1
|
Guerra A, Belinha J, Salgado C, Monteiro FJ, Natal Jorge R. Computational Insights into the Interplay of Mechanical Forces in Angiogenesis. Biomedicines 2024; 12:1045. [PMID: 38791007 PMCID: PMC11117778 DOI: 10.3390/biomedicines12051045] [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: 03/18/2024] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
This study employs a meshless computational model to investigate the impacts of compression and traction on angiogenesis, exploring their effects on vascular endothelial growth factor (VEGF) diffusion and subsequent capillary network formation. Three distinct initial domain geometries were defined to simulate variations in endothelial cell sprouting and VEGF release. Compression and traction were applied, and the ensuing effects on VEGF diffusion coefficients were analysed. Compression promoted angiogenesis, increasing capillary network density. The reduction in the VEGF diffusion coefficient under compression altered VEGF concentration, impacting endothelial cell migration patterns. The findings were consistent across diverse simulation scenarios, demonstrating the robust influence of compression on angiogenesis. This computational study enhances our understanding of the intricate interplay between mechanical forces and angiogenesis. Compression emerges as an effective mediator of angiogenesis, influencing VEGF diffusion and vascular pattern. These insights may contribute to innovative therapeutic strategies for angiogenesis-related disorders, fostering tissue regeneration and addressing diseases where angiogenesis is crucial.
Collapse
Affiliation(s)
- Ana Guerra
- INEGI—Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, 4200-465 Porto, Portugal
| | - Jorge Belinha
- ISEP—Instituto Superior de Engenharia do Porto, Departamento de Engenharia Mecânica, Politécnico do Porto, Rua Dr. António Bernardino de Almeida, 431, 4249-015 Porto, Portugal;
| | - Christiane Salgado
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.S.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Fernando Jorge Monteiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.S.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Renato Natal Jorge
- LAETA—Laboratório Associado de Energia, Transportes e Aeronáutica, Universidade do Porto, 4200-165 Porto, Portugal;
- FEUP—Faculdade de Engenharia, Departamento de Engenharia Mecânica, Universidade do Porto, 4200-165 Porto, Portugal
| |
Collapse
|
2
|
Yıldız S, Nursal AF, Yıgıt S, Tumer MK. Role of VEGF I/D variant in suspectibility to odontogenic cyst formation. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 42:308-316. [PMID: 36270022 DOI: 10.1080/15257770.2022.2136693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Odontogenic cysts, are located in the jawbones, filled with fluid surrounded by epithelial lining and fibrous connective tissue. Vascular endothelial growth factor (VEGF) can induce physiological and pathological angiogenesis and is an endothelial cell-specific mitogen. The aim of the present study was to investigate whether any possible association between the VEGF insertion/deletion (I/D) variant and odontogenic cyst in Turkish population. Clinical information and venous blood samples were collected from 62 odontogenic cyst patients and 98 healthy controls. DNA was isolated from peripheral blood leukocytes. Genotyping of the VEGF I/D variant was done by the polymerase chain reaction (PCR) method. There was a statistically differece in terms of VEGF I/D allele frequencies between patients and controls. VEGF I/D variant I allele frequency was more prevalant in patients compared to controls (p = 0.006411, OR: 2.08, 95%Cl: 1.322-3.272). A statistically significant association was observed when the patients were compared with the controls according to D/D + I/D versus I/I genotype (p = 0.0508, OR: 1.925, 95%Cl: 0.872-4.246). The genotype distribution of VEGF I/D was not statistically different between patients and controls (p > 0.05). For the first time, our results provided evidence supporting the odontogenic cyst formation associated with the I/D variant at the promoter region of the VEGF gene in a group of Turkish population. Although it was seen in our study that the I/D variant in the promoter region of the VEGF gene supports odontogenic cyst formation, large-scale studies are needed to elucidate the effect of this variant on odontogenic cysts.
Collapse
Affiliation(s)
- Serkan Yıldız
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Istanbul, Turkey
| | - Ayse Feyda Nursal
- Department of Medical Genetics, Faculty of Medicine, Hitit University, Corum, Turkey
| | - Serbulent Yıgıt
- Department of Veterinary Genetics, Faculty of Veterinary, Ondokuz Mayıs University, Samsun, Turkey
| | - Mehmet Kemal Tumer
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Alanya Alaaddin Keykubat University, Alanya, Turkey
| |
Collapse
|
3
|
Kim B, Bonassar LJ. Understanding the Influence of Local Physical Stimuli on Chondrocyte Behavior. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:31-44. [PMID: 37052844 DOI: 10.1007/978-3-031-25588-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Investigating the mechanobiology of chondrocytes is challenging due to the complex micromechanical environment of cartilage tissue. The innate zonal differences and poroelastic properties of the tissue combined with its heterogeneous composition create spatial- and temporal-dependent cell behavior, which further complicates the investigation. Despite the numerous challenges, understanding the mechanobiology of chondrocytes is crucial for developing strategies for treating cartilage related diseases as chondrocytes are the only cell type within the tissue. The effort to understand chondrocyte behavior under various mechanical stimuli has been ongoing over the last 50 years. Early studies examined global biosynthetic behavior under unidirectional mechanical stimulus. With the technological development in high-speed confocal imaging techniques, recent studies have focused on investigating real-time individual and collective cell responses to multiple / combined modes of mechanical stimuli. Such efforts have led to tremendous advances in understanding the influence of local physical stimuli on chondrocyte behavior. In addition, we highlight the wide variety of experimental techniques, spanning from static to impact loading, and analysis techniques, from biochemical assays to machine learning, that have been utilized to study chondrocyte behavior. Finally, we review the progression of hypotheses about chondrocyte mechanobiology and provide a perspective on the future outlook of chondrocyte mechanobiology.
Collapse
Affiliation(s)
- Byumsu Kim
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - Lawrence J Bonassar
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
4
|
Kurz B, Hart ML, Rolauffs B. Mechanical Articular Cartilage Injury Models and Their Relevance in Advancing Therapeutic Strategies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:107-124. [PMID: 37052850 DOI: 10.1007/978-3-031-25588-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
This chapter details how Alan Grodzinsky and his team unraveled the complex electromechanobiological structure-function relationships of articular cartilage and used these insights to develop an impressively versatile shear and compression model. In this context, this chapter focuses (i) on the effects of mechanical compressive injury on multiple articular cartilage properties for (ii) better understanding the molecular concept of mechanical injury, by studying gene expression, signal transduction and the release of potential injury biomarkers. Furthermore, we detail how (iii) this was used to combine mechanical injury with cytokine exposure or co-culture systems for generating a more realistic trauma model to (iv) investigate the therapeutic modulation of the injurious response of articular cartilage. Impressively, Alan Grodzinsky's research has been and will remain to be instrumental in understanding the proinflammatory response to injury and in developing effective therapies that are based on an in-depth understanding of complex structure-function relationships that underlay articular cartilage function and degeneration.
Collapse
Affiliation(s)
- Bodo Kurz
- Department of Anatomy, Christian-Albrechts-University, Kiel, Germany.
| | - Melanie L Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
5
|
Coveney CR, Samvelyan HJ, Miotla-Zarebska J, Carnegie J, Chang E, Corrin CJ, Coveney T, Stott B, Parisi I, Duarte C, Vincent TL, Staines KA, Wann AK. Ciliary IFT88 Protects Coordinated Adolescent Growth Plate Ossification From Disruptive Physiological Mechanical Forces. J Bone Miner Res 2022; 37:1081-1096. [PMID: 35038201 PMCID: PMC9304194 DOI: 10.1002/jbmr.4502] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/21/2021] [Accepted: 01/08/2022] [Indexed: 11/25/2022]
Abstract
Compared with our understanding of endochondral ossification, much less is known about the coordinated arrest of growth defined by the narrowing and fusion of the cartilaginous growth plate. Throughout the musculoskeletal system, appropriate cell and tissue responses to mechanical force delineate morphogenesis and ensure lifelong health. It remains unclear how mechanical cues are integrated into many biological programs, including those coordinating the ossification of the adolescent growth plate at the cessation of growth. Primary cilia are microtubule-based organelles tuning a range of cell activities, including signaling cascades activated or modulated by extracellular biophysical cues. Cilia have been proposed to directly facilitate cell mechanotransduction. To explore the influence of primary cilia in the mouse adolescent limb, we conditionally targeted the ciliary gene Intraflagellar transport protein 88 (Ift88fl/fl ) in the juvenile and adolescent skeleton using a cartilage-specific, inducible Cre (AggrecanCreERT2 Ift88fl/fl ). Deletion of IFT88 in cartilage, which reduced ciliation in the growth plate, disrupted chondrocyte differentiation, cartilage resorption, and mineralization. These effects were largely restricted to peripheral tibial regions beneath the load-bearing compartments of the knee. These regions were typified by an enlarged population of hypertrophic chondrocytes. Although normal patterns of hedgehog signaling were maintained, targeting IFT88 inhibited hypertrophic chondrocyte VEGF expression and downstream vascular recruitment, osteoclastic activity, and the replacement of cartilage with bone. In control mice, increases to physiological loading also impair ossification in the peripheral growth plate, mimicking the effects of IFT88 deletion. Limb immobilization inhibited changes to VEGF expression and epiphyseal morphology in Ift88cKO mice, indicating the effects of depletion of IFT88 in the adolescent growth plate are mechano-dependent. We propose that during this pivotal phase in adolescent skeletal maturation, ciliary IFT88 protects uniform, coordinated ossification of the growth plate from an otherwise disruptive heterogeneity of physiological mechanical forces. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Clarissa R Coveney
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Hasmik J Samvelyan
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Jadwiga Miotla-Zarebska
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Josephine Carnegie
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Emer Chang
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - C Jonty Corrin
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Trystan Coveney
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Bryony Stott
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Ida Parisi
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Claudia Duarte
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Tonia L Vincent
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Katherine A Staines
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Angus Kt Wann
- Centre for OA Pathogenesis Versus Arthritis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
6
|
The essential anti-angiogenic strategies in cartilage engineering and osteoarthritic cartilage repair. Cell Mol Life Sci 2022; 79:71. [PMID: 35029764 PMCID: PMC9805356 DOI: 10.1007/s00018-021-04105-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/01/2021] [Accepted: 12/18/2021] [Indexed: 01/16/2023]
Abstract
In the cartilage matrix, complex interactions occur between angiogenic and anti-angiogenic components, growth factors, and environmental stressors to maintain a proper cartilage phenotype that allows for effective load bearing and force distribution. However, as seen in both degenerative disease and tissue engineering, cartilage can lose its vascular resistance. This vascularization then leads to matrix breakdown, chondrocyte apoptosis, and ossification. Research has shown that articular cartilage inflammation leads to compromised joint function and decreased clinical potential for regeneration. Unfortunately, few articles comprehensively summarize what we have learned from previous investigations. In this review, we summarize our current understanding of the factors that stabilize chondrocytes to prevent terminal differentiation and applications of these factors to rescue the cartilage phenotype during cartilage engineering and osteoarthritis treatment. Inhibiting vascularization will allow for enhanced phenotypic stability so that we are able to develop more stable implants for cartilage repair and regeneration.
Collapse
|
7
|
Zhu YX, Huang JQ, Ming YY, Zhuang Z, Xia H. Screening of key biomarkers of tendinopathy based on bioinformatics and machine learning algorithms. PLoS One 2021; 16:e0259475. [PMID: 34714891 PMCID: PMC8555777 DOI: 10.1371/journal.pone.0259475] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/19/2021] [Indexed: 12/30/2022] Open
Abstract
Tendinopathy is a complex multifaceted tendinopathy often associated with overuse and with its high prevalence resulting in significant health care costs. At present, the pathogenesis and effective treatment of tendinopathy are still not sufficiently elucidated. The purpose of this research is to intensely explore the genes, functional pathways, and immune infiltration characteristics of the occurrence and development of tendinopathy. The gene expression profile of GSE106292, GSE26051 and GSE167226 are downloaded from GEO (NCBI comprehensive gene expression database) and analyzed by WGCNA software bag using R software, GSE26051, GSE167226 data set is combined to screen the differential gene analysis. We subsequently performed gene enrichment analysis of Gene Ontology (GO) and "Kyoto Encyclopedia of Genes and Genomes" (KEGG), and immune cell infiltration analysis. By constructing the LASSO regression model, Support vector machine (SVM-REF) and Gaussian mixture model (GMMs) algorithms are used to screen, to identify early diagnostic genes. We have obtained a total of 171 DEGs through WGCNA analysis and differentially expressed genes (DEGs) screening. By GO and KEGG enrichment analysis, it is found that these dysregulated genes were related to mTOR, HIF-1, MAPK, NF-κB and VEGF signaling pathways. Immune infiltration analysis showed that M1 macrophages, activated mast cells and activated NK cells had infiltration significance. After analysis of THE LASSO SVM-REF and GMMs algorithms, we found that the gene MACROD1 may be a gene for early diagnosis. We identified the potential of tendon disease early diagnosis way and immune gene regulation MACROD1 key infiltration characteristics based on comprehensive bioinformatics analysis. These hub genes and functional pathways may as early biomarkers of tendon injuries and molecular therapy level target is used to guide drug and basic research.
Collapse
Affiliation(s)
- Ya xi Zhu
- District 1, Department of Orthopedics, Xiangtan Central Hospital, Yuhu District, Xiangtan City, Hunan Province, China
- Nanhua University, Hengyang City, Hunan Province, China
| | - Jia qiang Huang
- District 1, Department of Orthopedics, Xiangtan Central Hospital, Yuhu District, Xiangtan City, Hunan Province, China
| | - Yu yang Ming
- Nanhua University, Hengyang City, Hunan Province, China
- Department of Orthopedics, Xiangtan Central Hospital, Yuhu District, Xiangtan City, Hunan Province, China
| | - Zhao Zhuang
- Academy of Anesthesiology, Weifang Medical University, Weifang, China
| | - Hong Xia
- Department of Orthopedics, Xiangtan Central Hospital, Yuhu District, Xiangtan City, Hunan Province, China
- * E-mail:
| |
Collapse
|
8
|
Heparan Sulfate Deficiency in Cartilage: Enhanced BMP-Sensitivity, Proteoglycan Production and an Anti-Apoptotic Expression Signature after Loading. Int J Mol Sci 2021; 22:ijms22073726. [PMID: 33918436 PMCID: PMC8038223 DOI: 10.3390/ijms22073726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) represents one major cause of disability worldwide still evading efficient pharmacological or cellular therapies. Severe degeneration of extracellular cartilage matrix precedes the loss of mobility and disabling pain perception in affected joints. Recent studies showed that a reduced heparan sulfate (HS) content protects cartilage from degradation in OA-animal models of joint destabilization but the underlying mechanisms remained unclear. We aimed to clarify whether low HS-content alters the mechano-response of chondrocytes and to uncover pathways relevant for HS-related chondro-protection in response to loading. Tissue-engineered cartilage with HS-deficiency was generated from rib chondrocytes of mice carrying a hypomorphic allele of Exostosin 1 (Ext1), one of the main HS-synthesizing enzymes, and wildtype (WT) littermate controls. Engineered cartilage matured for 2 weeks was exposed to cyclic unconfined compression in a bioreactor. The molecular loading response was determined by transcriptome profiling, bioinformatic data processing, and qPCR. HS-deficient chondrocytes expressed 3-6% of WT Ext1-mRNA levels. Both groups similarly raised Sox9, Col2a1 and Acan levels during maturation. However, HS-deficient chondrocytes synthesized and deposited 50% more GAG/DNA. TGFβ and FGF2-sensitivity of Ext1gt/gt chondrocytes was similar to WT cells but their response to BMP-stimulation was enhanced. Loading induced similar activation of mechano-sensitive ERK and P38-signaling in WT and HS-reduced chondrocytes. Transcriptome analysis reflected regulation of cell migration as major load-induced biological process with similar stimulation of common (Fosl1, Itgα5, Timp1, and Ngf) as well as novel mechano-regulated genes (Inhba and Dhrs9). Remarkably, only Ext1-hypomorphic cartilage responded to loading by an expression signature of negative regulation of apoptosis with pro-apoptotic Bnip3 being selectively down-regulated. HS-deficiency enhanced BMP-sensitivity, GAG-production and fostered an anti-apoptotic expression signature after loading, all of which may protect cartilage from load-induced erosion.
Collapse
|
9
|
Nakamura A, Ogi N, Sugita Y, Maeda H, Nagao T, Kurita K. Effects of Vascular Endothelial Growth Factor (VEGF) on the Progression of Osteoarthritis in the Mouse Temporomandibular Joint. J HARD TISSUE BIOL 2021. [DOI: 10.2485/jhtb.30.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Arihiro Nakamura
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Aichi Gakuin University
| | - Nobumi Ogi
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Aichi Gakuin University
| | - Yoshihiko Sugita
- Department of Oral Pathology, School of Dentistry, Aichi Gakuin University
| | - Hatsuhiko Maeda
- Department of Oral Pathology, School of Dentistry, Aichi Gakuin University
| | - Toru Nagao
- Department of Maxillofacial Surgery, School of Dentistry, Aichi Gakuin University
| | - Kenichi Kurita
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Aichi Gakuin University
| |
Collapse
|
10
|
Sperry MM, Yu YH, Kartha S, Ghimire P, Welch RL, Winkelstein BA, Granquist EJ. Intra-articular etanercept attenuates pain and hypoxia from TMJ loading in the rat. J Orthop Res 2020; 38:1316-1326. [PMID: 31903618 PMCID: PMC9118642 DOI: 10.1002/jor.24581] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/28/2019] [Indexed: 02/04/2023]
Abstract
Mechanical overloading of the temporomandibular joint (TMJ) and biochemical changes, like inflammation and hypoxia, contribute to cartilage degeneration and pain associated with osteoarthritis (OA). Yet, how overloading contributes to early dysregulation of chondrocytes is not understood, limiting the development of diagnostics and treatments for TMJ OA. Hypoxia-inducible factors (HIF)-1α/2α in chondrocytes were evaluated at Days 8 and 15 in a rat TMJ pain model induced by jaw loading (1 h/day for 7 days) using immunohistochemistry and compared between cases that induce persistent (3.5 N), acute (2 N), or no (0 N) sensitivity. Hypoxia was measured on Day 8 by immunolabeling of the tracer EF5 and 18 F-EF5 PET imaging. To assess the role of tumor necrosis factor (TNF) in painful TMJ loading, intra-articular etanercept was given before loading. Orofacial sensitivity was evaluated during and after loading. Facial grimace, TNF-α, HIF-2α, and hypoxia levels in the TMJ were measured after loading. HIF-2α was elevated (P = .03) after 3.5 N loading at Day 8, but HIF-1α was unchanged. EF5 uptake increased on Day 8 in the 3.5 N group (P < .048) by tissue assay and 18 F-EF5 PET. At Day 8, both HIF-2α (P = .01) and EF5 uptake (P = .005) were correlated with loading magnitude. Etanercept attenuated sensitivity (P < .01) and the facial grimace on Day 7 (P = .01). It also reduced (P < .01) HIF-2α and EF5 uptake on Day 8; but TNF-α levels were not different from controls at that time. Findings suggest that TMJ loading that induces persistent sensitivity upregulates the catabolic factor HIF-2α and reduces oxygen levels in the cartilage, which may be TNF-driven.
Collapse
Affiliation(s)
| | - Ya-Hsin Yu
- Department of Endodontics, University of Pennsylvania School of Dental Medicine
| | - Sonia Kartha
- Department of Bioengineering, University of Pennsylvania
| | | | | | - Beth A. Winkelstein
- Department of Bioengineering, University of Pennsylvania
- Department of Neurosurgery, University of Pennsylvania
| | - Eric J. Granquist
- Oral & Maxillofacial Surgery, University of Pennsylvania School of Medicine
| |
Collapse
|
11
|
Intrabody against prolyl hydroxylase 2 promotes angiogenesis by stabilizing hypoxia-inducible factor-1α. Sci Rep 2019; 9:11861. [PMID: 31413262 PMCID: PMC6694103 DOI: 10.1038/s41598-019-47891-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022] Open
Abstract
Hypoxia-inducible factor (HIF)-1α is a crucial transcription factor that regulates the expression of target genes involved in angiogenesis. Prolyl hydroxylase 2 (PHD2) dominantly hydroxylates two highly conserved proline residues of HIF-1α to promote its degradation. This study was designed to construct an intrabody against PHD2 that can inhibit PHD2 activity and promote angiogenesis. Single-chain variable fragment (scFv) against PHD2, INP, was isolated by phage display technique and was modified with an endoplasmic reticulum (ER) sequence to obtain ER-retained intrabody against PHD2 (ER-INP). ER-INP was efficiently expressed and bound to PHD2 in cells, significantly increased the levels of HIF-1α, and decreased hydroxylated HIF-1α in human embryonic kidney cell line (HEK293) cells and mouse mononuclear macrophage leukaemia cell line (RAW264.7) cells. ER-INP has shown distinct angiogenic activity both in vitro and in vivo, as ER-INP expression significantly promoted the migration and tube formation of human umbilical vein endothelial cells (HUVECs) and enhanced angiogenesis of chick chorioallantoic membranes (CAMs). Furthermore, ER-INP promoted distinct expression and secretion of a range of angiogenic factors. To the best of our knowledge, this is the first study to report an ER-INP intrabody enhancing angiogenesis by blocking PHD2 activity to increase HIF-1α abundance and activity. These results indicate that ER-INP may play a role in the clinical treatment of tissue injury and ischemic diseases in the future.
Collapse
|
12
|
Verhelst PJ, Van der Cruyssen F, De Laat A, Jacobs R, Politis C. The Biomechanical Effect of the Sagittal Split Ramus Osteotomy on the Temporomandibular Joint: Current Perspectives on the Remodeling Spectrum. Front Physiol 2019; 10:1021. [PMID: 31447704 PMCID: PMC6692453 DOI: 10.3389/fphys.2019.01021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 07/24/2019] [Indexed: 01/18/2023] Open
Abstract
The sagittal split ramus osteotomy is a key approach for treating dentofacial deformities. Although it delivers excellent results, the sagittal split ramus osteotomy is believed to induce stress to the temporomandibular joint. Potential stress inducers could be classified as intra- and postoperative factors resulting in an inflammatory response and molecular cascades, which initiate physiological remodeling. Occasionally, this process exceeds its capacity and causes pathological remodeling, through either degenerative joint disease or condylar resorption. Hard evidence on how orthognathic surgery causes inflammation and how this inflammation is linked to the spectrum of remodeling remains scarce. Current concepts on this matter are mainly based on clinical observations and molecular mechanisms are extrapolated from fundamental research in other body parts or joints. This perspective study provides an overview of current knowledge on molecular pathways and biomechanical effects in temporomandibular joint remodeling. It provides research directions that could lead to acquiring fundamental evidence of the relation of orthognathic surgery and inflammation and its role in remodeling. Performing osteotomies in animal models and identifying inflammatory mediators as well as their effect on the joint seem promising. Patients affected by pathological remodeling can also provide samples for histological as well as molecular analysis. Individual susceptibility analysis by linking certain suspect phenotypes to genetic variation could identify the cause and molecular pathway responsible for degenerative joint disease and condylar resorption, ultimately leading to clinically applicable treatment and prevention strategies.
Collapse
Affiliation(s)
- Pieter-Jan Verhelst
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Fréderic Van der Cruyssen
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Antoon De Laat
- Department of Oral Health Sciences, KU Leuven, Leuven, Belgium.,Department of Dentistry, University Hospitals Leuven, Leuven, Belgium
| | - Reinhilde Jacobs
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium.,Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Constantinus Politis
- OMFS IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Zhao R, Liu W, Xia T, Yang L. Disordered Mechanical Stress and Tissue Engineering Therapies in Intervertebral Disc Degeneration. Polymers (Basel) 2019; 11:polym11071151. [PMID: 31284436 PMCID: PMC6680713 DOI: 10.3390/polym11071151] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 12/11/2022] Open
Abstract
Low back pain (LBP), commonly induced by intervertebral disc degeneration, is a lumbar disease with worldwide prevalence. However, the mechanism of degeneration remains unclear. The intervertebral disc is a nonvascular organ consisting of three components: Nucleus pulposus, annulus fibrosus, and endplate cartilages. The disc is structured to support our body motion and endure persistent external mechanical pressure. Thus, there is a close connection between force and intervertebral discs in LBP. It is well established that with aging, disordered mechanical stress profoundly influences the fate of nucleus pulposus and the alignment of collagen fibers in the annulus fibrosus. These support a new understanding that disordered mechanical stress plays an important role in the degeneration of the intervertebral discs. Tissue-engineered regenerative and reparative therapies are being developed for relieving disc degeneration and symptoms of lower back pain. In this paper, we will review the current literature available on the role of disordered mechanical stress in intervertebral disc degeneration, and evaluate the existing tissue engineering treatment strategies of the current therapies.
Collapse
Affiliation(s)
- Runze Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Wanqian Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Tingting Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
14
|
Dane DM, Yilmaz C, Gyawali D, Iyer R, Menon J, Nguyen KT, Ravikumar P, Estrera AS, Hsia CCW. Erythropoietin inhalation enhances adult canine alveolar-capillary formation following pneumonectomy. Am J Physiol Lung Cell Mol Physiol 2019; 316:L936-L945. [PMID: 30785346 DOI: 10.1152/ajplung.00504.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Paracrine erythropoietin (EPO) signaling in the lung recruits endothelial progenitor cells, promotes cell maturation and angiogenesis, and is upregulated during canine postpneumonectomy (PNX) compensatory lung growth. To determine whether inhalational delivery of exogenous EPO augments endogenous post-PNX lung growth, adult canines underwent right PNX and received, via a permanent tracheal stoma, weekly nebulization of recombinant human EPO-containing nanoparticles or empty nanoparticles (control) for 16 wk. Lung function was assessed under anesthesia pre- and post-PNX. The remaining lobes were fixed for detailed morphometric analysis. Compared with control treatment, EPO delivery significantly increased serum EPO concentration without altering systemic hematocrit or hemoglobin concentration and abrogated post-PNX lipid oxidative stress damage. EPO delivery modestly increased post-PNX volume densities of the alveolar septum per unit of lung volume and type II epithelium and endothelium per unit of septal tissue volume in selected lobes. EPO delivery also augmented the post-PNX increase in alveolar double-capillary profiles, a marker of intussusceptive capillary formation, in all remaining lobes. EPO treatment did not significantly alter absolute resting lung volumes, lung and membrane diffusing capacities, alveolar-capillary blood volume, pulmonary blood flow, lung compliance, or extravascular alveolar tissue volumes or surface areas. Results established the feasibility of chronic inhalational delivery of growth-modifying biologics in a large animal model. Exogenous EPO selectively enhanced cytoprotection and alveolar angiogenesis in remaining lobes but not whole-lung extravascular tissue growth or resting function; the nonuniform response contributes to structure-function discrepancy, a major challenge for interventions aimed at amplifying the innate potential for compensatory lung growth.
Collapse
Affiliation(s)
- D Merrill Dane
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Cuneyt Yilmaz
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Dipendra Gyawali
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Roshni Iyer
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Jyothi Menon
- Department of Bioengineering, University of Texas at Arlington , Arlington, Texas
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington , Arlington, Texas
| | - Priya Ravikumar
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Aaron S Estrera
- Department of Cardiothoracic Surgery, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Connie C W Hsia
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| |
Collapse
|
15
|
Vaca-González JJ, Guevara JM, Moncayo MA, Castro-Abril H, Hata Y, Garzón-Alvarado DA. Biophysical Stimuli: A Review of Electrical and Mechanical Stimulation in Hyaline Cartilage. Cartilage 2019; 10:157-172. [PMID: 28933195 PMCID: PMC6425540 DOI: 10.1177/1947603517730637] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Hyaline cartilage degenerative pathologies induce morphologic and biomechanical changes resulting in cartilage tissue damage. In pursuit of therapeutic options, electrical and mechanical stimulation have been proposed for improving tissue engineering approaches for cartilage repair. The purpose of this review was to highlight the effect of electrical stimulation and mechanical stimuli in chondrocyte behavior. DESIGN Different information sources and the MEDLINE database were systematically revised to summarize the different contributions for the past 40 years. RESULTS It has been shown that electric stimulation may increase cell proliferation and stimulate the synthesis of molecules associated with the extracellular matrix of the articular cartilage, such as collagen type II, aggrecan and glycosaminoglycans, while mechanical loads trigger anabolic and catabolic responses in chondrocytes. CONCLUSION The biophysical stimuli can increase cell proliferation and stimulate molecules associated with hyaline cartilage extracellular matrix maintenance.
Collapse
Affiliation(s)
- Juan J. Vaca-González
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| | - Johana M. Guevara
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogota, Colombia
| | - Miguel A. Moncayo
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| | - Hector Castro-Abril
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| | - Yoshie Hata
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
| | - Diego A. Garzón-Alvarado
- Biomimetics Laboratory, Instituto de Biotecnología, Universidad Nacional de Colombia, Bogota, Colombia
- Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogota, Colombia
| |
Collapse
|
16
|
Costa NT, Iriyoda TMV, Alfieri DF, Simão ANC, Dichi I. Influence of disease-modifying antirheumatic drugs on oxidative and nitrosative stress in patients with rheumatoid arthritis. Inflammopharmacology 2018; 26:1151-1164. [DOI: 10.1007/s10787-018-0514-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/03/2018] [Indexed: 12/19/2022]
|
17
|
Galicia K, Thorson C, Banos A, Rondina M, Hopkinson W, Hoppensteadt D, Fareed J. Inflammatory Biomarker Profiling in Total Joint Arthroplasty and Its Relevance to Circulating Levels of Lubricin, a Novel Proteoglycan. Clin Appl Thromb Hemost 2018; 24:950-959. [PMID: 29683034 PMCID: PMC6714720 DOI: 10.1177/1076029618765090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Lubricin, also known as proteoglycan 4, acts as an antiadhesive and boundary lubricant to prevent cartilage damage in healthy joints. Following injury, a decrease in synovial fluid (SF) lubricin may lead to secondary osteoarthritis (OA). Inflammatory biomarkers, such as IL-1β and TNF-α, are also implicated in the pathophysiology of OA. Interestingly, they have been shown to suppress the expression and secretion of lubricin in SF. This study aims to compare circulating levels of inflammatory biomarkers and lubricin between total joint arthroplasty (TJA) patients and healthy individuals. Doing so may better elucidate their roles in OA and extend the understanding of inflammation as a regulator of lubricin. Deidentified plasma samples were obtained 1 day preoperatively and 1 day postoperatively from patients undergoing TJA. Utilizing biochip array technology, they were profiled for IL-2, IL-4, IL-6, IL-8, IL-10, VEGF, IFN-γ, IL-1α, IL-1β, MCP-1, EGF, and TNF-α. Circulating lubricin levels were also measured using enzyme-linked immunosorbent assay. Compared to healthy controls, IL-6, IL-8, VEGF, IL-1β, MCP-1, EGF, and TNF-α were significantly increased pre- and postoperatively. Lubricin was significantly decreased. This may indicate that elevations in inflammatory cytokines initiate a cascade of events, leading to decreased lubricin, which places the joint at increased risk of developing OA.
Collapse
Affiliation(s)
- Kevin Galicia
- 1 Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Chase Thorson
- 1 Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Andrew Banos
- 2 Department of Orthopedics, Loyola University Medical Center, Maywood, IL, USA
| | - Matthew Rondina
- 3 Department of Internal Medicine, University of Utah Hospital, Salt Lake City, UT, USA
| | - William Hopkinson
- 2 Department of Orthopedics, Loyola University Medical Center, Maywood, IL, USA
| | | | - Jawed Fareed
- 4 Department of Pathology, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
18
|
Mafu TS, September AV, Shamley D. The potential role of angiogenesis in the development of shoulder pain, shoulder dysfunction, and lymphedema after breast cancer treatment. Cancer Manag Res 2018; 10:81-90. [PMID: 29391829 PMCID: PMC5772395 DOI: 10.2147/cmar.s151714] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Shoulder morbidity is a well-documented sequela of breast cancer treatment, which includes various manifestations such as pain, reduced range of motion, and lymphedema, among others. The multifactorial nature of such morbidities has long been appreciated, and research on reliable risk predictors of development thereof still continues. Previous studies have demonstrated the potential of different types of physical therapy to treat such shoulder problems, and the integration of such interventions into routine care for breast cancer survivors is a requirement in most high-income countries. Although patients at risk for developing shoulder problems would most likely benefit from posttreatment physical therapy, currently, there is no gold standard for identifying this patient group. This is particularly important in low- and middle-income countries where scarce monetary resources need to be directed specifically to those most in need. Modulators of the angiogenesis pathway have been implicated in noncancer shoulder conditions such as rotator cuff disease, adhesive capsulitis, and tendon injuries. The present review summarizes the role of angiogenesis in the development of shoulder morbidity among breast cancer survivors and sets forth the rationale for our belief that angiogenesis signaling may help explain a proportion of the reported clinical variability noted in the development of shoulder pain and dysfunction and upper-limb lymphedema after breast cancer treatment.
Collapse
Affiliation(s)
- Trevor S Mafu
- Division of Exercise Science and Sports Medicine, Department of Human Biology, Faculty of Health Sciences, University of Cape Town
| | - Alison V September
- Division of Exercise Science and Sports Medicine, Department of Human Biology, Faculty of Health Sciences, University of Cape Town
| | - Delva Shamley
- Clinical Research Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
19
|
Yu J, Liang F, Huang H, Pirttiniemi P, Yu D. Effects of loading on chondrocyte hypoxia, HIF-1α and VEGF in the mandibular condylar cartilage of young rats. Orthod Craniofac Res 2017; 21:41-47. [PMID: 29271061 DOI: 10.1111/ocr.12212] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To investigate hypoxia-inducible factor 1-alpha (HIF-1α) and vascular endothelial growth factor (VEGF) expression under altered loading, and to explore the relationship between loading and hypoxia in the mandibular condylar cartilage of young rats. SETTING AND SAMPLE POPULATION Eighty Sprague-Dawley rats. MATERIAL AND METHODS The reduced loading group was fed soft food, and their incisors were cut to avoid occlusal contact. The increased loading group was fed hard food and had forced jaw-opening. Ten rats from each group (n = 10) were sacrificed at 12, 24, 48, and 96 hours after initiation of the experiment. Pimonidazole hydrochloride (Hypoxyprobe-1, HP-1) was used as a hypoxia marker to confirm the hypoxic state. Hypoxic chondrocytes as indicated by HP-1, HIF-1α and VEGF protein expressions were recognized by immunohistochemical detection. HIF-1α and VEGF mRNA expressions were detected by semi-quantitative RT-PCR. RESULTS Hypoxyprobe-1 was confined in the upper layers of cartilage, and was most strongly expressed in the weight-bearing area of TMJ at 12 and 96 hours. Staining of HIF-1α and VEGF was most strongly expressed in the chondrocytes of the fibrous and proliferative layer at all time points. Furthermore, expressions were also displayed in the hypertrophic and calcified layers at 48 and 96 hours. The expressions of HIF-1α and VEGF mRNA were higher in the increased loading group than in the reduced loading group at 48 and 96 hours (P < . 05). CONCLUSION Mechanical loading seems to directly induce weight-bearing area hypoxia followed by new vessel formation, which indicates that these factors are related and important for the development of cartilage.
Collapse
Affiliation(s)
- J Yu
- Department of Oral Development and Orthodontics, Institute of Dentistry, University of Oulu, Oulu, Finland
| | - F Liang
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, China
| | - H Huang
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, China
| | - P Pirttiniemi
- Department of Oral Development and Orthodontics, Institute of Dentistry, University of Oulu, Oulu, Finland
| | - D Yu
- Department of Stomatology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
20
|
Nagao M, Hamilton JL, Kc R, Berendsen AD, Duan X, Cheong CW, Li X, Im HJ, Olsen BR. Vascular Endothelial Growth Factor in Cartilage Development and Osteoarthritis. Sci Rep 2017; 7:13027. [PMID: 29026147 PMCID: PMC5638804 DOI: 10.1038/s41598-017-13417-w] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/21/2017] [Indexed: 02/07/2023] Open
Abstract
Genome wide studies indicate that vascular endothelial growth factor A (VEGF) is associated with osteoarthritis (OA), and increased VEGF expression correlates with increased disease severity. VEGF is also a chondrocyte survival factor during development and essential for bone formation, skeletal growth and postnatal homeostasis. This raises questions of how the important embryonic and postnatal functions of VEGF can be reconciled with an apparently destructive role in OA. Addressing these questions, we find that VEGF acts as a survival factor in growth plate chondrocytes during development but only up until a few weeks after birth in mice. It is also required for postnatal differentiation of articular chondrocytes and the timely ossification of bones in joint regions. In surgically induced knee OA in mice, a model of post-traumatic OA in humans, increased expression of VEGF is associated with catabolic processes in chondrocytes and synovial cells. Conditional knock-down of Vegf attenuates induced OA. Intra-articular anti-VEGF antibodies suppress OA progression, reduce levels of phosphorylated VEGFR2 in articular chondrocytes and synovial cells and reduce levels of phosphorylated VEGFR1 in dorsal root ganglia. Finally, oral administration of the VEGFR2 kinase inhibitor Vandetanib attenuates OA progression.
Collapse
Affiliation(s)
- Masashi Nagao
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA, 02115, USA.
- Department of Orthopaedic Surgery, Juntendo University School of Medicine 2-1-1 Hongo Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - John L Hamilton
- Department of Biochemistry, Rush University Medical Center, 1735 W, Harrison Street, Chicago, IL, 60612, USA
| | - Ranjan Kc
- Department of Biochemistry, Rush University Medical Center, 1735 W, Harrison Street, Chicago, IL, 60612, USA
| | - Agnes D Berendsen
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA, 02115, USA
| | - Xuchen Duan
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA, 02115, USA
| | - Chan Wook Cheong
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA, 02115, USA
| | - Xin Li
- Department of Biochemistry, Rush University Medical Center, 1735 W, Harrison Street, Chicago, IL, 60612, USA
| | - Hee-Jeong Im
- Jesse Brown Veterans Affairs (VA) Medical Center, 820S, Damen Avenue, Chicago, IL, 60612, USA.
- Department of Bioengineering, University of Illinois, Chicago, IL, 60612, USA.
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
21
|
Steinecker-Frohnwieser B, Kaltenegger H, Weigl L, Mann A, Kullich W, Leithner A, Lohberger B. Pharmacological treatment with diacerein combined with mechanical stimulation affects the expression of growth factors in human chondrocytes. Biochem Biophys Rep 2017; 11:154-160. [PMID: 28955780 PMCID: PMC5614688 DOI: 10.1016/j.bbrep.2017.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/19/2017] [Accepted: 06/21/2017] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) as the main chronic joint disease arises from a disturbed balance between anabolic and catabolic processes leading to destructions of articular cartilage of the joints. While mechanical stress can be disastrous for the metabolism of chondrocytes, mechanical stimulation at the physiological level is known to improve cell function. The disease modifying OA drug (DMOAD) diacerein functions as a slowly-acting drug in OA by exhibiting anti-inflammatory, anti-catabolic, and pro-anabolic properties on cartilage. Combining these two treatment options revealed positive effects on OA-chondrocytes. METHODS Cells were grown on flexible silicone membranes and mechanically stimulated by cyclic tensile loading. After seven days in the presence or absence of diacerein, inflammation markers and growth factors were analyzed using quantitative real-time PCR and enzyme linked immune assays. The influence of conditioned medium was tested on cell proliferation and cell migration. RESULTS Tensile strain and diacerein treatment reduced interleukin-6 (IL-6) expression, whereas cyclooxygenase-2 (COX2) expression was increased only by mechanical stimulation. The basic fibroblast growth factor (bFGF) was down regulated by the combined treatment modalities, whereas prostaglandin E2 (PGE2) synthesis was reduced only under OA conditions. The expression of platelet-derived growth factor (PDGF) and vascular endothelial growth factor A (VEGF-A) was down-regulated by both. CONCLUSIONS From our study we conclude that moderate mechanical stimulation appears beneficial for the fate of the cell and improves the pharmacological effect of diacerein based on cross-talks between different initiated pathways. GENERAL SIGNIFICANCE Combining two different treatment options broadens the perspective to treat OA and improves chondrocytes metabolism.
Collapse
Affiliation(s)
- Bibiane Steinecker-Frohnwieser
- Ludwig Boltzmann Department for Rehabilitation of Internal Diseases, Ludwig Boltzmann Cluster for Arthritis and Rehabilitation, Thorerstrasse 26, 5760 Saalfelden, Austria
| | - Heike Kaltenegger
- Department of Orthopaedic Surgery, Medical University of Graz, Graz, Austria
| | - Lukas Weigl
- Department of Special Anaesthesia and Pain Therapy, Medical University Vienna, Austria
| | - Anda Mann
- Department of Special Anaesthesia and Pain Therapy, Medical University Vienna, Austria
| | - Werner Kullich
- Ludwig Boltzmann Department for Rehabilitation of Internal Diseases, Ludwig Boltzmann Cluster for Arthritis and Rehabilitation, Thorerstrasse 26, 5760 Saalfelden, Austria
| | - Andreas Leithner
- Department of Orthopaedic Surgery, Medical University of Graz, Graz, Austria
| | - Birgit Lohberger
- Department of Orthopaedic Surgery, Medical University of Graz, Graz, Austria
| |
Collapse
|
22
|
Wang S, Zhou C, Zheng H, Zhang Z, Mei Y, Martin JA. Chondrogenic progenitor cells promote vascular endothelial growth factor expression through stromal-derived factor-1. Osteoarthritis Cartilage 2017; 25:742-749. [PMID: 27989872 PMCID: PMC6367939 DOI: 10.1016/j.joca.2016.10.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/14/2016] [Accepted: 10/18/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Vascular endothelial growth factor (VEGF) is elevated in joint fluids from patients diagnosed with osteoarthritis (OA). VEGF is known to contribute to vascular tidemark invasion and osteophyte formation, which are classic features of advanced OA. Among the factors that may drive VEGF accumulation in diseased joints, stromal cell-derived factor-1α (SDF-1α) is a likely culprit, as it is enriched in synovial fluids from osteoarthritic joints and is a potent inducer of VEGF expression. Chondrogenic progenitor cells (CPCs) that overexpress SDF-1α are abundant in osteoarthritic cartilage, implicating them in elevating synovial SDF-1α levels. Here we conducted a series of experiments to determine the potential for CPCs to stimulate VEGF expression via autocrine and paracrine mechanisms. DESIGN Immunohistochemistry, immunoblotting, and PCR were used to evaluate the effects of SDF-1α on VEGF expression in CPCs and chondrocytes, and the effects of CPC-conditioned medium on chondrocytes. An SDF-1α receptor antagonist and inhibitors of mitogen-activated protein kinases (MAPKs) were used to probe the pathway linking SDF-1 with VEGF expression in CPCs. RESULTS SDF-1α and CPC-conditioned medium stimulated VEGF expression in chondrocytes. In both chondrocytes and CPCs, SDF-1α stimulated increased VEGF expression via C-X-C chemokine receptor type 4 (CXCR4), a cell-surface SDF-1α receptor. This response in CPCs is dependent on p38 MAPK activation, but not on ERK or c-Jun N-terminal kinase (JNK) activation. CONCLUSIONS By secreting SDF-1α, CPCs stimulate VEGF expression in nearby cells. The co-expression of SDF-1 and its receptor by CPCs indicates they are capable of self-sustained VEGF expression via an autocrine mechanism.
Collapse
Affiliation(s)
- Shuya Wang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin China, 150001
| | - Cheng Zhou
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA,Department of Biomedical Engineering, University of Iowa, Iowa City, IA
| | - Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO
| | - Zhiyi Zhang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin China, 150001
| | - YiFang Mei
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, Harbin China, 150001
| | - James A. Martin
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA,Department of Biomedical Engineering, University of Iowa, Iowa City, IA
| |
Collapse
|
23
|
Qing L, Lei P, Liu H, Xie J, Wang L, Wen T, Hu Y. Expression of hypoxia-inducible factor-1α in synovial fluid and articular cartilage is associated with disease severity in knee osteoarthritis. Exp Ther Med 2016; 13:63-68. [PMID: 28123469 PMCID: PMC5244982 DOI: 10.3892/etm.2016.3940] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/09/2016] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to examine hypoxia-inducible factor 1α (HIF-1α) levels in the synovial fluid and articular cartilage of patients with primary knee osteoarthritis (OA) and to investigate their association with the severity of disease. A total of 36 patients with knee OA and ten healthy controls were enrolled. Anteroposterior knee radiographs and/or Mankin scores were assessed to determine the disease severity of the affected knee. Radiographic grading of OA in the knee was performed according to Kellgren-Lawrence criteria. HIF-1α levels in synovial fluid were measured using enzyme-linked immunosorbent assay, whereas HIF-1α levels in articular cartilage were assessed with immunohistochemical methods. Compared with healthy controls, OA patients exhibited an increased HIF-1α concentration in synovial fluid (218.17±25.12 vs. 156.66±7.74 pg/ml; P<0.001) and articular cartilage (P<0.05). Furthermore, synovial fluid HIF-1α levels demonstrated a positive correlation with articular cartilage HIF-1α levels (Pearson's P=0.815; P<0.001). Subsequent analysis showed that synovial fluid HIF-1α levels were significantly correlated with the severity of disease (Spearman's ρ=0.933; P<0.001). Furthermore, articular cartilage levels of HIF-1α also correlated with disease severity (Spearman's ρ=-0.967; P<0.001). The findings of the present study suggested that HIF-1α in synovial fluid and articular cartilage is associated with progressive joint damage and is likely to be a useful biomarker for determining disease severity and progression in knee OA.
Collapse
Affiliation(s)
- Liming Qing
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Pengfei Lei
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hao Liu
- Program of Biology and Biomedical Sciences, Division of Medical Sciences, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Jie Xie
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Long Wang
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ting Wen
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yihe Hu
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
24
|
Golding A, Guay JA, Herrera-Rincon C, Levin M, Kaplan DL. A Tunable Silk Hydrogel Device for Studying Limb Regeneration in Adult Xenopus Laevis. PLoS One 2016; 11:e0155618. [PMID: 27257960 PMCID: PMC4892606 DOI: 10.1371/journal.pone.0155618] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 05/02/2016] [Indexed: 01/08/2023] Open
Abstract
In certain amphibian models limb regeneration can be promoted or inhibited by the local wound bed environment. This research introduces a device that can be utilized as an experimental tool to characterize the conditions that promotes limb regeneration in the adult frog (Xenopus laevis) model. In particular, this device was designed to manipulate the local wound environment via a hydrogel insert. Initial characterization of the hydrogel insert revealed that this interaction had a significant influence on mechanical forces to the animal, due to the contraction of the hydrogel. The material and mechanical properties of the hydrogel insert were a factor in the device design in relation to the comfort of the animal and the ability to effectively manipulate the amputation site. The tunable features of the hydrogel were important in determining the pro-regenerative effects in limb regeneration, which was measured by cartilage spike formation and quantified by micro-computed tomography. The hydrogel insert was a factor in the observed morphological outcomes following amputation. Future work will focus on characterizing and optimizing the device's observed capability to manipulate biological pathways that are essential for limb regeneration. However, the present work provides a framework for the role of a hydrogel in the device and a path forward for more systematic studies.
Collapse
Affiliation(s)
- Anne Golding
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Justin A. Guay
- Department of Biology, Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Celia Herrera-Rincon
- Department of Biology, Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Michael Levin
- Department of Biology, Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts, United States of America
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| |
Collapse
|
25
|
Mousavizadeh R, Scott A, Lu A, Ardekani GS, Behzad H, Lundgreen K, Ghaffari M, McCormack RG, Duronio V. Angiopoietin-like 4 promotes angiogenesis in the tendon and is increased in cyclically loaded tendon fibroblasts. J Physiol 2016; 594:2971-83. [PMID: 26670924 PMCID: PMC4887665 DOI: 10.1113/jp271752] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/10/2015] [Indexed: 12/30/2022] Open
Abstract
KEY POINTS Angiopoietin-like 4 (ANGPTL4) modulates tendon neovascularization. Cyclic loading stimulates the activity of transforming growth factor-β and hypoxia-inducible factor 1α and thereby increases the expression and release of ANGPTL4 from human tendon cells. Targeting ANGPTL4 and its regulatory pathways is a potential avenue for regulating tendon vascularization to improve tendon healing or adaptation. ABSTRACT The mechanisms that regulate angiogenic activity in injured or mechanically loaded tendons are poorly understood. The present study examined the potential role of angiopoietin-like 4 (ANGPTL4) in the angiogenic response of tendons subjected to repetitive mechanical loading or injury. Cyclic stretching of human tendon fibroblasts stimulated the expression and release of ANGPTL4 protein via transforming growth factor-β (TGF-β) and hypoxia-inducible factor 1α (HIF-1α) signalling, and the released ANGPTL4 was pro-angiogenic. Angiogenic activity was increased following ANGPTL4 injection into mouse patellar tendons, whereas the patellar tendons of ANGPTL4 knockout mice displayed reduced angiogenesis following injury. In human rotator cuff tendons, the expression of ANGPTL4 was correlated with the density of tendon endothelial cells. To our knowledge, this is the first study characterizing a role of ANGPTL4 in the tendon. ANGPTL4 may assist in the regulation of vascularity in the injured or mechanically loaded tendon. TGF-β and HIF-1α comprise two signalling pathways that modulate the expression of ANGPTL4 by mechanically stimulated tendon fibroblasts and, in the future, these could be manipulated to influence tendon healing or adaptation.
Collapse
Affiliation(s)
- Rouhollah Mousavizadeh
- Jack Bell Research Centre, Department of Medicine, University of British Columbia, Vancouver, Canada
- Department of Physical Therapy, University of British Columbia, Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Alex Scott
- Department of Physical Therapy, University of British Columbia, Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Alex Lu
- Department of Physical Therapy, University of British Columbia, Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Gholamreza S Ardekani
- Jack Bell Research Centre, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Hayedeh Behzad
- Department of Physical Therapy, University of British Columbia, Centre for Hip Health and Mobility, Vancouver Coastal Health Research Institute, Vancouver, Canada
| | - Kirsten Lundgreen
- Department of Orthopaedic Surgery, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Mazyar Ghaffari
- Jack Bell Research Centre, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Robert G McCormack
- Department of Orthopedic Surgery, University of British Columbia, Vancouver, Canada
| | - Vincent Duronio
- Jack Bell Research Centre, Department of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
26
|
Zhang X, Crawford R, Xiao Y. Inhibition of vascular endothelial growth factor with shRNA in chondrocytes ameliorates osteoarthritis. J Mol Med (Berl) 2016; 94:787-98. [PMID: 27164955 DOI: 10.1007/s00109-016-1425-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 03/29/2016] [Accepted: 04/21/2016] [Indexed: 12/31/2022]
Abstract
UNLABELLED Osteoarthritis (OA) is a chronic, incurable and destructive joint disease that is characterized by chondrocyte hypertrophy and cartilage degradation. Angiogenesis, mediated by the action of vascular endothelial growth factor (VEGF), is known to be a contributing factor in the pathogenesis of OA. In this study, we use a lentivirus-based approach to investigate whether VEGF knockdown would be beneficial to chondrogenesis and could prevent or slow down OA progression. We first profiled cytokines in human OA cartilage using cytokine antibody arrays. This revealed that as many as 21 angiogenesis-related cytokines were significantly upregulated in severe OA cartilage compared to mild OA samples. Next, we infected chondrocytes with VEGF small hairpin RNA (shRNA) lentivirus (LV-VEGF shRNA) and treated these cells with tumour necrosis factor alpha (TNF-α) to induce hypertrophy. The results showed that inhibition of VEGF not only enhanced chondrogenic differentiation, but also protected chondrocytes from TNF-α-induced hypertrophy. We also found that knockdown of VEGF suppressed TNF-α-induced phosphorylation of ERK1/2 in chondrocytes. Furthermore, using a surgically induced OA rat model, we showed that VEGF inhibition delayed OA progression in animals given intra-articular injection of LV-VEGF shRNA. In conclusion, in this study, we have shown that VEGF knockdown can enhance chondrogenesis and prevent OA progression, thus providing evidence that inhibition of VEGF may be a potential therapeutic approach for OA patients. KEY MESSAGES Numerous pro-angiogenic factors are upregulated in severe OA cartilage. Inhibition of VEGF by shRNA protects chondrocytes from TNF-α-induced hypertrophy. Knockdown of VEGF suppresses TNF-α-induced phosphorylation of ERK1/2 in chondrocytes. VEGF inhibition delays OA progression in rat model in vivo. Inhibition of VEGF may be a potential therapeutic approach for OA patients.
Collapse
Affiliation(s)
- Xufang Zhang
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 60 Musk Ave, Kelvin Grove, Queensland, 4059, Australia.,Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Guangdong Province Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China
| | - Ross Crawford
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 60 Musk Ave, Kelvin Grove, Queensland, 4059, Australia
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 60 Musk Ave, Kelvin Grove, Queensland, 4059, Australia.
| |
Collapse
|
27
|
Chen YJ, Chan DC, Chiang CK, Wang CC, Yang TH, Lan KC, Chao SC, Tsai KS, Yang RS, Liu SH. Advanced glycation end-products induced VEGF production and inflammatory responses in human synoviocytes via RAGE-NF-κB pathway activation. J Orthop Res 2016; 34:791-800. [PMID: 26497299 DOI: 10.1002/jor.23083] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/22/2015] [Indexed: 02/04/2023]
Abstract
Aging and diabetes are known to be the major cause to affect the progression of osteoarthritis (OA). Advanced glycation end products (AGEs) have been observed to accumulate in various organs especially in joint tissue and do damage to the joint tissue during aging and diabetes. Synovial angiogenesis and inflammation are observed across the full range of OA severity. The signaling pathway of AGEs on vascular endothelial growth factor (VEGF) production and inflammatory responses in synoviocytes are still unclear. Here, we investigated the role of receptor for AGEs (RAGE) and the signaling pathway involved in AGEs-induced VEGF production and inflammatory responses in human synoviocytes. Human synoviocytes were cultured and treated with AGEs (25-100 µg/ml). AGEs significantly induced the protein expressions of cyclooxygenase-2 (COX-2) and VEGF and the productions of prostaglandin-E2 (PGE2), VEGF, interleukin-6 (IL-6), and metalloproteinase-13 (MMP-13) in human synoviocytes in a dose-dependent manner. Moreover, AGEs markedly activated the phosphorylations of IκB kinase (IKK)α/β, IκBα, and nuclear factor (NF)-κB-p65 proteins in human synoviocytes in a time-dependent manner. Treatment with neutralizing antibody for RAGE statistically significantly decreased the AGEs-induced increase in COX-2, VEGF, PGE2, IL-6, and MMP13 and AGEs-activated NF-κB pathway activation. Taken together, these findings indicate that AGEs are capable of inducing VEGF production and inflammatory responses via RAGE-NF-κB pathway activation in human synoviocytes. © 2015 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:791-800, 2016.
Collapse
Affiliation(s)
- Ying-Ju Chen
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ding-Cheng Chan
- Department of Geriatrics and Gerontology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Kang Chiang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Chia Wang
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ting-Hua Yang
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Sung-Chuan Chao
- Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Keh-Sung Tsai
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Rong-Sen Yang
- Department of Orthopaedics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
28
|
Hamilton JL, Nagao M, Levine BR, Chen D, Olsen BR, Im HJ. Targeting VEGF and Its Receptors for the Treatment of Osteoarthritis and Associated Pain. J Bone Miner Res 2016; 31:911-24. [PMID: 27163679 PMCID: PMC4863467 DOI: 10.1002/jbmr.2828] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 01/15/2023]
Abstract
Increased vascular endothelial growth factor (VEGF) levels are associated with osteoarthritis (OA) progression. Indeed, VEGF appears to be involved in OA-specific pathologies including cartilage degeneration, osteophyte formation, subchondral bone cysts and sclerosis, synovitis, and pain. Moreover, a wide range of studies suggest that inhibition of VEGF signaling reduces OA progression. This review highlights both the potential significance of VEGF in OA pathology and pain, as well as potential benefits of inhibition of VEGF and its receptors as an OA treatment. With the emergence of the clinical use of anti-VEGF therapy outside of OA, both as high-dose systemic treatments and low-dose local treatments, these particular therapies are now more widely understood. Currently, there is no established disease-modifying drug available for patients with OA, which warrants continued study of the inhibition of VEGF signaling in OA, as stand-alone or adjuvant therapy. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- John L. Hamilton
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - Masashi Nagao
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, 02115, USA
| | - Brett R. Levine
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - Bjorn R. Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, 02115, USA
| | - Hee-Jeong Im
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
- Internal Medicine Section of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Bioengineering, University of Illinois, Chicago, IL, 60612, USA
- Jesse Brown Veterans Affairs, Chicago, IL 60612, USA
| |
Collapse
|
29
|
Kuyinu EL, Narayanan G, Nair LS, Laurencin CT. Animal models of osteoarthritis: classification, update, and measurement of outcomes. J Orthop Surg Res 2016; 11:19. [PMID: 26837951 PMCID: PMC4738796 DOI: 10.1186/s13018-016-0346-5] [Citation(s) in RCA: 347] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/11/2016] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is one of the most commonly occurring forms of arthritis in the world today. It is a debilitating chronic illness causing pain and immense discomfort to the affected individual. Significant research is currently ongoing to understand its pathophysiology and develop successful treatment regimens based on this knowledge. Animal models have played a key role in achieving this goal. Animal models currently used to study osteoarthritis can be classified based on the etiology under investigation, primary osteoarthritis, and post-traumatic osteoarthritis, to better clarify the relationship between these models and the pathogenesis of the disease. Non-invasive animal models have shown significant promise in understanding early osteoarthritic changes. Imaging modalities play a pivotal role in understanding the pathogenesis of OA and the correlation with pain. These imaging studies would also allow in vivo surveillance of the disease as a function of time in the animal model. This review summarizes the current understanding of the disease pathogenesis, invasive and non-invasive animal models, imaging modalities, and pain assessment techniques in the animals.
Collapse
Affiliation(s)
- Emmanuel L Kuyinu
- Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, USA. .,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA.
| | - Ganesh Narayanan
- Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, USA. .,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA.
| | - Lakshmi S Nair
- Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, USA. .,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA. .,Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA. .,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, USA. .,Institute of Materials Science, University of Connecticut, Storrs, CT, USA.
| | - Cato T Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, USA. .,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA. .,Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA. .,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, USA. .,Institute of Materials Science, University of Connecticut, Storrs, CT, USA. .,Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA. .,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
30
|
Kim JS, Ali MH, Wydra F, Li X, Hamilton JL, An HS, Cs-Szabo G, Andrews S, Moric M, Xiao G, Wang JHC, Chen D, Cavanaugh JM, Im HJ. Characterization of degenerative human facet joints and facet joint capsular tissues. Osteoarthritis Cartilage 2015; 23:2242-2251. [PMID: 26117175 PMCID: PMC4663154 DOI: 10.1016/j.joca.2015.06.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 05/27/2015] [Accepted: 06/09/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Lumbar facet joint degeneration (FJD) may be an important cause of low back pain (LBP) and sciatica. The goal of this study was to characterize cellular alterations of inflammatory factor expression and neovascularization in human degenerative facet joint capsular (FJC) tissue. These alterations in FJC tissues in pain stimulation were also assessed. DESIGN FJs were obtained from consented patients undergoing spinal reconstruction surgery and cadaveric donors with no history of back pain. Histological analyses of the FJs were performed. Cytokine antibody array and quantitative real-time polymerase chain reaction (qPCR) were used to determine the production of inflammatory cytokines, and western blotting analyses (WB) were used to assay for cartilage-degrading enzymes and pain mediators. Ex vivo rat dorsal root ganglion (DRG) co-culture with human FJC tissues was also performed. RESULTS Increased neovascularization, inflammatory cell infiltration, and pain-related axonal-promoting factors were observed in degenerative FJCs surgically obtained from symptomatic subjects. Increased VEGF, (NGF/TrkA), and sensory neuronal distribution were also detected in degenerative FJC tissues from subjects with LBP. qPCR and WB results demonstrated highly upregulated inflammatory cytokines, pain mediators, and cartilage-degrading enzymes in degenerative FJCs. Results from ex vivo co-culture of the DRG and FJC tissue demonstrated that degenerative FJCs increased the expression of inflammatory pain molecules in the sensory neurons. CONCLUSION Degenerative FJCs possess greatly increased inflammatory and angiogenic features, suggesting that these factors play an important role in the progression of FJD and serve as a link between joint degeneration and neurological stimulation of afferent pain fibers.
Collapse
Affiliation(s)
- Jae-Sung Kim
- Department of Biochemistry, Rush University at Rush University Medical Center, Chicago, IL 60612, USA,The Division of Natural Medical Sciences, College of Health Science, Chosun University, Gwangju, 501-759, Republic of Korea
| | - Mir H. Ali
- Department of Orthopedic Surgery, Rush University at Rush University Medical Center, Chicago, IL 60612, USA
| | - Frank Wydra
- Department of Biochemistry, Rush University at Rush University Medical Center, Chicago, IL 60612, USA
| | - Xin Li
- Department of Biochemistry, Rush University at Rush University Medical Center, Chicago, IL 60612, USA
| | - John L. Hamilton
- Department of Biochemistry, Rush University at Rush University Medical Center, Chicago, IL 60612, USA
| | - Howard S. An
- Department of Orthopedic Surgery, Rush University at Rush University Medical Center, Chicago, IL 60612, USA
| | - Gabriella Cs-Szabo
- Department of Biochemistry, Rush University at Rush University Medical Center, Chicago, IL 60612, USA,Department of Orthopedic Surgery, Rush University at Rush University Medical Center, Chicago, IL 60612, USA
| | | | - Mario Moric
- Department of Anesthesiology, Rush University at Rush University Medical Center, Chicago, IL 60612, USA
| | - Guozhi Xiao
- Department of Biochemistry, Rush University at Rush University Medical Center, Chicago, IL 60612, USA,Department of Biology and Shenzhen Key Laboratory of Cell Microenvironment, South University of Science and Technology of China, Shenzhen, 518055, China
| | - James H-C Wang
- MechanoBiology Laboratory Departments of Orthopaedic Surgery, Bioengineering, and Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Di Chen
- Department of Biochemistry, Rush University at Rush University Medical Center, Chicago, IL 60612, USA
| | - John M. Cavanaugh
- Bioengineering Center, Wayne State University, Detroit, MI 48202, USA
| | - Hee-Jeong Im
- Department of Biochemistry, Rush University at Rush University Medical Center, Chicago, IL 60612, USA,Department of Orthopedic Surgery, Rush University at Rush University Medical Center, Chicago, IL 60612, USA,Department of Internal Medicine, Section of Rheumatology, Rush University at Rush University Medical Center, Chicago, IL 60612, USA,Department of Bioengineering, University of Illinois, Chicago, IL 60612, USA,Jesse Brown Veterans Affair, Chicago IL 60612, USA,Address correspondence to: Dr. Hee-Jeong Im Sampen, Rush University Medical Center, Cohn Research BD 516, 1735 W. Harrison St., Chicago, IL 60612, Tel: 312-942-3091, Fax: 312-942-3053,
| |
Collapse
|
31
|
Injecting vascular endothelial growth factor into the temporomandibular joint induces osteoarthritis in mice. Sci Rep 2015; 5:16244. [PMID: 26531672 PMCID: PMC4632030 DOI: 10.1038/srep16244] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/12/2015] [Indexed: 11/08/2022] Open
Abstract
It is unclear whether vascular endothelial growth factor (VEGF) can initiate osteoarthritis (OA) in the temporomandibular joint (TMJ). In this study we evaluated the effects of intra-articular injection of exogenous VEGF in the TMJ in mice on the early stage. Forty-eight male Sprague-Dawley mice were equally divided into 3 groups. In the vegf group, the mice received an injection of VEGF solution (50 μL) in the TMJ once a week over a period of 4 weeks. In the sham group, the mice received an injection of saline (50 μL). The control group did not receive any injection. Four mice from each group were sacrificed at 1, 2, 4, and 8 weeks. Gradual prominent cartilage degeneration was observed in the vegf group. Additionally, this group showed higher expressions of metalloproteinase (MMP)-9, MMP-13, receptor activator of nuclear factor-kappa-B ligand (RANKL), and a higher number of apoptotic chondrocytes and VEGF receptor 2 (VEGFR2)-positive chondrocytes. Micro-computed tomography (CT) revealed prominent subchondral bone resorption in the vegf group, with a high number of osteoclasts in the subchondral bone. In vitro study demonstrated that VEGF can promote osteoclast differentiation. In conclusion, our study found that VEGF can initiate TMJ OA by destroying cartilage and subchondral bone.
Collapse
|
32
|
Caron JP, Gandy JC, Schmidt M, Hauptman JG, Sordillo LM. Influence of Corticosteroids on Interleukin-1β-Stimulated Equine Chondrocyte Gene Expression. Vet Surg 2015; 42:231-7. [DOI: 10.1111/j.1532-950x.2012.01025.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- John P. Caron
- Department of Large Animal, College of Veterinary Medicine; Michigan State University; East Lansing MI
| | - Jeff C. Gandy
- Department of Large Animal, College of Veterinary Medicine; Michigan State University; East Lansing MI
| | - Michelle Schmidt
- Department of Large Animal, College of Veterinary Medicine; Michigan State University; East Lansing MI
| | - Joseph G. Hauptman
- Department of Small Animal Clinical Sciences; College of Veterinary Medicine, Michigan State University; East Lansing MI
| | - Lorraine M. Sordillo
- Department of Large Animal, College of Veterinary Medicine; Michigan State University; East Lansing MI
| |
Collapse
|
33
|
Wehland M, Aleshcheva G, Schulz H, Saar K, Hübner N, Hemmersbach R, Braun M, Ma X, Frett T, Warnke E, Riwaldt S, Pietsch J, Corydon TJ, Infanger M, Grimm D. Differential gene expression of human chondrocytes cultured under short-term altered gravity conditions during parabolic flight maneuvers. Cell Commun Signal 2015; 13:18. [PMID: 25889719 PMCID: PMC4369370 DOI: 10.1186/s12964-015-0095-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/02/2015] [Indexed: 01/06/2023] Open
Abstract
Background Chondrocytes are the main cellular component of articular cartilage. In healthy tissue, they are embedded in a strong but elastic extracelluar matrix providing resistance against mechanical forces and friction for the joints. Osteoarthritic cartilage, however, disrupted by heavy strain, has only very limited potential to heal. One future possibility to replace damaged cartilage might be the scaffold-free growth of chondrocytes in microgravity to form 3D aggregates. Results To prepare for this, we have conducted experiments during the 20th DLR parabolic flight campaign, where we fixed the cells after the first (1P) and the 31st parabola (31P). Furthermore, we subjected chondrocytes to isolated vibration and hypergravity conditions. Microarray and quantitative real time PCR analyses revealed that hypergravity regulated genes connected to cartilage integrity (BMP4, MMP3, MMP10, EDN1, WNT5A, BIRC3). Vibration was clearly detrimental to cartilage (upregulated inflammatory IL6 and IL8, downregulated growth factors EGF, VEGF, FGF17). The viability of the cells was not affected by the parabolic flight, but showed a significantly increased expression of anti-apoptotic genes after 31 parabolas. The IL-6 release of chondrocytes cultured under conditions of vibration was not changed, but hypergravity (1.8 g) induced a clear elevation of IL-6 protein in the supernatant compared with corresponding control samples. Conclusion Taken together, this study provided new insights into the growth behavior of chondrocytes under short-term microgravity. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0095-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Markus Wehland
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Ganna Aleshcheva
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Herbert Schulz
- Max-Delbrück-Center for Molecular Medicine, 13092, Berlin, Germany. .,University of Cologne, Cologne Center for Genomics (CCG), 50931, Cologne, Germany.
| | - Katrin Saar
- Max-Delbrück-Center for Molecular Medicine, 13092, Berlin, Germany.
| | - Norbert Hübner
- Max-Delbrück-Center for Molecular Medicine, 13092, Berlin, Germany.
| | - Ruth Hemmersbach
- DLR German Aerospace Center, Biomedical Research, Gravitational Biology, 51147, Köln, Germany.
| | - Markus Braun
- Institute for Molecular Physiology and Biotechnology of Plants (IMBIO), University of Bonn, Gravitational Biology Group, 53115, Bonn, Germany.
| | - Xiao Ma
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000, Aarhus C, Denmark.
| | - Timo Frett
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Biomedical Research, 51147, Köln, Germany.
| | - Elisabeth Warnke
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Stefan Riwaldt
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Jessica Pietsch
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Thomas Juhl Corydon
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000, Aarhus C, Denmark.
| | - Manfred Infanger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Daniela Grimm
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, DK-8000, Aarhus C, Denmark.
| |
Collapse
|
34
|
Liechti EF, Ferguson SJ, Tannast M. Protrusio acetabuli: joint loading with severe pincer impingement and its theoretical implications for surgical therapy. J Orthop Res 2015; 33:106-13. [PMID: 25297821 DOI: 10.1002/jor.22724] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 07/24/2014] [Indexed: 02/04/2023]
Abstract
Severe pincer impingement (acetabular protrusio) is an established cause of hip pain and osteoarthritis. The proposed underlying pathomechanism is a dynamic pathological contact of the prominent acetabular rim with the femoral head-neck junction. However, this cannot explain the classically described medial osteoarthritis in these hips. We therefore asked: (1) Does an overload exist in the medial aspect of the protrusio joint? and (2) What is the influence of three contemporary joint-preserving procedures on load distribution in protrusio hips? In vivo force and motion data for walking and standing to sitting were applied to six 3D finite element models (normal, dysplasia, protrusio, acetabular rim trimming, acetabular reorientation, and combined reorientation/rim trimming). Compared with dysplasia, the protrusio joint resulted in opposite patterns of von Mises stress and contact pressure during walking. In protrusio hips, we found an overload at the medial margin of the lunate surface (54% higher than normal). Isolated rim trimming further increased the medial overload (up to 28% higher than protrusio), whereas acetabular reorientation with/without rim trimming reduced stresses by up to 25%. Our results can be used as an adjunct for surgical decision making in the treatment of acetabular protrusio.
Collapse
Affiliation(s)
- Emanuel F Liechti
- Department of Orthopaedic Surgery, Inselspital, University of Bern, Switzerland; Institute for Surgical Technology and Biomechanics, University of Bern, Switzerland
| | | | | |
Collapse
|
35
|
Liu SC, Chuang SM, Hsu CJ, Tsai CH, Wang SW, Tang CH. CTGF increases vascular endothelial growth factor-dependent angiogenesis in human synovial fibroblasts by increasing miR-210 expression. Cell Death Dis 2014; 5:e1485. [PMID: 25341039 PMCID: PMC4649533 DOI: 10.1038/cddis.2014.453] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/21/2014] [Accepted: 09/04/2014] [Indexed: 01/15/2023]
Abstract
Connective tissue growth factor (CTGF, a.k.a. CCN2) is inflammatory mediator and abundantly expressed in osteoarthritis (OA). Angiogenesis is essential for OA progression. Here, we investigated the role of CTGF in vascular endothelial growth factor (VEGF) production and angiogenesis in OA synovial fibroblasts (OASFs). We showed that expression of CTGF and VEGF in synovial fluid were higher in OA patients than in controls. Directly applying CTGF to OASFs increased VEGF production then promoted endothelial progenitor cells tube formation and migration. CTGF induced VEGF by raising miR-210 expression via PI3K, AKT, ERK, and nuclear factor-κB (NF-κB)/ELK1 pathways. CTGF-mediating miR-210 upregulation repressed glycerol-3-phosphate dehydrogenase 1-like (GPD1L) expression and PHD activity and subsequently promoted hypoxia-inducible factor (HIF)-1α-dependent VEGF expression. Knockdown of CTGF decreased VEGF expression and abolished OASF-conditional medium-mediated angiogenesis in vitro as well as angiogenesis in chick chorioallantoic membrane and Matrigel-plug nude mice model in vivo. Taken together, our results suggest CTGF activates PI3K, AKT, ERK, and NF-κB/ELK1 pathway, leading to the upregulation of miR-210, contributing to inhibit GPD1L expression and prolyl hydroxylases 2 activity, promoting HIF-1α-dependent VEGF expression and angiogenesis in human synovial fibroblasts.
Collapse
Affiliation(s)
- S-C Liu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - S-M Chuang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - C-J Hsu
- 1] School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan [2] Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - C-H Tsai
- 1] Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan [2] Department of Medicine, Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - S-W Wang
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan
| | - C-H Tang
- 1] Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan [2] Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan [3] Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
36
|
Signaling pathways in cartilage repair. Int J Mol Sci 2014; 15:8667-98. [PMID: 24837833 PMCID: PMC4057753 DOI: 10.3390/ijms15058667] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/28/2014] [Accepted: 05/04/2014] [Indexed: 12/29/2022] Open
Abstract
In adult healthy cartilage, chondrocytes are in a quiescent phase characterized by a fine balance between anabolic and catabolic activities. In ageing, degenerative joint diseases and traumatic injuries of cartilage, a loss of homeostatic conditions and an up-regulation of catabolic pathways occur. Since cartilage differentiation and maintenance of homeostasis are finely tuned by a complex network of signaling molecules and biophysical factors, shedding light on these mechanisms appears to be extremely relevant for both the identification of pathogenic key factors, as specific therapeutic targets, and the development of biological approaches for cartilage regeneration. This review will focus on the main signaling pathways that can activate cellular and molecular processes, regulating the functional behavior of cartilage in both physiological and pathological conditions. These networks may be relevant in the crosstalk among joint compartments and increased knowledge in this field may lead to the development of more effective strategies for inducing cartilage repair.
Collapse
|
37
|
Moriyama H, Tobimatsu Y, Ozawa J, Kito N, Tanaka R. Amount of torque and duration of stretching affects correction of knee contracture in a rat model of spinal cord injury. Clin Orthop Relat Res 2013; 471:3626-36. [PMID: 23893364 PMCID: PMC3792286 DOI: 10.1007/s11999-013-3196-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 07/17/2013] [Indexed: 01/31/2023]
Abstract
BACKGROUND Joint contractures are a common complication of many neurologic conditions, and stretching often is advocated to prevent and treat these contractures. However, the magnitude and duration of the stretching done in practice usually are guided by subjective clinical impressions. QUESTIONS/PURPOSES Using an established T8 spinal cord injury rat model of knee contracture, we sought to determine what combination of static or intermittent stretching, varied by magnitude (high or low) and duration (long or short), leads to the best (1) improvement in the limitation in ROM; (2) restoration of the muscular and articular factors leading to contractures; and (3) prevention and treatment of contracture-associated histologic alterations of joint capsule and articular cartilage. METHODS Using a rat animal model, the spinal cord was transected completely at the level of T8. The rats were randomly assigned to seven treatment groups (n = 4 per group), which were composed of static or intermittent stretching in combination with different amounts of applied torque magnitude and duration. We assessed the effect of stretching by measuring the ROM and evaluating the histologic alteration of the capsule and cartilage. RESULTS Contractures improved in all treated groups except for the low-torque and short-duration static stretching conditions. High-torque stretching was effective against shortening of the synovial membrane and adhesions in the posterosuperior regions. Collagen Type II and VEGF in the cartilage were increased by stretching. CONCLUSIONS High-torque and long-duration static stretching led to greater restoration of ROM than the other torque and duration treatment groups. Stretching was more effective in improving articular components of contractures compared with the muscular components. Stretching in this rat model prevented shortening and adhesion of the joint capsule, and affected biochemical composition, but did not change morphologic features of the cartilage. CLINICAL RELEVANCE This animal study tends to support the ideas that static stretching can influence joint ROM and histologic qualities of joint tissues, and that the way stretching is performed influences its efficacy. However, further studies are warranted to determine if our findings are clinically applicable.
Collapse
Affiliation(s)
- Hideki Moriyama
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Tomogaoka 7-10-2, Suma-ku, Kobe, Hyogo, 654-0142, Japan,
| | | | | | | | | |
Collapse
|
38
|
Lugo JP, Saiyed ZM, Lau FC, Molina JPL, Pakdaman MN, Shamie AN, Udani JK. Undenatured type II collagen (UC-II®) for joint support: a randomized, double-blind, placebo-controlled study in healthy volunteers. J Int Soc Sports Nutr 2013; 10:48. [PMID: 24153020 PMCID: PMC4015808 DOI: 10.1186/1550-2783-10-48] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 10/10/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND UC-II contains a patented form of undenatured type II collagen derived from chicken sternum. Previous preclinical and clinical studies support the safety and efficacy of UC-II in modulating joint discomfort in osteoarthritis and rheumatoid arthritis. The purpose of this study was to assess the efficacy and tolerability of UC-II in moderating joint function and joint pain due to strenuous exercise in healthy subjects. METHODS This randomized, double-blind, placebo-controlled study was conducted in healthy subjects who had no prior history of arthritic disease or joint pain at rest but experienced joint discomfort with physical activity. Fifty-five subjects who reported knee pain after participating in a standardized stepmill performance test were randomized to receive placebo (n = 28) or the UC-II (40 mg daily, n = 27) product for 120 days. Joint function was assessed by changes in degree of knee flexion and knee extension as well as measuring the time to experiencing and recovering from joint pain following strenuous stepmill exertion. RESULTS After 120 days of supplementation, subjects in the UC-II group exhibited a statistically significant improvement in average knee extension compared to placebo (81.0 ± 1.3º vs 74.0 ± 2.2º; p = 0.011) and to baseline (81.0 ± 1.3º vs 73.2 ± 1.9º; p = 0.002). The UC-II cohort also demonstrated a statistically significant change in average knee extension at day 90 (78.8 ± 1.9º vs 73.2 ± 1.9º; p = 0.045) versus baseline. No significant change in knee extension was observed in the placebo group at any time. It was also noted that the UC-II group exercised longer before experiencing any initial joint discomfort at day 120 (2.8 ± 0.5 min, p = 0.019), compared to baseline (1.4 ± 0.2 min). By contrast, no significant changes were seen in the placebo group. No product related adverse events were observed during the study. At study conclusion, five individuals in the UC-II cohort reported no pain during or after the stepmill protocol (p = 0.031, within visit) as compared to one subject in the placebo group. CONCLUSIONS Daily supplementation with 40 mg of UC-II was well tolerated and led to improved knee joint extension in healthy subjects. UC-II also demonstrated the potential to lengthen the period of pain free strenuous exertion and alleviate the joint pain that occasionally arises from such activities.
Collapse
Affiliation(s)
- James P Lugo
- InterHealth Nutraceuticals, Benicia, CA 94510, USA
| | | | | | | | - Michael N Pakdaman
- Medicus Research LLC, 28720 Roadside Drive, Suite 310, Agoura Hills, CA 91301, USA
| | | | - Jay K Udani
- Medicus Research LLC, 28720 Roadside Drive, Suite 310, Agoura Hills, CA 91301, USA
- Northridge Hospital Integrative Medicine Program, Northridge, CA 91325, USA
| |
Collapse
|
39
|
Wu M, Xu T, Zhou Y, Lu H, Gu Z. Pressure and inflammatory stimulation induced increase of cadherin-11 is mediated by PI3K/Akt pathway in synovial fibroblasts from temporomandibular joint. Osteoarthritis Cartilage 2013; 21:1605-12. [PMID: 23916685 DOI: 10.1016/j.joca.2013.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 07/12/2013] [Accepted: 07/24/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The goal of the study was to investigate the expression of cadherin-11 in synovial fibroblasts (SFs) under mechanical or inflammatory stimuli, and its potential relationship with PI3K/Akt signaling pathway. METHODS SFs separated from rat temporomandibular joint (TMJ) were treated with hydrostatic pressures (HP) of 30, 60, 90, and 120 kPa, as well as tumor necrosis factor-α (TNF-α) for 12, 24, 48, and 72 h. The location of cadherin-11 was observed by immunofluorescence microscopy, and its expression was detected by real-time PCR and Western blot. We also studied the activation of PI3K/Akt signaling pathway in SFs with HP or TNF-α stimulation. RESULTS The results showed that increased expression of cadherin-11 could be found in the cell-cell contact site of SFs in response to HP and inflammatory stimulation. The mRNA and protein expression of cadherin-11 was positively correlated with the intensity of HP and the duration time of TNF-α treatment. Increased expression of vascular endothelial growth factor-D (VEGF-D) and activation of Akt were also found. Treatment with PI3K inhibitor LY294002 attenuated the pressure or inflammatory cytokine induction increases of cadherin-11, VEGF-D, and FGF-2 both in mRNA and protein levels. CONCLUSIONS These findings suggest that cadherin-11 may play important roles in SFs following exposure to mechanical loading and inflammatory stimulation. In addition, PI3K/Akt pathway was associated with pressure or inflammation-induced cadherin-11 expression, which may involve in the pathogenesis of temporomandibular diseases.
Collapse
Affiliation(s)
- M Wu
- Department of Orthodontics, Hospital of Stomatology, Zhejiang University, 395 Yan'an Road, Hangzhou 310006, China
| | | | | | | | | |
Collapse
|
40
|
Tsai CH, Chiang YC, Chen HT, Huang PH, Hsu HC, Tang CH. High glucose induces vascular endothelial growth factor production in human synovial fibroblasts through reactive oxygen species generation. Biochim Biophys Acta Gen Subj 2013; 1830:2649-58. [PMID: 23274526 DOI: 10.1016/j.bbagen.2012.12.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 12/03/2012] [Accepted: 12/16/2012] [Indexed: 11/28/2022]
Abstract
BACKGROUND Diabetes is an independent risk factor of osteoarthritis (OA). Angiogenesis is essential for the progression of OA. Here, we investigated the intracellular signaling pathways involved in high glucose (HG)-induced vascular endothelial growth factor (VEGF) expression in human synovial fibroblast cells. METHODS HG-mediated VEGF expression was assessed with qPCR and ELISA. The mechanisms of action of HG in different signaling pathways were studied using Western blotting. Knockdown of proteins was achieved by transfection with siRNA. Chromatin immunoprecipitation assays were used to study in vivo binding of c-Jun to the VEGF promoter. RESULTS Stimulation of OA synovial fibroblasts (OASF) with HG induced concentration- and time-dependent increases in VEGF expression. Treatment of OASF with HG increased reactive oxygen species (ROS) generation. Pretreatment with NADPH oxidase inhibitor (APO or DPI), ROS scavenger (NAC), PI3K inhibitor (Ly294002 or wortmannin), Akt inhibitor, or AP-1 inhibitor (curcumin or tanshinone IIA) blocked the HG-induced VEGF production. HG also increased PI3K and Akt activation. Treatment of OASF with HG increased the accumulation of phosphorylated c-Jun in the nucleus, AP-1-luciferase activity, and c-Jun binding to the AP-1 element on the VEGF promoter. CONCLUSIONS Our results suggest that the HG increases VEGF expression in human synovial fibroblasts via the ROS, PI3K, Akt, c-Jun and AP-1 signaling pathway. GENERAL SIGNIFICANCE We link high glucose on VEGF expression in osteoarthritis.
Collapse
Affiliation(s)
- Chun-Hao Tsai
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
41
|
Ludin A, Sela JJ, Schroeder A, Samuni Y, Nitzan DW, Amir G. Injection of vascular endothelial growth factor into knee joints induces osteoarthritis in mice. Osteoarthritis Cartilage 2013; 21:491-7. [PMID: 23257244 DOI: 10.1016/j.joca.2012.12.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 11/13/2012] [Accepted: 12/07/2012] [Indexed: 02/02/2023]
Abstract
UNLABELLED Osteoarthritis (OA) is a common joint disorder affecting circa 2% of the population. OBJECTIVES It has been suggested that secretion of vascular endothelial growth factor (VEGF) could play a role in the chain of events leading to OA. METHODS In the present study, healthy mice were injected intra-articularly with VEGF. RESULTS Shortly after the administration of VEGF, synovial hyperplasia, increased calcification of the articular cartilage and bone sclerosis were observed. Consequently, cartilage degradation characteristic of OA was found. These changes were seen to a lesser degree in the opposite knees of VEGF-injected mice and did not occur in the control mice. CONCLUSIONS The findings suggest an active role of VEGF in the pathogenesis of OA and render support to a possible role for subchondral bone sclerosis in the pathogenesis of cartilage degradation.
Collapse
Affiliation(s)
- A Ludin
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | |
Collapse
|
42
|
Wang G, Chen C, Yang R, Cao X, Lai S, Luo X, Feng Y, Xia X, Gong J, Hu J. p55PIK-PI3K stimulates angiogenesis in colorectal cancer cell by activating NF-κB pathway. Angiogenesis 2013; 16:561-73. [PMID: 23354733 DOI: 10.1007/s10456-013-9336-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 01/15/2013] [Indexed: 12/19/2022]
Abstract
Vascular growth factor (VEGF) is an important mediator of angiogenesis. PI3K plays essential roles in angiogenesis; however, the mechanisms and specific functions of individual isoforms of PI3K members in tumor angiogenesis regulation are still not fully understood. In this study, we evaluate the role of p55PIK, a PI3K regulatory subunit encoded by PIK3R3 gene, in tumor angiogenesis. We reported that overexpression of p55PIK in cancer cells up-regulated HIF-1α expression and increased VEGF expression. Furthermore, overexpression of p55PIK increased tumor angiogenesis in vivo and in vitro. Moreover, data indicated enhanced HIF-1α expression by p55PIK-PI3K depended on its ability to activate NF-кB signaling pathways, especially to increase the phosphorylation of p65 subunits of NF-κB. Our study suggested that p55PIK-PI3K was essential in regulating cancer cell-mediated angiogenesis and contributed to tumor growth and that the p55PIK provides a potential and specific target for new anti-angiogenesis drug development.
Collapse
Affiliation(s)
- Guihua Wang
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Okubo M, Kimura T, Fujita Y, Mochizuki S, Niki Y, Enomoto H, Suda Y, Toyama Y, Okada Y. Semaphorin 3A is expressed in human osteoarthritic cartilage and antagonizes vascular endothelial growth factor 165-promoted chondrocyte migration: an implication for chondrocyte cloning. ACTA ACUST UNITED AC 2013; 63:3000-9. [PMID: 21953086 DOI: 10.1002/art.30482] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Vascular endothelial growth factor 165 (VEGF165) and its receptors, including neuropilin 1 (NRP-1), are overexpressed in human osteoarthritic (OA) articular cartilage, although their functional roles in the cartilage are not fully understood. An axon-guidance molecule, semaphorin 3A (Sema3A), which binds to NRP-1, acts as an antagonist of VEGF signaling in endothelial cells. The aim of this study was to examine the expression of Sema3A and the functions of the VEGF165/Sema3A/NRP-1 axis in OA cartilage. METHODS The expression of Sema3A in OA and normal cartilage samples was examined by real-time polymerase chain reaction and immunohistochemical analyses. Functional analyses of VEGF165 and Sema3A were carried out using OA chondrocytes in culture. The migration activity of chondrocytes was examined in a monolayer wound assay. The effects of Sema3A on VEGF165-induced up-regulation of matrix metalloproteinases (MMPs) and intracellular signaling were also studied in cultured chondrocytes. RESULTS Sema3A expression was significantly elevated in OA cartilage as compared to normal cartilage. Sema3A immunoreactivity directly correlated with the Mankin score and with chondrocyte cloning. VEGF165 promoted the migration of chondrocytes, and this activity was suppressed by VEGF receptor 2 tyrosine kinase inhibitors. Sema3A antagonized the chondrocyte migration promoted by VEGF165, and the activity was blocked by a selective inhibitor of, or small interfering RNA for, Sema3A. VEGF165-induced overexpression of MMPs and phosphorylation of ERK and focal adhesion kinase in chondrocytes were inhibited by Sema3A. CONCLUSION Our findings provide the first evidence that Sema3A is overexpressed, with a direct correlation with cloning, in OA cartilage and that it suppresses the VEGF165-promoted migration of chondrocytes. Our findings also suggest that Sema3A plays a role in chondrocyte cloning through inhibition of cell migration in OA cartilage.
Collapse
Affiliation(s)
- Masashi Okubo
- Department of Pathology, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pulsatelli L, Addimanda O, Brusi V, Pavloska B, Meliconi R. New findings in osteoarthritis pathogenesis: therapeutic implications. Ther Adv Chronic Dis 2013; 4:23-43. [PMID: 23342245 DOI: 10.1177/2040622312462734] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This review focuses on the new perspectives which can provide insight into the crucial pathways that drive cartilage-bone physiopathology. In particular, we discuss the critical signaling and effector molecules that can activate cellular and molecular processes in both cartilage and bone cells and which may be relevant in cross talk among joint compartments: growth factors (bone morphogenetic proteins and transforming growth factor), hypoxia-related factors, cell-matrix interactions [discoidin domain receptor 2 (DDR2) and syndecan 4], signaling molecules [WNT, Hedgehog (Hh)]. With the continuous progression of our knowledge on the molecular pathways involved in cartilage and bone changes in osteoarthritis (OA), an increasing number of potentially effective candidates for OA therapy are already under scrutiny in clinical trials to ascertain their possible safe use in an attempt to identify molecules active in slowing or halting OA progression and reducing joint pain. We then review the principal molecules currently under clinical investigation.
Collapse
Affiliation(s)
- Lia Pulsatelli
- Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopaedic Institute, Bologna, Italy
| | | | | | | | | |
Collapse
|
45
|
Sahin H, Tholema N, Petersen W, Raschke MJ, Stange R. Impaired biomechanical properties correlate with neoangiogenesis as well as VEGF and MMP-3 expression during rat patellar tendon healing. J Orthop Res 2012; 30:1952-7. [PMID: 22615070 DOI: 10.1002/jor.22147] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 04/30/2012] [Indexed: 02/04/2023]
Abstract
Recent studies reveal an important role of vascular endothelial growth factor (VEGF)-induced angiogenesis in degenerative tendon diseases. The way how VEGF influences mechanical properties of the tendons is not well understood yet. We here hypothesized that tendinopathy results in a hypoxia-mediated stimulation of VEGF and that the mechanical stability of the tendon is impaired in an angiogenic process by VEGF-induced matrix metalloproteinases (MMPs). A modified in situ freezing model of patellar tendon was used to create a tendinopathy. 0, 7, 14, and 28 days post-surgical animals were sacrificed and patellar tendons were dissected for biomechanical and immunohistochemical analysis. Native tendons were used as controls. Immunohistochemical staining revealed a peak in HIF-1α stabilization immediately after surgery. Both VEGF and MMP-3 were increased 7 days after surgery. Angiogenesis was also abundant 7 days after surgery. In contrast, biomechanical stability of the tendon was decreased 7 days after surgery. The current results reveal a time-dependent correlation of HIF-1/VEGF-induced and MMP-3-supported angiogenesis with decreased biomechanical properties during tendon healing. The therapeutical modulation of neoangiogenesis by influencing the level of VEGF and MMP-3 might be a promising target for new approaches in degenerative tendon diseases. 30:1952-1957, 2012.
Collapse
Affiliation(s)
- Hacer Sahin
- Department of Trauma, Hand and Reconstructive Surgery, WWU Muenster, Germany
| | | | | | | | | |
Collapse
|
46
|
Jansen H, Meffert R, Birkenfeld F, Petersen W, Pufe T. Detection of vascular endothelial growth factor (VEGF) in moderate osteoarthritis in a rabbit model. Ann Anat 2012; 194:452-6. [DOI: 10.1016/j.aanat.2012.01.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 01/19/2012] [Accepted: 01/31/2012] [Indexed: 01/06/2023]
|
47
|
Weinans H, Siebelt M, Agricola R, Botter SM, Piscaer TM, Waarsing JH. Pathophysiology of peri-articular bone changes in osteoarthritis. Bone 2012; 51:190-6. [PMID: 22343134 DOI: 10.1016/j.bone.2012.02.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 02/01/2012] [Accepted: 02/02/2012] [Indexed: 12/01/2022]
Abstract
Osteoarthritis (OA) is a disease that involves the entire joint, but its pathophysiology is not well described. Alterations in peri-articular bone are an integral part of the OA disease process and different aspects of bone changes have been described in different patient (sub)groups and animal models. In this review we will discuss the osteoarthritis pathophysiology from the perspective of periarticular bone changes, which can be considered at three hierarchical levels: the bone (or joint) shape, the subchondral bone architecture and its cellular and molecular phenotype. In this review we try to provide an overview of the current knowledge of peri-articular bone changes in OA and what it could possibly imply for the initiation of OA and its progression. This article is part of a Special Issue entitled "Osteoarthritis".
Collapse
Affiliation(s)
- H Weinans
- Department of Orthopedics, Orthopaedic Research Laboratory, Erasmus MC, The Netherlands.
| | | | | | | | | | | |
Collapse
|
48
|
Masuko K, Murata M, Beppu M, Nakamura H, Kato T, Yudoh K. Sphingosine-1-phosphate modulates expression of vascular endothelial growth factor in human articular chondrocytes: a possible new role in arthritis. Int J Rheum Dis 2012; 15:366-73. [PMID: 22898216 DOI: 10.1111/j.1756-185x.2012.01756.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AIM Although sphingosine-1-phosphate (S1P) is suggested to have an important role in arthritis, its function in chondrocytes remains unknown. In contrast, vascular endothelial growth factor (VEGF) has been speculated to contribute to the pathogenesis of osteoarthritis (OA), most likely by regulating angiogenesis. We here investigated the in vitro effect of S1P on VEGF expression in human articular chondrocytes from OA patients. METHODS Human articular cartilage samples were obtained from patients with OA under informed consent. Chondrocytes were isolated by an enzymatic procedure, grown in monolayer culture, and then stimulated with S1P in the presence or absence of mitogen-activated protein kinase (MAPK) inhibitors or the Gi protein inhibitor pertussis toxin (PTX). VEGF expression and secretion in culture supernatants were analyzed using real-time polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS Although S1P did not enhance basal secretion of matrix metalloproteinase (MMP)-1 and MMP-13, it stimulated VEGF expression in human articular chondrocytes, both at the messenger RNA and protein levels. MAPK inhibitors SB203580 and PD98059 were not effective at suppressing VEGF induction; rather, blocking extracellular signal-regulated kinase (ERK) MAPK enhanced VEGF expression. The Gi protein inhibitor PTX partially attenuated S1P-induced VEGF secretion. CONCLUSION Our results suggest that S1P may contribute to the regulation of VEGF expression in human chondrocytes. S1P may therefore play a unique role in the pathophysiology of OA by regulating VEGF expression in chondrocytes.
Collapse
Affiliation(s)
- Kayo Masuko
- Department of Biochemistry, St. Marianna University School of Medicine, Kanagawa, Japan.
| | | | | | | | | | | |
Collapse
|
49
|
Fan W, Wu C, Miao X, Liu G, Saifzadeh S, Sugiyama S, Afara I, Crawford R, Xiao Y. Biomaterial scaffolds in cartilage–subchondral bone defects influencing the repair of autologous articular cartilage transplants. J Biomater Appl 2012; 27:979-89. [DOI: 10.1177/0885328211431310] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The repair of articular cartilage typically involves the repair of cartilage–subchondral bone tissue defects. Although various bioactive materials have been used to repair bone defects, how these bioactive materials in subchondral bone defects influence the repair of autologous cartilage transplant remains unclear. The aim of this study was to investigate the effects of different subchondral biomaterial scaffolds on the repair of autologous cartilage transplant in a sheep model. Cylindrical cartilage–subchondral bone defects were created in the right femoral knee joint of each sheep. The subchondral bone defects were implanted with hydroxyapatite–β-tricalcium phosphate (HA–TCP), poly lactic-glycolic acid (PLGA)-HA–TCP dual-layered composite scaffolds (PLGA/HA–TCP scaffolds), or autologous bone chips. The autologous cartilage layer was placed on top of the subchondral materials. After 3 months, the effect of different subchondral scaffolds on the repair of autologous cartilage transplant was systematically studied by investigating the mechanical strength, structural integration, and histological responses. The results showed that the transplanted cartilage layer supported by HA–TCP scaffolds had better structural integration and higher mechanical strength than that supported by PLGA/HA–TCP scaffolds. Furthermore, HA–TCP-supported cartilage showed higher expression of acid mucosubstances and glycol-amino-glycan contents than that supported by PLGA/HA–TCP scaffolds. Our results suggested that the physicochemical properties, including the inherent mechanical strength and material chemistry of the scaffolds, play important roles in influencing the repair of autologous cartilage transplants. The study may provide useful information for the design and selection of proper subchondral biomaterials to support the repair of both subchondral bone and cartilage defects.
Collapse
Affiliation(s)
- Wei Fan
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
- Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, People’s Republic of China
| | - Chengtie Wu
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| | - Xigeng Miao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| | - Gang Liu
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| | - Siamak Saifzadeh
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| | - Sadahiro Sugiyama
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| | - Isaac Afara
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| | - Ross Crawford
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| |
Collapse
|
50
|
The combined effects of continuous passive motion treatment and acellular PLGA implants on osteochondral regeneration in the rabbit. Biomaterials 2012; 33:3153-63. [PMID: 22264523 DOI: 10.1016/j.biomaterials.2011.12.054] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 12/31/2011] [Indexed: 11/21/2022]
Abstract
We investigated the active role of clinical rehabilitation in osteochondral regeneration using continuous passive motion (CPM) treatment together with acellular PLGA implants. CPM treatment was performed and compared with immobilization (Imm) treatment and intermittent active motion (IAM) treatment upon full-thickness osteochondral defects either with or without an PLGA implant in the PI (PLGA-implanted) and ED (empty defect) models. The PI and ED tests were performed in 38 rabbits for 4 and 12 weeks. At the end of testing, the PI-CPM group had the best regeneration with nearly normal articular surfaces and no joint contracture or inflammatory reaction. In contrast, degenerated joints, abrasion cartilage surfaces and synovitis were observed in the Imm and IAM groups. The achieved bone volume/tissue volume (BV/TV) ratio, which was measured using micro-CT, was significantly higher in the CPM group compared with the Imm and IAM groups; in particular, the performance of the PI-CPM group exceeds that of the ED-CPM group. The thickness of the trabecular (subchondral) bone was visibly increased in all of the groups from 4 through 12 weeks of testing. However, a histological analysis revealed differences in cartilage regeneration. At week 4, compared with the ED samples, all of the PI groups exhibited better collagen alignment and higher GAG content in the core of their repaired tissues, particularly in the PI-CPM group. At week 12, sound osteochondral repair and hyaline cartilaginous regeneration was observed in the PI-CPM group, and this was marked by type II collagen expression, osteocyte maturation, and trabecular boney deposition. In contrast, the PI-Imm and PI-IAM groups exhibited fibrocartilaginous tissues that had modest GAG content. In summary, this study demonstrates that early CPM treatment together with acellular PLGA implantation has significant positive effects on osteochondral regeneration in rabbit knee joint models.
Collapse
|