However, no significant treatment effects were seen at the conclusion of the study at 12 weeks. This somewhat contradicts previously published results. Nevertheless, the choice of OA model, outcome measures, or lack of additional medication might explain the differences. Our results indicate that OA might benefit from intra-articular MSC injection, but further studies are needed to refine patient selection and injection parameters for a more substantially improved outcome.
Collapse
Affiliation(s)
- Uta Delling
- University of Leipzig, Faculty of Veterinary Medicine, Large Animal Clinic for Surgery, Leipzig, Germany
| | - Walter Brehm
- University of Leipzig, Faculty of Veterinary Medicine, Large Animal Clinic for Surgery, Leipzig, Germany
| | - Eberhard Ludewig
- University of Leipzig, Faculty of Veterinary Medicine, Department of Small Animal Medicine, Leipzig, Germany
| | - Karsten Winter
- University of Leipzig, Translational Centre for Regenerative Medicine (TRM), Leipzig, Germany
| | - Henriette Jülke
- University of Leipzig, Translational Centre for Regenerative Medicine (TRM), Leipzig, Germany
| |
Collapse
|
181
|
Kim YS, Kwon OR, Choi YJ, Suh DS, Heo DB, Koh YG. Comparative Matched-Pair Analysis of the Injection Versus Implantation of Mesenchymal Stem Cells for Knee Osteoarthritis. Am J Sports Med 2015; 43:2738-46. [PMID: 26337418 DOI: 10.1177/0363546515599632] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The mesenchymal stem cell (MSC)-based tissue engineering approach has been developed to address the problem of articular cartilage repair in knee osteoarthritis (OA). However, the most effective method of MSC application has not yet been established. PURPOSE To compare the injection and implantation of MSCs in patients with knee OA in terms of clinical and second-look arthroscopic outcomes. STUDY DESIGN Cohort study; Level of evidence, 3. METHODS Among 182 patients treated with arthroscopic surgery using MSCs for knee OA from October 2010 to August 2012, patients treated with an injection of MSCs in combination with platelet-rich plasma (injection group; n = 20) were pair-matched with patients who underwent MSC implantation on a fibrin glue scaffold (implantation group; n = 20) based on sex, age, and lesion size. Clinical outcomes were evaluated using the International Knee Documentation Committee (IKDC) score and Tegner activity scale, and cartilage repair was assessed arthroscopically with the International Cartilage Repair Society (ICRS) grading system. RESULTS The mean (±SD) IKDC and Tegner activity scores significantly improved from 38.5 ± 9.2 to 55.2 ± 15.0 and from 2.5 ± 1.2 to 3.5 ± 1.2, respectively, in the injection group and from 36.6 ± 4.9 to 62.7 ± 14.1 and from 2.3 ± 0.9 to 3.6 ± 1.1, respectively, in the implantation group at the time of second-look arthroscopic surgery (mean, 12.6 months postoperatively) (P < .001 in all cases). At final follow-up (mean, 28.6 months postoperatively), the mean IKDC and Tegner activity scores in the implantation group had improved further to 64.8 ± 13.4 and 3.9 ± 1.0, respectively (P < .001 and P = .035, respectively), while no significant improvements were found in the injection group (P = .130 and P = .655, respectively). At final follow-up, there was a significant difference in the mean IKDC score between groups (P = .049). Significant correlations between the number of administered MSCs and the postoperative clinical outcomes were found only in the injection group. Significant correlations between the clinical outcomes and the ICRS grades were found in both groups. The ICRS grades were significantly better in the implantation group (P = .041). In the injection group, 2 of the 20 lesions (10%) were grade I (normal), 5 (25%) were grade II (near normal), 8 (40%) were grade III (abnormal), and 5 (25%) were grade IV (severely abnormal). In the implantation group, 6 of the 20 lesions (30%) were grade I, 7 (35%) were grade II, 4 (20%) were grade III, and 3 (15%) were grade IV. CONCLUSION Utilizing the described method, MSC implantation for knee OA resulted in better clinical and second-look arthroscopic outcomes than an MSC injection.
Collapse
Affiliation(s)
- Yong Sang Kim
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Korea
| | - Oh Ryong Kwon
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Korea
| | - Yun Jin Choi
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Korea
| | - Dong Suk Suh
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Korea
| | - Dong Beom Heo
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Korea
| | - Yong Gon Koh
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Korea
| |
Collapse
|
182
|
Pers YM, Ruiz M, Noël D, Jorgensen C. Mesenchymal stem cells for the management of inflammation in osteoarthritis: state of the art and perspectives. Osteoarthritis Cartilage 2015; 23:2027-35. [PMID: 26521749 DOI: 10.1016/j.joca.2015.07.004] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 06/29/2015] [Accepted: 07/07/2015] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is the most common form of degenerative arthritis, mainly characterized by the degradation of articular cartilage and associated with subchondral bone lesions. Novel therapeutic approaches for OA include cell-based therapies that have become thriving areas of research and development. In this context, mesenchymal stem or stromal cells (MSCs) have gained much interest based on their trophic and immunomodulatory properties that can help tissue repair/regeneration. The present review article discusses the interest of using MSCs in cell-therapy approaches with a focus on the mechanisms by which MSCs might exhibit a therapeutic potential in OA. Special attention is given to the anti-inflammatory function of MSCs and on miRNA modulation in OA for possible future innovative strategies. The paper also presents the current data on the undergoing MSCs-based clinical trials in OA.
Collapse
Affiliation(s)
- Y-M Pers
- Inserm U1183, Hôpital Saint-Eloi, Montpellier, F-34295, France; Université Montpellier, UFR de Médecine, Montpellier, F-34000, France; Service d'immuno-Rhumatologie, Hôpital Lapeyronie, Montpellier, F-34295, France
| | - M Ruiz
- Inserm U1183, Hôpital Saint-Eloi, Montpellier, F-34295, France; Université Montpellier, UFR de Médecine, Montpellier, F-34000, France
| | - D Noël
- Inserm U1183, Hôpital Saint-Eloi, Montpellier, F-34295, France; Université Montpellier, UFR de Médecine, Montpellier, F-34000, France.
| | - C Jorgensen
- Inserm U1183, Hôpital Saint-Eloi, Montpellier, F-34295, France; Université Montpellier, UFR de Médecine, Montpellier, F-34000, France; Service d'immuno-Rhumatologie, Hôpital Lapeyronie, Montpellier, F-34295, France
| |
Collapse
|
183
|
Ruiz M, Cosenza S, Maumus M, Jorgensen C, Noël D. Therapeutic application of mesenchymal stem cells in osteoarthritis. Expert Opin Biol Ther 2015; 16:33-42. [PMID: 26413975 DOI: 10.1517/14712598.2016.1093108] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Osteoarthritis (OA) is a degenerative disease characterized by cartilage degradation and subchondral bone alterations. This disease represents a global public health problem whose prevalence is rapidly growing with the increasing aging of the population. With the discovery of mesenchymal stem cells (MSC) as possible therapeutic agents, their potential for repairing cartilage damage in OA is under investigation. AREAS COVERED Characterization of MSCs and their functional properties are mentioned with an insight into their trophic function and secretory profile. We present a special focus on the types of extracellular vesicles (EVs) that are produced by MSCs and their role in the paracrine activity of MSCs. We then discuss the therapeutic approaches that have been evaluated in pre-clinical models of OA and the results coming out from the clinical trials in patients with OA. EXPERT OPINION MSC-based therapy seems a promising approach for the treatment of patients with OA. Further research is still needed to demonstrate their efficacy in clinical trials using controlled, prospective studies. However, the emergence of MSC-derived EVs as possible therapeutic agents could be an alternative to cell-based therapy.
Collapse
Affiliation(s)
- Maxime Ruiz
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France
| | - Stella Cosenza
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France
| | - Marie Maumus
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France
| | - Christian Jorgensen
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France.,c 3 Hôpital Lapeyronie, Service d'immuno-Rhumatologie , Montpellier, F-34295, France
| | - Danièle Noël
- a 1 Inserm, U1183, CHRU Saint Eloi, Hôpital Saint-Eloi , 80 avenue Augustin Fliche, Montpellier, F-34295, France +33 4 67 33 04 73 ; +33 4 67 33 01 13 ; .,b 2 Université Montpellier, UFR de Médecine , Montpellier, F-34000, France.,c 3 Hôpital Lapeyronie, Service d'immuno-Rhumatologie , Montpellier, F-34295, France
| |
Collapse
|
184
|
Mesenchymal stem cells in regenerative medicine: Focus on articular cartilage and intervertebral disc regeneration. Methods 2015; 99:69-80. [PMID: 26384579 DOI: 10.1016/j.ymeth.2015.09.015] [Citation(s) in RCA: 320] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 08/10/2015] [Accepted: 09/15/2015] [Indexed: 01/15/2023] Open
Abstract
Musculoskeletal disorders represent a major cause of disability and morbidity globally and result in enormous costs for health and social care systems. Development of cell-based therapies is rapidly proliferating in a number of disease areas, including musculoskeletal disorders. Novel biological therapies that can effectively treat joint and spine degeneration are high priorities in regenerative medicine. Mesenchymal stem cells (MSCs) isolated from bone marrow (BM-MSCs), adipose tissue (AD-MSCs) and umbilical cord (UC-MSCs) show considerable promise for use in cartilage and intervertebral disc (IVD) repair. This review article focuses on stem cell-based therapeutics for cartilage and IVD repair in the context of the rising global burden of musculoskeletal disorders. We discuss the biology MSCs and chondroprogenitor cells and specifically focus on umbilical cord/Wharton's jelly derived MSCs and examine their potential for regenerative applications. We also summarize key components of the molecular machinery and signaling pathways responsible for the control of chondrogenesis and explore biomimetic scaffolds and biomaterials for articular cartilage and IVD regeneration. This review explores the exciting opportunities afforded by MSCs and discusses the challenges associated with cartilage and IVD repair and regeneration. There are still many technical challenges associated with isolating, expanding, differentiating, and pre-conditioning MSCs for subsequent implantation into degenerate joints and the spine. However, the prospect of combining biomaterials and cell-based therapies that incorporate chondrocytes, chondroprogenitors and MSCs leads to the optimistic view that interdisciplinary approaches will lead to significant breakthroughs in regenerating musculoskeletal tissues, such as the joint and the spine in the near future.
Collapse
|
185
|
D'souza N, Rossignoli F, Golinelli G, Grisendi G, Spano C, Candini O, Osturu S, Catani F, Paolucci P, Horwitz EM, Dominici M. Mesenchymal stem/stromal cells as a delivery platform in cell and gene therapies. BMC Med 2015; 13:186. [PMID: 26265166 PMCID: PMC4534031 DOI: 10.1186/s12916-015-0426-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 07/17/2015] [Indexed: 02/07/2023] Open
Abstract
Regenerative medicine relying on cell and gene therapies is one of the most promising approaches to repair tissues. Multipotent mesenchymal stem/stromal cells (MSC), a population of progenitors committing into mesoderm lineages, are progressively demonstrating therapeutic capabilities far beyond their differentiation capacities. The mechanisms by which MSC exert these actions include the release of biomolecules with anti-inflammatory, immunomodulating, anti-fibrogenic, and trophic functions. While we expect the spectra of these molecules with a therapeutic profile to progressively expand, several human pathological conditions have begun to benefit from these biomolecule-delivering properties. In addition, MSC have also been proposed to vehicle genes capable of further empowering these functions. This review deals with the therapeutic properties of MSC, focusing on their ability to secrete naturally produced or gene-induced factors that can be used in the treatment of kidney, lung, heart, liver, pancreas, nervous system, and skeletal diseases. We specifically focus on the different modalities by which MSC can exert these functions. We aim to provide an updated understanding of these paracrine mechanisms as a prerequisite to broadening the therapeutic potential and clinical impact of MSC.
Collapse
Affiliation(s)
- Naomi D'souza
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Filippo Rossignoli
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Giulia Golinelli
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Giulia Grisendi
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Carlotta Spano
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Olivia Candini
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Satoru Osturu
- The Division of Hematology/Oncology/BMT, Nationwide Children's Hospital, Departments of Pediatrics and Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Fabio Catani
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Paolo Paolucci
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Edwin M Horwitz
- The Division of Hematology/Oncology/BMT, Nationwide Children's Hospital, Departments of Pediatrics and Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy.
| |
Collapse
|
186
|
Use of Adult Stem Cells for Cartilage Tissue Engineering: Current Status and Future Developments. Stem Cells Int 2015; 2015:438026. [PMID: 26246809 PMCID: PMC4515346 DOI: 10.1155/2015/438026] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 03/16/2015] [Accepted: 03/17/2015] [Indexed: 02/06/2023] Open
Abstract
Due to their low self-repair ability, cartilage defects that result from joint injury, aging, or osteoarthritis, are the most often irreversible and are a major cause of joint pain and chronic disability. So, in recent years, researchers and surgeons have been working hard to elaborate cartilage repair interventions for patients who suffer from cartilage damage. However, current methods do not perfectly restore hyaline cartilage and may lead to the apparition of fibro- or hypertrophic cartilage. In the next years, the development of new strategies using adult stem cells, in scaffolds, with supplementation of culture medium and/or culture in low oxygen tension should improve the quality of neoformed cartilage. Through these solutions, some of the latest technologies start to bring very promising results in repairing cartilage from traumatic injury or chondropathies. This review discusses the current knowledge about the use of adult stem cells in the context of cartilage tissue engineering and presents clinical trials in progress, as well as in the future, especially in the field of bioprinting stem cells.
Collapse
|
187
|
|
188
|
Abstract
Elastin is the dominant mammalian elastic protein found in soft tissue. Elastin-based biomaterials have the potential to repair elastic tissues by improving local elasticity and providing appropriate cellular interactions and signaling. Studies that combine these biomaterials with mesenchymal stem cells have demonstrated their capacity to also regenerate non-elastic tissue. Mesenchymal stem cell differentiation can be controlled by their immediate environment, and their sensitivity to elasticity makes them an ideal candidate for combining with elastin-based biomaterials. With the growing accessibility of the elastin precursor, tropoelastin, and elastin-derived materials, the amount of research interest in combining these two fields has increased and, subsequently, is leading to the realization of a potentially new strategy for regenerative medicine.
Collapse
Affiliation(s)
- Jazmin Ozsvar
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia ; Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
| | - Suzanne M Mithieux
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia ; Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
| | - Richard Wang
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia ; Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
| | - Anthony S Weiss
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia ; Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
| |
Collapse
|
189
|
Grässel S, Lorenz J. Tissue-engineering strategies to repair chondral and osteochondral tissue in osteoarthritis: use of mesenchymal stem cells. Curr Rheumatol Rep 2015; 16:452. [PMID: 25182680 PMCID: PMC4182613 DOI: 10.1007/s11926-014-0452-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Focal chondral or osteochondral lesions can be painful and disabling because they have insufficient intrinsic repair potential, and constitute one of the major extrinsic risk factors for osteoarthritis (OA). Attention has, therefore, been paid to regenerative therapeutic procedures for the early treatment of cartilaginous defects. Current treatments for OA are not regenerative and have little effect on the progressive degeneration of joint tissue. One major reason for this underrepresentation of regenerative therapy is that approaches to treating OA with cell-based strategies have to take into consideration the larger sizes of the defects, as compared with isolated focal articular-cartilage defects, and the underlying disease process. Here, we review current treatment strategies using mesenchymal stem cells (MSCs) for chondral and osteochondral tissue repair in trauma and OA-affected joints. We discuss tissue-engineering approaches, in preclinical large-animal models and clinical studies in humans, which use crude bone-marrow aspirates and MSCs from different tissue sources in combination with bioactive agents and materials.
Collapse
Affiliation(s)
- Susanne Grässel
- Experimental Orthopedics, Centre for Medical Biotechnology, BioPark 1, Department of Orthopedic Surgery, University Hospital Regensburg, Regensburg, Germany,
| | | |
Collapse
|
190
|
Davatchi F, Sadeghi Abdollahi B, Mohyeddin M, Nikbin B. Mesenchymal stem cell therapy for knee osteoarthritis: 5 years follow-up of three patients. Int J Rheum Dis 2015; 19:219-25. [PMID: 25990685 DOI: 10.1111/1756-185x.12670] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIM Osteoarthritis is a degenerative joint disease characterized by the destruction of joint cartilage. Mesenchymal stem cells (MSCs) are found in low numbers in normal cartilage, mainly in the superficial layer, acting as repairing agents. In OA, MSCs are seen in larger numbers, but act chaotic and are unable to repair the cartilage. The synovial membrane becomes inflamed and interacts with the cartilage. Transplanted MSC have the ability to normalize them, redirecting them to their normal function. In a preliminary study, we showed that MSC could improve knee OA in four patients at 6 months. This report shows their long-term follow-up at 5 years. METHODS One patient was lost to follow-up at 2 years and three were followed for 5 years. They were aged 55, 57, 65 and 54 years, and had moderate to severe knee osteoarthritis. The worse knee of each patient was injected with 8-9 × 10(6) MSC. RESULTS As previously reported, all parameters improved in transplant knees at 6 months (walking time, stair climbing, gelling pain, patella crepitus, flection contracture and the visual analogue score on pain). Then, they started gradually to deteriorate, but at 5 years they were still better than at baseline. PGA (Patient Global Assessment) improved from baseline to 5 years. The better knee at baseline (no MSC), continued its progression toward aggravation and at 5 years became the worse knee. CONCLUSION Transplant knees were all in a rather advanced stage of OA. Earlier transplantation may give better results in long-term follow-up. This is what future studies have to demonstrate.
Collapse
Affiliation(s)
- Fereydoun Davatchi
- Rheumatology Research Center, Tehran University for Medical Sciences, Shariati Hospital, Tehran, Iran
| | - Bahar Sadeghi Abdollahi
- Rheumatology Research Center, Tehran University for Medical Sciences, Shariati Hospital, Tehran, Iran
| | - Mandana Mohyeddin
- Molecular Immunology Research Center, Tehran University for Medical Sciences, Shariati Hospital, Tehran, Iran
| | - Behrooz Nikbin
- Molecular Immunology Research Center, Tehran University for Medical Sciences, Shariati Hospital, Tehran, Iran
| |
Collapse
|
191
|
Abstract
The most common diseases of the joints and its tissues are osteoarthritis and rheumatoid arthritis, with osteoarthritis being anticipated to be the fourth leading cause of disability by the year 2020. To date, no truly causal therapies are available, and this has promoted tissue engineering attempts mainly involving mesenchymal stem cells. The goal of all tissue repairs would be to restore a fully functional tissue, here a hyaline articular cartilage. The hyaline cartilage is the most affected in osteoarthritis, where altered cell–matrix interactions gradually destroy tissue integrity. In rheumatoid arthritis, the inflammatory aspect is more important, and the cartilage tissue is destroyed by the invasion of tumor-like pannus tissue arising from the inflamed synovia. Furthermore, the fibrocartilage of the meniscus is clearly involved in the initiation of osteoarthritis, especially after trauma. Recent investigations have highlighted the role of migratory progenitor cells found in diseased tissues in situ. In osteoarthritis and rheumatoid arthritis, these chondrogenic progenitor cells are involved in regeneration efforts that are largely unsuccessful in diseased cartilage tissue. However, these progenitor cells are interesting targets for a cell-based regenerative therapy for joint diseases.
Collapse
Affiliation(s)
- Boris Schminke
- Tissue Regeneration Work Group, Department of Prosthodontics, Georg August University, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | | |
Collapse
|
192
|
Xia P, Wang X, Lin Q, Li X. Efficacy of mesenchymal stem cells injection for the management of knee osteoarthritis: a systematic review and meta-analysis. INTERNATIONAL ORTHOPAEDICS 2015; 39:2363-72. [DOI: 10.1007/s00264-015-2785-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 03/29/2015] [Indexed: 01/22/2023]
|
193
|
Tissue Engineering and Regenerative Medicine in Iran: Current State of Research and Future Outlook. Mol Biotechnol 2015; 57:589-605. [DOI: 10.1007/s12033-015-9865-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
194
|
Ren J, Wang H, Tran K, Civini S, Jin P, Castiello L, Feng J, Kuznetsov SA, Robey PG, Sabatino M, Stroncek DF. Human bone marrow stromal cell confluence: effects on cell characteristics and methods of assessment. Cytotherapy 2015; 17:897-911. [PMID: 25882666 DOI: 10.1016/j.jcyt.2015.03.607] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/09/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND AIMS Ex vivo expansion and serial passage of human bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) is required to obtain sufficient quantities for clinical therapy. The BMSC confluence criteria used to determine passage and harvest timing vary widely, and the impact of confluence on BMSC properties remains controversial. The effects of confluence on BMSC properties were studied and confluence-associated markers were identified. METHODS BMSC characteristics were analyzed as they grew from 50% to 100% confluence, including viability, population doubling time, apoptosis, colony formation, immunosuppression, surface marker expression, global gene expression and microRNA expression. In addition, culture supernatant protein, glucose, lactate and pH levels were analyzed. RESULTS Confluence-dependent changes were detected in the expression of several cell surface markers: 39 culture supernatant proteins, 26 microRNAs and 2078 genes. Many of these surface markers, proteins, microRNAs and genes have been reported to be important in BMSC function. The pigment epithelium-derived factor/vascular endothelial growth factor ratio increased with confluence, but 80% and 100% confluent BMSCs demonstrated a similar level of immunosuppression of mixed lymphocyte reactions. In addition, changes in lactate and glucose levels correlated with BMSC density. CONCLUSIONS BMSC characteristics change as confluence increases. 100% confluent BMSCs may have compromised pro-angiogenesis properties but may retain their immunomodulatory properties. Supernatant lactate and glucose levels can be used to estimate confluence and ensure consistency in passage and harvest timing. Flow cytometry or microRNA expression can be used to confirm that the BMSCs have been harvested at the appropriate confluence.
Collapse
Affiliation(s)
- Jiaqiang Ren
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Huan Wang
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine Tran
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara Civini
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Ping Jin
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Luciano Castiello
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Ji Feng
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Sergei A Kuznetsov
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Marianna Sabatino
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - David F Stroncek
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
195
|
Sampson S, Botto-van Bemden A, Aufiero D. Stem Cell Therapies for Treatment of Cartilage and Bone Disorders: Osteoarthritis, Avascular Necrosis, and Non-union Fractures. PM R 2015; 7:S26-S32. [DOI: 10.1016/j.pmrj.2015.01.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 01/21/2015] [Accepted: 01/23/2015] [Indexed: 12/16/2022]
|
196
|
Muhonen V, Narcisi R, Nystedt J, Korhonen M, van Osch GJVM, Kiviranta I. Recombinant human type II collagen hydrogel provides a xeno-free 3D micro-environment for chondrogenesis of human bone marrow-derived mesenchymal stromal cells. J Tissue Eng Regen Med 2015; 11:843-854. [PMID: 25643647 DOI: 10.1002/term.1983] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 10/02/2014] [Accepted: 12/09/2014] [Indexed: 12/12/2022]
Abstract
Recombinant human type II collagen (rhCII) hydrogel was tested as a xeno-free micro-environment for the chondrogenesis of human bone marrow-derived mesenchymal stromal cells (BM-MSCs). The rhCII hydrogels were seeded with BM-MSCs and cultured in a xeno-free chondro-inductive medium for 14, 28 and 84 days. High-density pellet cultures served as controls. The samples were subjected to biochemical, histological and gene expression analyses. Although the cells deposited glycosaminoglycans into the extracellular space significantly more slowly in the rhCII hydrogels compared to the high-density pellets, a similar potential of matrix deposition was reached by the end of the 84-day culture. At day 28 of culture, the gene expression level for cartilage marker genes (i.e. genes encoding for Sox9 transcription factor, Collagen type II and Aggrecan) were considerably lower in the rhCII hydrogels than in the high-density pellets, but at the end of the 84-day culture period, all the cartilage marker genes analysed were expressed at a similar level. Interestingly, the expression of the matrix metallopeptidases (MMP)-13, MMP-14 and MMP-8, i.e. extracellular collagen network-degrading enzymes, were transiently upregulated in the rhCII hydrogel, indicating active matrix reorganization. This study demonstrated that the rhCII hydrogel functions as a xeno-free platform for BM-MSC chondrogenesis, although the process is delayed. The reversible catabolic reaction evoked by the rhCII hydrogel might be beneficial in graft integration in vivo and pinpoints the need to further explore the use of hydrogels containing recombinant extracellular matrix (ECM) proteins to induce the chondrogenesis of MSCs. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Virpi Muhonen
- Department of Surgery, Institute of Clinical Medicine, University of Helsinki, Finland.,Department of Orthopaedics and Traumatology, Hospital District of Helsinki and Uusimaa, Helsinki, Finland
| | - Roberto Narcisi
- Department of Orthopaedics, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Johanna Nystedt
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Matti Korhonen
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands.,Department of Otorhinolaryngology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Ilkka Kiviranta
- Department of Surgery, Institute of Clinical Medicine, University of Helsinki, Finland.,Department of Orthopaedics and Traumatology, Hospital District of Helsinki and Uusimaa, Helsinki, Finland
| |
Collapse
|
197
|
Wei CC, Lin AB, Hung SC. Mesenchymal stem cells in regenerative medicine for musculoskeletal diseases: bench, bedside, and industry. Cell Transplant 2015; 23:505-12. [PMID: 24816447 DOI: 10.3727/096368914x678328] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (MSCs) can self-renew and differentiate into osteoblasts, chondrocytes, and adipocytes. MSCs have effectively emerged as a promising tool for clinical applications, specifically in musculoskeletal diseases. This article reviews the status of preclinical animal studies, clinical trials, and the efforts of the industry in using MSCs to treat musculoskeletal diseases such as bone fractures, bone defects, focal chondral lesions, osteoarthritis, spinal diseases, and tendon injuries. We also discuss the current problems encountered and potential of using MSCs in future clinical studies.
Collapse
Affiliation(s)
- Chih-Chang Wei
- Stem Cell Laboratory, Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | |
Collapse
|
198
|
Van Pham P, Bui KHT, Ngo DQ, Doan TTP, Vu NB, Truong NH, Le DM, Phan NK. Expanded Adipose Tissue-Derived Stem Cells for Articular Cartilage Injury Treatment: A Safety and Efficacy Evaluation. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
199
|
Makris EA, Gomoll AH, Malizos KN, Hu JC, Athanasiou KA. Repair and tissue engineering techniques for articular cartilage. Nat Rev Rheumatol 2015; 11:21-34. [PMID: 25247412 PMCID: PMC4629810 DOI: 10.1038/nrrheum.2014.157] [Citation(s) in RCA: 805] [Impact Index Per Article: 89.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chondral and osteochondral lesions due to injury or other pathology commonly result in the development of osteoarthritis, eventually leading to progressive total joint destruction. Although current progress suggests that biologic agents can delay the advancement of deterioration, such drugs are incapable of promoting tissue restoration. The limited ability of articular cartilage to regenerate renders joint arthroplasty an unavoidable surgical intervention. This Review describes current, widely used clinical repair techniques for resurfacing articular cartilage defects; short-term and long-term clinical outcomes of these techniques are discussed. Also reviewed is a developmental pipeline of acellular and cellular regenerative products and techniques that could revolutionize joint care over the next decade by promoting the development of functional articular cartilage. Acellular products typically consist of collagen or hyaluronic-acid-based materials, whereas cellular techniques use either primary cells or stem cells, with or without scaffolds. Central to these efforts is the prominent role that tissue engineering has in translating biological technology into clinical products; therefore, concomitant regulatory processes are also discussed.
Collapse
Affiliation(s)
- Eleftherios A Makris
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Andreas H Gomoll
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Konstantinos N Malizos
- Department of Orthopaedic Surgery and Musculoskeletal Trauma, University of Thessaly, Biopolis, Larisa 41110, Greece
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Kyriacos A Athanasiou
- Department of Orthopaedic Surgery, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
200
|
Zwolanek D, Flicker M, Kirstätter E, Zaucke F, van Osch GJVM, Erben RG. β1 Integrins Mediate Attachment of Mesenchymal Stem Cells to Cartilage Lesions. Biores Open Access 2015; 4:39-53. [PMID: 26309781 PMCID: PMC4497673 DOI: 10.1089/biores.2014.0055] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSC) may have great potential for cell-based therapies of osteoarthritis. However, after injection in the joint, only few cells adhere to defective articular cartilage and contribute to cartilage regeneration. Little is known about the molecular mechanisms of MSC attachment to defective articular cartilage. Here, we developed an ex vivo attachment system, using rat osteochondral explants with artificially created full-thickness cartilage defects in combination with genetically labeled MSC isolated from bone marrow of human placental alkaline phosphatase transgenic rats. Binding of MSC to full-thickness cartilage lesions was improved by serum, but not hyaluronic acid, and was dependent on the presence of divalent cations. Additional in vitro tests showed that rat MSC attach, in a divalent cation-dependent manner, to collagen I, collagen II, and fibronectin, but not to collagen XXII or cartilage oligomeric matrix protein (COMP). RGD peptides partially blocked the adhesion of MSC to fibronectin in vitro and to cartilage lesions ex vivo. Furthermore, the attachment of MSC to collagen I and II in vitro and to cartilage lesions ex vivo was almost completely abolished in the presence of a β1 integrin blocking antibody. In conclusion, our data suggest that attachment of MSC to ex vivo full-thickness cartilage lesions is almost entirely β1 integrin-mediated, whereby both RGD- and collagen-binding integrins are involved. These findings suggest a key role of integrins during MSC attachment to defective cartilage and may pave the way for improved MSC-based therapies in the future.
Collapse
Affiliation(s)
- Daniela Zwolanek
- Department of Biomedical Sciences, University of Veterinary Medicine , Vienna, Austria
| | - Magdalena Flicker
- Department of Biomedical Sciences, University of Veterinary Medicine , Vienna, Austria
| | - Elisabeth Kirstätter
- Department of Biomedical Sciences, University of Veterinary Medicine , Vienna, Austria
| | - Frank Zaucke
- Center for Biochemistry and Cologne Center for Musculoskeletal Biomechanics, Medical Faculty, University of Cologne , Cologne, Germany
| | - Gerjo J V M van Osch
- Department of Orthopaedics and Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Reinhold G Erben
- Department of Biomedical Sciences, University of Veterinary Medicine , Vienna, Austria
| |
Collapse
|