1
|
Shi Y, Shen S, Huang D, Yilamu K, Chen Z, Wang K, Chen P, Pan N, Jin Z, Guo L, Xu W, Zhao X. Delivery of FBXO6 with highly branched poly(β-amino ester)s to modulate the inflammatory environment for the treatment of osteoarthritis. J Control Release 2024; 378:294-305. [PMID: 39674232 DOI: 10.1016/j.jconrel.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/21/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by the progressive degradation of articular cartilage. Delivering functional genes to chondrocytes to modulate the inflammatory environment offers a promising approach to treating OA. However, the dense extracellular matrix (ECM) in the OA microenvironment and the rapid clearance of naked nucleic acids from synovial fluid present significant challenges. Here we report the development of highly branched poly(β-amino ester)s (HPAEs) for effective delivery of F-box protein 6 (FBXO6) gene to treat OA. Four HPAEs were synthesized using an "A2 + B4 + C2" Michael addition strategy. By optimizing the chemical compositions and topological structures, the optimal HMDA-2 was identified to exhibit superior transfection efficiency, outperforming the commercial reagents Lipofectamine 3000 and branched polyethyleneimine (PEI). HMDA-2 was further employed to deliver FBXO6 plasmid to effectively regulate H2O2 expression, improve oxidative stress injury, and reduce the expression of MMP-13 and COX-2 in chondrocytes, leading to significant reductions in synovial inflammatory cell infiltration, cartilage loss, and bone erosion. After intra-articular injection in an anterior cruciate ligament transection (ACLT) rat model, HMDA-2/FBXO6 polyplexes substantially reduced synovitis and cartilage damage, improved cartilage surface integrity, and restored proteoglycan levels. The delivery of FBXO6 with HMDA-2 to chondrocytes offers a potential treatment for OA.
Collapse
Affiliation(s)
- Yongxiang Shi
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City 310009, Zhejiang Province, China
| | - Shuying Shen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City 310009, Zhejiang Province, China; Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou City 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopedic Trauma and Aging Diseases of Zhejiang Province, Zhejiang 310016, China
| | - Dongdong Huang
- Pooling Institutes of Translational Medicine, Hangzhou 311000, China
| | - Kamuran Yilamu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou City 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopedic Trauma and Aging Diseases of Zhejiang Province, Zhejiang 310016, China
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing 100006, China
| | - Kefan Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou City 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopedic Trauma and Aging Diseases of Zhejiang Province, Zhejiang 310016, China
| | - Pengyu Chen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou City 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopedic Trauma and Aging Diseases of Zhejiang Province, Zhejiang 310016, China
| | - Nan Pan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou City 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopedic Trauma and Aging Diseases of Zhejiang Province, Zhejiang 310016, China
| | - Zhanping Jin
- Nursing Department, the First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Lei Guo
- Pooling Institutes of Translational Medicine, Hangzhou 311000, China.
| | - Wenbin Xu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou City 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopedic Trauma and Aging Diseases of Zhejiang Province, Zhejiang 310016, China.
| | - Xing Zhao
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou City 310016, China; Key Laboratory of Mechanism Research and Precision Repair of Orthopedic Trauma and Aging Diseases of Zhejiang Province, Zhejiang 310016, China.
| |
Collapse
|
2
|
Snuggs JW, Senter RK, Whitt JP, Jackson JD, Le Maitre CL. PCRX-201, a novel IL-1Ra gene therapy treatment approach for low back pain resulting from intervertebral disc degeneration. Gene Ther 2024:10.1038/s41434-024-00504-7. [PMID: 39572769 DOI: 10.1038/s41434-024-00504-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
Low back pain is the leading cause of global disability with intervertebral disc (IVD) degeneration a major cause. However, no current treatments target the underlying pathophysiological causes. PCRX-201 presents a novel gene therapy approach that addresses this issue. PCRX-201 codes for interleukin-1 receptor antagonist, the signalling inhibitor of the pro-inflammatory cytokine interleukin-1, which orchestrates the catabolic degeneration of the IVD. Here, the ability of PCRX-201 to transduce human nucleus pulposus cells to increase IL-1Ra production was assessed together with effects on catabolic pathways. When transduced with PCRX-201, the production and release of IL-1Ra was increased in degenerate human nucleus pulposus cells and tissue. Whereas, the production of downstream proteins, including IL-1β, IL-6, MMP3, ADAMTS4 and VEGF were decreased in both cells and tissue, indicating a reduction in IL-1-induced catabolic signalling. Here, a novel gene therapy vector, PCRX-201, was shown to transduce degenerate NP cells and tissue, increasing the production of IL-1Ra. The increased IL-1Ra resulted in decreased production of catabolic cytokines, enzymes and angiogenic factors, whilst also increasing aggrecan expression. This demonstrates PCRX-201 enables the inhibition of IL-1-driven IVD degeneration. The ability of PCRX-201 to elicit anti-catabolic responses is promising and warrants further development to determine the efficacy of this exciting, novel gene therapy.
Collapse
Affiliation(s)
- Joseph W Snuggs
- Division of Clinical Medicine, The University of Sheffield, Sheffield, UK
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Rebecca K Senter
- Former employee of Flexion Therapeutics, a wholly owned subsidiary of Pacira Biosciences, Tampa, Florida, USA
| | - Joshua P Whitt
- Former employee of Flexion Therapeutics, a wholly owned subsidiary of Pacira Biosciences, Tampa, Florida, USA
| | | | - Christine L Le Maitre
- Division of Clinical Medicine, The University of Sheffield, Sheffield, UK.
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK.
| |
Collapse
|
3
|
McKune CM. Clinical Management and Pharmacologic Treatment of Pain. VETERINARY ANESTHESIA AND ANALGESIA 2024:1010-1022. [DOI: 10.1002/9781119830306.ch48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
4
|
Defois A, Bon N, Mével M, Deniaud D, Maugars Y, Guicheux J, Adjali O, Vinatier C. Gene therapies for osteoarthritis: progress and prospects. JOURNAL OF CARTILAGE & JOINT PRESERVATION 2024; 4:100186. [DOI: 10.1016/j.jcjp.2024.100186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Grol MW. The evolving landscape of gene therapy strategies for the treatment of osteoarthritis. Osteoarthritis Cartilage 2024; 32:372-384. [PMID: 38199296 DOI: 10.1016/j.joca.2023.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/05/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
OBJECTIVES Significant advances have been made in our understanding of osteoarthritis (OA) pathogenesis; however, no disease-modifying therapies have been identified. This review will summarize the gene therapy landscape, its initial successes for OA, and possible challenges using recent studies and examples of gene therapies in clinical trials. DESIGN This narrative review has three major sections: 1) vector systems for OA gene therapy, 2) current and emerging targets for OA gene therapy, and 3) considerations and future directions. RESULTS Gene therapy is the strategy by which nucleic acids are delivered to treat and reverse disease progression. Specificity and prolonged expression of these nucleic acids are achieved by manipulating promoters, genes, and vector systems. Certain vector systems also allow for the development of combinatorial nucleic acid strategies that can be delivered in a single intraarticular injection - an approach likely required to treat the complexity of OA pathogenesis. Several viral and non-viral vector-based gene therapies are in clinical trials for OA, and many more are being evaluated in the preclinical arena. CONCLUSIONS In a post-coronavirus disease 2019 (COVID-19) era, the future of gene therapy for OA is certainly promising; however, the majority of preclinical validation continues to focus heavily on post-traumatic models and changes in only cartilage and subchondral bone. To ensure successful translation, new candidates in the preclinical arena should be examined against all joint tissues as well as pain using diverse models of injury-, obesity-, and age-induced disease. Lastly, consideration must be given to strategies for repeat administration and the cost of treatment owing to the chronic nature of OA.
Collapse
Affiliation(s)
- Matthew W Grol
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
6
|
Wilken F, Buschner P, Benignus C, Behr AM, Rieger J, Beckmann J. Pharmatherapeutic Treatment of Osteoarthrosis-Does the Pill against Already Exist? A Narrative Review. J Pers Med 2023; 13:1087. [PMID: 37511701 PMCID: PMC10381646 DOI: 10.3390/jpm13071087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
The aim of this narrative review is to summarize the current pharmacotherapeutic treatment options for osteoarthritis (OA). Is therapy still mainly symptomatic or does the pill against arthrosis already exist? Causal and non-causal, as well as future therapeutic approaches, are discussed. Various surgical and non-surgical treatment options are available that can help manage symptoms, slow down progression, and improve quality of life. To date, however, therapy is still mainly symptomatic, often using painkilling and anti-inflammatory drugs until the final stage, which is usually joint replacement. These "symptomatic pills against" have side effects and do not alter the progression of OA, which is caused by an imbalance between degenerative and regenerative processes. Next to resolving mechanical issues, the goal must be to gain a better understanding of the cellular and molecular basis of OA. Recently, there has been a lot of interest in cartilage-regenerative medicine and in the current style of treating rheumatoid arthritis, where drug therapy ("the pill against") has been established to slow down or even stop the progression of rheumatoid arthritis and has banned the vast majority of former almost regular severe joint destructions. However, the "causal pill against" OA does not exist so far. First, the early detection of osteoarthritis by means of biomarkers and imaging should therefore gain more focus. Second, future therapeutic approaches have to identify innovative therapeutic approaches influencing inflammatory and metabolic processes. Several pharmacologic, genetic, and even epigenetic attempts are promising, but none have clinically improved causal therapy so far, unfortunately.
Collapse
Affiliation(s)
- Frauke Wilken
- Department of Orthopedic Surgery and Traumatology, Hospital Barmherzige Brüder Munich, Romanstr. 93, 80639 München, Germany
| | - Peter Buschner
- Department of Orthopedic Surgery and Traumatology, Hospital Barmherzige Brüder Munich, Romanstr. 93, 80639 München, Germany
| | - Christian Benignus
- Department of Traumatology and Orthopedic Surgery, Hospital Ludwigsburg, Posilipostr. 4, 71640 Ludwigsburg, Germany
| | - Anna-Maria Behr
- Department of Orthopedic Surgery and Traumatology, Hospital Barmherzige Brüder Munich, Romanstr. 93, 80639 München, Germany
| | - Johannes Rieger
- Department of Orthopedic Surgery and Traumatology, Hospital Barmherzige Brüder Munich, Romanstr. 93, 80639 München, Germany
| | - Johannes Beckmann
- Department of Orthopedic Surgery and Traumatology, Hospital Barmherzige Brüder Munich, Romanstr. 93, 80639 München, Germany
| |
Collapse
|
7
|
Colantuoni M, Jofra Hernandez R, Pettinato E, Basso-Ricci L, Magnani L, Andolfi G, Rigamonti C, Finardi A, Romeo V, Soldi M, Sergi Sergi L, Rocchi M, Scala S, Hoffman HM, Gregori S, Kajaste-Rudnitski A, Sanvito F, Muzio L, Naldini L, Aiuti A, Mortellaro A. Constitutive IL-1RA production by modified immune cells protects against IL-1-mediated inflammatory disorders. Sci Transl Med 2023; 15:eade3856. [PMID: 37256935 DOI: 10.1126/scitranslmed.ade3856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 05/10/2023] [Indexed: 06/02/2023]
Abstract
Dysregulation of the interleukin-1 (IL-1) pathway leads to immune diseases that can result in chronic tissue and organ inflammation. Although IL-1 blockade has shown promise in ameliorating these symptoms and improving patients' quality of life, there is an urgent need for more effective, long-lasting treatments. We developed a lentivirus (LV)-mediated gene transfer strategy using transplanted autologous hematopoietic stem/progenitor cells (HSPCs) as a source of IL-1 receptor antagonist (IL-1RA) for systemic delivery to tissues and organs. Transplantation of mouse and human HSPCs transduced with an IL-1RA-encoding LV ensured stable IL-1RA production while maintaining the clonogenic and differentiation capacities of HSPCs in vivo. We examined the efficacy of cell-mediated IL-1RA delivery in three models of IL-1-dependent inflammation, for which treatment hindered neutrophil recruitment in an inducible model of gout, prevented systemic and multi-tissue inflammation in a genetic model of cryopyrin-associated periodic syndromes, and reduced disease severity in an experimental autoimmune encephalomyelitis model of multiple sclerosis. Our findings demonstrate HSPC-mediated IL-1RA delivery as a potential therapeutic modality that can be exploited to suppress tissue and organ inflammation in diverse immune-related diseases involving IL-1-driven inflammation.
Collapse
Affiliation(s)
- Mariasilvia Colantuoni
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Raisa Jofra Hernandez
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuela Pettinato
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Basso-Ricci
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Magnani
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Grazia Andolfi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Rigamonti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Annamaria Finardi
- Neuroimmunology Unit, INSpe, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Romeo
- Neuroimmunology Unit, INSpe, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Monica Soldi
- Processing Developmental Laboratory, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Sergi Sergi
- Processing Developmental Laboratory, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Rocchi
- GLP Test Facility, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Scala
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Hal M Hoffman
- Department of Pediatrics, University of California at San Diego, La Jolla, CA 92093, USA
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Francesca Sanvito
- GLP Test Facility, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pathology Unit, Department of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Muzio
- Vita-Salute San Raffaele University, Milan, Italy
- Neuroimmunology Unit, INSpe, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Mortellaro
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
8
|
Floramo JS, Molchanov V, Liu H, Liu Y, Craig SEL, Yang T. An Integrated View of Stressors as Causative Agents in OA Pathogenesis. Biomolecules 2023; 13:721. [PMID: 37238590 PMCID: PMC10216563 DOI: 10.3390/biom13050721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Cells in the body are exposed to dynamic external and internal environments, many of which cause cell damage. The cell's response to this damage, broadly called the stress response, is meant to promote survival and repair or remove damage. However, not all damage can be repaired, and sometimes, even worse, the stress response can overtax the system itself, further aggravating homeostasis and leading to its loss. Aging phenotypes are considered a manifestation of accumulated cellular damage and defective repair. This is particularly apparent in the primary cell type of the articular joint, the articular chondrocytes. Articular chondrocytes are constantly facing the challenge of stressors, including mechanical overloading, oxidation, DNA damage, proteostatic stress, and metabolic imbalance. The consequence of the accumulation of stress on articular chondrocytes is aberrant mitogenesis and differentiation, defective extracellular matrix production and turnover, cellular senescence, and cell death. The most severe form of stress-induced chondrocyte dysfunction in the joints is osteoarthritis (OA). Here, we summarize studies on the cellular effects of stressors on articular chondrocytes and demonstrate that the molecular effectors of the stress pathways connect to amplify articular joint dysfunction and OA development.
Collapse
Affiliation(s)
| | | | | | | | | | - Tao Yang
- Laboratory of Skeletal Biology, Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| |
Collapse
|
9
|
Uebelhoer M, Lambert C, Grisart J, Guse K, Plutizki S, Henrotin Y. Interleukins, growth factors, and transcription factors are key targets for gene therapy in osteoarthritis: A scoping review. Front Med (Lausanne) 2023; 10:1148623. [PMID: 37077668 PMCID: PMC10106745 DOI: 10.3389/fmed.2023.1148623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023] Open
Abstract
ObjectiveOsteoarthritis (OA) is the most common degenerative joint disease, characterized by a progressive loss of cartilage associated with synovitis and subchondral bone remodeling. There is however no treatment to cure or delay the progression of OA. The objective of this manuscript was to provide a scoping review of the preclinical and clinical studies reporting the effect of gene therapies for OA.MethodThis review followed the JBI methodology and was reported in accordance with the PRISMA-ScR checklist. All research studies that explore in vitro, in vivo, or ex vivo gene therapies that follow a viral or non-viral gene therapy approach were considered. Only studies published in English were included in this review. There were no limitations to their date of publication, country of origin, or setting. Relevant publications were searched in Medline ALL (Ovid), Embase (Elsevier), and Scopus (Elsevier) in March 2023. Study selection and data charting were performed by two independent reviewers.ResultsWe found a total of 29 different targets for OA gene therapy, including studies examining interleukins, growth factors and receptors, transcription factors and other key targets. Most articles were on preclinical in vitro studies (32 articles) or in vivo animal models (39 articles), while four articles were on clinical trials related to the development of TissueGene-C (TG-C).ConclusionIn the absence of any DMOAD, gene therapy could be a highly promising treatment for OA, even though further development is required to bring more targets to the clinical stage.
Collapse
Affiliation(s)
| | - Cécile Lambert
- musculoSKeletal Innovative Research Lab (mSKIL), Center for Interdisciplinary Research on Medicines, University of Liège, Liège, Belgium
| | | | - Kilian Guse
- GeneQuine Biotherapeutics GmbH, Hamburg, Germany
| | | | - Yves Henrotin
- Artialis S.A., Liège, Belgium
- musculoSKeletal Innovative Research Lab (mSKIL), Center for Interdisciplinary Research on Medicines, University of Liège, Liège, Belgium
- Department of Physical Therapy and Rehabilitation, Princess Paola Hospital, Vivalia, Marche-en-Famenne, Belgium
| |
Collapse
|
10
|
Agas D, Marchegiani A, Laus F, Gabai V, Sufianov AA, Shneider A, Sabbieti MG. p62/SQSTM1 indirectly mediates remote multipotent mesenchymal cells and rescues bone loss and bone marrow integrity in ovariectomized rats. J Cell Physiol 2023; 238:407-419. [PMID: 36565474 DOI: 10.1002/jcp.30937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022]
Abstract
Intramuscular administration of p62/SQSTM1 (sequestosome1)-encoding plasmid demonstrated an anticancer effect in rodent models and dogs as well as a high safety profile and the first evidence of clinical benefits in humans. Also, an anti-inflammatory effect of the plasmid was reported in several rodent disease models. Yet, the mechanisms of action for the p62 plasmid remain unknown. Here, we tested a hypothesis that the p62-plasmid can act through the modulation of bone marrow multipotent mesenchymal cells (MSCs). We demonstrated that a p62 plasmid can affect MSCs indirectly by stimulating p62-transfected cells to secrete an active ingredient(s) sensed by untransfected MSCs. When we transfected MSCs with the p62-plasmid, collected their supernatant, and added it to an untransfected MSCs culture, it switched the differentiation state and prompt osteogenic responses of the untransfected MSCs. According to an accepted viewpoint, ovariectomy leads to bone pathology via dysregulation of MSCs, and restoring the MSC homeostasis would restore ovariectomy-induced bone damage. To validate our in vitro observations in a clinically relevant in vivo model, we administered the p62 plasmid to ovariectomized rats. It partially reversed bone loss and notably reduced adipogenesis with concurrent reestablishing of the MSC subpopulation pool within the bone marrow. Overall, our study suggests that remote modulation of progenitor MSCs via administering a p62-encoding plasmid may constitute a mechanism for its previously reported effects and presents a feasible disease-preventing and/or therapeutic strategy.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | - Andrea Marchegiani
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | - Fulvio Laus
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino (MC), Italy
| | | | - Albert A Sufianov
- Federal Center of Neurosurgery, Tyumen, Russian Federation.,Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Alexander Shneider
- CureLab Oncology Inc., Dedham, Massachusetts, USA.,Department of Molecular Biology, Ariel University, Ariel, Israel
| | | |
Collapse
|
11
|
Haughan J, Ortved KF, Robinson MA. Administration and detection of gene therapy in horses: A systematic review. Drug Test Anal 2023; 15:143-162. [PMID: 36269665 DOI: 10.1002/dta.3394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/30/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022]
Abstract
Gene therapy uses genetic modification of cells to produce a therapeutic effect. Defective or missing genes can be repaired or replaced, or gene expression can be modified using a variety of technologies. Repair of defective genes can be achieved using specialized gene editing tools. Gene addition promotes gene expression by introducing synthetic copies of genes of interest (transgenes) into cells where they are transcribed and translated into therapeutic proteins. Protein production can also be modified using therapies that regulate gene expression. Gene therapy is currently prohibited in both human and equine athletes because of the potential to induce production of performance-enhancing proteins in the athlete's body, also referred to as "gene doping." Detection of gene doping is challenging and necessitates development of creative, novel analytical methods for doping control. Methods for detection of gene doping must be specific to and will vary depending on the type of gene therapy. The purpose of this paper is to present the results of a systematic review of gene editing, gene therapy, and detection of gene doping in horses. Based on the published literature, gene therapy has been administered to horses in a large number of experimental studies and a smaller number of clinical cases. Detection of gene therapy is possible using a combination of PCR and sequencing technologies. This summary can provide a basis for discussion of appropriate and inappropriate uses for gene therapy in horses by the veterinary community and guide expansion of methods to detect inappropriate uses by the regulatory community.
Collapse
Affiliation(s)
- Joanne Haughan
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania, USA
| | - Kyla F Ortved
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania, USA
| | - Mary A Robinson
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania, USA.,Pennsylvania Equine Toxicology & Research Center, West Chester University, West Chester, Pennsylvania, USA
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW To assess the present status of gene therapy for osteoarthritis (OA). RECENT FINDINGS An expanding list of cDNAs show therapeutic activity when introduced into the joints of animals with experimental models of OA. In vivo delivery with adenovirus or adeno-associated virus is most commonly used for this purpose. The list of encoded products includes cytokines, cytokine antagonists, enzymes, enzyme inhibitors, growth factors and noncoding RNA. Elements of CRISPR-Cas have also been delivered to mouse knees to ablate key genes. Several human trials have been initiated, using transgenes encoding transforming growth factor-β1, interleukin-1 receptor antagonist, interferon-β, the NKX3.2 transcription factor or variant interleukin-10. The first of these, using ex vivo delivery with allogeneic chondrocytes, gained approval in Korea which was subsequently retracted. However, it is undergoing Phase III clinical trials in the United States. The other trials are in Phase I or II. No gene therapy for OA has current marketing approval in any jurisdiction. SUMMARY Extensive preclinical data support the use of intra-articular gene therapy for treating OA. Translation is beginning to accelerate and six gene therapeutics are in clinical trials. Importantly, venture capital has begun to flow and at least seven companies are developing products. Significant progress in the future can be expected.
Collapse
|
13
|
Khoramjouy M, Bayanati M, Noori S, Faizi M, Zarghi A. Effects of Ziziphus Jujuba Extract Alone and Combined with Boswellia Serrata Extract on Monosodium Iodoacetate Model of Osteoarthritis in Mice. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e134338. [PMID: 36896317 PMCID: PMC9990515 DOI: 10.5812/ijpr-134338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/31/2023]
Abstract
Background As a chronic joint condition, osteoarthritis (OA) is a common problem among older people. Pain, aching, stiffness, swelling, decreased flexibility, reduced function, and disability are the symptoms of arthritis. Objectives In this study, we tested the extracts of Ziziphus jujuba (ZJE) and Boswellia serrata (BSE) to reduce OA symptoms as an alternative treatment. Methods NMRI mice were administered an intra-articular injection of monosodium iodoacetate (MIA; 1 mg/10 mL) in the left knee joint cavity for the induction of OA. Hydroalcoholic extracts of ZJE (250 and 500 mg/kg), BSE (100 and 200 mg/kg), and combined ZJE and BSE were orally administered daily for 21 days. Following behavioral tests, plasma samples were collected to detect inflammatory factors. To screen for general toxicity, acute oral toxicity was evaluated. Results Oral administration of all the hydroalcoholic extracts significantly increased the locomotor activity, pixel values of the foot-print area, paw withdrawal threshold, the latency of the withdrawal response to heat stimulation, and decreased the difference between pixel values of hind limbs compared to the vehicle group. Also, the elevated levels of IL-1β, IL-6, and TNF-α were reduced. As tested in this study, ZJE and BSE were practically nontoxic and had a high degree of safety. Conclusions This study demonstrated that the oral administration of ZJE and BSE slows the progression of OA through anti-nociceptive and anti-inflammatory properties. Oral co-administration of ZJE and BSE extracts can be used as herbal medicine to inhibit OA progression.
Collapse
Affiliation(s)
- Mona Khoramjouy
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Bayanati
- Department of Food Technology Research, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shokoofe Noori
- Department of Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Faizi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Zarghi
- Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Department of Pharmaceutical Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Treatment of Periodontal Inflammation in Diabetic Rats with IL-1ra Thermosensitive Hydrogel. Int J Mol Sci 2022; 23:ijms232213939. [PMID: 36430410 PMCID: PMC9693501 DOI: 10.3390/ijms232213939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Periodontitis is a chronic inflammatory disease that is considered to be the main cause of adult tooth loss. Diabetes mellitus (DM) has a bidirectional relationship with periodontitis. Interleukin-1β (IL-1β) is an important pre-inflammatory factor, which participates in the pathophysiological process of periodontitis and diabetes. The interleukin-1 receptor antagonist (IL-1ra) is a natural inhibitor of IL-1, and the balance between IL-1ra and IL-1β is one of the main factors affecting chronic periodontitis (CP) and diabetes. The purpose of this study is to develop a drug carrier that is safe and nontoxic and can effectively release IL-1ra, which can effectively slow down the inflammation of periodontal tissues with diabetes, and explore the possibility of lowering the blood sugar of this drug carrier. Therefore, in this experiment, a temperature-sensitive hydrogel loaded with IL-1ra was prepared and characterized, and its anti-inflammatory effect in high-sugar environments in vivo and in vitro was evaluated. The results showed that the hydrogel could gel after 5 min at 37 °C, the pore size was 5-70 μm, and the cumulative release of IL-1ra reached 83.23% on the 21st day. Real-time polymerase chain reaction (qRT-PCR) showed that the expression of IL-1β, Interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α) inflammatory factors decreased after the treatment with IL-1ra-loaded thermosensitive hydrogel. Histological evaluation and micro-computed tomography (Micro-CT) showed that IL-1ra-loaded thermosensitive hydrogel could effectively inhibit periodontal inflammation and reduce alveolar bone absorption in rats with diabetic periodontitis. It is worth mentioning that this hydrogel also plays a role in relieving hyperglycemia. Therefore, the temperature-sensitive hydrogel loaded with IL-1ra may be an effective method to treat periodontitis with diabetes.
Collapse
|
15
|
Thampi P, Samulski RJ, Grieger JC, Phillips JN, McIlwraith CW, Goodrich LR. Gene therapy approaches for equine osteoarthritis. Front Vet Sci 2022; 9:962898. [PMID: 36246316 PMCID: PMC9558289 DOI: 10.3389/fvets.2022.962898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/08/2022] [Indexed: 01/24/2023] Open
Abstract
With an intrinsically low ability for self-repair, articular cartilage injuries often progress to cartilage loss and joint degeneration resulting in osteoarthritis (OA). Osteoarthritis and the associated articular cartilage changes can be debilitating, resulting in lameness and functional disability both in human and equine patients. While articular cartilage damage plays a central role in the pathogenesis of OA, the contribution of other joint tissues to the pathogenesis of OA has increasingly been recognized thus prompting a whole organ approach for therapeutic strategies. Gene therapy methods have generated significant interest in OA therapy in recent years. These utilize viral or non-viral vectors to deliver therapeutic molecules directly into the joint space with the goal of reprogramming the cells' machinery to secrete high levels of the target protein at the site of injection. Several viral vector-based approaches have demonstrated successful gene transfer with persistent therapeutic levels of transgene expression in the equine joint. As an experimental model, horses represent the pathology of human OA more accurately compared to other animal models. The anatomical and biomechanical similarities between equine and human joints also allow for the use of similar imaging and diagnostic methods as used in humans. In addition, horses experience naturally occurring OA and undergo similar therapies as human patients and, therefore, are a clinically relevant patient population. Thus, further studies utilizing this equine model would not only help advance the field of human OA therapy but also benefit the clinical equine patients with naturally occurring joint disease. In this review, we discuss the advancements in gene therapeutic approaches for the treatment of OA with the horse as a relevant patient population as well as an effective and commonly utilized species as a translational model.
Collapse
Affiliation(s)
- Parvathy Thampi
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Joshua C. Grieger
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Jennifer N. Phillips
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - C. Wayne McIlwraith
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - Laurie R. Goodrich
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States,*Correspondence: Laurie R. Goodrich
| |
Collapse
|
16
|
Aman ZS, DePhillipo NN, Familiari F, Dickens JF, LaPrade RF, Dekker TJ. Acute Intervention With Selective Interleukin-1 Inhibitor Therapy May Reduce the Progression of Posttraumatic Osteoarthritis of the Knee: A Systematic Review of Current Evidence. Arthroscopy 2022; 38:2543-2556. [PMID: 35189307 DOI: 10.1016/j.arthro.2022.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE To evaluate the efficacy of selective interleukin (IL)-1 inhibitor therapy in the reduction of posttraumatic osteoarthritis (PTOA) progression following knee ligament or meniscal injury. METHODS A systematic review was conducted evaluating the disease-modifying efficacy of selective IL-1 inhibition in the setting of knee PTOA. RESULTS The literature search identified 364 articles and 11 studies were included (n = 10 preclinical, n = 1 clinical). Drug delivery in preclinical studies was administered using IL-1Ra-encoded helper-dependent adenovirus particles (n = 3), synovial cells transfected with an IL-1Ra-encoded retroviral vector (n = 3), or varying chemical compositions of nonviral microcapsule gene carriers (n = 4). Intervention with selective IL-1 inhibitor therapy within 2 weeks of injury provided the greatest protective benefits in reducing the progression of PTOA regardless of drug delivery methodology in preclinical models. The majority of studies reported significantly better cartilage integrity and reduction in lesion size in animals treated with gene therapy with the greatest effects seen in those treated within 5 to 7 days of injury. CONCLUSIONS Early intervention with selective IL-1 inhibitor therapy were effective in reducing proinflammatory IL-1β levels in the acute and subacute phases following traumatic knee injury in preclinical animal model studies, while significantly reducing cartilage damage, lesion size, and PTOA progression at short-term follow-up. However, it was found that the effect of these therapies diminished over time. CLINICAL RELEVANCE Acute, intra-articular injection of selective IL-1 inhibitors may reduce PTOA progression, supporting the need for additional basic and clinical investigation.
Collapse
Affiliation(s)
- Zachary S Aman
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, U.S.A
| | | | - Filippo Familiari
- Department of Orthopaedics and Trauma Surgery, Magna Graecia University, Catanzaro, Italy
| | | | | | | |
Collapse
|
17
|
Buchanan MW, Furman BD, McNulty AL, Olson SA. Combination of Lidocaine and IL-1Ra Is Effective at Reducing Degradation of Porcine Cartilage Explants. Am J Sports Med 2022; 50:1997-2006. [PMID: 35482438 DOI: 10.1177/03635465221090611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Posttraumatic inflammation after joint injury, ranging from sprains to articular fracture, contributes to the development of arthritis, and the administration of interleukin 1 (IL-1) receptor antagonist (IL-1Ra) is a potential intervention to mitigate this response. Although IL-1Ra mitigates cartilage degenerative changes induced by IL-1, lidocaine is used for local pain management in acute joint injury. Intra-articular delivery of both drugs in combination would be a novel and possibly disease-modifying treatment. However, it is not known whether the interaction with lidocaine at clinical concentrations (1%) would alter the efficacy of IL-1Ra to protect cartilage from the catabolic effects of IL-1. HYPOTHESIS Treatment of articular cartilage with IL-1Ra in combination with a clinically relevant concentration of lidocaine (1%) will inhibit the catabolic effects of IL-1α in a manner similar to treatment with IL-1Ra alone. STUDY DESIGN Controlled laboratory study. METHODS Fresh porcine cartilage explants were harvested, challenged with IL-1α, and incubated for 72 hours with IL-1Ra or a combination of IL-1Ra and lidocaine. The primary outcome was total sulfated glycosaminoglycan (sGAG) release. Additional experiments assessed the effect of storage temperature and premixing of IL-1Ra and lidocaine on sGAG release. All explants were histologically assessed for cartilage degradation using a modified Mankin grading scale. RESULTS The combination of IL-1Ra and lidocaine, premixed at various time points and stored at room temperature or 4°C, was as effective as IL-1Ra alone at inhibiting IL-1α-mediated sGAG release. Mankin histopathology scores supported these findings. CONCLUSION Our hypothesis was supported, and results indicated that the combination of IL-1Ra and lidocaine was as efficacious as IL-1Ra treatment alone in acutely mitigating biological cartilage injury due to IL-1α in an explant model. CLINICAL SIGNIFICANCE The combination of IL-1Ra and lidocaine is stable when reagents are stored in advance of administration at varying temperatures, providing clinically relevant information about storage of medications. The ability to premix and store this drug combination for intra-articular delivery may provide a novel treatment after joint injury to provide pain relief and block inflammation-induced catabolism of joint tissues.
Collapse
Affiliation(s)
- Michael W Buchanan
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Bridgette D Furman
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Amy L McNulty
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA.,Department of Pathology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Steven A Olson
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
18
|
Genetic Therapy in Veterinary Medicine. BIONANOSCIENCE 2022. [DOI: 10.1007/s12668-022-00986-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Senter R, Boyce R, Repic M, Martin EW, Chabicovsky M, Langevin-Carpentier G, Bédard A, Bodick N. Efficacy and Safety of FX201, a Novel Intra-Articular IL-1Ra Gene Therapy for Osteoarthritis Treatment, in a Rat Model. Hum Gene Ther 2022; 33:541-549. [PMID: 34963343 PMCID: PMC9142767 DOI: 10.1089/hum.2021.131] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022] Open
Abstract
Osteoarthritis (OA) is a disabling, degenerative disease characterized by progressive cartilage and bone damage. There remains a need for local therapies that, following a single injection, can provide long-term pain relief and functional improvement and potentially delay disease progression. FX201 is a novel, intra-articular (IA), interleukin-1 receptor antagonist (IL-1Ra) gene therapy in development for the treatment of OA. In this study, we assessed the efficacy, biodistribution, and safety of helper-dependent adenovirus (HDAd)-ratIL-1Ra, the rat surrogate of FX201, and the biodistribution of FX201, in the anterior cruciate ligament transection (ACLT) rat OA model. A single IA injection of HDAd-ratIL-1Ra administered 7 days post-ACLT mitigated OA-related changes to cartilage, bone, and the synovial membrane at week 12 following surgery. Furthermore, FX201 and HDAd-ratIL-1Ra persisted for at least 92 days in the injected joint and proximal tissues with minimal evidence of vector spreading peripherally. Finally, HDAd-ratIL-1Ra showed a favorable safety profile without any local or systemic adverse effects. In conclusion, HDAd-ratIL-1Ra demonstrated local therapeutic and disease-modifying effects and was well tolerated, supporting further clinical development of FX201.
Collapse
Affiliation(s)
- Rebecca Senter
- Flexion Therapeutics, Inc., Burlington, Massachusetts, USA
| | - Rogely Boyce
- Beechy Ridge ToxPath, LLC, Clay, West Virginia, USA
| | | | | | | | | | | | - Neil Bodick
- Gate Science, Inc., Moultonborough, New Hampshire, USA
| |
Collapse
|
20
|
Coşkun HS, Yurtbay A, Say F. Platelet Rich Plasma Versus Autologous Conditioned Serum in Osteoarthritis of the Knee: Clinical Results of a Five-Year Retrospective Study. Cureus 2022; 14:e24500. [PMID: 35651374 PMCID: PMC9135591 DOI: 10.7759/cureus.24500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 11/28/2022] Open
Abstract
Objectives: There is no consensus on the effectiveness of platelet-rich plasma (PRP) and autologous conditioned serum (ACS) in the treatment of knee osteoarthritis (OA). Also, the group of patients who will benefit most from this treatment is not clear. This study aims to understand the effects of two treatment modalities: ACS and PRP on pain and clinical scores in the treatment of OA. For this reason, we compared the long-term (five-year follow up) clinical results of the patients to whom these two treatment methods were applied. Materials and methods: Eighty-two knee osteoarthritis cases, selected from a database prospectively maintained in our tertiary university hospital after institutional ethics committee approval, examined between January 2013 and September 2020 and treated with ACS and PRP by the same orthobiological treatment team, were retrospectively analyzed. The clinical results of group A (n=40) treated with ACS and group B (n=42) treated with PRP were statistically analyzed. Clinical evaluations were made pre-injection and at one, six, 12, 24 and 60 months post-treatment, using the knee injury and osteoarthritis result score (KOOS) for the evaluation of function and a visual analog scale (VAS) for the evaluation of pain. Results: Side effects were noted in two patients (5%) in group A and 16 patients (38.1%) in group B. More side effects were seen in group B compared to group A (p<0.001). The better VAS scores in both groups were detected in the sixth and 12th months. When VAS scores were examined, better results were obtained in group A in the 12th and 24th months (p<0.05). When KOOS scores were examined, the superiority of ACS to PRP at 12 and 24 months was shown in KOOS.S, KOOS.P and KOOS.ADL scores (p<0.05). There was no statistically significant difference between the two groups in terms of all scores and baseline scores at 60 months. Conclusion: The effectiveness of ACS and PRP treatments can last up to two years. After two years, the effectiveness of both treatments decreases. Comparing the two treatments, ACS treatment showed better results on VAS and KOOS scores compared to PRP treatment.
Collapse
|
21
|
Kumar N, Saraber P, Ding Z, Kusumbe AP. Diversity of Vascular Niches in Bones and Joints During Homeostasis, Ageing, and Diseases. Front Immunol 2021; 12:798211. [PMID: 34975909 PMCID: PMC8718446 DOI: 10.3389/fimmu.2021.798211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/19/2021] [Indexed: 12/29/2022] Open
Abstract
The bones and joints in the skeletal system are composed of diverse cell types, including vascular niches, bone cells, connective tissue cells and mineral deposits and regulate whole-body homeostasis. The capacity of maintaining strength and generation of blood lineages lies within the skeletal system. Bone harbours blood and immune cells and their progenitors, and vascular cells provide several immune cell type niches. Blood vessels in bone are phenotypically and functionally diverse, with distinct capillary subtypes exhibiting striking changes with age. The bone vasculature has a special impact on osteogenesis and haematopoiesis, and dysregulation of the vasculature is associated with diverse blood and bone diseases. Ageing is associated with perturbed haematopoiesis, loss of osteogenesis, increased adipogenesis and diminished immune response and immune cell production. Endothelial and perivascular cells impact immune cell production and play a crucial role during inflammation. Here, we discuss normal and maladapted vascular niches in bone during development, homeostasis, ageing and bone diseases such as rheumatoid arthritis and osteoarthritis. Further, we discuss the role of vascular niches during bone malignancy.
Collapse
Affiliation(s)
| | | | | | - Anjali P. Kusumbe
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), Tissue and Tumor Microenvironments Group, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Wang QS, Xu BX, Fan KJ, Fan YS, Teng H, Wang TY. Dexamethasone-loaded thermo-sensitive hydrogel attenuates osteoarthritis by protecting cartilage and providing effective pain relief. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1120. [PMID: 34430561 PMCID: PMC8350682 DOI: 10.21037/atm-21-684] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022]
Abstract
Background We utilized the destabilization of medial meniscus (DMM)-induced mice to illustrate the osteoarthritis (OA) suppressing and pain-relieving effects of a novel prolonged-release intra-articular (IA)-dexamethasone-loaded thermo-sensitive hydrogel (DLTH). Methods The effects of temperature and pH on DLTH formation and in vitro DLTH release profile were assessed. C57BL/6J mice were randomly divided into three groups: Ctrl group, Model group and DLTH group. The DLTH group received joint injections of 10 µL DLTH (1 mg/kg) into the right knee once a week from week 2 to week 11. We performed micro-computed tomography (Micro-CT) and histological analyses of safranin O-fast green, hematoxylin and eosin, and tartrate-resistant acid phosphatase in knee joints. We also carried out immunohistochemical (IHC) staining for matrix metalloproteinase-9 (MMP-9), MMP-13, and a disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5) in cartilage and Ki-67 in synovia. Pain behavioral testing was carried out in all mice. The serum content of prostaglandin E2 (PGE2) and real-time polymerase chain reaction (PCR) of inflammatory cytokines and pain-related factors in dorsal root ganglia (DRGs) were evaluated. Results It took 20 minutes to form DLTH at pH 7.0 and 37 °C. The cumulative release profiles of dexamethasone (Dex) from DLTH at 37 °C revealed a rapid release in the first 24 h and a sustained slow release for 7 days. In vivo study illustrated that DLTH attenuated OA bone destruction and ameliorated synovitis and progression of OA in DMM-induced mice. The chondroprotective effects of DLTH were mediated by decreased expressions of MMP-9, MMP-13, and ADAMTS-5. The results showed that IA-DLTH exerted pain-relieving effects in OA mice. Upregulation of nociceptive response time (NRT) and downregulations of serum PGE2, inflammatory factors, and pain-related mediators in DRGs of mice in the DLTH group were recorded. Conclusions Data presented in this study elucidated that DLTH exhibited a long and lasting Dex release and it is a potential sustainable drug delivery system (DDS) to treat OA locally. IA-DLTH injection exerted chondroprotective and pain-relieving effects in DMM-induced arthritis. The involvement of MMP-9, MMP-13, ADAMTS-5, and inflammatory and pain-related factors, may account for the suppression of OA progression and pain.
Collapse
Affiliation(s)
- Qi-Shan Wang
- Departments of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing-Xin Xu
- Departments of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai-Jian Fan
- Departments of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun-Shan Fan
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Hui Teng
- Departments of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting-Yu Wang
- Departments of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Jiang P, Mao L, Qiao L, Lei X, Zheng Q, Li D. Efficacy and safety of mesenchymal stem cell injections for patients with osteoarthritis: a meta-analysis and review of RCTs. Arch Orthop Trauma Surg 2021; 141:1241-1251. [PMID: 33507375 DOI: 10.1007/s00402-020-03703-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Osteoarthritis (OA), which has a high incidence in the elderly, brings a huge economic burden to society. MSCs (Mesenchymal Stem Cells) have shown great multidirectional differentiation potential which are expected to treat OA, and numerous clinical trials have been conducted. However, the efficacy and safety of the MSCs still need to be further integrated and analyzed. MATERIALS AND METHODS We searched several databases (PubMed, EMBASE, Scopus, Web of Science, Cochrane Library, Ovid, and ScienceDirect) for assessing eligible trials that randomized controlled trials, hyaluronic acid as control, and MSCs injection to treat OA. Vitro studies and animal studies were excluded. Search terms were: "cartilage," "clinical trial," "mesenchymal," "stromal" and "stem cell", "osteoarthritis". The preliminary guidelines and study protocol were published online at PROSPERO. RESULTS Many assessment scales could not be improved significantly after 6 months. However, most of the scales were significantly improved after 12 months, indicating that compared with hyaluronic acid, stem cells could relieve OA symptoms significantly. No serious adverse effect was found. CONCLUSION There are significant therapeutic effects on joint function, symptoms, and no permanent adverse effect has been found after stem cell treatment. It is promising to apply intro-articular injection of stem cells for OA to clinical application. More researches are needed to supplement present deficiencies.
Collapse
Affiliation(s)
- Pan Jiang
- Affiliated Hospital of Jiangsu University, No.438, Jiefang road, Zhenjiang, 212000, China
| | - Lianghao Mao
- Affiliated Hospital of Jiangsu University, No.438, Jiefang road, Zhenjiang, 212000, China
| | - Longwei Qiao
- The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan Lei
- Affiliated Hospital of Jiangsu University, No.438, Jiefang road, Zhenjiang, 212000, China
| | - Qiping Zheng
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Dapeng Li
- Affiliated Hospital of Jiangsu University, No.438, Jiefang road, Zhenjiang, 212000, China.
| |
Collapse
|
24
|
Grol MW, Haelterman NA, Lim J, Munivez EM, Archer M, Hudson DM, Tufa SF, Keene DR, Lei K, Park D, Kuzawa CD, Ambrose CG, Eyre DR, Lee BH. Tendon and motor phenotypes in the Crtap-/- mouse model of recessive osteogenesis imperfecta. eLife 2021; 10:e63488. [PMID: 34036937 PMCID: PMC8186905 DOI: 10.7554/elife.63488] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 05/24/2021] [Indexed: 01/22/2023] Open
Abstract
Osteogenesis imperfecta (OI) is characterized by short stature, skeletal deformities, low bone mass, and motor deficits. A subset of OI patients also present with joint hypermobility; however, the role of tendon dysfunction in OI pathogenesis is largely unknown. Using the Crtap-/- mouse model of severe, recessive OI, we found that mutant Achilles and patellar tendons were thinner and weaker with increased collagen cross-links and reduced collagen fibril size at 1- and 4-months compared to wildtype. Patellar tendons from Crtap-/- mice also had altered numbers of CD146+CD200+ and CD146-CD200+ progenitor-like cells at skeletal maturity. RNA-seq analysis of Achilles and patellar tendons from 1-month Crtap-/- mice revealed dysregulation in matrix and tendon marker gene expression concomitant with predicted alterations in TGF-β, inflammatory, and metabolic signaling. At 4-months, Crtap-/- mice showed increased αSMA, MMP2, and phospho-NFκB staining in the patellar tendon consistent with excess matrix remodeling and tissue inflammation. Finally, a series of behavioral tests showed severe motor impairments and reduced grip strength in 4-month Crtap-/- mice - a phenotype that correlates with the tendon pathology.
Collapse
Affiliation(s)
- Matthew William Grol
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Nele A Haelterman
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Joohyun Lim
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Elda M Munivez
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Marilyn Archer
- Department of Orthopaedics and Sports Medicine, University of WashingtonSeattleUnited States
| | - David M Hudson
- Department of Orthopaedics and Sports Medicine, University of WashingtonSeattleUnited States
| | - Sara F Tufa
- Shriners Hospital for ChildrenPortlandUnited States
| | | | - Kevin Lei
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Dongsu Park
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Cole D Kuzawa
- Department of Orthopaedic Surgery, UT Health Sciences CenterHoustonUnited States
| | - Catherine G Ambrose
- Department of Orthopaedic Surgery, UT Health Sciences CenterHoustonUnited States
| | - David R Eyre
- Department of Orthopaedics and Sports Medicine, University of WashingtonSeattleUnited States
| | - Brendan H Lee
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
25
|
Abstract
Osteoarthritis affects hundreds of millions of people worldwide, and its prevalence is constantly increasing. While there is currently no treatment that can alter the course of the disease, promising therapeutic strategies and novel targets are being investigated. Innovative cell therapies are already reaching clinical trials, and recent progress in our understanding of the disease is opening new routes for gene therapy. In the long term, the development of new biofabrication tools, such as 3D bioprinting, may pave the way for personalized mini-joint models that could be used to screen drugs and to personalize treatments. This review provides an overview of the most promising therapeutic approaches in the field of osteoarthritis, from upcoming treatments to those that are yet to be discovered.
Collapse
|
26
|
Candidates for Intra-Articular Administration Therapeutics and Therapies of Osteoarthritis. Int J Mol Sci 2021; 22:ijms22073594. [PMID: 33808364 PMCID: PMC8036705 DOI: 10.3390/ijms22073594] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) of the knee is a disease that significantly decreases the quality of life due to joint deformation and pain caused by degeneration of articular cartilage. Since the degeneration of cartilage is irreversible, intervention from an early stage and control throughout life is important for OA treatment. For the treatment of early OA, the development of a disease-modifying osteoarthritis drug (DMOAD) for intra-articular (IA) injection, which is attracting attention as a point-of-care therapy, is desired. In recent years, the molecular mechanisms involved in OA progression have been clarified while new types of drug development methods based on gene sequences have been established. In addition to conventional chemical compounds and protein therapeutics, the development of DMOAD from the new modalities such as gene therapy and oligonucleotide therapeutics is accelerating. In this review, we have summarized the current status and challenges of DMOAD for IA injection, especially for protein therapeutics, gene therapy, and oligonucleotide therapeutics.
Collapse
|
27
|
Shakouri SK, Dolati S, Santhakumar J, Thakor AS, Yarani R. Autologous conditioned serum for degenerative diseases and prospects. Growth Factors 2021; 39:59-70. [PMID: 34886733 DOI: 10.1080/08977194.2021.2012467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autologous conditioned serum (ACS) is a blood-derived product that is prepared by the incubation of whole blood with medical-grade glass beads, resulting in serum enrichment in interleukin-1 receptor antagonist (IL-1Ra), anti-inflammatory cytokines (IL-4, IL-10, and IL-13), and high concentrations of growth factors. ACS has shown qualitatively and quantitatively better therapeutic effects than most established pharmacological treatments and surgery for joint diseases given its ability to both target the inflammatory cascade to decrease cartilage destruction as well as improve endogenous repair mechanisms. ACS application is simple and safe with limited adverse effects. This article reviews the role of ACS in degenerative joint disease, in addition to other inflammatory and autoimmune diseases, given its regenerative and immune-modulating properties.
Collapse
Affiliation(s)
- Seyed Kazem Shakouri
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jessica Santhakumar
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Reza Yarani
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|
28
|
Dowgray N, Comerford E. Feline musculoskeletal ageing: How are we diagnosing and treating musculoskeletal impairment? J Feline Med Surg 2020; 22:1069-1083. [PMID: 33100170 PMCID: PMC10814220 DOI: 10.1177/1098612x20965832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PRACTICAL RELEVANCE An understanding of the process of musculoskeletal ageing - which all senior and geriatric cats will experience - is vital to maintaining the health and welfare of our ageing cat population. CLINICAL CHALLENGES Assessment of the feline musculoskeletal system is not always straightforward. Diagnosis of impairment relies on input from owners and veterinarians in terms of visual observation, and clinical and orthopaedic examination, in addition to diagnostic imaging. AUDIENCE This review is written for the primary care veterinary team. AIMS The goals are to raise awareness and improve clinical diagnosis of musculoskeletal impairment as a result of ageing. The article also reviews therapeutic options and considers the evidence available for the prevention/deceleration of musculoskeletal ageing and impairment. EVIDENCE BASE There is good evidence of a high prevalence of osteoarthritis (OA) and degenerative joint disease (DJD) in older cats. There is also good evidence to indicate that functional impairment and chronic pain are sequelae of musculoskeletal disease. However, there is a paucity of information for what is best practice for the management and treatment of musculoskeletal impairment in a clinical situation. There is also a lack of evidence on how prevention of central stimulation of the nervous system caused by musculoskeletal impairment and, in turn the development of chronic pain, can be avoided.
Collapse
Affiliation(s)
| | - Eithne Comerford
- Institute of Life Course and Medical Sciences and School of Veterinary Science, University of Liverpool, UK
| |
Collapse
|
29
|
Biological strategies for osteoarthritis: from early diagnosis to treatment. INTERNATIONAL ORTHOPAEDICS 2020; 45:335-344. [PMID: 33078204 DOI: 10.1007/s00264-020-04838-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE To provide an updated review of the literature on the use of orthobiologics as a potential treatment option to alleviate symptoms associated with osteoarthritis (OA), slow the progression of the disease, and aid in cartilage regeneration. METHODS A comprehensive review of the literature was performed to identify basic science and clinical studies examining the role of orthobiologics in the diagnosis and management of osteoarthritis. RESULTS Certain molecules (such as interleukin-6 (IL-6), interleukin-8 (IL-8), matrix metalloproteinase (MMPs), cartilage oligomeric matrix protein (COMP), and tumor necrosis factor (TNF), microRNAs, growth differentiation factor 11 (GDF-11)) have been recognized as biomarkers that are implicated in the pathogenesis and progression of degenerative joint disease (DJD). These biomarkers have been used to develop newer diagnostic applications and targeted biologic therapies for DJD. Local injection therapy with biologic agents such as platelet-rich plasma or stem cell-based preparations has been associated with significant improvement in joint pain and function in patients with OA and has increased in popularity during the last decade. The combination of PRP with kartogenin or TGF-b3 may also enhance its biologic effect. The mesenchymal stem cell secretome has been recognized as a potential target for the development of OA therapies due to its role in mediating the chondroprotective effects of these cells. Recent experiments have also suggested the modification of gut microbiome as a newer method to prevent OA or alter the progression of the disease. CONCLUSIONS The application of orthobiologics for the diagnosis and treatment of DJD is a rapidly evolving field that will continue to expand. The identification of OA-specific and joint-specific biomarker molecules for early diagnosis of OA would be extremely useful for the development of preventive and therapeutic protocols. Local injection therapies with HA, PRP, BMAC, and other stem cell-based preparations are currently being used to improve pain and function in patients with early OA or those with progressed disease who are not surgical candidates. Although the clinical outcomes of these therapies seem to be promising in clinical studies, future research will determine the true role of orthobiologic applications in the field of DJS.
Collapse
|
30
|
Jayaram P, Liu C, Dawson B, Ketkar S, Patel SJ, Lee BH, Grol MW. Leukocyte-dependent effects of platelet-rich plasma on cartilage loss and thermal hyperalgesia in a mouse model of post-traumatic osteoarthritis. Osteoarthritis Cartilage 2020; 28:1385-1393. [PMID: 32629163 PMCID: PMC7787501 DOI: 10.1016/j.joca.2020.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 06/14/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Platelet-rich plasma (PRP) is an emerging therapeutic strategy for treatment of osteoarthritis (OA); however, there is a lack of preclinical and clinical evidence for its efficacy and its mechanism of action is unclear. In the current study, we utilized leukocyte poor-PRP (LP-PRP) and leukocyte rich-PRP (LR-PRP) to mimic clinical point of care formulations and assessed their potential to alter disease progression in a mouse model of post-traumatic OA. METHOD Three-month-old wild-type male FVB/N mice received destabilization of the medial meniscus (DMM) surgery to induce OA. To assess the efficacy of LP-PRP and LR-PRP, mice were given intraarticular injections at 2-, 7- and 28-days post-surgery. Mice were then assessed at 5-, 9-, and 13-weeks post-surgery for changes in chronic pain using the hot plate nociceptive assay. At 14-weeks, OA pathogenesis was evaluated using histology and phase-contrast μCT. RESULTS Treatment with LP-PRP and to a lesser extent LR-PRP preserved cartilage volume and surface area compared to phosphate-buffered saline (PBS) as measured by phase-contrast μCT. However, both treatments had higher Osteoarthritis Research Society International (OARSI) and synovitis scores compared to sham, and neither substantially improved scores compared to PBS controls. With respect to thermal hyperalgesia, PBS-treated mice displayed reduced latency to response compared to sham, and LR-PRP but not LP-PRP improved latency to response at 5-, 9- and 13-weeks post-surgery compared to PBS. CONCLUSION The results of this study suggest that effects of PRP therapy on OA progression and disease-induced hyperalgesia may be leukocyte-dependent. And while LP-PRP and to a lesser extent LR-PRP protect from volume and surface loss, significant pathology is still seen within OA joints. Future work is needed to understand how the different components of PRP effect OA pathogenesis and pain, and how these could be modified to achieve greater therapeutic efficacy.
Collapse
Affiliation(s)
- P Jayaram
- H. Ben Taub Department of Physical Medicine & Rehabilitation, Baylor College of Medicine, Houston, TX, USA; Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | - C Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - B Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - S Ketkar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - S J Patel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - B H Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| | - M W Grol
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
31
|
Lamplot JD, Rai MF, Tompkins WP, Friedman MV, Schmidt EJ, Sandell LJ, Brophy RH. Gene Expression in Meniscal Tears at the Time of Arthroscopic Partial Meniscectomy Predicts the Progression of Osteoarthritis Within 6 Years of Surgery. Orthop J Sports Med 2020; 8:2325967120936275. [PMID: 32923494 PMCID: PMC7446268 DOI: 10.1177/2325967120936275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Background: While knees with meniscal tears are associated with a heightened risk of developing osteoarthritis (OA), it is difficult to predict which patients are at the greatest risk for OA. Gene signatures in menisci that are resected during arthroscopic partial meniscectomy (APM) may provide insight into the risk of OA progression. Hypothesis: Meniscal gene signatures at the time of APM will predict radiographic OA progression. Study Design: Case series; Level of evidence, 4. Methods: Meniscal fragments were collected from 38 patients without OA during clinically indicated APM of the medial meniscus. The expression of 28 candidate genes with known roles in cartilage homeostasis, OA, extracellular matrix degradation, and obesity was assessed by quantitative real-time polymerase chain reaction. Weightbearing radiographs obtained before surgery and at final follow-up were graded by a musculoskeletal radiologist using the Kellgren-Lawrence classification of OA. The association of meniscal gene expression at baseline with the progression of radiographic OA was determined. Results: Gene expression and baseline and follow-up radiographic data were available from 31 patients (81.6%) at a mean follow-up of 6.2 ± 1.3 years. Patients without OA progression had significantly higher expression of 7 genes: MMP9 (5.1-fold; P = .002), IL8 (2.9-fold; P = .016), CCL3 (3.7-fold; P = .032), CCL3L1 (4.5-fold; P = .008), CXCL6 (6.2-fold; P = .010), LEP (5.2-fold; P = .004), and RETN (46-fold; P = .008). Conclusion: Gene expression in the meniscus at the time of APM may be associated with the risk for progression of OA after surgery. Elevated expression of the aforementioned genes may reflect a chondroprotective response. Stratifying the risk for OA progression after APM could facilitate targeted interventions to delay or prevent the development of OA. Further studies in a larger cohort with an extended follow-up, and inclusion of additional genes, are warranted to better characterize this association.
Collapse
Affiliation(s)
- Joseph D Lamplot
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, Missouri, USA.,Department of Cell Biology & Physiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - William P Tompkins
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Michael V Friedman
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Eric J Schmidt
- Department of Health Sciences, University of Lynchburg, Lynchburg, Virginia, USA
| | - Linda J Sandell
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, Missouri, USA.,Department of Cell Biology & Physiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Robert H Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
32
|
Primorac D, Molnar V, Rod E, Jeleč Ž, Čukelj F, Matišić V, Vrdoljak T, Hudetz D, Hajsok H, Borić I. Knee Osteoarthritis: A Review of Pathogenesis and State-Of-The-Art Non-Operative Therapeutic Considerations. Genes (Basel) 2020; 11:E854. [PMID: 32722615 PMCID: PMC7464436 DOI: 10.3390/genes11080854] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/11/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
Being the most common musculoskeletal progressive condition, osteoarthritis is an interesting target for research. It is estimated that the prevalence of knee osteoarthritis (OA) among adults 60 years of age or older is approximately 10% in men and 13% in women, making knee OA one of the leading causes of disability in elderly population. Today, we know that osteoarthritis is not a disease characterized by loss of cartilage due to mechanical loading only, but a condition that affects all of the tissues in the joint, causing detectable changes in tissue architecture, its metabolism and function. All of these changes are mediated by a complex and not yet fully researched interplay of proinflammatory and anti-inflammatory cytokines, chemokines, growth factors and adipokines, all of which can be measured in the serum, synovium and histological samples, potentially serving as biomarkers of disease stage and progression. Another key aspect of disease progression is the epigenome that regulates all the genetic expression through DNA methylation, histone modifications, and mRNA interference. A lot of work has been put into developing non-surgical treatment options to slow down the natural course of osteoarthritis to postpone, or maybe even replace extensive surgeries such as total knee arthroplasty. At the moment, biological treatments such as platelet-rich plasma, bone marrow mesenchymal stem cells and autologous microfragmented adipose tissue containing stromal vascular fraction are ordinarily used. Furthermore, the latter two mentioned cell-based treatment options seem to be the only methods so far that increase the quality of cartilage in osteoarthritis patients. Yet, in the future, gene therapy could potentially become an option for orthopedic patients. In the following review, we summarized all of the latest and most important research in basic sciences, pathogenesis, and non-operative treatment.
Collapse
Affiliation(s)
- Dragan Primorac
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Eberly College of Science, The Pennsylvania State University, University Park, State College, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
- Medical School, University of Split, 21000 Split, Croatia
- School of Medicine, Faculty of Dental Medicine and Health, University “Josip Juraj Strossmayer”, 31000 Osijek, Croatia
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96 450 Coburg, Germany
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Vilim Molnar
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Eduard Rod
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Željko Jeleč
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Nursing, University North, 48 000 Varaždin, Croatia
| | - Fabijan Čukelj
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Medical School, University of Split, 21000 Split, Croatia
| | - Vid Matišić
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
| | - Trpimir Vrdoljak
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Department of Orthopedics, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Damir Hudetz
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Orthopedics, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Hana Hajsok
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Medical School, University of Zagreb, 10000 Zagreb, Croatia
| | - Igor Borić
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Medical School, University of Split, 21000 Split, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| |
Collapse
|
33
|
Abstract
Being the most common musculoskeletal progressive condition, osteoarthritis is an interesting target for research. It is estimated that the prevalence of knee osteoarthritis (OA) among adults 60 years of age or older is approximately 10% in men and 13% in women, making knee OA one of the leading causes of disability in elderly population. Today, we know that osteoarthritis is not a disease characterized by loss of cartilage due to mechanical loading only, but a condition that affects all of the tissues in the joint, causing detectable changes in tissue architecture, its metabolism and function. All of these changes are mediated by a complex and not yet fully researched interplay of proinflammatory and anti-inflammatory cytokines, chemokines, growth factors and adipokines, all of which can be measured in the serum, synovium and histological samples, potentially serving as biomarkers of disease stage and progression. Another key aspect of disease progression is the epigenome that regulates all the genetic expression through DNA methylation, histone modifications, and mRNA interference. A lot of work has been put into developing non-surgical treatment options to slow down the natural course of osteoarthritis to postpone, or maybe even replace extensive surgeries such as total knee arthroplasty. At the moment, biological treatments such as platelet-rich plasma, bone marrow mesenchymal stem cells and autologous microfragmented adipose tissue containing stromal vascular fraction are ordinarily used. Furthermore, the latter two mentioned cell-based treatment options seem to be the only methods so far that increase the quality of cartilage in osteoarthritis patients. Yet, in the future, gene therapy could potentially become an option for orthopedic patients. In the following review, we summarized all of the latest and most important research in basic sciences, pathogenesis, and non-operative treatment.
Collapse
|
34
|
Vitali M, Ometti M, Drossinos A, Pironti P, Santoleri L, Salini V. Autologous conditioned serum: clinical and functional results using a novel disease modifying agent for the management of knee osteoarthritis. J Drug Assess 2020; 9:43-51. [PMID: 32284907 PMCID: PMC7144201 DOI: 10.1080/21556660.2020.1734009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/30/2020] [Indexed: 02/06/2023] Open
Abstract
Objective: The purpose of this study was to investigate the potential ability of autologous conditioned serum (ACS) to decrease pain and improve joint functionality in patients affected by knee osteoarthritis (OA). Methods: Fifteen patients with clinical and radiological signs of OA of the knee were recruited for this study. Each patient received 4 injections of ACS (Orthokine; orthogen, Dusseldorf, Germany) at the site of OA once per week for 4 weeks. Clinical and functional evaluation was performed using the VAS scale for pain, WOMAC scale and KSS functional and clinical scores before the first injection, at one week, at two weeks, at three weeks, at one month and at six months. Statistical analysis was done with the Wilcoxon Signed-Rank Test. Results: Our results show an improvement of all the evaluation scales at 6 months follow-up. Particularly, VAS scales among all patients decreased by 35.8% (p = .00148), KSS functional scores improved by 38.2% (p = .00148), KSS clinical scores improved by 28.9% (p = .00236) and WOMAC scores were reduced by 19.8% (p = .00188). Few adverse effects were observed in our sample. The most common complaint was pain and swelling in the subsequent days after performing the intra-articular injection. Only one patient reported rigidity following the injection of the ACS. Conclusion: Our results, in conjunction with preexisting studies in the medical literature regarding ACS, demonstrate the viability of this therapy for the treatment of knee OA, showing positive influence on pain and joint function without significant adverse effects.
Collapse
Affiliation(s)
- Matteo Vitali
- Department of Orthopedics and Traumatology, San Raffaele Hospital Scientific Institute, Milan, Italy
| | - Marco Ometti
- Department of Orthopedics and Traumatology, San Raffaele Hospital Scientific Institute, Milan, Italy
| | - Andreas Drossinos
- Department of Orthopedics and Traumatology, San Raffaele Hospital Scientific Institute, Milan, Italy
| | - Pierluigi Pironti
- Department of Orthopedics and Traumatology, San Raffaele Hospital Scientific Institute, Milan, Italy
| | - Luca Santoleri
- Immunohematology and Transfusion Medicine, IRRCS Ospedale San Raffaele, Milan, Italy
| | - Vincenzo Salini
- Department of Orthopedics and Traumatology, San Raffaele Hospital Scientific Institute, Milan, Italy
| |
Collapse
|
35
|
Salameh JW, Zhou L, Ward SM, Santa Chalarca CF, Emrick T, Figueiredo ML. Polymer-mediated gene therapy: Recent advances and merging of delivery techniques. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1598. [PMID: 31793237 PMCID: PMC7676468 DOI: 10.1002/wnan.1598] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/02/2019] [Accepted: 09/19/2019] [Indexed: 01/01/2023]
Abstract
The ability to safely and precisely deliver genetic materials to target sites in complex biological environments is vital to the success of gene therapy. Numerous viral and nonviral vectors have been developed and evaluated for their safety and efficacy. This study will feature progress in synthetic polymers as nonviral vectors, which benefit from their chemical versatility, biocompatibility, and ability to carry both therapeutic cargo and targeting moieties. The combination of synthetic gene carrying constructs with advanced delivery techniques promises new therapeutic options for treating and curing genetic disorders. This article is characterized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Janelle W. Salameh
- The Weldon School of Biomedical Engineering and the
Interdisciplinary Biomedical Sciences Program, Purdue University, West Lafayette,
Indiana
| | - Le Zhou
- Polymer Science and Engineering Department, University of
Massachusetts, Amherst, Massachusetts
| | - Sarah M. Ward
- Polymer Science and Engineering Department, University of
Massachusetts, Amherst, Massachusetts
| | | | - Todd Emrick
- Polymer Science and Engineering Department, University of
Massachusetts, Amherst, Massachusetts
| | - Marxa L. Figueiredo
- Department of Basic Medical Sciences and the
Interdisciplinary Biomedical Sciences Program, Purdue University, West Lafayette,
Indiana
| |
Collapse
|
36
|
Ji Q, Qiao X, Liu Y, Wang D, Yan J. Silencing of long‑chain non‑coding RNA GAS5 in osteoarthritic chondrocytes is mediated by targeting the miR‑34a/Bcl‑2 axis. Mol Med Rep 2019; 21:1310-1319. [PMID: 31894330 DOI: 10.3892/mmr.2019.10900] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/18/2019] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effects of the long non‑coding RNA (lncRNA) growth arrest‑specific transcript 5 (GAS5) on proliferation, apoptosis and the inflammatory response of osteoarthritic chondrocytes (OACs) and its associated mechanism of action. Primary chondrocytes were isolated from cartilage tissues of osteoarthritis (OA) patients for subculture. GAS5 was silenced in OACs by liposome transfection. The effects of GAS5 silencing on proliferation, apoptosis, stromal metabolism and inflammatory response of OACs were analyzed. The association of GAS5 with its target microRNA‑34a (miR‑34a) and the downstream target gene Bcl‑2 was verified by luciferase reporter assays. The results indicated that GAS5 silencing promoted the proliferation, inhibited cell apoptosis and caused G1 arrest of OACs compared with the control group (P<0.05). The expression levels of TNF‑α and IL‑6 in the supernatant of OACs in the si‑GAS5 group were significantly lower than those of the control group (P<0.05). The results of the double luciferase reporter assays indicated that overexpression of GAS5 downregulated miR‑34a and upregulated Bcl‑2 levels (P<0.05) compared with the expression levels of these markers in the control group. In contrast to GAS5 overexpression, knockdown of this RNA caused a significant upregulation of miR‑34a levels and a significant downregulation in the levels of Bcl‑2 (P<0.05). Moreover, GAS5 overexpression could counteract the inhibition of apoptosis by overexpression of miR‑34a (P<0.05). The data indicated that GAS5 participated in the development of OA by regulating the biological behavior of chondrocytes via the miR‑34a/Bcl‑2 axis.
Collapse
Affiliation(s)
- Qinghui Ji
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Xiaofeng Qiao
- Department of Orthopedics, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154000, P.R. China
| | - Yongxiang Liu
- Department of Orthopedics, Hegang People's Hospital, Hegang, Heilongjiang 154100, P.R. China
| | - Dawei Wang
- Department of Orthopedics, First Hospital of Zhangjiakou, Zhangjiakou, Hebei 075000, P.R. China
| | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
37
|
Moss KL, Jiang Z, Dodson ME, Linardi RL, Haughan J, Gale AL, Grzybowski C, Engiles JE, Stefanovski D, Robinson MA, Ortved KF. Sustained Interleukin-10 Transgene Expression Following Intra-Articular AAV5-IL-10 Administration to Horses. Hum Gene Ther 2019; 31:110-118. [PMID: 31773987 DOI: 10.1089/hum.2019.195] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Joint trauma leads to post-traumatic inflammation with upregulation of inflammatory cytokines and degradative enzymes. If severe enough, this response can lead to irreversible post-traumatic osteoarthritis. Interleukin-10 (IL-10), a cytokine with potent anti-inflammatory effects, has been shown to have chondroprotective effects. A gene therapy approach using a vector to overexpress IL-10 in the joint represents a feasible method of delivering sustained high doses of IL-10 to post-traumatic joints. We hypothesized that an AAV5 vector overexpressing IL-10 would result in rapid and sustained IL-10 expression following direct intra-articular injection and that this increase would not be reflected in systemic circulation. In addition, we hypothesized that intra-articular AAV5-IL-10 injection would not induce a local inflammatory response. Twelve horses were assigned to either treatment (AAV5-IL-10-injected) or control (PBS-injected) groups. Middle carpal joints were injected with 1012 vector genomes/joint or phosphate-buffered saline (PBS) alone (3 mL). Serial synovial fluid samples were analyzed for inflammatory changes, IL-10 concentration, and vector genome copy number. Serum samples were also analyzed for IL-10 concentration and vector genome copy number. Synovial membrane was collected on day 84. Synovial fluid IL-10 was significantly increased within 48 h of AAV5-IL-10 injection and remained increased, compared to PBS-injected joints, until day 84. Serum IL-10 was not different between groups. Vector administration did not cause a significant synovial inflammatory response. Vector genomes were detectable in the plasma, synovial fluid, and synovial membrane of AAV5-IL-10-injected horses only. IL-10 has the potential to modulate the articular inflammatory response, thereby protecting cartilage from degradation and osteoarthritis. This study demonstrates the feasibility and efficiency of intra-articular AAV5-IL-10, and future studies investigating the chondroprotective effects of IL-10 in inflamed joints in vivo are warranted.
Collapse
Affiliation(s)
- Kaitlyn L Moss
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Zibin Jiang
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Michael E Dodson
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Renata L Linardi
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Joanne Haughan
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Alexis L Gale
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Cara Grzybowski
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Julie E Engiles
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| | - Darko Stefanovski
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| | - Mary A Robinson
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania.,Pennsylvania Equine Toxicology & Research Center, West Chester University, West Chester, Pennsylvania
| | - Kyla F Ortved
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania
| |
Collapse
|
38
|
Cheng F, Yan FF, Liu YP, Cong Y, Sun KF, He XM. Dexmedetomidine inhibits the NF-κB pathway and NLRP3 inflammasome to attenuate papain-induced osteoarthritis in rats. PHARMACEUTICAL BIOLOGY 2019; 57:649-659. [PMID: 31545916 PMCID: PMC6764405 DOI: 10.1080/13880209.2019.1651874] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/15/2019] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Context: Dexmedetomidine (Dex) has been reported to have an anti-inflammatory effect. However, its role on osteoarthritis (OA) has not been explored. Objective: This study investigates the effect of Dex on OA rat model induced by papain. Materials and methods: The OA Wistar rat model was induced by intraluminal injection of 20 mL of papain mixed solution (4% papain 0.2 mL mixed with 0.03 mol L-1 l-cysteine 0.1 mL) into the right knee joint. Two weeks after papain injection, OA rats were treated by intra-articular injection of Dex (5, 10, or 20 μg kg-1) into the right knee (once a day, continuously for 4 weeks). Articular cartilage tissue was obtained after Dex treatment was completed. Results: The gait behavior scores (2.83 ± 0.49), PWMT (15.2 ± 1.78) and PTWL (14.81 ± 0.92) in H-DEX group were higher than that of OA group, while Mankin score (5.5 ± 0.81) was decreased (p < 0.05). Compared with the OA group, the IL-1β (153.11 ± 16.05 pg mg-1), IL-18 (3.71 ± 0.7 pg mg-1), IL-6 (14.15 ± 1.94 pg/mg) and TNF-α (40.45 ± 10.28 pg mg-1) levels in H-DEX group were decreased (p < 0.05). MMP-13, NLRP3, and caspase-1 p10 expression in Dex groups were significantly lower than that of OA group (p < 0.05), while collagen II was increased (p < 0.05). p65 in the nucleus of Dex groups was significantly down-regulated than that of OA group (p < 0.05). Discussion and Conclusions: Dex can improve pain symptoms and cartilage tissue damage of OA rats, which may be related to its inhibition of the activation of NF-κB and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Pain Clinic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Feng-Feng Yan
- Department of Pain Clinic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Yue-Peng Liu
- Center for Clinical Research and Translational Medicine, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Yan Cong
- Department of Traditional Chinese and Western Medicine, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Ke-Fu Sun
- Department of Orthopedic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Xue-Ming He
- Department of Geratology, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| |
Collapse
|
39
|
Evans C. Editorial: Arthritis Gene Therapy Using Interleukin-1 Receptor Antagonist. Arthritis Rheumatol 2019; 70:1699-1701. [PMID: 30035385 DOI: 10.1002/art.40675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 07/17/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Christopher Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
40
|
Zhou Y, Ming J, Li Y, Deng M, Chen Q, Ma Y, Chen Z, Zhang Y, Liu S. Ligustilide attenuates nitric oxide-induced apoptosis in rat chondrocytes and cartilage degradation via inhibiting JNK and p38 MAPK pathways. J Cell Mol Med 2019; 23:3357-3368. [PMID: 30770640 PMCID: PMC6484328 DOI: 10.1111/jcmm.14226] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/22/2019] [Accepted: 01/27/2019] [Indexed: 12/13/2022] Open
Abstract
Ligustilide (LIG) is the main lipophilic component of the Umbelliferae family of pharmaceutical plants, including Radix angelicae sinensis and Ligusticum chuanxiong. LIG shows various pharmacological properties associated with anti‐inflammation and anti‐apoptosis in several kinds of cell lines. However, the therapeutic effects of LIG on chondrocyte apoptosis remain unknown. In this study, we investigated whether LIG had an anti‐apoptotic activity in sodium nitroprusside (SNP)‐stimulated chondrocyte apoptosis and could delay cartilage degeneration in a surgically induced rat OA model, and elucidated the potential mechanisms. In vitro studies revealed that LIG significantly suppressed chondrocyte apoptosis and cytoskeletal remodelling, which maintained the nuclear morphology and increased the mitochondrial membrane potential. In terms of SNP, LIG treatment considerably reduced the expression levels of cleaved caspase‐3, Bax and inducible nitric oxide synthase and increased the expression level of Bcl‐2 in a dose‐dependent manner. The LIG‐treated groups presented a significantly suppressed expression of activating transcription factor 2 and phosphorylation of Jun N‐terminal kinase (JNK) and p38 mitogen‐activated protein kinase (MAPK). The inhibitory effect of LIG was enhanced by the p38 MAPK inhibitor SB203580 or the JNK inhibitor SP600125 and offset by the agonist anisomycin. In vivo studies demonstrated that LIG attenuated osteoarthritic cartilage destruction by inhibiting the cartilage chondrocyte apoptosis and suppressing the phosphorylation levels of activating transcription factor 2, JNK and p38 MAPK. This result was confirmed by histological analyses, micro‐CT, TUNEL assay and immunohistochemical analyses. Collectively, our studies indicated that LIG protected chondrocytes against SNP‐induced apoptosis and delayed articular cartilage degeneration by suppressing JNK and p38 MAPK pathways.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Orthopedics, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jianghua Ming
- Department of Orthopedics, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yaming Li
- Department of Orthopedics, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ming Deng
- Department of Orthopedics, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Chen
- Department of Orthopedics, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yonggang Ma
- Department of Orthopedics, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhonghui Chen
- Department of Orthopedics, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yubiao Zhang
- Department of Orthopedics, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiqing Liu
- Department of Orthopedics, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
41
|
Stone A, Grol MW, Ruan MZC, Dawson B, Chen Y, Jiang MM, Song IW, Jayaram P, Cela R, Gannon F, Lee BHL. Combinatorial Prg4 and Il-1ra Gene Therapy Protects Against Hyperalgesia and Cartilage Degeneration in Post-Traumatic Osteoarthritis. Hum Gene Ther 2018; 30:225-235. [PMID: 30070147 DOI: 10.1089/hum.2018.106] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative disease of synovial joints characterized by progressive loss of articular cartilage, subchondral bone remodeling, and intra-articular inflammation with synovitis that results in chronic pain and motor impairment. Despite the economic and health impacts, current medical therapies are targeted at symptomatic relief of OA and fail to alter its progression. Given the complexity of OA pathogenesis, we hypothesized that a combinatorial gene therapy approach, designed to inhibit inflammation with interleukin-1 receptor antagonist (IL-1Ra) while promoting chondroprotection using lubricin (PRG4), would improve preservation of the joint compared to monotherapy alone. Employing two surgical techniques to model mild, moderate and severe posttraumatic OA, we found that combined delivery of helper-dependent adenoviruses (HDVs), expressing IL-1Ra and PRG4, preserved articular cartilage better than either monotherapy in both models as demonstrated by preservation of articular cartilage volume and surface area. This improved protection was associated with increased expression of proanabolic and cartilage matrix genes together with decreased expression of catabolic genes and inflammatory mediators. In addition to improvements in joint tissues, this combinatorial gene therapy prolonged protection against thermal hyperalgesia compared to either monotherapy. Taken together, our results show that a combinatorial strategy is superior to monotherapeutic approaches for treatment of posttraumatic OA.
Collapse
Affiliation(s)
- Adrianne Stone
- 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,2 Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, Texas
| | - Matthew W Grol
- 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Merry Z C Ruan
- 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Brian Dawson
- 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Yuqing Chen
- 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Ming-Ming Jiang
- 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - I-Wen Song
- 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Prathap Jayaram
- 3 H. Ben Taub Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, Texas.,4 Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas
| | - Racel Cela
- 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Francis Gannon
- 5 Department of Pathology, Baylor College of Medicine, Houston, Texas
| | - Brendan H L Lee
- 1 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|