1
|
Atwal A, Mahnavi A, Davoodi P. Unlocking the potential of injectable hydrogels for cartilage repair. Regen Med 2025:1-10. [PMID: 40528501 DOI: 10.1080/17460751.2025.2520708] [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: 04/16/2025] [Accepted: 06/12/2025] [Indexed: 06/20/2025] Open
Abstract
Osteoarthritis (OA), a prevalent degenerative joint disease, poses significant global health implications and is characterized by progressive degradation of articular cartilage, resulting in debilitating pain and disability. Current treatments often provide only symptomatic relief or require multiple invasive open-joint procedures that are both costly and surgically complex. This review intends to evaluate key design considerations, drawing inspiration from clinically approved therapeutics and recent advances in the field that can truly unlock the potential of injectable hydrogels in cartilage tissue engineering. These include the choice of cargo, hydrogel architecture, and critical factors that are often overlooked in injectable hydrogels, such as ensuring integration with native tissue and restoring joint lubrication. The manuscript also addresses key barriers to clinical translation, including navigating stringent regulatory pathways and optimizing hydrogel performance to meet the mechanical and biological demands of the joint environment. Finally, the review outlines future research directions with emphasis on acellular off-the-shelf strategies, and personalized medicine approaches informed by advances in -omics and high-throughput screening data to accelerate the clinical translation of next-generation injectable hydrogels for effective cartilage repair.
Collapse
Affiliation(s)
- Arjan Atwal
- School of Allied Health Professions and Pharmacy, Keele University, Staffordshire, UK
- Guy Hilton Research Centre, School of Allied Health Professions and Pharmacy, Keele University, Staffordshire, UK
| | - Ali Mahnavi
- School of Allied Health Professions and Pharmacy, Keele University, Staffordshire, UK
- Guy Hilton Research Centre, School of Allied Health Professions and Pharmacy, Keele University, Staffordshire, UK
| | - Pooya Davoodi
- School of Allied Health Professions and Pharmacy, Keele University, Staffordshire, UK
- Guy Hilton Research Centre, School of Allied Health Professions and Pharmacy, Keele University, Staffordshire, UK
| |
Collapse
|
2
|
Gomes Velasque Gama F, Casciani C, Dutra EH. FGF18 induces chondrogenesis and anti-osteoarthritic effects in a mouse model for TMJ degeneration. PLoS One 2025; 20:e0317816. [PMID: 40273050 PMCID: PMC12021239 DOI: 10.1371/journal.pone.0317816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/06/2025] [Indexed: 04/26/2025] Open
Abstract
OBJECTIVE Temporomandibular Joint Osteoarthritis (TMJ-OA) is a degenerative disease characterized by progressive loss of cartilage and subchondral bone sclerosis. Currently there are no effective treatments for TMJ-OA. FGF18 is a member of the fibroblast growth factor family with essential roles for chondrogenesis, selectively binding to FGFR3 receptor. Studies have reported FGF18 attenuates cartilage degradation. Whereas the anti-osteoarthritic effects of FGF18 in the articular cartilage are known, the effects of FGF18 in a TMJ fibrocartilage degeneration mouse model remain to be determined. The goal of this project was to determine the effects of intra-articular injections of FGF18 in a mouse model for TMJ degeneration. METHOD Prosthesis tubes were bonded at the left lower incisor of 6-week-old triple collagen transgenic mice (Col1a1XCol2a1XCol10a1), creating unilateral crossbite and degeneration of the TMJ fibrocartilage. Six weeks after placement of prosthesis tubes, experimental and control mice received intra-articular injections of rmFGF18 (5µg/week) or saline, respectively, for 3 weeks. RESULTS Mice receiving saline intra-articular injections presented with a thinner cartilage layer with decreased proteoglycan distribution and Edu positive cells (chondrocyte proliferation marker), while mice injected with rmFGF18 presented with significant increased fibrocartilage thickness, remarkable proteoglycan distribution and chondrocyte proliferation, suggesting healing of the induced degeneration. Furthermore, reversal of the TMJ degeneration achieved by rmFGF18 injection was accompanied by a substantial reduction in Noggin (antagonist of BMP signaling), increase in TIMP1 (inhibitor of metalloproteinases such as MMP13) and decrease in MMP13 expression. CONCLUSION Our results postulate FGF18 as a powerful growth factor for the healing of TMJ fibrocartilage.
Collapse
Affiliation(s)
| | - Christina Casciani
- Department of Orthodontics, UConn Health, Farmington, Connecticut, United States of America
| | - Eliane Hermes Dutra
- Department of Orthodontics, UConn Health, Farmington, Connecticut, United States of America
| |
Collapse
|
3
|
Shanto PC, Park S, Fahad MAA, Park M, Lee BT. 3D bio-printed proteinaceous bioactive scaffold loaded with dual growth factor enhanced chondrogenesis and in situ cartilage regeneration. Bioact Mater 2025; 46:365-385. [PMID: 39845130 PMCID: PMC11751550 DOI: 10.1016/j.bioactmat.2024.12.021] [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: 09/13/2024] [Revised: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Articular cartilage has a limited self-healing capacity, leading to joint degeneration and osteoarthritis over time. Therefore, bioactive scaffolds are gaining attention as a promising approach to regenerating and repairing damaged articular cartilage through tissue engineering. In this study, we reported on a novel 3D bio-printed proteinaceous bioactive scaffolds combined with natural porcine cancellous bone dECM, tempo-oxidized cellulose nanofiber (TOCN), and alginate carriers for TGF-β1, FGF-18, and ADSCs to repair cartilage defects. The characterization results demonstrate that the 3D scaffolds are physically stable and facilitate a controlled dual release of TGF-β1 and FGF-18. Moreover, the key biological proteins within the bioactive scaffold actively interact with the biological systems to create a favorable microenvironment for cartilage regeneration. Importantly, the in vitro, in vivo, and in silico simulation showed that the scaffolds promote stem cell recruitment, migration, proliferation, and ECM deposition, and synergistic effects of TGF-β1/FGF-18 with the bioactive scaffolds significantly regulate stem cell chondrogenesis by activating the PI3K/AKT and TGFβ1/Smad4 signaling pathways. After implantation, the proteinaceous bioactive scaffold led to the regeneration of mechanically robust, full-thickness cartilage tissue that closely resembles native cartilage. Thus, these findings may provide a promising approach for regulating stem cell chondrogenesis and treating in situ cartilage regeneration.
Collapse
Affiliation(s)
- Prayas Chakma Shanto
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Seongsu Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Md Abdullah Al Fahad
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Myeongki Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
- Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| |
Collapse
|
4
|
Fu PJ, Zheng SY, Luo Y, Ren ZQ, Li ZH, Wang YP, Lu BB. Prg4 and Osteoarthritis: Functions, Regulatory Factors, and Treatment Strategies. Biomedicines 2025; 13:693. [PMID: 40149669 PMCID: PMC11940178 DOI: 10.3390/biomedicines13030693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Proteoglycan 4 (PRG4), also known as lubricin, plays a critical role in maintaining joint homeostasis by reducing friction between articular cartilage surfaces and preventing cartilage degradation. Its deficiency leads to early-onset osteoarthritis (OA), while overexpression can protect against cartilage degeneration. Beyond its lubricating properties, PRG4 exerts anti-inflammatory effects by interacting with Toll-like receptors, modulating inflammatory responses within the joint. The expression of Prg4 is regulated by various factors, including mechanical stimuli, inflammatory cytokines, transcription factors such as Creb5 and FoxO, and signaling pathways like TGF-β, EGFR, and Wnt/β-catenin. Therapeutic strategies targeting PRG4 in OA have shown promising results, including recombinant PRG4 protein injections, gene therapies, and small molecules that enhance endogenous Prg4 expression or mimic its function. Further research into the molecular mechanisms regulating Prg4 expression will be essential in developing more effective OA treatments. Understanding the interplay between Prg4 and other signaling pathways could reveal novel therapeutic targets. Additionally, advancements in gene therapy and biomaterials designed to deliver PRG4 in a controlled manner may hold potential for the long-term management of OA, improving patient outcomes and delaying disease progression.
Collapse
Affiliation(s)
- Peng-Jie Fu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; (P.-J.F.); (S.-Y.Z.); (Y.L.); (Z.-Q.R.); (Z.-H.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha 410013, China
| | - Sheng-Yuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; (P.-J.F.); (S.-Y.Z.); (Y.L.); (Z.-Q.R.); (Z.-H.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha 410013, China
| | - Yan Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; (P.-J.F.); (S.-Y.Z.); (Y.L.); (Z.-Q.R.); (Z.-H.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha 410013, China
| | - Zhuo-Qun Ren
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; (P.-J.F.); (S.-Y.Z.); (Y.L.); (Z.-Q.R.); (Z.-H.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha 410013, China
| | - Zi-Han Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; (P.-J.F.); (S.-Y.Z.); (Y.L.); (Z.-Q.R.); (Z.-H.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha 410013, China
| | - Ya-Ping Wang
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Bang-Bao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; (P.-J.F.); (S.-Y.Z.); (Y.L.); (Z.-Q.R.); (Z.-H.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
5
|
Pill SG, Ahearn B, Tokish JM, Cook C, Siffri PJ, Mercuri JJ, Burnikel B, Cassas KJ, Wyland DJ, Sawvell E, Wright N, Hutchinson J, Bynarowicz T, Adams KJ, Kissenberth MJ. Amniotic Tissue Injections Are an Effective Alternative to Corticosteroid Injections for Pain Relief and Function in Patients With Severe Knee Osteoarthritis: A Double-Blind, Randomized, Prospective Study. J Am Acad Orthop Surg Glob Res Rev 2025; 9:01979360-202501000-00009. [PMID: 39813395 PMCID: PMC11723680 DOI: 10.5435/jaaosglobal-d-23-00276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 11/03/2024] [Indexed: 01/18/2025]
Abstract
INTRODUCTION The use of corticosteroid injections for short-term pain relief for knee osteoarthritis can have deleterious adverse effects. Amniotic tissue has shown promise in vitro; therefore, this study compared a morcellized injectable amniotic tissue allograft to corticosteroid injection. METHODS Eighty-one patients with symptomatic severe knee osteoarthritis (Kellgren-Lawrence grade 3 to 4) were prospectively randomized to either a double-blinded single injection of BioDRestore (Integra LifeSciences; n = 39) or triamcinolone acetonide (n = 42). Knee Injury and Osteoarthritis Outcome Score (KOOS), Single Alpha Numeric Evaluation, visual analog scale (VAS) pain, Lysholm Rating, and Veterans-Rand-12 scales at baseline, 6 weeks, 3, 6, and 12 months were analyzed. RESULTS No differences were observed in adverse reactions or patient reported outcomes (PROs); however, a notable continued improvement was found in the amnion group from 6 weeks to 1 year for Single Alpha Numeric Evaluation, Lysholm, and KOOS Symptoms, Pain, activities of daily living [ADL], QofL. The minimal clinically important difference (MCID) was met for Lysholm, KOOS ADL, and KOOS pain. DISCUSSION Both amnion and steroid injections showed an initial improvement in pain relief and function at 6 weeks; however, more patients in the amniotic tissue group maintained pain relief and function at the 1-year follow-up. The mixed results suggest that amniotic tissue injections may be a safe and effective alternative to corticosteroid injections.
Collapse
Affiliation(s)
- Stephan G. Pill
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Briggs Ahearn
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - John M. Tokish
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Chad Cook
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Paul J. Siffri
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Jeremy J. Mercuri
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Brian Burnikel
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Kyle J. Cassas
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Douglas J. Wyland
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Emily Sawvell
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Noah Wright
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Josh Hutchinson
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Taylor Bynarowicz
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Kyle J. Adams
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| | - Michael J. Kissenberth
- From the Steadman Hawkins Clinic of the Carolinas, Prisma Health-Upstate, Greenville, SC (Dr. Pill, Dr. Ahearn, Dr. Siffri, Dr. Burnikel, Dr. Cassas, Dr. Wyland, and Dr. Kissenberth); the Mayo Clinic Arizona, Scottsdale, AZ (Dr. Tokish); the Department of Orthopaedics, Duke University, Durham NC (Dr. Cook); the Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Department of Bioengineering, Clemson University, Clemson, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); the Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, SC (Dr. Mercuri, Mr. Sawvell, and Mr. Wright); and the Hawkins Foundation, Greenville, SC (Dr. Hutchinson, Dr. Bynarowicz, and Dr. Adams)
| |
Collapse
|
6
|
Sun M, Ma B, Pan Z, Zhao Y, Tian L, Fan Y, Kong W, Wang J, Xu B, Ao Y, Guo Q, Wang X, Peng X, Li X, Cheng J, Miao L, Wang K, Hu X. Targeted Therapy of Osteoarthritis via Intra-Articular Delivery of Lipid-Nanoparticle-Encapsulated Recombinant Human FGF18 mRNA. Adv Healthc Mater 2024; 13:e2400804. [PMID: 39363784 PMCID: PMC11582510 DOI: 10.1002/adhm.202400804] [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: 03/01/2024] [Revised: 07/08/2024] [Indexed: 10/05/2024]
Abstract
Fibroblast growth factor 18 (FGF18) emerges as a promising therapeutic target for osteoarthritis (OA). In this study, a novel articular cavity-localized lipid nanoparticle (LNP) named WG-PL14 is developed. This optimized formulation has a nearly 30-fold increase in mRNA expression as well as better articular cavity enrichment compared to commercial lipids MC3 when performing intra-articular injection. Then, a mRNA sequence encoding recombinant human FGF18 (rhFGF18) for potential mRNA therapy in OA is optimized. In vitro assays confirm the translation of rhFGF18 mRNA into functional proteins within rat and human chondrocytes, promoting cell proliferation and extracellular matrix (ECM) synthesis. Subsequently, the therapeutic efficacy of the LNP-rhFGF18 mRNA complex is systematically assessed in a mouse OA model. The administration exhibits several positive outcomes, including an improved pain response, upregulation of ECM-related genes (e.g., AGRN and HAS2), and remodeling of subchondral bone homeostasis compared to a control group. Taken together, these findings underscore the potential of localized LNP-rhFGF18 mRNA therapy in promoting the regeneration of cartilage tissue and mitigating the progression of OA.
Collapse
Affiliation(s)
- Mengze Sun
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Bin Ma
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Beijing Key Laboratory of Molecular PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Zihang Pan
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Yun Zhao
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Liangliang Tian
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Yifei Fan
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Weijing Kong
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Junyan Wang
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Boyang Xu
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Yingfang Ao
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Quanyi Guo
- Institute of OrthopedicsThe Fourth Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLANo. 28 Fuxing Road, Haidian DistrictBeijing100853China
| | - Xi Wang
- State Key Laboratory of Female Fertility PromotionClinical Stem Cell Research CenterPeking University Third HospitalBeijing100191China
| | - Xiaohong Peng
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Xiaoxia Li
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Jin Cheng
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Lei Miao
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Beijing Key Laboratory of Molecular PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Kai Wang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Xiaoqing Hu
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| |
Collapse
|
7
|
Xie B, Ma H, Yang F, Chen H, Guo Y, Zhang H, Li T, Huang X, Zhao Y, Li X, Du J. Development and evaluation of 3D composite scaffolds with piezoelectricity and biofactor synergy for enhanced articular cartilage regeneration. J Mater Chem B 2024; 12:10416-10433. [PMID: 39291892 DOI: 10.1039/d4tb01319k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The inability of articular cartilage to self-repair following injuries frequently precipitates osteoarthritis, profoundly affecting patients' quality of life. Given the limitations inherent in current clinical interventions, an urgent need exists for more effective cartilage regeneration methodologies. Previous studies have underscored the potential of electrical stimulation in cartilage repair, thus motivating the investigation of innovative strategies. The present study introduces a three-dimensional scaffold fabricated through a composite technique that leverages the synergy between piezoelectricity and biofactors to enhance cartilage repair. This scaffold is composed of polylactic acid (PLLA) and barium titanate (BT) for piezoelectric stimulation and at the bottom with a collagen-coated layer infused with fibroblast growth factor-18 (FGF-18) for biofactor delivery. Designed to emulate the properties of natural cartilage, the scaffold enables controlled generation of piezoelectric charges and the sustained release of biofactors. In vitro tests confirm that the scaffold promotes chondrocyte proliferation, matrix hyperplasia, cellular migration, and the expression of genes associated with cartilage formation. Moreover, in vivo studies on rabbits have illustrated its efficacy in catalyzing the in situ regeneration of articular cartilage defects and remodeling the extracellular matrix. This innovative approach offers significant potential for enhancing cartilage repair and holds profound implications for regenerative medicine.
Collapse
Affiliation(s)
- Bowen Xie
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
- Air Force Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, China.
| | - Hebin Ma
- Medical School of the PLA General Hospital, Beijing 100853, China
- Senior Department of Orthopedics, The Fourth Medical Center of the PLA General Hospital, Beijing 100048, China.
| | - Fengyuan Yang
- Graduate School of Medicine, China Medical University, Shenyang 110122, China
| | - Hongguang Chen
- Senior Department of Orthopedics, The Fourth Medical Center of the PLA General Hospital, Beijing 100048, China.
| | - Ya'nan Guo
- Senior Department of Orthopedics, The Fourth Medical Center of the PLA General Hospital, Beijing 100048, China.
| | - Hongxing Zhang
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
| | - Tengfei Li
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
| | - Xiaogang Huang
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
| | - Yantao Zhao
- Senior Department of Orthopedics, The Fourth Medical Center of the PLA General Hospital, Beijing 100048, China.
| | - Xiaojie Li
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
| | - Junjie Du
- Department of Orthopedics, Air Force Medical Center, Beijing 100142, China.
- Air Force Clinical College, The Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, China.
- Graduate School of Medicine, China Medical University, Shenyang 110122, China
| |
Collapse
|
8
|
Goldschagg MGE, Hockman D. FGF18. Differentiation 2024; 139:100735. [PMID: 38007374 DOI: 10.1016/j.diff.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/25/2023] [Accepted: 10/25/2023] [Indexed: 11/27/2023]
Abstract
FGF18 was discovered in 1998. It is a pleiotropic growth factor that stimulates major signalling pathways involved in cell proliferation and growth, and is involved in the development and homeostasis of many tissues such as bone, lung, and central nervous system. The gene consists of five exons that code for a 207 amino acid glycosylated protein. FGF18 is widely expressed in developing and adult chickens, mice, and humans, being seen in the mesenchyme, brain, skeleton, heart, and lungs. Knockout studies of FGF18 in mice lead to perinatal death, characterised by distinct phenotypes such as cleft palate, smaller body size, curved long bones, deformed ribs, and reduced crania. As can be expected from a protein involved in so many functions FGF18 is associated with various diseases such as idiopathic pulmonary fibrosis, congenital diaphragmatic hernia, and most notably various types of cancer such as breast, lung, and ovarian cancer.
Collapse
Affiliation(s)
- Michael G E Goldschagg
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Dorit Hockman
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
9
|
Li HZ, Zhang JL, Yuan DL, Xie WQ, Ladel CH, Mobasheri A, Li YS. Role of signaling pathways in age-related orthopedic diseases: focus on the fibroblast growth factor family. Mil Med Res 2024; 11:40. [PMID: 38902808 PMCID: PMC11191355 DOI: 10.1186/s40779-024-00544-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Fibroblast growth factor (FGF) signaling encompasses a multitude of functions, including regulation of cell proliferation, differentiation, morphogenesis, and patterning. FGFs and their receptors (FGFR) are crucial for adult tissue repair processes. Aberrant FGF signal transduction is associated with various pathological conditions such as cartilage damage, bone loss, muscle reduction, and other core pathological changes observed in orthopedic degenerative diseases like osteoarthritis (OA), intervertebral disc degeneration (IVDD), osteoporosis (OP), and sarcopenia. In OA and IVDD pathologies specifically, FGF1, FGF2, FGF8, FGF9, FGF18, FGF21, and FGF23 regulate the synthesis, catabolism, and ossification of cartilage tissue. Additionally, the dysregulation of FGFR expression (FGFR1 and FGFR3) promotes the pathological process of cartilage degradation. In OP and sarcopenia, endocrine-derived FGFs (FGF19, FGF21, and FGF23) modulate bone mineral synthesis and decomposition as well as muscle tissues. FGF2 and other FGFs also exert regulatory roles. A growing body of research has focused on understanding the implications of FGF signaling in orthopedic degeneration. Moreover, an increasing number of potential targets within the FGF signaling have been identified, such as FGF9, FGF18, and FGF23. However, it should be noted that most of these discoveries are still in the experimental stage, and further studies are needed before clinical application can be considered. Presently, this review aims to document the association between the FGF signaling pathway and the development and progression of orthopedic diseases. Besides, current therapeutic strategies targeting the FGF signaling pathway to prevent and treat orthopedic degeneration will be evaluated.
Collapse
Affiliation(s)
- Heng-Zhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing-Lve Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Dong-Liang Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | - Ali Mobasheri
- Faculty of Medicine, Research Unit of Health Sciences and Technology, University of Oulu, 90014, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406, Vilnius, Lithuania.
- Department of Rheumatology and Clinical Immunology, Universitair Medisch Centrum Utrecht, Utrecht, 3508, GA, the Netherlands.
- Department of Joint Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000, Liège, Belgium.
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
10
|
Chen M, Lu Y, Liu Y, Liu Q, Deng S, Liu Y, Cui X, Liang J, Zhang X, Fan Y, Wang Q. Injectable Microgels with Hybrid Exosomes of Chondrocyte-Targeted FGF18 Gene-Editing and Self-Renewable Lubrication for Osteoarthritis Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312559. [PMID: 38266145 DOI: 10.1002/adma.202312559] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Abnormal silencing of fibroblast growth factor (FGF) signaling significantly contributes to joint dysplasia and osteoarthritis (OA); However, the clinical translation of FGF18-based protein drugs is hindered by their short half-life, low delivery efficiency and the need for repeated articular injections. This study proposes a CRISPR/Cas9-based approach to effectively activate the FGF18 gene of OA chondrocytes at the genome level in vivo, using chondrocyte-affinity peptide (CAP) incorporated hybrid exosomes (CAP/FGF18-hyEXO) loaded with an FGF18-targeted gene-editing tool. Furthermore, CAP/FGF18-hyEXO are encapsulated in methacrylic anhydride-modified hyaluronic (HAMA) hydrogel microspheres via microfluidics and photopolymerization to create an injectable microgel system (CAP/FGF18-hyEXO@HMs) with self-renewable hydration layers to provide persistent lubrication in response to frictional wear. Together, the injectable CAP/FGF18-hyEXO@HMs, combined with in vivo FGF18 gene editing and continuous lubrication, have demonstrated their capacity to synergistically promote cartilage regeneration, decrease inflammation, and prevent ECM degradation both in vitro and in vivo, holding great potential for clinical translation.
Collapse
Affiliation(s)
- Manyu Chen
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yan Lu
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yuhan Liu
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, P. R. China
| | - Quanying Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Siyan Deng
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yuan Liu
- Orthopedics Research Institute, Department of Orthopedics, West China Hospital Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaolin Cui
- School of medicine the Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
- Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine University of Otago, Christchurch, 8140, New Zealand
| | - Jie Liang
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| |
Collapse
|
11
|
Sun K, Sun J, Yan C, Sun J, Xu X, Shi J. Sympathetic Neurotransmitter, VIP, Delays Intervertebral Disc Degeneration via FGF18/FGFR2-Mediated Activation of Akt Signaling Pathway. Adv Biol (Weinh) 2024; 8:e2300250. [PMID: 38047500 DOI: 10.1002/adbi.202300250] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/28/2023] [Indexed: 12/05/2023]
Abstract
Neuromodulation-related intervertebral disc degeneration (IVDD) is a novel IVDD pattern and are proposed recently. However, the mechanistic basis of neuromodulation and intervertebral disc (IVD) homeostasis remains unclear. Here, this study aimed to investigate the expression of postganglionic sympathetic nerve fiber-derived vasoactive intestinal peptide (VIP) system in human IVD tissue, and to assess the role of VIP-related neuromodulation in IVDD. Patient samples and in vitro cell experiments showed that the expression of receptors for VIP is negatively correlated with the severity of IVDD, and the administration of exogenous VIP can ameliorate interleukin 1β-induced nucleus pulposus (NP) cell apoptosis and inflammation. Further mRNA-seq analysis revealed that fibroblast growth factor 18- (FGF18)-mediated activation of V-akt murine thymoma viral oncogene homolog signaling pathway is involved in the protective effects of VIP on inflammation-induced NP cell degeneration. Further analysis identified VIP via its receptor vasoactive intestinal peptide receptor 2 can directly result in decreased expression of miR-15a-5p, which targeted FGF18. Finally, in vivo mice lumbar IVDD model confirmed that focally exogenous administration of VIP can effectively ameliorated the progression of IVDD, as shown by the radiological and histological analysis. In conclusion, these results indicated that sympathetic neurotransmitter, VIP, delayed IVDD via FGF18/FGFR2-mediated activation of V-akt murine thymoma viral oncogene homolog signaling pathway, which will broaden the horizon concerning how the neuromodulation correlates with IVDD and shed new light on novel therapeutical alternatives to IVDD.
Collapse
Affiliation(s)
- Kaiqiang Sun
- Department of Orthopedic Surgery, Changzheng Hospital, Navy Medical University, No.415 Fengyang Road, Shanghai, 200003, China
- Department of Orthopedics, Naval Medical Center of PLA, Navy Medical University, No.338 Western HuaiHai Road, Shanghai, 200003, China
| | - Jiuyi Sun
- Department of Orthopedics, Naval Medical Center of PLA, Navy Medical University, No.338 Western HuaiHai Road, Shanghai, 200003, China
| | - Chen Yan
- Department of Orthopedic Surgery, Changzheng Hospital, Navy Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Jingchuan Sun
- Department of Orthopedic Surgery, Changzheng Hospital, Navy Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Ximing Xu
- Department of Orthopedic Surgery, Changzheng Hospital, Navy Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Jiangang Shi
- Department of Orthopedic Surgery, Changzheng Hospital, Navy Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| |
Collapse
|
12
|
Chen M, Jiang Z, Zou X, You X, Cai Z, Huang J. Advancements in tissue engineering for articular cartilage regeneration. Heliyon 2024; 10:e25400. [PMID: 38352769 PMCID: PMC10862692 DOI: 10.1016/j.heliyon.2024.e25400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024] Open
Abstract
Articular cartilage injury is a prevalent clinical condition resulting from trauma, tumors, infection, osteoarthritis, and other factors. The intrinsic lack of blood vessels, nerves, and lymphatic vessels within cartilage tissue severely limits its self-regenerative capacity after injury. Current treatment options, such as conservative drug therapy and joint replacement, have inherent limitations. Achieving perfect regeneration and repair of articular cartilage remains an ongoing challenge in the field of regenerative medicine. Tissue engineering has emerged as a key focus in articular cartilage injury research, aiming to utilize cultured and expanded tissue cells combined with suitable scaffold materials to create viable, functional tissues. This review article encompasses the latest advancements in seed cells, scaffolds, and cytokines. Additionally, the role of stimulatory factors including cytokines and growth factors, genetic engineering techniques, biophysical stimulation, and bioreactor systems, as well as the role of scaffolding materials including natural scaffolds, synthetic scaffolds, and nanostructured scaffolds in the regeneration of cartilage tissues are discussed. Finally, we also outline the signaling pathways involved in cartilage regeneration. Our review provides valuable insights for scholars to address the complex problem of cartilage regeneration and repair.
Collapse
Affiliation(s)
- Maohua Chen
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhiyuan Jiang
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiuyuan Zou
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiaobo You
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhen Cai
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jinming Huang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Li Z, Chen H, Chen C. Pain sensitivity genes as therapeutic targets in knee osteoarthritis: A comprehensive analysis. Mol Pain 2024; 20:17448069241289961. [PMID: 39313491 PMCID: PMC11456193 DOI: 10.1177/17448069241289961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024] Open
Abstract
Pain sensitivity is a significant factor in knee osteoarthritis (KOA), influencing patient outcomes and complicating treatment. Genetic differences, particularly in pain-sensing genes (PSRGs), are known to contribute to the variability in pain experiences among KOA patients. This study aims to systematically analyze PSRGs in KOA to better understand their role and potential as therapeutic targets. We utilized bulk RNA-seq data from the GSE114007 and GSE169077 datasets to identify differentially expressed genes, with 20 genes found to be significantly altered. Key PSRGs, including PENK, NGF, HOXD1, and TRPA1, were identified using LASSO, SVM, and random forest algorithms. Further, KEGG and GO enrichment analyses revealed pathways such as "Neuroactive ligand-receptor interaction" and "ECM-receptor interaction," which were validated through external datasets. Single-cell RNA-seq analysis from GSE152805, GSE133449, and GSE104782 datasets demonstrated the heterogeneity and dynamic expression of PSRGs across different cell subpopulations in synovium, meniscus, and cartilage samples. UMAP and pseudotime analyses were used to visualize spatial distribution and developmental trajectories of these genes. The findings emphasize the critical roles of PSRGs in KOA, highlighting their potential as therapeutic targets and suggesting that integrating genetic information into clinical practice could significantly improve pain management and treatment strategies for KOA.
Collapse
Affiliation(s)
- Zirui Li
- School of Physical Education and Health, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haicheng Chen
- The Department of Orthopedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chujie Chen
- School of Physical Education and Health, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
14
|
Yu L, Cavelier S, Hannon B, Wei M. Recent development in multizonal scaffolds for osteochondral regeneration. Bioact Mater 2023; 25:122-159. [PMID: 36817819 PMCID: PMC9931622 DOI: 10.1016/j.bioactmat.2023.01.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/30/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Osteochondral (OC) repair is an extremely challenging topic due to the complex biphasic structure and poor intrinsic regenerative capability of natural osteochondral tissue. In contrast to the current surgical approaches which yield only short-term relief of symptoms, tissue engineering strategy has been shown more promising outcomes in treating OC defects since its emergence in the 1990s. In particular, the use of multizonal scaffolds (MZSs) that mimic the gradient transitions, from cartilage surface to the subchondral bone with either continuous or discontinuous compositions, structures, and properties of natural OC tissue, has been gaining momentum in recent years. Scrutinizing the latest developments in the field, this review offers a comprehensive summary of recent advances, current hurdles, and future perspectives of OC repair, particularly the use of MZSs including bilayered, trilayered, multilayered, and gradient scaffolds, by bringing together onerous demands of architecture designs, material selections, manufacturing techniques as well as the choices of growth factors and cells, each of which possesses its unique challenges and opportunities.
Collapse
Affiliation(s)
- Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Sacha Cavelier
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Brett Hannon
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
| | - Mei Wei
- Biomedical Engineering Program, Ohio University, Athens, OH, 45701, USA
- Department of Mechanical Engineering, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
15
|
Michelacci YM, Baccarin RYA, Rodrigues NNP. Chondrocyte Homeostasis and Differentiation: Transcriptional Control and Signaling in Healthy and Osteoarthritic Conditions. Life (Basel) 2023; 13:1460. [PMID: 37511835 PMCID: PMC10381434 DOI: 10.3390/life13071460] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/13/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Chondrocytes are the main cell type in articular cartilage. They are embedded in an avascular, abundant, and specialized extracellular matrix (ECM). Chondrocytes are responsible for the synthesis and turnover of the ECM, in which the major macromolecular components are collagen, proteoglycans, and non-collagen proteins. The crosstalk between chondrocytes and the ECM plays several relevant roles in the regulation of cell phenotype. Chondrocytes live in an avascular environment in healthy cartilage with a low oxygen supply. Although chondrocytes are adapted to anaerobic conditions, many of their metabolic functions are oxygen-dependent, and most cartilage oxygen is supplied by the synovial fluid. This review focuses on the transcription control and signaling responsible for chondrocyte differentiation, homeostasis, senescence, and cell death and the changes that occur in osteoarthritis. The effects of chondroitin sulfate and other molecules as anti-inflammatory agents are also approached and analyzed.
Collapse
Affiliation(s)
- Yara M Michelacci
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, SP, Brazil
| | - Raquel Y A Baccarin
- Faculdade de Medicina Veterinária e Zootecnia, Universidade São Paulo, São Paulo 05508-270, SP, Brazil
| | - Nubia N P Rodrigues
- Faculdade de Medicina Veterinária e Zootecnia, Universidade São Paulo, São Paulo 05508-270, SP, Brazil
| |
Collapse
|
16
|
Zhang X, Pu X, Pi C, Xie J. The role of fibroblast growth factor 7 in cartilage development and diseases. Life Sci 2023:121804. [PMID: 37245839 DOI: 10.1016/j.lfs.2023.121804] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Fibroblast growth factor 7 (FGF7), also known as keratinocyte growth factor (KGF), shows a crucial biological significance in tissue development, wound repair, tumorigenesis, and immune reconstruction. In the skeletal system, FGF7 directs the cellular synaptic extension of individual cells and facilities functional gap junction intercellular communication of a collective of cells. Moreover, it promotes the osteogenic differentiation of stem cells via a cytoplasmic signaling network. For cartilage, reports have indicated the potential role of FGF7 on the regulation of key molecules Cx43 in cartilage and Runx2 in hypertrophic cartilage. However, the molecular mechanism of FGF7 in chondrocyte behaviors and cartilage pathological process remains largely unknown. In this review, we systematically summarize the recent biological function of FGF7 and its regulatory role on chondrocytes and cartilage diseases, especially through the hot focus of two key molecules, Runx2 and Cx43. The current knowledge of FGF7 on the physiological and pathological processes of chondrocytes and cartilage provides us new cues for wound repair of cartilage defect and therapy of cartilage diseases.
Collapse
Affiliation(s)
- Xinyue Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaohua Pu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
17
|
Yao Q, Wu X, Tao C, Gong W, Chen M, Qu M, Zhong Y, He T, Chen S, Xiao G. Osteoarthritis: pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther 2023; 8:56. [PMID: 36737426 PMCID: PMC9898571 DOI: 10.1038/s41392-023-01330-w] [Citation(s) in RCA: 494] [Impact Index Per Article: 247.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder that leads to disability and affects more than 500 million population worldwide. OA was believed to be caused by the wearing and tearing of articular cartilage, but it is now more commonly referred to as a chronic whole-joint disorder that is initiated with biochemical and cellular alterations in the synovial joint tissues, which leads to the histological and structural changes of the joint and ends up with the whole tissue dysfunction. Currently, there is no cure for OA, partly due to a lack of comprehensive understanding of the pathological mechanism of the initiation and progression of the disease. Therefore, a better understanding of pathological signaling pathways and key molecules involved in OA pathogenesis is crucial for therapeutic target design and drug development. In this review, we first summarize the epidemiology of OA, including its prevalence, incidence and burdens, and OA risk factors. We then focus on the roles and regulation of the pathological signaling pathways, such as Wnt/β-catenin, NF-κB, focal adhesion, HIFs, TGFβ/ΒΜP and FGF signaling pathways, and key regulators AMPK, mTOR, and RUNX2 in the onset and development of OA. In addition, the roles of factors associated with OA, including MMPs, ADAMTS/ADAMs, and PRG4, are discussed in detail. Finally, we provide updates on the current clinical therapies and clinical trials of biological treatments and drugs for OA. Research advances in basic knowledge of articular cartilage biology and OA pathogenesis will have a significant impact and translational value in developing OA therapeutic strategies.
Collapse
Affiliation(s)
- Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chu Tao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Minghao Qu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
18
|
Pan YN, Jia C, Yu JP, Wu ZW, Xu GC, Huang YX. Fibroblast growth factor 9 reduces TBHP-induced oxidative stress in chondrocytes and diminishes mouse osteoarthritis by activating ERK/Nrf2 signaling pathway. Int Immunopharmacol 2023; 114:109606. [PMID: 36700776 DOI: 10.1016/j.intimp.2022.109606] [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: 08/10/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Osteoarthritis (OA) is a degenerative and progressive disease that affects joints. Pathologically, it is characterized by oxidative stress-mediated excessive chondrocyte apoptosis and mitochondrial dysfunction. Fibroblast growth factor 9 (FGF9) has been shown to exert antioxidant effects and prevent degenerative diseases by activating ERK-related signaling pathways. However, the mechanism of FGF9 in the pathogenesis of OA and its relationship with anti-oxidative stress and related pathways are unclear. In this study, mice with medial meniscus instability (DMM) were used as the in vivo model whereas TBHP-induced chondrocytes served as the in vitro model to explore the mechanism underlying the effects of FGF9 in OA and its association with anti-oxidative stress. Results showed that FGF9 reduced oxidative stress, apoptosis, and mitochondrial dysfunction in TBHP-treated chondrocytes and promoted the nuclear translocation of Nrf2 to activate the Nrf2/HO1 signaling pathway. Interestingly, silencing the Nrf2 gene or blocking the ERK signaling pathway abolished the antioxidant effects of FGF9. FGF9 treatment reduced joint space narrowing, cartilage ossification, and synovial thickening in the DMM model mice. In conclusion, the present findings demonstrate that FGF9 can inhibit TBHP-induced oxidative stress in chondrocytes through the ERK and Nrf2-HO1 signaling pathways and prevent the progression of OA in vivo.
Collapse
Affiliation(s)
- Yi-Nan Pan
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Chao Jia
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jia-Pei Yu
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhou-Wei Wu
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Guo-Chao Xu
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yi-Xing Huang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
19
|
Kuhns BD, Reuter JM, Hansen VL, Soles GL, Jonason JH, Ackert-Bicknell CL, Wu CL, Giordano BD. Whole-genome RNA sequencing identifies distinct transcriptomic profiles in impingement cartilage between patients with femoroacetabular impingement and hip osteoarthritis. J Orthop Res 2022. [PMID: 36463522 DOI: 10.1002/jor.25485] [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] [Received: 09/30/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022]
Abstract
Femoroacetabular impingement (FAI) has a strong clinical association with the development of hip osteoarthritis (OA); however, the pathobiological mechanisms underlying the transition from focal impingement to global joint degeneration remain poorly understood. The purpose of this study is to use whole-genome RNA sequencing to identify and subsequently validate differentially expressed genes (DEGs) in femoral head articular cartilage samples from patients with FAI and hip OA secondary to FAI. Thirty-seven patients were included in the study with whole-genome RNA sequencing performed on 10 gender-matched patients in the FAI and OA cohorts and the remaining specimens were used for validation analyses. We identified a total of 3531 DEGs between the FAI and OA cohorts with multiple targets for genes implicated in canonical OA pathways. Quantitative reverse transcription-polymerase chain reaction validation confirmed increased expression of FGF18 and WNT16 in the FAI samples, while there was increased expression of MMP13 and ADAMTS4 in the OA samples. Expression levels of FGF18 and WNT16 were also higher in FAI samples with mild cartilage damage compared to FAI samples with severe cartilage damage or OA cartilage. Our study further expands the knowledge regarding distinct genetic reprogramming in the cartilage between FAI and hip OA patients. We independently validated the results of the sequencing analysis and found increased expression of anabolic markers in patients with FAI and minimal histologic cartilage damage, suggesting that anabolic signaling may be increased in early FAI with a transition to catabolic and inflammatory gene expression as FAI progresses towards more severe hip OA. Clinical significance:Cam-type FAI has a strong clinical association with hip OA; however, the cellular pathophysiology of disease progression remains poorly understood. Several previous studies have demonstrated increased expression of inflammatory markers in FAI cartilage samples, suggesting the involvement of these inflammatory pathways in the disease progression. Our study further expands the knowledge regarding distinct genetic reprogramming in the cartilage between FAI and hip OA patients. In addition to differences in inflammatory gene expression, we also identified differential expression in multiple pathways involved in hip OA progression.
Collapse
Affiliation(s)
- Benjamin D Kuhns
- Center for Regenerative and Personalized Medicine, Steadman-Philippon Research Institute, Vail, Colorado, USA
| | - John M Reuter
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| | - Victoria L Hansen
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| | - Gillian L Soles
- Department of Orthopedic Surgery, University of California Davis Health System, Sacramento, California, USA
| | - Jennifer H Jonason
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| | - Cheryl L Ackert-Bicknell
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chia-Lung Wu
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| | - Brian D Giordano
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
20
|
Zhao XX, Xie WQ, Xiao WF, Li HZ, Naranmandakh S, Bruyere O, Reginster JY, Li YS. Perlecan: Roles in osteoarthritis and potential treating target. Life Sci 2022; 312:121190. [PMID: 36379311 DOI: 10.1016/j.lfs.2022.121190] [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: 09/10/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Osteoarthritis (OA) is the most common joint disease, affecting hundreds of millions of people globally, which leads to a high cost of treatment and further medical care and an apparent decrease in patient prognosis. The recent view of OA pathogenesis is that increased vascularity, bone remodeling, and disordered turnover are influenced by multivariate risk factors, such as age, obesity, and overloading. The view also reveals the gap between the development of these processes and early stage risk factors. This review presents the latest research on OA-related signaling pathways and analyzes the potential roles of perlecan, a typical component of the well-known protective structure against osteoarthritic pericellular matrix (PCM). Based on the experimental results observed in end-stage OA models, we summarized and analyzed the role of perlecan in the development of OA. In normal cartilage, it plays a protective role by maintaining the integrin of PCM and sequesters growth factors. Second, perlecan in cartilage is required to not only activate vascular epithelium growth factor receptor (VEGFR) signaling of endothelial cells for vascular invasion and catabolic autophagy, but also for different signaling pathways for the catabolic and anabolic actions of chondrocytes. Finally, perlecan may participate in pain sensitization pathways.
Collapse
Affiliation(s)
- Xiao-Xuan Zhao
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya School of Medicine, Central South University, Changsha 410083, Hunan, China
| | - Wen-Qing Xie
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Wen-Feng Xiao
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Heng-Zhen Li
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shinen Naranmandakh
- School of Arts and Sciences, National University of Mongolia, Sukhbaatar district, 14201 Ulaanbaatar, Mongolia
| | - Olivier Bruyere
- Department of Public Health, Epidemiology and Health Economics, University of Liège, CHU Sart Tilman B23, 4000 Liège, Belgium
| | - Jean-Yves Reginster
- Department of Public Health, Epidemiology and Health Economics, University of Liège, CHU Sart Tilman B23, 4000 Liège, Belgium.
| | - Yu-Sheng Li
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
21
|
The synergistic mechanism of fibroblast growth factor 18 and integrin β1 in rat abdominal aortic aneurysm repair. BMC Cardiovasc Disord 2022; 22:415. [PMID: 36115958 PMCID: PMC9482292 DOI: 10.1186/s12872-022-02851-y] [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] [Received: 06/02/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
Background Abdominal aortic aneurysms have a high mortality rate. While surgery is the preferred treatment method, the biological repair of abdominal aortic aneurysms is being increasingly studied. We performed cellular and animal experiments to investigate the simultaneous function and mechanism of fibroblast growth factor 18 and integrin β1 in the biological repair of abdominal aortic aneurysms. Methods Endothelial and smooth muscle cells of rat arteries were used for the cellular experiments. Intracellular integrin β1 expression was regulated through lentiviral transfection. Interventions with fibroblast growth factor 18 were determined according to the experimental protocol. Several methods were used to detect the expression of elastic fiber component proteins, cell proliferation, and migratory activity of endothelial and smooth muscle cells after different treatments. For animal experiments, abdominal aortic aneurysms were induced in rats by wrapping the abdominal aortae in sterile cotton balls soaked with CaCl2 solution. Fibroblast growth factor 18 was administered through tail vein injections. The local expression of integrin β1 was regulated through lentiviral injections into the adventitia of the abdominal aortic aneurysms. The abdominal aortae were harvested for pathological examinations and tensile mechanical tests. Results The expression of integrin β1 in endothelial and smooth muscle cells could be regulated effectively through lentiviral transfection. Animal and cellular experiments showed that fibroblast growth factor 18 + integrin β1 could improve the expression of elastic fiber component proteins and enhance the migratory and proliferative activities of smooth muscle and endothelial cells. Moreover, animal experiments showed that fibroblast growth factor 18 + integrin β1 could enhance the aortic integrity to withstand stretch of aortic aneurysm tissue. Conclusion Fibroblast growth factor 18 + integrin β1 improved the biological repair of abdominal aortic aneurysms in rats by increasing the expression of elastic proteins, improving the migratory and proliferative abilities of endothelial and smooth muscle cells, and improving aortic remodeling. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02851-y.
Collapse
|
22
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
23
|
Pothiawala A, Sahbazoglu BE, Ang BK, Matthias N, Pei G, Yan Q, Davis BR, Huard J, Zhao Z, Nakayama N. GDF5+ chondroprogenitors derived from human pluripotent stem cells preferentially form permanent chondrocytes. Development 2022; 149:dev196220. [PMID: 35451016 PMCID: PMC9245189 DOI: 10.1242/dev.196220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 04/07/2022] [Indexed: 12/02/2023]
Abstract
It has been established in the mouse model that during embryogenesis joint cartilage is generated from a specialized progenitor cell type, distinct from that responsible for the formation of growth plate cartilage. We recently found that mesodermal progeny of human pluripotent stem cells gave rise to two types of chondrogenic mesenchymal cells in culture: SOX9+ and GDF5+ cells. The fast-growing SOX9+ cells formed in vitro cartilage that expressed chondrocyte hypertrophy markers and readily underwent mineralization after ectopic transplantation. In contrast, the slowly growing GDF5+ cells derived from SOX9+ cells formed cartilage that tended to express low to undetectable levels of chondrocyte hypertrophy markers, but expressed PRG4, a marker of embryonic articular chondrocytes. The GDF5+-derived cartilage remained largely unmineralized in vivo. Interestingly, chondrocytes derived from the GDF5+ cells seemed to elicit these activities via non-cell-autonomous mechanisms. Genome-wide transcriptomic analyses suggested that GDF5+ cells might contain a teno/ligamento-genic potential, whereas SOX9+ cells resembled neural crest-like progeny-derived chondroprogenitors. Thus, human pluripotent stem cell-derived GDF5+ cells specified to generate permanent-like cartilage seem to emerge coincidentally with the commitment of the SOX9+ progeny to the tendon/ligament lineage.
Collapse
Affiliation(s)
- Azim Pothiawala
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Berke E. Sahbazoglu
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Bryan K. Ang
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nadine Matthias
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Guangsheng Pei
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Qing Yan
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Brian R. Davis
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Johnny Huard
- Department of Orthopedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Naoki Nakayama
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Orthopedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
24
|
Abstract
The superficial zone of articular cartilage contributes to smooth joint motion through the production of proteoglycan 4 (PRG4), also known as lubricin. Recent studies indicate novel effects of PRG4 as a signaling molecule, other than a simple extracellular matrix protein. Additionally, the accumulating evidence displays that various molecules and signaling pathways are involved in regulating the superficial zone and PRG4 expression. In addition, Prg4-expressing cells include a progenitor population of articular chondrocytes. Several non-clinical and clinical studies have shown that PRG4 and related molecules are promising candidates for disease-modifying drugs for treating osteoarthritis. Since PRG4 is also expressed in the synovium, tendons, and ligaments, further studies of PRG4-related pathways and PRG4-positive cells may elucidate the mechanisms underlying joint homeostasis.
Collapse
|
25
|
Schwartz NB, Domowicz MS. Roles of Chondroitin Sulfate Proteoglycans as Regulators of Skeletal Development. Front Cell Dev Biol 2022; 10:745372. [PMID: 35465334 PMCID: PMC9026158 DOI: 10.3389/fcell.2022.745372] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
The extracellular matrix (ECM) is critically important for most cellular processes including differentiation, morphogenesis, growth, survival and regeneration. The interplay between cells and the ECM often involves bidirectional signaling between ECM components and small molecules, i.e., growth factors, morphogens, hormones, etc., that regulate critical life processes. The ECM provides biochemical and contextual information by binding, storing, and releasing the bioactive signaling molecules, and/or mechanical information that signals from the cell membrane integrins through the cytoskeleton to the nucleus, thereby influencing cell phenotypes. Using these dynamic, reciprocal processes, cells can also remodel and reshape the ECM by degrading and re-assembling it, thereby sculpting their environments. In this review, we summarize the role of chondroitin sulfate proteoglycans as regulators of cell and tissue development using the skeletal growth plate model, with an emphasis on use of naturally occurring, or created mutants to decipher the role of proteoglycan components in signaling paradigms.
Collapse
Affiliation(s)
- Nancy B. Schwartz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, United States
- Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, Chicago, IL, United States
- *Correspondence: Nancy B. Schwartz,
| | - Miriam S. Domowicz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
26
|
Chen H, Cui Y, Zhang D, Xie J, Zhou X. The role of fibroblast growth factor 8 in cartilage development and disease. J Cell Mol Med 2022; 26:990-999. [PMID: 35001536 PMCID: PMC8831980 DOI: 10.1111/jcmm.17174] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factor 8 (FGF‐8), also known as androgen‐induced growth factor (AIGF), is presumed to be a potent mitogenic cytokine that plays important roles in early embryonic development, brain formation and limb development. In the bone environment, FGF‐8 produced or received by chondrocyte precursor cells binds to fibroblast growth factor receptor (FGFR), causing different levels of activation of downstream signalling pathways, such as phospholipase C gamma (PLCγ)/Ca2+, RAS/mitogen‐activated protein kinase‐extracellular regulated protein kinases (RAS/MAPK‐MEK‐ERK), and Wnt‐β‐catenin‐Axin2 signalling, and ultimately controlling chondrocyte proliferation, differentiation, cell survival and migration. However, the molecular mechanism of FGF‐8 in normal or pathological cartilage remains unclear, and thus, FGF‐8 represents a novel exploratory target for studies of chondrocyte development and cartilage disease progression. In this review, studies assessing the relationship between FGF‐8 and chondrocytes that have been published in the past 5 years are systematically summarized to determine the probable mechanism and physiological effect of FGF‐8 on chondrocytes. Based on the existing research results, a therapeutic regimen targeting FGF‐8 is proposed to explore the possibility of treating chondrocyte‐related diseases.
Collapse
Affiliation(s)
- Haoran Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujia Cui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Song Z, Li Y, Shang C, Shang G, Kou H, Li J, Chen S, Liu H. Sprifermin: Effects on Cartilage Homeostasis and Therapeutic Prospects in Cartilage-Related Diseases. Front Cell Dev Biol 2022; 9:786546. [PMID: 34970547 PMCID: PMC8712868 DOI: 10.3389/fcell.2021.786546] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/23/2021] [Indexed: 11/15/2022] Open
Abstract
When suffering from osteoarthritis (OA), articular cartilage homeostasis is out of balance and the living quality declines. The treatment of knee OA has always been an unsolved problem in the world. At present, symptomatic treatment is mainly adopted for OA. Drug therapy is mainly used to relieve pain symptoms, but often accompanied with adverse reactions; surgical treatment involves the problem of poor integration between the repaired or transplanted tissues and the natural cartilage, leading to the failure of repair. Biotherapy which aims to promote cartilage in situ regeneration and to restore endochondral homeostasis is expected to be an effective method for the prevention and treatment of OA. Disease-modifying osteoarthritis drugs (DMOADs) are intended for targeted treatment of OA. The DMOADs prevent excessive destruction of articular cartilage through anti-catabolism and stimulate tissue regeneration via excitoanabolic effects. Sprifermin (recombinant human FGF18, rhFGF18) is an effective DMOAD, which can not only promote the proliferation of articular chondrocyte and the synthesis of extracellular matrix, increase the thickness of cartilage in a dose-dependent manner, but also inhibit the activity of proteolytic enzymes and remarkedly slow down the degeneration of cartilage. This paper reviews the unique advantages of Sprifermin in repairing cartilage injury and improving cartilage homeostasis, aiming to provide an important strategy for the effective prevention and treatment of cartilage injury-related diseases.
Collapse
Affiliation(s)
- Zongmian Song
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Chunfeng Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowei Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongwei Kou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinfeng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
Shestovskaya MV, Bozhkova SA, Sopova JV, Khotin MG, Bozhokin MS. Methods of Modification of Mesenchymal Stem Cells and Conditions of Their Culturing for Hyaline Cartilage Tissue Engineering. Biomedicines 2021; 9:biomedicines9111666. [PMID: 34829895 PMCID: PMC8615732 DOI: 10.3390/biomedicines9111666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
The use of mesenchymal stromal cells (MSCs) for tissue engineering of hyaline cartilage is a topical area of regenerative medicine that has already entered clinical practice. The key stage of this procedure is to create conditions for chondrogenic differentiation of MSCs, increase the synthesis of hyaline cartilage extracellular matrix proteins by these cells and activate their proliferation. The first such works consisted in the indirect modification of cells, namely, in changing the conditions in which they are located, including microfracturing of the subchondral bone and the use of 3D biodegradable scaffolds. The most effective methods for modifying the cell culture of MSCs are protein and physical, which have already been partially introduced into clinical practice. Genetic methods for modifying MSCs, despite their effectiveness, have significant limitations. Techniques have not yet been developed that allow studying the effectiveness of their application even in limited groups of patients. The use of MSC modification methods allows precise regulation of cell culture proliferation, and in combination with the use of a 3D biodegradable scaffold, it allows obtaining a hyaline-like regenerate in the damaged area. This review is devoted to the consideration and comparison of various methods used to modify the cell culture of MSCs for their use in regenerative medicine of cartilage tissue.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Svetlana A. Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
| | - Julia V. Sopova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Mikhail G. Khotin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Mikhail S. Bozhokin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
29
|
Lohova E, Vitenberga-Verza Z, Kazoka D, Pilmane M. Local Defence System in Healthy Lungs. Clin Pract 2021; 11:728-746. [PMID: 34698129 PMCID: PMC8544484 DOI: 10.3390/clinpract11040088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/23/2021] [Accepted: 09/08/2021] [Indexed: 11/22/2022] Open
Abstract
Background: The respiratory system is one of the main entrance gates for infection. The aim of this work was to compare the appearance of specific mucosal pro-inflammatory and common anti-microbial defence factors in healthy lung tissue, from an ontogenetic point of view. Materials and methods: Healthy lung tissues were collected from 15 patients (three females and 12 males) in the age range from 18 to 86. Immunohistochemistry to human β defensin 2 (HBD-2), human β defensin 3 (HBD-3), human β defensin 4 (HBD-4), cathelicidine (LL-37) and interleukine 17A (IL-17A) were performed. Results: The lung tissue material contained bronchial and lung parenchyma material in which no histological changes, connected with the inflammatory process, were detected. During the study, various statistically significant differences were detected in immunoreactive expression between different factors in all lung tissue structures. Conclusion: All healthy lung structures, but especially the cartilage, alveolar epithelium and the alveolar macrophages, are the main locations for the baseline synthesis of antimicrobial proteins and IL-17A. Cartilage shows high functional plasticity of this structure, including significant antimicrobial activity and participation in local lung protection response. Interrelated changes between antimicrobial proteins in different tissue confirm baseline synergistical cooperation of all these factors in healthy lung host defence.
Collapse
|
30
|
Aoqierbatu, Luo A, Shi Y, Na Y, Tuo Y. Microarray analysis of hub genes and pathways in damaged cartilage tissues of knee. Medicine (Baltimore) 2021; 100:e27183. [PMID: 34664844 PMCID: PMC8448002 DOI: 10.1097/md.0000000000027183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 08/21/2021] [Indexed: 11/25/2022] Open
Abstract
The aim of this study was to identify genes and functional pathways associated with damaged cartilage tissues of knee using microarray analysis.The gene expression profile GSE129147 including including 10 knee cartilage tissues from damaged side and 10 knee nonweight-bearing healthy cartilage was downloaded and bioinformatics analysis was made.A total of 182 differentially-expressed genes including 123 up-regulated and 59 down-regulated genes were identified from the GSE129147 dataset. Gene ontology and pathway enrichment analysis confirmed that extracellular matrix organization, collagen catabolic process, antigen processing and presentation of peptide or polysaccharide antigen, and endocytic vesicle membrane were strongly associated with cartilage injury. Furthermore, 10 hub differentially-expressed genes with a higher connectivity degree in protein-protein interactions network were found such as POSTN, FBN1, LOX, insulin-like growth factor binding proteins3, C3AR1, MMP2, ITGAM, CDKN2A, COL1A1, COL5A1.These hub genes and pathways provide a new perspective for revealing the potential pathological mechanisms and therapy strategy of cartilage injury.
Collapse
Affiliation(s)
- Aoqierbatu
- Department of Mongolian Osteopath, International Hospital of Mongolian Medicine, Saihan District, Hohhot, Inner Mongolia Autonomous Region, China
| | - Aqilatu Luo
- Department of Mongolian Osteopath, International Hospital of Mongolian Medicine, Saihan District, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yuting Shi
- Cardiac Function Department, Cadre Health Care Center, Inner Mongolia Autonomous Region People's Hospital, Saihan District, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yuyan Na
- Department of Arthroscopy and Sports Medicine, the Second Affiliated Hospital of Inner Mongolia Medical University, Huimin District, Hohhot, Inner Mongolia Autonomous Region, China
| | - Ya Tuo
- Department of Anesthesia, the Second Affiliated Hospital of Inner Mongolia Medical University, Huimin District, Hohhot, Inner Mongolia Autonomous Region, China
| |
Collapse
|
31
|
Li J, Wang X, Ruan G, Zhu Z, Ding C. Sprifermin: a recombinant human fibroblast growth factor 18 for the treatment of knee osteoarthritis. Expert Opin Investig Drugs 2021; 30:923-930. [PMID: 34427483 DOI: 10.1080/13543784.2021.1972970] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Osteoarthritis (OA) is a serious and incurable disease leading the disability. Surgical treatment is the last but not necessarily the best approach for patients with high risks and costs. However, there are no disease-modifying OA drugs (DMOADs) developed for the disease so far, leaving a huge unmet need for drug treatments. Sprifermin is a recombinant human fibroblast growth factor 18 (rhFGF18) and has been confirmed to have anabolic effects on articular cartilage, which makes it a promising DMOAD. AREAS COVERED The content of this review includes overview of the market, discovery and development, molecular mechanism, preclinical studies, clinical efficacy, safety, and tolerability of sprifermin. It examines the potential of sprifermin as a disease modifying drug for the treatment of knee OA. EXPERT OPINION Sprifermin could be one of the most promising DMOADs, especially for cartilage phenotype. Current studies show good tolerability and no safety concerns. Well-designed phase 3 clinical trials are required to examine its effects on symptoms and cartilage loss in knee OA.
Collapse
Affiliation(s)
- Jia Li
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoshuai Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guangfeng Ruan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.,Clinical Research Centre, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
32
|
Rejuvenated Stem/Progenitor Cells for Cartilage Repair Using the Pluripotent Stem Cell Technology. Bioengineering (Basel) 2021; 8:bioengineering8040046. [PMID: 33920285 PMCID: PMC8070387 DOI: 10.3390/bioengineering8040046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 01/19/2023] Open
Abstract
It is widely accepted that chondral defects in articular cartilage of adult joints are never repaired spontaneously, which is considered to be one of the major causes of age-related degenerative joint disorders, such as osteoarthritis. Since mobilization of subchondral bone (marrow) cells and addition of chondrocytes or mesenchymal stromal cells into full-thickness defects show some degrees of repair, the lack of self-repair activity in adult articular cartilage can be attributed to lack of reparative cells in adult joints. In contrast, during a fetal or embryonic stage, joint articular cartilage has a scar-less repair activity, suggesting that embryonic joints may contain cells responsible for such activity, which can be chondrocytes, chondroprogenitors, or other cell types such as skeletal stem cells. In this respect, the tendency of pluripotent stem cells (PSCs) to give rise to cells of embryonic characteristics will provide opportunity, especially for humans, to obtain cells carrying similar cartilage self-repair activity. Making use of PSC-derived cells for cartilage repair is still in a basic or preclinical research phase. This review will provide brief overviews on how human PSCs have been used for cartilage repair studies.
Collapse
|
33
|
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: 15] [Impact Index Per Article: 3.8] [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
|
34
|
Schulze-Tanzil G. Experimental Therapeutics for the Treatment of Osteoarthritis. J Exp Pharmacol 2021; 13:101-125. [PMID: 33603501 PMCID: PMC7887204 DOI: 10.2147/jep.s237479] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) therapy remains a large challenge since no causative treatment options are so far available. Despite some main pathways contributing to OA are identified its pathogenesis is still rudimentary understood. A plethora of therapeutically promising agents are currently tested in experimental OA research to find an opportunity to reverse OA-associated joint damage and prevent its progression. Hence, this review aims to summarize novelly emerging experimental approaches for OA. Due to the diversity of strategies shown only main aspects could be summarized here including herbal medicines, nanoparticular compounds, growth factors, hormones, antibody-, cell- and extracellular vesicle (EV)-based approaches, optimized tools for joint viscosupplementation, genetic regulators such as si- or miRNAs and promising combinations. An abundant multitude of compounds obtained from plants, environmental, autologous or synthetic sources have been identified with anabolic, anti-inflammatory, -catabolic and anti-apoptotic properties. Some ubiquitous signaling pathways such as wingless and Integration site-1 (Wnt), Sirtuin, Toll-like receptor (TLR), mammalian target of rapamycin (mTOR), Nuclear Factor (NF)-κB and complement are involved in OA and addressed by them. Hyaluronan (HA) provided benefit in OA since many decades, and novel HA formulations have been developed now with higher HA content and long-term stability achieved by cross-linking suitable to be combined with other agents such as components from herbals or chemokines to attract regenerative cells. pH- or inflammation-sensitive nanoparticular compounds could serve as versatile slow-release systems of active compounds, for example, miRNAs. Some light has been brought into the intimate regulatory network of small RNAs in the pathogenesis of OA which might be a novel avenue for OA therapy in future. Attraction of autologous regenerative cells by chemokines and exosome-based treatment strategies could also innovate OA therapy.
Collapse
Affiliation(s)
- Gundula Schulze-Tanzil
- Department of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg, Bavaria, Germany
| |
Collapse
|
35
|
McClurg O, Tinson R, Troeberg L. Targeting Cartilage Degradation in Osteoarthritis. Pharmaceuticals (Basel) 2021; 14:ph14020126. [PMID: 33562742 PMCID: PMC7916085 DOI: 10.3390/ph14020126] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis is a common, degenerative joint disease with significant socio-economic impact worldwide. There are currently no disease-modifying drugs available to treat the disease, making this an important area of pharmaceutical research. In this review, we assessed approaches being explored to directly inhibit metalloproteinase-mediated cartilage degradation and to counteract cartilage damage by promoting growth factor-driven repair. Metalloproteinase-blocking antibodies are discussed, along with recent clinical trials on FGF18 and Wnt pathway inhibitors. We also considered dendrimer-based approaches being developed to deliver and retain such therapeutics in the joint environment. These may reduce systemic side effects while improving local half-life and concentration. Development of such targeted anabolic therapies would be of great benefit in the osteoarthritis field.
Collapse
|
36
|
Abstract
The prevalence of osteoarthritis (OA) and the burden associated with the disease are steadily increasing worldwide, representing a major public health challenge for the coming decades. The lack of specific treatments for OA has led to it being recognized as a serious disease that has an unmet medical need. Advances in the understanding of OA pathophysiology have enabled the identification of a variety of potential therapeutic targets involved in the structural progression of OA, some of which are promising and under clinical investigation in randomized controlled trials. Emerging therapies include those targeting matrix-degrading proteases or senescent chondrocytes, promoting cartilage repair or limiting bone remodelling, local low-grade inflammation or Wnt signalling. In addition to these potentially disease-modifying OA drugs (DMOADs), several targets are being explored for the treatment of OA-related pain, such as nerve growth factor inhibitors. The results of these studies are expected to considerably reshape the landscape of OA management over the next few years. This Review describes the pathophysiological processes targeted by emerging therapies for OA, along with relevant clinical data and discussion of the main challenges for the further development of these therapies, to provide context for the latest advances in the field of pharmaceutical therapies for OA.
Collapse
|
37
|
Fibroblast growth factor signalling in osteoarthritis and cartilage repair. Nat Rev Rheumatol 2020; 16:547-564. [PMID: 32807927 DOI: 10.1038/s41584-020-0469-2] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
Abstract
Regulated fibroblast growth factor (FGF) signalling is a prerequisite for the correct development and homeostasis of articular cartilage, as evidenced by the fact that aberrant FGF signalling contributes to the maldevelopment of joints and to the onset and progression of osteoarthritis. Of the four FGF receptors (FGFRs 1-4), FGFR1 and FGFR3 are strongly implicated in osteoarthritis, and FGFR1 antagonists, as well as agonists of FGFR3, have shown therapeutic efficacy in mouse models of spontaneous and surgically induced osteoarthritis. FGF18, a high affinity ligand for FGFR3, is the only FGF-based drug currently in clinical trials for osteoarthritis. This Review covers the latest advances in our understanding of the molecular mechanisms that regulate FGF signalling during normal joint development and in the pathogenesis of osteoarthritis. Strategies for FGF signalling-based treatment of osteoarthritis and for cartilage repair in animal models and clinical trials are also introduced. An improved understanding of FGF signalling from a structural biology perspective, and of its roles in skeletal development and diseases, could unlock new avenues for discovery of modulators of FGF signalling that can slow or stop the progression of osteoarthritis.
Collapse
|
38
|
Nakayama N, Pothiawala A, Lee JY, Matthias N, Umeda K, Ang BK, Huard J, Huang Y, Sun D. Human pluripotent stem cell-derived chondroprogenitors for cartilage tissue engineering. Cell Mol Life Sci 2020; 77:2543-2563. [PMID: 31915836 PMCID: PMC11104892 DOI: 10.1007/s00018-019-03445-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
The cartilage of joints, such as meniscus and articular cartilage, is normally long lasting (i.e., permanent). However, once damaged, especially in large animals and humans, joint cartilage is not spontaneously repaired. Compensating the lack of repair activity by supplying cartilage-(re)forming cells, such as chondrocytes or mesenchymal stromal cells, or by transplanting a piece of normal cartilage, has been the basis of therapy for biological restoration of damaged joint cartilage. Unfortunately, current biological therapies face problems on a number of fronts. The joint cartilage is generated de novo from a specialized cell type, termed a 'joint progenitor' or 'interzone cell' during embryogenesis. Therefore, embryonic chondroprogenitors that mimic the property of joint progenitors might be the best type of cell for regenerating joint cartilage in the adult. Pluripotent stem cells (PSCs) are expected to differentiate in culture into any somatic cell type through processes that mimic embryogenesis, making human (h)PSCs a promising source of embryonic chondroprogenitors. The major research goals toward the clinical application of PSCs in joint cartilage regeneration are to (1) efficiently generate lineage-specific chondroprogenitors from hPSCs, (2) expand the chondroprogenitors to the number needed for therapy without loss of their chondrogenic activity, and (3) direct the in vivo or in vitro differentiation of the chondroprogenitors to articular or meniscal (i.e., permanent) chondrocytes rather than growth plate (i.e., transient) chondrocytes. This review is aimed at providing the current state of research toward meeting these goals. We also include our recent achievement of successful generation of "permanent-like" cartilage from long-term expandable, hPSC-derived ectomesenchymal chondroprogenitors.
Collapse
Affiliation(s)
- Naoki Nakayama
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA.
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA.
| | - Azim Pothiawala
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - John Y Lee
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Nadine Matthias
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - Katsutsugu Umeda
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Department of Pediatrics, Kyoto University School of Medicine, Kyoto, Japan
| | - Bryan K Ang
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Weil Cornell Medicine, New York, NY, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Yun Huang
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Deqiang Sun
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
39
|
Sprifermin (rhFGF18) versus vehicle induces a biphasic process of extracellular matrix remodeling in human knee OA articular cartilage ex vivo. Sci Rep 2020; 10:6011. [PMID: 32265494 PMCID: PMC7138815 DOI: 10.1038/s41598-020-63216-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Sprifermin, recombinant human fibroblast growth factor 18 (rhFGF18), induces cartilage regeneration in knees of patients with osteoarthritis (OA). We hypothesized that a temporal multiphasic process of extracellular matrix (ECM) degradation and formation underlie this effect. We aimed to characterize the temporal ECM remodeling of human knee OA articular cartilage in response to sprifermin treatment. Articular cartilage explants from patients with knee OA (npatients = 14) were cultured for 70 days, with permanent exposure to sprifermin (900, 450, 225 ng/mL), FGF18 (450 ng/mL), insulin-like growth factor-1 (100 ng/mL, positive control) or vehicle (nreplicates/treatment/patient = 2). Metabolic activity (AlamarBlue) and biomarkers of type IIB collagen (PIIBNP) formation (Pro-C2 enzyme-linked immunosorbent assay [ELISA]) and aggrecanase-mediated aggrecan neo-epitope NITEGE (AGNx1 ELISA) were quantified once a week. At end of culture (day 70), gene expression (quantitative reverse transcription polymerase chain reaction) and proteoglycan content (Safranin O/Fast green staining) were quantified. The cartilage had continuously increased metabolic activity, when treated with sprifermin/FGF18 compared to vehicle. During days 7-28 PIIBNP was decreased and NITEGE was increased, and during days 35-70 PIIBNP was increased. At end of culture, the cartilage had sustained proteoglycan content and relative expression of ACAN < COL2A1 < SOX9 < COL1A1, indicating that functional chondrocytes remained in the explants. Sprifermin induces a temporal biphasic cartilage remodeling in human knee OA articular cartilage explants, with early-phase increased aggrecanase activity and late-phase increased type II collagen formation.
Collapse
|
40
|
Antunes BP, Vainieri ML, Alini M, Monsonego-Ornan E, Grad S, Yayon A. Enhanced chondrogenic phenotype of primary bovine articular chondrocytes in Fibrin-Hyaluronan hydrogel by multi-axial mechanical loading and FGF18. Acta Biomater 2020; 105:170-179. [PMID: 31982592 DOI: 10.1016/j.actbio.2020.01.032] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
Abstract
Current treatments for cartilage lesions are often associated with fibrocartilage formation and donor site morbidity. Mechanical and biochemical stimuli play an important role in hyaline cartilage formation. Biocompatible scaffolds capable of transducing mechanical loads and delivering bioactive instructive factors may better support cartilage regeneration. In this study we aimed to test the interplay between mechanical and FGF-18 mediated biochemical signals on the proliferation and differentiation of primary bovine articular chondrocytes embedded in a chondro-conductive Fibrin-Hyaluronan (FB/HA) based hydrogel. Chondrocytes seeded in a Fibrin-HA hydrogel, with or without a chondro-inductive, FGFR3 selective FGF18 variant (FGF-18v) were loaded into a joint-mimicking bioreactor applying controlled, multi-axial movements, simulating the natural movements of articular joints. Samples were evaluated for DNA content, sulphated glycosaminoglycan (sGAG) accumulation, key chondrogenic gene expression markers and histology. Under moderate loading, samples produced particularly significant amounts of sGAG/DNA compared to unloaded controls. Interestingly there was no significant effect of FGF-18v on cartilage gene expression at rest. Following moderate multi-axial loading, FGF-18v upregulated the expression of Aggrecan (ACAN), Cartilage Oligomeric Matrix Protein (COMP), type II collagen (COL2) and Lubricin (PRG4). Moreover, the combination of load and FGF-18v, significantly downregulated Matrix Metalloproteinase-9 (MMP-9) and Matrix Metaloproteinase-13 (MMP-13), two of the most important factors contributing to joint destruction in OA. Biomimetic mechanical signals and FGF-18 may work in concert to support hyaline cartilage regeneration and repair. STATEMENT OF SIGNIFICANCE: Articular cartilage has very limited repair potential and focal cartilage lesions constitute a challenge for current standard clinical procedures. The aim of the present research was to explore novel procedures and constructs, based on biomaterials and biomechanical algorithms that can better mimic joints mechanical and biochemical stimulation to promote regeneration of damaged cartilage. Using a hydrogel-based platform for chondrocyte 3D culture revealed a synergy between mechanical forces and growth factors. Exploring the mechanisms underlying this mechano-biochemical interplay may enhance our understanding of cartilage remodeling and the development of new strategies for cartilage repair and regeneration.
Collapse
|
41
|
Evaluation of Relationship Between Common Variants in FGF18 Gene and Knee Osteoarthritis Susceptibility. Arch Med Res 2020; 51:76-81. [PMID: 32109713 DOI: 10.1016/j.arcmed.2019.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/26/2019] [Accepted: 12/10/2019] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Osteoarthritis (OA) is the most common type of arthritis disease. Previous etiological studies indicated that both environmental and genetic factors play important roles in the occurrence and development of knee OA (KOA). In the present study, we aimed to investigate the association between the FGF18 gene and susceptibility to KOA in Han Chinese population. METHODS A total of 2,556 unrelated Han Chinese individuals, including 866 KOA patients and 1,688 healthy controls, were recruited. Nine tag SNPs located within the gene region of FGF18 were selected for genotyping. Logistic models were fitted for single marker-based association analyses. Age, gender and BMI were included in each model as covariates. To investigate the functional consequences of significant SNP, we extracted expression quantitative trait loci (eQTL) data from the GTEx database. RESULTS SNP rs3884606 was significantly associated with the risk of KOA (OR [95% CI] = 1.25 [1.11-1.41], p = 0.0002) after adjusting for age, gender and BMI. The G allele was significantly associated with an increased risk of KOA. No significant eQTL signals could be identified for SNP rs3884606 on FGF18. CONCLUSION In this study, we identified a genetic polymorphism in FGF18 that was significantly associated with the risk of KOA based on samples with Chinese Han ancestry. Our findings replicated the recent GWAS report and supported an association between KOA and FGF18.
Collapse
|
42
|
Chijimatsu R, Saito T. Mechanisms of synovial joint and articular cartilage development. Cell Mol Life Sci 2019; 76:3939-3952. [PMID: 31201464 PMCID: PMC11105481 DOI: 10.1007/s00018-019-03191-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/30/2019] [Accepted: 06/11/2019] [Indexed: 12/29/2022]
Abstract
Articular cartilage is formed at the end of epiphyses in the synovial joint cavity and permanently contributes to the smooth movement of synovial joints. Most skeletal elements develop from transient cartilage by a biological process known as endochondral ossification. Accumulating evidence indicates that articular and growth plate cartilage are derived from different cell sources and that different molecules and signaling pathways regulate these two kinds of cartilage. As the first sign of joint development, the interzone emerges at the presumptive joint site within a pre-cartilage tissue. After that, joint cavitation occurs in the center of the interzone, and the cells in the interzone and its surroundings gradually form articular cartilage and the synovial joint. During joint development, the interzone cells continuously migrate out to the epiphyseal cartilage and the surrounding cells influx into the joint region. These complicated phenomena are regulated by various molecules and signaling pathways, including GDF5, Wnt, IHH, PTHrP, BMP, TGF-β, and FGF. Here, we summarize current literature and discuss the molecular mechanisms underlying joint formation and articular development.
Collapse
Affiliation(s)
- Ryota Chijimatsu
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
43
|
Hagan AS, Boylan M, Smith C, Perez-Santamarina E, Kowalska K, Hung IH, Lewis RM, Hajihosseini MK, Lewandoski M, Ornitz DM. Generation and validation of novel conditional flox and inducible Cre alleles targeting fibroblast growth factor 18 (Fgf18). Dev Dyn 2019; 248:882-893. [PMID: 31290205 PMCID: PMC7029619 DOI: 10.1002/dvdy.85] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/24/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Fibroblast growth factor 18 (FGF18) functions in the development of several tissues, including the lung, limb bud, palate, skeleton, central nervous system, and hair follicle. Mice containing a germline knockout of Fgf18 (Fgf18 -/- ) die shortly after birth. Postnatally, FGF18 is being evaluated for pathogenic roles in fibrosis and several types of cancer. The specific cell types that express FGF18 have been difficult to identify, and the function of FGF18 in postnatal development and tissue homeostasis has been hampered by the perinatal lethality of Fgf18 null mice. RESULTS We engineered a floxed allele of Fgf18 (Fgf18 flox ) that allows conditional gene inactivation and a CreERT2 knockin allele (Fgf18 CreERT2 ) that allows the precise identification of cells that express Fgf18 and their lineage. We validated the Fgf18 flox allele by targeting it in mesenchymal tissue and primary mesoderm during embryonic development, resulting in similar phenotypes to those observed in Fgf18 null mice. We also use the Fgf18 CreERT2 allele, in combination with a conditional fluorescent reporter to confirm known and identify new sites of Fgf18 expression. CONCLUSION These alleles will be useful to investigate FGF18 function during organogenesis and tissue homeostasis, and to target specific cell lineages at embryonic and postnatal time points.
Collapse
Affiliation(s)
- Andrew S. Hagan
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri
| | - Michael Boylan
- Cancer and Developmental Biology Lab, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Craig Smith
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri
| | | | - Karolina Kowalska
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Irene H. Hung
- Department of Neurobiology & Anatomy, University of Utah School of Medicine, Salt Lake City, Utah
| | - Renate M. Lewis
- Department of Neurology, Washington University School of Medicine, Saint Louis, Missouri
| | | | - Mark Lewandoski
- Cancer and Developmental Biology Lab, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - David M. Ornitz
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
44
|
Meloni GR, Farran A, Mohanraj B, Guehring H, Cocca R, Rabut E, Mauck RL, Dodge GR. Recombinant human FGF18 preserves depth-dependent mechanical inhomogeneity in articular cartilage. Eur Cell Mater 2019; 38:23-34. [PMID: 31393594 PMCID: PMC7273689 DOI: 10.22203/ecm.v038a03] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Articular cartilage is a specialised tissue that has a relatively homogenous endogenous cell population but a diverse extracellular matrix (ECM), with depth-dependent mechanical properties. Repair of this tissue remains an elusive clinical goal, with biological interventions preferred to arthroplasty in younger patients. Osteochondral transplantation (OCT) has emerged for the treatment of cartilage defects and osteoarthritis. Fresh allografts stored at 4 °C have been utilised, though matrix and cell viability loss remains an issue. To address this, several studies have developed media formulations to maintain cartilage explants in vitro. One promising factor for these applications is sprifermin, a human-recombinant fibroblast growth factor-18, which stimulates chondrocyte proliferation and matrix synthesis and is in clinical trials for the treatment of osteoarthritis. The study hypothesis was that addition of sprifermin during storage would maintain the unique depth-dependent mechanical profile of articular cartilage explants, a feature not often evaluated. Explants were maintained for up to 6 weeks with or without a weekly 24 h exposure to sprifermin (100 ng/mL) and the compressive modulus was assessed. Results showed that sprifermin-treated samples maintained their depth-dependent mechanical profile through 3 weeks, whereas untreated samples lost their mechanical integrity over 1 week of culture. Sprifermin also affected ECM balance by maintaining the levels of extracellular collagen and suppressing matrix metalloproteinase production. These findings support the use of sprifermin as a medium additive for OCT allografts during in vitro storage and present a potential mechanism where sprifermin may impact a functional characteristic of articular cartilage in repair strategies.
Collapse
Affiliation(s)
- Gregory R. Meloni
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Alexandra Farran
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bhavana Mohanraj
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Ryan Cocca
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emilie Rabut
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA,Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George R. Dodge
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA,Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
45
|
Yao X, Zhang J, Jing X, Ye Y, Guo J, Sun K, Guo F. Fibroblast growth factor 18 exerts anti-osteoarthritic effects through PI3K-AKT signaling and mitochondrial fusion and fission. Pharmacol Res 2018; 139:314-324. [PMID: 30273654 DOI: 10.1016/j.phrs.2018.09.026] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/22/2018] [Accepted: 09/24/2018] [Indexed: 12/18/2022]
Abstract
Osteoarthritis (OA) is a degenerative disease characterized by progressive loss of cartilage, osteophyte formation and subchondral bone sclerosis. Although some animal experiments have reported that fibroblast growth factor 18 (FGF18) attenuates cartilage degradation, the effect of FGF18 on chondrocytes and its underlying mechanism at the cellular level remain largely unknown. In this study, we found that an intra-articular injection of FGF18 attenuates cartilage degradation, increases Collagen II deposition and suppresses matrix metallopeptidase 13 (MMP13) expression in rat post-traumatic osteoarthritis (PTOA). At the cellular level, FGF18 promotes chondrocyte proliferation through PI3K-AKT signaling and migration through PI3K signaling. We found that FGF18 attenuates IL-1β-induced apoptosis, restores mitochondrial function and reduces Reactive Oxygen Species (ROS) production through PI3K-AKT signaling. Moreover, the mitochondrial fusion and fission of chondrocytes were enhanced by a short duration of treatment (within 24 h) of IL-1β and suppressed by prolonged treatment (48 h). FGF18 significantly enhances the mitochondrial fusion and fission, restoring mitochondrial function and morphology, and reduces ROS production. We also found that the FGFR1/FGFR3 ratio, which might contribute to the progression of osteoarthritis, was upregulated by IL-1β and downregulated by FGF18. To the best of our knowledge, our data demonstrated the anti-osteoarthritic effect of FGF18 at the cellular level for the first time and suggested that PI3K-AKT signaling and mitochondrial fusion and fission might play critical roles during the process. Our study proved that FGF18 might be a promising drug for the treatment of early stage osteoarthritis and is worth further study.
Collapse
Affiliation(s)
- Xudong Yao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Jiaming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Xingzhi Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yaping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Jiachao Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
46
|
Shkhyan R, Van Handel B, Bogdanov J, Lee S, Yu Y, Scheinberg M, Banks NW, Limfat S, Chernostrik A, Franciozi CE, Alam MP, John V, Wu L, Ferguson GB, Nsair A, Petrigliano FA, Vangsness CT, Vadivel K, Bajaj P, Wang L, Liu NQ, Evseenko D. Drug-induced modulation of gp130 signalling prevents articular cartilage degeneration and promotes repair. Ann Rheum Dis 2018; 77:760-769. [PMID: 29436471 PMCID: PMC8444286 DOI: 10.1136/annrheumdis-2017-212037] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/03/2018] [Accepted: 01/16/2018] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Human adult articular cartilage (AC) has little capacity for repair, and joint surface injuries often result in osteoarthritis (OA), characterised by loss of matrix, hypertrophy and chondrocyte apoptosis. Inflammation mediated by interleukin (IL)-6 family cytokines has been identified as a critical driver of proarthritic changes in mouse and human joints, resulting in a feed-forward process driving expression of matrix degrading enzymes and IL-6 itself. Here we show that signalling through glycoprotein 130 (gp130), the common receptor for IL-6 family cytokines, can have both context-specific and cytokine-specific effects on articular chondrocytes and that a small molecule gp130 modulator can bias signalling towards anti-inflammatory and antidegenerative outputs. METHODS High throughput screening of 170 000 compounds identified a small molecule gp130 modulator termed regulator of cartilage growth and differentiation (RCGD 423) that promotes atypical homodimeric signalling in the absence of cytokine ligands, driving transient increases in MYC and pSTAT3 while suppressing oncostatin M- and IL-6-mediated activation of ERK and NF-κB via direct competition for gp130 occupancy. RESULTS This small molecule increased proliferation while reducing apoptosis and hypertrophic responses in adult chondrocytes in vitro. In a rat partial meniscectomy model, RCGD 423 greatly reduced chondrocyte hypertrophy, loss and degeneration while increasing chondrocyte proliferation beyond that observed in response to injury. Moreover, RCGD 423 improved cartilage healing in a rat full-thickness osteochondral defect model, increasing proliferation of mesenchymal cells in the defect and also inhibiting breakdown of cartilage matrix in de novo generated cartilage. CONCLUSION These results identify a novel strategy for AC remediation via small molecule-mediated modulation of gp130 signalling.
Collapse
Affiliation(s)
- Ruzanna Shkhyan
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Ben Van Handel
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
- Department of Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, California, USA
| | - Jacob Bogdanov
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Siyoung Lee
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Yifan Yu
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
- Department of Orthopaedic Surgery, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Mila Scheinberg
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Nicholas W Banks
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Sean Limfat
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Arthur Chernostrik
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Carlos Eduardo Franciozi
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
- Department of Orthoapedic Surgery, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Mohammad Parvez Alam
- Drug Discovery Laboratory, Department of Neurology, University of California at Los Angeles, Los Angeles, California, USA
| | - Varghese John
- Drug Discovery Laboratory, Department of Neurology, University of California at Los Angeles, Los Angeles, California, USA
| | - Ling Wu
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Gabriel B Ferguson
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Ali Nsair
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine (DGSOM), University of California, Los Angeles, California, USA
| | - Frank A Petrigliano
- Department of Orthopaedic Surgery, David Geffen School of Medicine (DGSOM), University of California at Los Angeles, Los Angeles, California, USA
| | - C Thomas Vangsness
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Kanagasabai Vadivel
- Department of Orthopaedic Surgery, David Geffen School of Medicine (DGSOM), University of California at Los Angeles, Los Angeles, California, USA
| | - Paul Bajaj
- Department of Orthopaedic Surgery, David Geffen School of Medicine (DGSOM), University of California at Los Angeles, Los Angeles, California, USA
| | - Liming Wang
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Nancy Q Liu
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, University of Southern California (USC), Los Angeles, California, USA
- Department of Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, California, USA
- Department of Orthopaedic Surgery, David Geffen School of Medicine (DGSOM), University of California at Los Angeles, Los Angeles, California, USA
| |
Collapse
|
47
|
Reker D, Kjelgaard-Petersen CF, Siebuhr AS, Michaelis M, Gigout A, Karsdal MA, Ladel C, Bay-Jensen AC. Sprifermin (rhFGF18) modulates extracellular matrix turnover in cartilage explants ex vivo. J Transl Med 2017; 15:250. [PMID: 29233174 PMCID: PMC5727954 DOI: 10.1186/s12967-017-1356-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/04/2017] [Indexed: 12/19/2022] Open
Abstract
Background Sprifermin (recombinant human fibroblast growth factor 18) is in clinical development as a potential disease-modifying osteoarthritis drug (DMOAD). In vitro studies have shown that cartilage regenerative properties of sprifermin involve chondrocyte proliferation and extracellular matrix (ECM) production. To gain further insight into the process of sprifermin in the cartilage tissue, this study aimed at investigating the ECM turnover of articular cartilage explants in a longitudinal manner. Methods Bovine full-depth articular cartilage explants were stimulated with sprifermin or placebo at weekly intervals, similar to the dosing regimen used in clinical trials. Pre-culturing with oncostatin M and tumour necrosis factor-α, was also used to induce an inflammatory state before treatment. Metabolic activity was measured using AlamarBlue, and chondrocyte proliferation was visualized by immuno-histochemical detection of proliferating cell nuclear antigen. ECM turnover was quantified by biomarker ELISAs; ProC2 reflecting type II collagen formation, CS846 reflecting aggrecan formation, active MMP9, C2M and AGNx2 reflecting matrix metalloproteinase activity, and AGNx1 reflecting aggrecanase activity. Results Sprifermin was able to reach the chondrocytes through the extracellular matrix, as it increased cell proliferation and metabolic activity of explants. ProC2 and CS846 was dose-dependently increased (P < 0.05) by sprifermin compared to placebo, while C2M and AGNx2 were unaffected, active MMP9 was slightly decreased, and AGNx1 was slightly increased. Over the course of treatment, the temporal order of ECM turnover responses was AGNx1, then ProC2, followed by CS846 and MMP9. Pro-inflammatory activation of the explants diminished the ECM turnover responses otherwise observed under non-inflammatory conditions. Conclusions The data suggest that sprifermin has chondrogenic effects on articular cartilage ex vivo, exerted through a sequential process of ECM turnover; aggrecan degradation seems to occur first, while type II collagen and aggrecan production increased at a later time point. In addition, it was observed that these chondrogenic effects are dependent on the inflammatory status of the cartilage prior to treatment. Electronic supplementary material The online version of this article (10.1186/s12967-017-1356-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ditte Reker
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark. .,Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen Ø, Denmark.
| | - Cecilie F Kjelgaard-Petersen
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark.,Technical University of Denmark, Anker Engelunds Vej 1, 2800, Kgs. Lyngby, Denmark
| | - Anne Sofie Siebuhr
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Martin Michaelis
- Osteoarthritis Research, Merck-KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Anne Gigout
- Osteoarthritis Research, Merck-KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Morten A Karsdal
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Christoph Ladel
- Osteoarthritis Research, Merck-KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Anne C Bay-Jensen
- Biomarkers and Research Rheumatology, Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark.
| |
Collapse
|
48
|
Gigout A, Guehring H, Froemel D, Meurer A, Ladel C, Reker D, Bay-Jensen AC, Karsdal MA, Lindemann S. Sprifermin (rhFGF18) enables proliferation of chondrocytes producing a hyaline cartilage matrix. Osteoarthritis Cartilage 2017; 25:1858-1867. [PMID: 28823647 DOI: 10.1016/j.joca.2017.08.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/24/2017] [Accepted: 08/08/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Fibroblast growth factor (FGF) 18 has been shown to increase cartilage volume when injected intra-articularly in animal models of osteoarthritis (OA) and in patients with knee OA (during clinical development of the recombinant human FGF18, sprifermin). However, the exact nature of this effect is still unknown. In this study, we aimed to investigate the effects of sprifermin at the cellular level. DESIGN A combination of different chondrocyte culture systems was used and the effects of sprifermin on proliferation, the phenotype and matrix production were evaluated. The involvement of MAPKs in sprifermin signalling was also studied. RESULTS In monolayer, we observed that sprifermin promoted a round cell morphology and stimulated both cellular proliferation and Sox9 expression while strongly decreasing type I collagen expression. In 3D culture, sprifermin increased the number of matrix-producing chondrocytes, improved the type II:I collagen ratio and enabled human OA chondrocytes to produce a hyaline extracellular matrix (ECM). Furthermore, we found that sprifermin displayed a 'hit and run' mode of action, with intermittent exposure required for the compound to fully exert its anabolic effect. Finally, sprifermin appeared to signal through activation of ERK. CONCLUSIONS Our results indicate that intermittent exposure to sprifermin leads to expansion of hyaline cartilage-producing chondrocytes. These in vitro findings are consistent with the increased cartilage volume observed in the knees of OA patients after intra-articular injection with sprifermin in clinical studies.
Collapse
Affiliation(s)
- A Gigout
- Osteoarthritis Research, Merck KGaA, Darmstadt, Germany.
| | - H Guehring
- Osteoarthritis Research, Merck KGaA, Darmstadt, Germany.
| | - D Froemel
- Orthopaedic University Hospital Friedrichsheim, Frankfurt, Germany.
| | - A Meurer
- Orthopaedic University Hospital Friedrichsheim, Frankfurt, Germany.
| | - C Ladel
- Osteoarthritis Research, Merck KGaA, Darmstadt, Germany.
| | - D Reker
- Rheumatology, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark.
| | - A C Bay-Jensen
- Rheumatology, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark.
| | - M A Karsdal
- Rheumatology, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark.
| | - S Lindemann
- Osteoarthritis Research, Merck KGaA, Darmstadt, Germany.
| |
Collapse
|
49
|
Tang J, Su N, Zhou S, Xie Y, Huang J, Wen X, Wang Z, Wang Q, Xu W, Du X, Chen H, Chen L. Fibroblast Growth Factor Receptor 3 Inhibits Osteoarthritis Progression in the Knee Joints of Adult Mice. Arthritis Rheumatol 2017; 68:2432-43. [PMID: 27159076 DOI: 10.1002/art.39739] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 04/26/2016] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Fibroblast growth factor (FGF) signaling is involved in articular cartilage homeostasis. This study was undertaken to investigate the role and mechanisms of FGF receptor 3 (FGFR-3) in the pathogenesis of osteoarthritis (OA) caused by surgery and aging in mice. METHODS FGFR-3 was conditionally deleted or activated in articular chondrocytes in adult mice subjected to surgical destabilization of the medial meniscus (DMM). A mouse model of human achondroplasia was also used to assess the role of FGFR-3 in age-associated spontaneous OA. Knee joint cartilage was histologically evaluated and scored using the Osteoarthritis Research Society International system. The expression of genes associated with articular cartilage maintenance was quantitatively evaluated in hip cartilage explants. The effect of inhibiting Indian hedgehog (IHH) signaling in Fgfr3-deficient explants was analyzed. RESULTS Conditional Fgfr3 deletion in mice aggravated DMM-induced cartilage degeneration. Matrix metalloproteinase 13 and type X collagen levels were up-regulated, while type II collagen levels were down-regulated, in the articular cartilage of these mice. Conversely, FGFR-3 activation attenuated cartilage degeneration induced by DMM surgery and age. IHH signaling and runt-related transcription factor 2 levels in mouse articular chondrocytes were up-regulated in the absence of Fgfr3, while inhibition of IHH signaling suppressed the increases in the expression of Runx2, Mmp13, and other factors in Fgfr3-deficient mouse cartilage explants. CONCLUSION Our findings indicate that FGFR-3 delays OA progression in mouse knee joints at least in part via down-regulation of IHH signaling in articular chondrocytes.
Collapse
Affiliation(s)
- Junzhou Tang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Nan Su
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Siru Zhou
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Junlan Huang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xuan Wen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zuqiang Wang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Quan Wang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Wei Xu
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiaolan Du
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Hangang Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns, and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
50
|
Wang Y, Yang T, Liu Y, Zhao W, Zhang Z, Lu M, Zhang W. Decrease of miR-195 Promotes Chondrocytes Proliferation and Maintenance of Chondrogenic Phenotype via Targeting FGF-18 Pathway. Int J Mol Sci 2017; 18:ijms18050975. [PMID: 28471382 PMCID: PMC5454888 DOI: 10.3390/ijms18050975] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 02/06/2023] Open
Abstract
Slow growth and rapid loss of chondrogenic phenotypes are the major problems affecting chronic cartilage lesions. The role of microRNA-195 (miR-195) and its detailed working mechanism in the fore-mentioned process remains unknown. Fibroblastic growth factor 18 (FGF-18) plays a key role in cartilage homeostasis; whether miR-195 could regulate FGF-18 and its downstream signal pathway in chondrocyte proliferation and maintenance of chondrogenic phenotypes still remains unclear. The present research shows elevated miR-195 but depressed FGF-18 expressed in joint fluid specimens of 20 patients with chronic cartilage lesions and in CH1M and CH3M chondrocytes when compared with that in joint fluid specimens without cartilage lesions and in CH1W and CH2W chondrocytes, respectively. The following loss of function test revealed that downregulation of miR-195 by transfection of miR-195 inhibitors promoted chondrocyte proliferation and expression of a type II collagen α I chain (Col2a1)/aggrecan. Through the online informatics analysis we theoretically predicted that miR-195 could bind to a FGF-18 3' untranslated region (3'UTR), also, we verified that a miR-195 could regulate the FGF-18 and its downstream pathway. The constructed dual luciferase assay further confirmed that FGF-18 was a direct target of miR-195. The executed anti-sense experiment displayed that miR-195 could regulate chondrocyte proliferation and Col2a1/aggrecan expression via the FGF-18 pathway. Finally, through an in vivo anterior cruciate ligament transection (ACLT) model, downregulation of miR-195 presented a significantly protective effect on chronic cartilage lesions. Evaluating all of the outcomes of the current research revealed that a decrease of miR-195 protected chronic cartilage lesions by promoting chondrocyte proliferation and maintenance of chondrogenic phenotypes via the targeting of the FGF-18 pathway and that the miR-195/FGF-18 axis could be a potential target in the treatment of cartilage lesions.
Collapse
Affiliation(s)
- Yong Wang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
- The 4th Department of Orthopedic Surgery, The Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China.
| | - Tao Yang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Yadong Liu
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Wei Zhao
- The 4th Department of Orthopedic Surgery, The Central Hospital Affiliated to Shenyang Medical College, Shenyang 110024, China.
| | - Zhen Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Ming Lu
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Weiguo Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|