1
|
Kumar B, Ghaytidak AB, Pandey AK, Somepalli RR, Sarda P, Raychaudhuri SP, Rokkam MP. A Standardized Boswellia serrata Extract Improves Knee Joint Function and Cartilage Morphology in Human Volunteers with Mild to Moderate Osteoarthritis in a Randomized Placebo-Controlled Study. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2024:1-12. [PMID: 39700461 DOI: 10.1080/27697061.2024.2438894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND AND OBJECTIVE Boswellia serrata Roxb. ex Colebr. (Family: Burseraceae; Genus: Boswellia) gum resin (Salai guggul) has profound therapeutic value in Ayurvedic and Unani medicines in alleviating several chronic inflammatory illnesses, including arthritis, asthma, skin and blood diseases, fever, etc. SN13108F (Aflapin®) is a proprietary, standardized Boswellia serrata gum resin extract. This 180-day randomized, placebo-controlled clinical study aimed to evaluate cartilage morphology using magnetic resonance imaging (MRI), pain and joint function and long-term safety in the SN13108F-supplemented volunteers with knee osteoarthritis (KOA). MATERIALS AND METHODS Eighty adult male and female subjects with the Kellgren-Lawrence grade II - III KOA were supplemented with SN13108F (100 mg/day) or a matched placebo for 180 consecutive days. RESULTS SN13108F reduced (p < 0.001; vs. baseline and placebo) Western Ontario and McMaster Universities Osteoarthritis Index, Visual Analogue Scale, Lequesne's Functional Index scores, improved six-minute walk test, and stair climb test. Post-trial MRI assessments of the tibiofemoral joints revealed that the cartilage volume, thickness, and joint space width were increased (p < 0.001; vs. placebo), and levels of high-sensitivity C-reactive protein, matrix metalloproteinase-3, Fibulin-3, type II collagen degradation peptide in serum, and cross-linked C-terminal telopeptide of type II collagen in urine were significantly reduced (p < 0.001; vs. baseline and placebo) in the SN13108F-supplemented subjects. Hematology, complete serum biochemistry, urine analysis, and the participants' vital signs did not alter between the groups. CONCLUSION SN13108F supplementation is safe, and it mitigates joint pain and improves musculoskeletal function and cartilage morphology in KOA.
Collapse
Affiliation(s)
- Brijesh Kumar
- Department of Orthopedics, Lakshmi Hospital, Varanasi, India
| | | | - Abhinav Kumar Pandey
- Department of Radiology, Autonomous State Medical College Pilibhit, Bithaura Kalan, India
| | - Raghu Ram Somepalli
- Department of Orthopedics, MJ Naidu Super Speciality Hospital, Vijayawada, India
| | - Praveen Sarda
- Shreyam Specialist Orthopaedic Centre, Chaitanya Nagar, Ahmedabad, India
| | | | | |
Collapse
|
2
|
Emanuel KS, Huang L, Haartmans MJJ, Sanmartin Martinez J, Zijta F, Heeren RMA, Kerkhoffs GMMJ, Emans PJ, Cillero-Pastor B. Patient-responsive protein biomarkers for cartilage degeneration and repair identified in the infrapatellar fat pad. Expert Rev Proteomics 2024:1-11. [PMID: 39635821 DOI: 10.1080/14789450.2024.2438774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/13/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVES Cartilage defects (CDs) are regarded as early manifestation of osteoarthritis (OA). The infrapatellar fat pad (IPFP) is an important mediator in maintaining joint homeostasis, disease progression and tissue repair, with a crucial role of its secreted proteins. Here, we investigate the proteome of the IPFP in relation to clinical status and response to surgical treatment of CDs. METHODS In order to characterize the proteome of the IPFP, samples from a cohort of 53 patients who received surgical treatment for knee CDs were analyzed with label-free proteomics. Patients were divided based on validated outcome scores for pain and knee function, preoperatively and at 1-year postoperatively, and on MRI assessment of the defect severity, fibrosis and synovitis. RESULTS Specific proteins were differentially abundant in patients with MRI features and better clinical outcome after CD surgery, including a downregulation of cartilage intermediate layer protein 2 (CILP-2) and microsomal glutathione s-transferase 1 (MGST1), and an upregulation of aggrecan (ACAN), and proteoglycan 4 (PRG4). Pathways related to cell interaction, oxidation and matrix remodeling were altered. CONCLUSION Proteins in the IPFP that have a function in extracellular matrix, inflammation and immunomodulation were identified as potentially relevant markers for cartilage repair monitoring.
Collapse
Affiliation(s)
- Kaj S Emanuel
- Department of Orthopedic Surgery and Sports Medicine, Amsterdam Movement Sciences, Academic Center for Evidence Based Sports Medicine (ACES), Amsterdam Collaboration on Health & Safety in Sports (ACHSS), IOC Research Center, Amsterdam UMC, Amsterdam, The Netherlands
- Joint-Preserving Clinic, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University, Medical Center, Maastricht, The Netherlands
| | - Luojiao Huang
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering (cBITE), Maastricht University, Maastricht, The Netherlands
- Maastricht MultiModal Molecular Imaging Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, The Netherlands
| | - Mirella J J Haartmans
- Joint-Preserving Clinic, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University, Medical Center, Maastricht, The Netherlands
- Maastricht MultiModal Molecular Imaging Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, The Netherlands
| | - Javier Sanmartin Martinez
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering (cBITE), Maastricht University, Maastricht, The Netherlands
| | - Frank Zijta
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ron M A Heeren
- Maastricht MultiModal Molecular Imaging Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, The Netherlands
| | - Gino M M J Kerkhoffs
- Department of Orthopedic Surgery and Sports Medicine, Amsterdam Movement Sciences, Academic Center for Evidence Based Sports Medicine (ACES), Amsterdam Collaboration on Health & Safety in Sports (ACHSS), IOC Research Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - Pieter J Emans
- Joint-Preserving Clinic, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University, Medical Center, Maastricht, The Netherlands
| | - Berta Cillero-Pastor
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering (cBITE), Maastricht University, Maastricht, The Netherlands
- Maastricht MultiModal Molecular Imaging Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
3
|
Herren AOF, Amin AK, Hall AC. A Disintegrin and Metalloproteinase with Thrombospondin Motifs-4 Levels in Chondrocytes of Different Morphology within Nondegenerate and Early Osteoarthritic Human Femoral Head Cartilage. Cartilage 2024; 15:278-282. [PMID: 37272453 PMCID: PMC11418412 DOI: 10.1177/19476035231176532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/02/2023] [Indexed: 06/06/2023] Open
Affiliation(s)
- Arthur O. F. Herren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anish K. Amin
- Edinburgh Orthopaedics, Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, UK
| | - Andrew C. Hall
- Deanery of Biomedical Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Gao S, Jia M, Wang J, Sun Q, Liu F, Yu L, Guo Y, Li N, Wei L. Association of ADAMTS-5 gene polymorphisms with the susceptibility to knee osteoarthritis in a Chinese Han population. J Orthop Surg Res 2024; 19:513. [PMID: 39192347 PMCID: PMC11348706 DOI: 10.1186/s13018-024-05023-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most prevalent type of arthritis and the main reason for progressive disability in middle-aged and older people. Studies of candidate genes may provide a novel insight and treatment strategy for knee osteoarthritis (KOA). The aim of this study was to investigate the relationship between KOA susceptibility and single-nucleotide polymorphism (SNP) of the ADAMTS-5 gene. MATERIALS AND METHODS The case group included 188 patients from Luoyang Orthopedic Hospital with clinically and radiographically diagnosed primary KOA, and the control group included 100 age-matched individuals without KOA. Fifteen ADAMTS-5 SNPs were assayed using MALDI-TOF MS. Allelic and haplotypic frequencies were compared between the groups. The relationship between genotype distribution and risk of KOA was analyzed by multivariate logistic regression. RESULTS The frequency of A allele in rs2249350 site in the KOA group was significantly lower (odds ratio [OR]: 0.761; 95% confidence interval [95% CI]: 0.612-0.947; P = 0.016), while that of C allele was higher than that in the control group (OR: 1.176; 95% CI: 1.025-1.351; P = 0.016). AA genotype and gene model, especially recessive gene model at rs2249350 locus, negatively correlated with KOA risk after adjustment for sex, body mass index, age, and occupation (AA vs. CC: OR: 0.288; 95% CI: 0.124-0.669; P = 0.004; AA vs. CA + CC: OR: 0.348; 95% CI: 0.162-0.749; P = 0.007). Meanwhile, one protective haplotype, GA (rs229054, rs2249350) (OR: 0.763; 95% CI: 0.614-0.949; P = 0.017), and one high-risk haplotype, GC (rs229054, rs2249350) (OR: 1.259; 95% CI: 1.032-1.537; P = 0.019), were found in this study. CONCLUSION Despite a limited sample size, our study suggests that the rs2249350 polymorphism in the ADAMTS-5 gene is one of the genetic factors influencing the risk of KOA. The A allele and AA genotype of rs2249350 may protect from KOA, whereas C allele and CC genotype increase the risk of KOA. In addition, the GA haplotype (rs229054, rs2249350) might be associated with a decreased risk of KOA, whereas the GC haplotype (rs229054, rs2249350) may be a risk factor for KOA. Additional larger-sized studies in more ethnically diverse populations are needed to confirm these findings.
Collapse
Affiliation(s)
- Shan Gao
- Department of Spine, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong Province, 261041, China
| | - Menglong Jia
- Department of Spine, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong Province, 261041, China
| | - Jingwei Wang
- Department of Orthopedics, Luoyang Orthopedic Hospital of Henan Province, Luoyang, Henan Province, 471000, China
| | - Qiankun Sun
- Emergency Department, Luoyang No. 1 Traditional Chinese Medicine Hospital, Luoyang, Henan Province, 471000, China
| | - Fangxiu Liu
- Patient Service Center, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong Province, 261041, China
| | - Longtan Yu
- Department of Spine, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong Province, 261041, China
| | - YanXing Guo
- Department of Orthopedics, Luoyang Orthopedic Hospital of Henan Province, Luoyang, Henan Province, 471000, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Lei Wei
- Stomatology Department, Weifang People's Hospital, Weifang, Shandong Province, 261041, China.
| |
Collapse
|
5
|
Primorac D, Molnar V, Tsoukas D, Uzieliene I, Tremolada C, Brlek P, Klarić E, Vidović D, Zekušić M, Pachaleva J, Bernotiene E, Wilson A, Mobasheri A. Tissue engineering and future directions in regenerative medicine for knee cartilage repair: a comprehensive review. Croat Med J 2024; 65:268-287. [PMID: 38868973 PMCID: PMC11157252 DOI: 10.3325/cmj.2024.65.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/26/2024] [Indexed: 01/06/2025] Open
Abstract
This review evaluates the current landscape and future directions of regenerative medicine for knee cartilage repair, with a particular focus on tissue engineering strategies. In this context, scaffold-based approaches have emerged as promising solutions for cartilage regeneration. Synthetic scaffolds, while offering superior mechanical properties, often lack the biological cues necessary for effective tissue integration. Natural scaffolds, though biocompatible and biodegradable, frequently suffer from inadequate mechanical strength. Hybrid scaffolds, combining elements of both synthetic and natural materials, present a balanced approach, enhancing both mechanical support and biological functionality. Advances in decellularized extracellular matrix scaffolds have shown potential in promoting cell infiltration and integration with native tissues. Additionally, bioprinting technologies have enabled the creation of complex, bioactive scaffolds that closely mimic the zonal organization of native cartilage, providing an optimal environment for cell growth and differentiation. The review also explores the potential of gene therapy and gene editing techniques, including CRISPR-Cas9, to enhance cartilage repair by targeting specific genetic pathways involved in tissue regeneration. The integration of these advanced therapies with tissue engineering approaches holds promise for developing personalized and durable treatments for knee cartilage injuries and osteoarthritis. In conclusion, this review underscores the importance of continued multidisciplinary collaboration to advance these innovative therapies from bench to bedside and improve outcomes for patients with knee cartilage damage.
Collapse
Affiliation(s)
- Dragan Primorac
- Dragan Primorac, Poliklinika Sv. Katarina, Branimirova 71E, 10000 Zagreb, Croatia,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Dubuc J, Schneider MJ, Dubuc V, Richard H, Pinsard M, Bancelin S, Legare F, Girard C, Laverty S. Degradation of Proteoglycans and Collagen in Equine Meniscal Tissues. Int J Mol Sci 2024; 25:6439. [PMID: 38928148 PMCID: PMC11203490 DOI: 10.3390/ijms25126439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Investigate meniscal extracellular matrix degradation. Equine menisci (n = 34 from 17 horses) were studied. Site-matched sections were cut and scored from three regions (ROIs; n = 102) and stained for histology, proteoglycan (safranin O and fast green), aggrecan, and collagen cleavage (NITEGE, DIPEN, and C1,2C antibodies, respectively). Picrosirius red and second harmonic generation microscopy were performed to investigate collagen ultrastructure. A total of 42 ROIs met the inclusion criteria and were included in the final analysis. The median (range) ROI histological score was 3 (0-9), providing a large spectrum of pathology. The median (range) proteoglycan score was 1 (0-3), representing superficial and central meniscal loss. The median (range) of DIPEN, NITEGE, and C1,2C scores was 1 (0-3), revealing immunostaining of the femoral and tibial surfaces. The proteoglycan scores exhibited significant positive associations with both histologic evaluation (p = 0.03) and DIPEN scores (p = 0.02). Additionally, a robust positive association (p = 0.007) was observed between the two aggrecanolysis indicators, NITEGE and DIPEN scores. A negative association (p = 0.008) was identified between NITEGE and histological scores. The C1,2C scores were not associated with any other scores. Picrosirius red and second harmonic generation microscopy (SHGM) illustrated the loss of the collagen matrix and structure centrally. Proteoglycan and collagen degradation commonly occur superficially in menisci and less frequently centrally. The identification of central meniscal proteoglycan and collagen degradation provides novel insight into central meniscal degeneration. However, further research is needed to elucidate the etiology and sequence of degradative events.
Collapse
Affiliation(s)
- Julia Dubuc
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Melodie Jil Schneider
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Valerie Dubuc
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Helene Richard
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Maxime Pinsard
- Institut National de la Recherche Scientifique, Université du Quebec, 1650 Bd Lionel-Boulet, Varennes, Quebec, QC J3X1P7, Canada
| | - Stephane Bancelin
- Institut National de la Recherche Scientifique, Université du Quebec, 1650 Bd Lionel-Boulet, Varennes, Quebec, QC J3X1P7, Canada
| | - Francois Legare
- Institut National de la Recherche Scientifique, Université du Quebec, 1650 Bd Lionel-Boulet, Varennes, Quebec, QC J3X1P7, Canada
| | - Christiane Girard
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| | - Sheila Laverty
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte Saint-Hyacinthe, Quebec, QC J2S2M2, Canada; (J.D.); (M.J.S.); (V.D.); (H.R.); (C.G.)
| |
Collapse
|
7
|
Alharbi K, Asnayanti A, Do ADT, Perera R, Al-Mitib L, Shwani A, Rebollo MA, Kidd MT, Alrubaye AAK. Identifying Dietary Timing of Organic Trace Minerals to Reduce the Incidence of Osteomyelitis Lameness in Broiler Chickens Using the Aerosol Transmission Model. Animals (Basel) 2024; 14:1526. [PMID: 38891572 PMCID: PMC11171233 DOI: 10.3390/ani14111526] [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: 04/19/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Our prior research demonstrated a 20% to 25% reduction in bacterial chondronecrosis with osteomyelitis (BCO) lameness in broilers with organic Zn, Mn, and Cu (Availa® ZMC) supplementation. Expanding on this, we investigated the optimal timing for Availa® ZMC feeding to mitigate BCO lameness and reduce feed additive costs in the poultry industry. In this study, we compared the application of 0.15% Availa® ZMC for 56 days, the first 28 days, and the last 28 days. The experimental design was a randomized block design involving 1560 one-day-old chicks distributed across two wire-floor pens as BCO source infection and four treatment groups with six replicates. The source of BCO infection exhibited a cumulative lameness incidence of 83%, whereas the negative control group showed a 77% cumulative incidence of lameness (p = 0.125). Administering 0.15% of Availa® ZMC during the initial 28 d resulted in a 41.3% reduction in BCO incidence, significantly different from the supplementation during the last 28 d (p < 0.05). However, this reduction did not differ substantially (p > 0.05) from the 56d application period. Hence, administering 0.15% Availa® ZMC during the first four weeks emerges as the optimal timing protocol, providing a defense against lameness comparable to the continuous supplementation throughout the complete production duration. Implementing this feeding approach reduces the cost of feed additive, promotes the health of skeletal bones, and effectively protects against BCO lameness in broilers, offering a valuable consideration for producers seeking optimal outcomes in the poultry industry.
Collapse
Affiliation(s)
- Khawla Alharbi
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (K.A.); (A.A.); (A.D.T.D.); (R.P.); (L.A.-M.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Andi Asnayanti
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (K.A.); (A.A.); (A.D.T.D.); (R.P.); (L.A.-M.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
- National Agency of Drug and Food Control, Jakarta 10520, Indonesia
| | - Anh Dang Trieu Do
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (K.A.); (A.A.); (A.D.T.D.); (R.P.); (L.A.-M.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Ruvindu Perera
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (K.A.); (A.A.); (A.D.T.D.); (R.P.); (L.A.-M.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Layla Al-Mitib
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (K.A.); (A.A.); (A.D.T.D.); (R.P.); (L.A.-M.)
| | - Abdulkarim Shwani
- Southeast Poultry Research Laboratory, U.S. National Poultry Research Center, ARS, USDA, Athens, GA 30605, USA;
| | | | - Michael T. Kidd
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Adnan Ali Khalaf Alrubaye
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA; (K.A.); (A.A.); (A.D.T.D.); (R.P.); (L.A.-M.)
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
| |
Collapse
|
8
|
Kim H, Jung J, Lee M, Kim M, Kang N, Kim OK, Lee J. Curcuma longa L. extract exhibits anti-inflammatory and cytoprotective functions in the articular cartilage of monoiodoacetate-injected rats. Food Nutr Res 2024; 68:10402. [PMID: 38571919 PMCID: PMC10989232 DOI: 10.29219/fnr.v68.10402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 04/05/2024] Open
Abstract
Background Osteoarthritis (OA), the most prevalent form of arthritis, is a degenerative joint disease marked by the progressive deterioration of articular cartilage, leading to clinical manifestations such as joint pain. Objective This study investigated the effects of Curcuma longa L. extract (CL) containing curcumin, demethoxycurcumin, and bisdemethoxycurcumin on monosodium iodoacetate (MIA)-induced OA rats. Design Sprague-Dawley rats with MIA-induced OA received CL supplementation at doses of 5, 25, and 40 mg/kg body weight. Results CL extract administration suppressed mineralisation parameters and morphological modifications and decreased arachidonate5-lipoxygenase and leukotriene B4 levels in articular cartilage. Additionally, it decreased serum prostaglandin E2, NO, and glycosaminoglycanlevels as well as the protein expression of phosphorylated inhibitor kappa B-alpha, phosphorylated p65, cyclooxygenase-2, and inducible nitric oxide synthase in the cartilage of MIA-injected rats. Furthermore, it also reduced matrix metalloproteinases and elevated SMAD family member 3 phosphorylation, tissue inhibitor of metalloproteinases, aggrecan, collagen type I, and collagen type II levels in the articular cartilage of MIA-induced OA rats. Conclusions This study's findings suggest that CL supplementation helps prevent OA development and is an effective therapy for OA.
Collapse
Affiliation(s)
- Hyelim Kim
- Department of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Jaeeun Jung
- Department of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Minhee Lee
- Department of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea
- Department of Food Innovation and Health, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Minha Kim
- Nutrione Co., Ltd, Seoul 05510, Republic of Korea
| | - Namgil Kang
- Nutrione Co., Ltd, Seoul 05510, Republic of Korea
| | - Ok-Kyung Kim
- Division of Food and Nutrition and Human Ecology Research Institute, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jeongmin Lee
- Department of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea
- Department of Food Innovation and Health, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, Republic of Korea
| |
Collapse
|
9
|
Davis S, Zekonyte J, Karali A, Roldo M, Blunn G. Early Degenerative Changes in a Spontaneous Osteoarthritis Model Assessed by Nanoindentation. Bioengineering (Basel) 2023; 10:995. [PMID: 37760097 PMCID: PMC10525236 DOI: 10.3390/bioengineering10090995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Understanding early mechanical changes in articular cartilage (AC) and subchondral bone (SB) is crucial for improved treatment of osteoarthritis (OA). The aim of this study was to develop a method for nanoindentation of fresh, unfixed osteochondral tissue to assess the early changes in the mechanical properties of AC and SB. Nanoindentation was performed throughout the depth of AC and SB in the proximal tibia of Dunkin Hartley guinea pigs at 2 months, 3 months, and 2 years of age. The contralateral tibias were either histologically graded for OA or analyzed using immunohistochemistry. The results showed an increase in the reduced modulus (Er) in the deep zone of AC during early-stage OA (6.0 ± 1.75 MPa) compared to values at 2 months (4.04 ± 1.25 MPa) (*** p < 0.001). In severe OA (2-year) specimens, there was a significant reduction in Er throughout the superficial and middle AC zones, which correlated to increased ADAMTS 4 and 5 staining, and proteoglycan loss in these regions. In the subchondral bone, a 35.0% reduction in stiffness was observed between 2-month and 3-month specimens (*** p < 0.001). The severe OA age group had significantly increased SB stiffness of 36.2% and 109.6% compared to 2-month and 3-month-old specimens respectively (*** p < 0.001). In conclusion, this study provides useful information about the changes in the mechanical properties of both AC and SB during both early- and late-stage OA and indicates that an initial reduction in stiffness of the SB and an increase in stiffness in the deep zone of AC may precede early-stage cartilage degeneration.
Collapse
Affiliation(s)
- Sarah Davis
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth PO1 2DT, UK; (M.R.); (G.B.)
- School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth PO1 3DJ, UK; (J.Z.); (A.K.)
| | - Jurgita Zekonyte
- School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth PO1 3DJ, UK; (J.Z.); (A.K.)
| | - Aikaterina Karali
- School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth PO1 3DJ, UK; (J.Z.); (A.K.)
| | - Marta Roldo
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth PO1 2DT, UK; (M.R.); (G.B.)
| | - Gordon Blunn
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth PO1 2DT, UK; (M.R.); (G.B.)
| |
Collapse
|
10
|
Shenegelegn Mern D, Thomé C. Synergetic enrichment of aggrecan in nucleus pulposus cells by scAAV6-shRNA-mediated knockdown of aggrecanase-1 and aggrecanase-2. Exp Biol Med (Maywood) 2023; 248:1134-1144. [PMID: 37354087 PMCID: PMC10583755 DOI: 10.1177/15353702231171905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/07/2023] [Indexed: 06/26/2023] Open
Abstract
Degenerative disk disease (DDD) that aggravates structural deterioration of intervertebral disks (IVDs) can be accompanied by painful inflammation and immunopathological progressions. Current surgical or pharmacological therapies cannot repair the structure and function of IVDs. Nucleus pulposus (NP) cells are crucial for the preservation or restoration of IVDs by balancing the anabolic and catabolic factors affecting the extracellular matrix. Imbalanced anabolic and catabolic factors cause increased degradation of aggrecan. Aggrecanases A Disintegrin And Metalloproteinase with ThromboSpondin motifs (ADAMTS)4 and ADAMTS5 are the main degrading enzymes of aggrecan. Previously, we characterized adeno-associated virus (AAV6) as the most suitable serotype with marked NP cellular tropism and demonstrated that ADAMTS4 could be silenced by self-complementary adeno-associated virus grade 6 small helix ribonucleic acid (scAAV6-shRNA) in NP cells of degeneration grade III, which resulted in enrichment of aggrecan. Nonetheless, neither scAAV6-shRNA-mediated inhibition of ADAMTS5 nor joint inhibitions of ADAMTS4 and ADAMTS5 have been investigated, although both enzymes are regulated by analogous proinflammatory cytokines and have the same cleavage sites in aggrecan. Therefore, we attempted scAAV6-shRNA-mediated inhibitions of both enzymes in NP cells of degeneration grade IV to increase efficacies in treatments of DDD. The degeneration grade of IVDs in patients was determined by magnetic resonance imaging (MRI) before surgical operations. After isolation and culturing of NP cells, cells were transduced with scAAV6-shRNAs targeting ADAMTS4 or ADAMTS5. Transduced cells were analyzed by reverse transcription quantitative polymerase chain reaction (RT-qPCR), fluorescence microscopy, flow cytometry-assisted cell sorting (FACS), MTT assay (3-(4, 5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide assay), immunoblotting, and enzyme-linked immunosorbent assay (ELISA). Joint transduction of NP cells exhibited high transduction efficacies (98.1%), high transduction units (TU) (1381 TU/Cell), and no effect on cell viability or proliferation. Above all joint treatments resulted in effective knockdown of ADAMTS4 (92.8%) and ADAMTS5 (93.4%) along with additive enrichment of aggrecan (113.9%). Treatment effects were significant for more than 56 days after transduction (P < 0.001). In conclusion, scAAV6-shRNA-mediated combined molecular therapy could be very valuable for more effective, durable, and less immunogenic treatment approaches in DDD.
Collapse
Affiliation(s)
| | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck 6020, Austria
| |
Collapse
|
11
|
Ochoa M, Yang A, Kollias C, Bakir C, Carsen S, Lazier J, Innes AM, Pagé M, Dawrant J, Robinson ME, Koujok K, Shenouda N, Rauch F, Ward LM. From " ACAN" to "I CAN": Restoring wellness in a boy with severe osteochondritis dissecans through diagnostic precision combined with optimal medical, surgical and rehabilitation management. Bone Rep 2023; 18:101663. [PMID: 36950254 PMCID: PMC10025132 DOI: 10.1016/j.bonr.2023.101663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 01/29/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023] Open
Abstract
Osteochondritis dissecans (OCD) is a disease of the joints characterized by idiopathic focal subchondral lesions. Aggrecan, a proteoglycan encoded by the ACAN gene, is important for cartilage structure and function. We describe the clinical evolution of a patient with short stature, multi-focal OCD, and subchondral osteopenia that appeared linked to a novel pathogenic ACAN variant. A multi-disciplinary approach including medical (bisphosphonate) therapy, surgical intervention and rehabilitation were successful in restoring wellness and physical function.
Collapse
Affiliation(s)
- Maria Ochoa
- The Ottawa Pediatric Bone Health Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Endocrinology Unit, Division of Pediatrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Pediatric Genetic and Metabolic Bone Disorders Program, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Ashlee Yang
- Department of Pediatrics, Division of Endocrinology, University of Alberta, Edmonton, Alberta, Canada
| | - Carrie Kollias
- Royal Children's Hospital Melbourne, Melbourne, Australia
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Christina Bakir
- Department of Orthopedic Surgery, Chinook Regional Hospital, Lethbridge, Alberta, Canada
| | - Sasha Carsen
- The Ottawa Pediatric Bone Health Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Surgery, University of Ottawa, Division of Pediatric Orthopedic Surgery, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Joanna Lazier
- Department of Medical Genetics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - A. Micheil Innes
- Department of Medical Genetics, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Marika Pagé
- The Ottawa Pediatric Bone Health Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Pediatric Genetic and Metabolic Bone Disorders Program, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Jonathan Dawrant
- Department of Pediatrics, Division of Endocrinology, University of Calgary, Calgary, Alberta, Canada
| | - Marie-Eve Robinson
- The Ottawa Pediatric Bone Health Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Pediatrics, University Of Ottawa, Division of Endocrinology, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- Pediatric Genetic and Metabolic Bone Disorders Program, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Khaldoun Koujok
- The Ottawa Pediatric Bone Health Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Medical Imaging, University of Ottawa, Division of Pediatric Radiology, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Nazih Shenouda
- The Ottawa Pediatric Bone Health Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Medical Imaging, University of Ottawa, Division of Pediatric Radiology, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Frank Rauch
- Shriners Hospital for Children, McGill University, Montreal, Quebec, Canada
| | - Leanne M. Ward
- The Ottawa Pediatric Bone Health Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Department of Pediatrics, University Of Ottawa, Division of Endocrinology, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- Pediatric Genetic and Metabolic Bone Disorders Program, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
- Corresponding author at: Tier 1 Clinical Research Chair in Pediatric Bone Disorders, University of Ottawa and Pediatric Endocrinologist Division of Endocrinology and Metabolism, Children's Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, Ontario KIH 8L1, Canada.
| |
Collapse
|
12
|
Król M, Kupnicka P, Bosiacki M, Chlubek D. Mechanisms Underlying Anti-Inflammatory and Anti-Cancer Properties of Stretching-A Review. Int J Mol Sci 2022; 23:ijms231710127. [PMID: 36077525 PMCID: PMC9456560 DOI: 10.3390/ijms231710127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 02/07/2023] Open
Abstract
Stretching is one of the popular elements in physiotherapy and rehabilitation. When correctly guided, it can help minimize or slow down the disabling effects of chronic health conditions. Most likely, the benefits are associated with reducing inflammation; recent studies demonstrate that this effect from stretching is not just systemic but also local. In this review, we present the current body of knowledge on the anti-inflammatory properties of stretching at a molecular level. A total of 22 papers, focusing on anti-inflammatory and anti-cancer properties of stretching, have been selected and reviewed. We show the regulation of oxidative stress, the expression of pro- and anti-inflammatory genes and mediators, and remodeling of the extracellular matrix, expressed by changes in collagen and matrix metalloproteinases levels, in tissues subjected to stretching. We point out that a better understanding of the anti-inflammatory properties of stretching may result in increasing its importance in treatment and recovery from diseases such as osteoarthritis, systemic sclerosis, and cancer.
Collapse
Affiliation(s)
- Małgorzata Król
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Correspondence:
| | - Mateusz Bosiacki
- Chair and Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University, Żołnierska 54, 71-210 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
13
|
Smith MM, Hayes AJ, Melrose J. Pentosan Polysulphate (PPS), a Semi-Synthetic Heparinoid DMOAD With Roles in Intervertebral Disc Repair Biology emulating The Stem Cell Instructive and Tissue Reparative Properties of Heparan Sulphate. Stem Cells Dev 2022; 31:406-430. [PMID: 35102748 DOI: 10.1089/scd.2022.0007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review highlights the attributes of pentosan polysulphate (PPS) in the promotion of intervertebral disc (IVD) repair processes. PPS has been classified as a disease modifying osteoarthritic drug (DMOAD) and many studies have demonstrated its positive attributes in the countering of degenerative changes occurring in cartilaginous tissues during the development of osteoarthritis (OA). Degenerative changes in the IVD also involve inflammatory cytokines, degradative proteases and cell signalling pathways similar to those operative in the development of OA in articular cartilage. PPS acts as a heparan sulphate (HS) mimetic to effect its beneficial effects in cartilage. The IVD contains small cell membrane HS-proteoglycans (HSPGs) such as syndecan, and glypican and a large multifunctional HS/chondroitin sulphate (CS) hybrid proteoglycan (HSPG2/perlecan) that have important matrix stabilising properties and sequester, control and present growth factors from the FGF, VEGF, PDGF and BMP families to cellular receptors to promote cell proliferation, differentiation and matrix synthesis. HSPG2 also has chondrogenic properties and stimulates the synthesis of extracellular matrix (ECM) components, expansion of cartilaginous rudiments and has roles in matrix stabilisation and repair. Perlecan is a perinuclear and nuclear proteoglycan in IVD cells with roles in chromatin organisation and control of transcription factor activity, immunolocalises to stem cell niches in cartilage, promotes escape of stem cells from quiescent recycling, differentiation and attainment of pluripotency and migratory properties. These participate in tissue development and morphogenesis, ECM remodelling and repair. PPS also localises in the nucleus of stromal stem cells, promotes development of chondroprogenitor cell lineages, ECM synthesis and repair and discal repair by resident disc cells. The availability of recombinant perlecan and PPS offer new opportunities in repair biology. These multifunctional agents offer welcome new developments in repair strategies for the IVD.
Collapse
Affiliation(s)
- Margaret M Smith
- The University of Sydney Raymond Purves Bone and Joint Research Laboratories, 247198, St Leonards, New South Wales, Australia;
| | - Anthony J Hayes
- Cardiff School of Biosciences, University of Cardiff, UK, Bioimaging Unit, Cardiff, Wales, United Kingdom of Great Britain and Northern Ireland;
| | - James Melrose
- Kolling Institute, University of Sydney, Royal North Shore Hospital, Raymond Purves Lab, Sydney Medical School Northern, Level 10, Kolling Institute B6, Royal North Shore Hospital, St. Leonards, New South Wales, Australia, 2065.,University of New South Wales, 7800, Graduate School of Biomedical Engineering, University of NSW, Sydney, New South Wales, Australia, 2052;
| |
Collapse
|
14
|
Gurram S, Anchi P, Panda B, Tekalkar SS, Mahajan RB, Godugu C. Amelioration of experimentally induced inflammatory arthritis by intra-articular injection of visnagin. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100114. [PMID: 35992378 PMCID: PMC9389203 DOI: 10.1016/j.crphar.2022.100114] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/04/2022] [Accepted: 06/16/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
| | | | | | | | | | - Chandraiah Godugu
- Corresponding author. Department of Biological Sciences (Regulatory Toxicology), National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad Balanagar, Hyderabad, Telangana State, India.
| |
Collapse
|
15
|
Assessment of serum concentrations of matrix metalloproteinase 1, matrix metalloproteinase 2 and tissue inhibitors of metalloproteinases 1 in atopic dermatitis in correlation with disease severity and epidermal barrier parameters. Postepy Dermatol Alergol 2021; 38:773-779. [PMID: 34849123 PMCID: PMC8610038 DOI: 10.5114/ada.2021.110043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 04/25/2020] [Indexed: 11/17/2022] Open
Abstract
Introduction Matrix metalloproteinases (MMPs) are a group of proteolytic enzymes, conditioning the integrity of skin cells, however, their role in the inflammatory process of atopic dermatitis (AD) and the direct effect on the epidermal barrier parameters remain unexplained. Aim To assess MMP-1, MMP-2, tissue inhibitors of metalloproteinases (TIMP)-1 concentrations in blood serum in the context of transepidermal water loss (TEWL) and stratum corneum hydration in AD. Moreover, serum levels of MMPs and TIMP-1 were analysed in relation to the Eczema Area and Severity Index (EASI). Material and methods Forty-three AD patients and 22 control group subjects have been investigated. Serum concentrations of MMP-1, MMP-2, and TIMP-1 have been evaluated with ELISA. TEWL and stratum corneum hydration have been assessed with a TM300 Tewameter and a CM825 Corneometer. Skin lesions in patients with AD have been evaluated with the Eczema Area and Severity Index. Results MMP-1 and MMP-2 serum concentrations were significantly higher in the AD group. The results of TIMP-1 serum concentration were similar for both groups. The correlation between the serum concentration and the EASI was demonstrated only for MMP-2 for patients with severe and moderate AD. Patients with AD and TIMP-1 serum concentration greater than MMP-1 presented lower TEWL and higher epidermal hydration. Conclusions The results of this study warrant further investigation. The predominance of TIMP-1 over MMP-1 in blood serum can potentially limit TEWL and maintain the proper water content of the epidermis. Future work is necessary to establish how reliable the role of MMP-2 concentration is as an indicator of the severity of AD.
Collapse
|
16
|
Kubanov AA, Karamova AE, Chikin VV, Verbenko DA, Znamenskaya LF, Artamonova OG. Genetic markers for psoriatic arthritis in patients with psoriasis. Part I: non-HLA genes. VESTNIK DERMATOLOGII I VENEROLOGII 2021. [DOI: 10.25208/vdv1260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Psoriatic arthritis often develops in patients with psoriasis and can lead to joint deformity, stiffness, dysfunction, and disability. Psoriatic arthritis is a polygenic disease. and the issue of personalizing the prognosis of its development can only be resolved taking into account the variability of plenty genomic loci associated with the development of the disease. The personification of the prognosis of the disease can be solved taking into account the variability of the set of genomic loci with which its development is associated. The review examines genomic polymorphisms associated with the development of psoriatic arthritis not psoriasis, except of HLA polymorphisms. Genome regions containing polymorphisms, allelic variants of which are associated both with the development of psoriatic arthritis and reducing the likelihood of its occurrence, are described. It has been reported that the predisposition to the development of psoriatic arthritis in patients with psoriasis is determined by genes encoding proteins involved in inflammation and bone metabolism.
Collapse
|
17
|
Ferreira A, Duarte T, Marques S, Costa P, Neves S, dos Santos T, Granja P, Porto G. Iron triggers the early stages of cartilage degeneration in vitro: The role of articular chondrocytes. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100145. [DOI: 10.1016/j.ocarto.2021.100145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
|
18
|
Bartoli DMF, Felizatti AL, do Bomfim FRC, Bovo JL, de Aro AA, do Amaral MEC, Esquisatto MAM. Laser treatment of synovial inflammatory process in experimentally induced microcrystalline arthritis in Wistar rats. Lasers Med Sci 2021; 36:529-540. [PMID: 32519204 DOI: 10.1007/s10103-020-03055-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 06/01/2020] [Indexed: 01/11/2023]
Abstract
The presence of intra-articular crystals is detected in different articular pathologies of acute or chronic nature. The aim of this work was to analyze the action of the indium gallium aluminum and phosphorus (InGaAlP) (λ = 670 nm) laser on the synovial membrane present in the knee joint in experimentally induced microcrystalline arthritis in male adult Wistar rats. The animals were divided into three experimental groups (n = 24): control (A), experimentally induced arthritis (B), experimentally induced arthritis+InGaAlP laser therapy (C). The laser treatment was made daily in the patellar region of the right knee after 48 h of the experimental induction. After 7, 14, and 21 days of therapy, the rats were euthanized and the right knees were removed and processed for histomorphometric, immunohistochemical, ultrastructural, and biochemical investigation of the synovium. The number of granulocytes on the 14th and 21st days was higher in B and lower in C and, lastly, in A. The number of fibroblasts on the 14th and 21st days was similar between A and C and below B. The number of blood vessels on the 21st day was higher in B than in the other groups. The positive number of cells for the TUNEL test was higher on the 14th and 21st days in B compared to the others. The percentage of tissue area occupied by birefringent collagen fibers was higher in B on the 21st day than in the others. The ultrastructure of cells showed fibroblast-like morphology in all groups and periods evaluated. The quantification of glycosaminoglycans did not present significant differences between the groups in all the experimental periods. The amount of hydroxyproline was higher in B compared to the other groups on the 14th and 21st days. The content of non-collagen proteins was higher in B on the 21st day in relation to the other groups. Quantification of TNF-α on the 21st day was higher in A and B than in C. For TGF-β on the 21st day, groups B and C presented similar and higher values than A. For MMP-13, groups A and B presented data similar to and above C. In relation to ADAMT-S4, on the 21st day, groups B and C presented data similar to and lower than A. InGaAlP-670 nm therapy reduced the inflammatory process and tissue injuries of the synovial membrane in comparison to the untreated group, indicating its potential utilization in clinical studies aiming in the recovery of acute arthritis in patients.
Collapse
Affiliation(s)
- Débora Mara Fortes Bartoli
- Graduate Program in Biomedical Sciences, University Center of Herminio Ometto Foundation - FHO, Av. Dr. Maximiliano Baruto, 500, Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Airton Luiz Felizatti
- Graduate Program in Biomedical Sciences, University Center of Herminio Ometto Foundation - FHO, Av. Dr. Maximiliano Baruto, 500, Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Fernando Russo Costa do Bomfim
- Graduate Program in Biomedical Sciences, University Center of Herminio Ometto Foundation - FHO, Av. Dr. Maximiliano Baruto, 500, Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Julia Leme Bovo
- Graduate Program in Biomedical Sciences, University Center of Herminio Ometto Foundation - FHO, Av. Dr. Maximiliano Baruto, 500, Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Andrea Aparecida de Aro
- Graduate Program in Biomedical Sciences, University Center of Herminio Ometto Foundation - FHO, Av. Dr. Maximiliano Baruto, 500, Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Maria Esméria Corezzola do Amaral
- Graduate Program in Biomedical Sciences, University Center of Herminio Ometto Foundation - FHO, Av. Dr. Maximiliano Baruto, 500, Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Marcelo Augusto Marretto Esquisatto
- Graduate Program in Biomedical Sciences, University Center of Herminio Ometto Foundation - FHO, Av. Dr. Maximiliano Baruto, 500, Jd. Universitário, Araras, SP, 13607-339, Brazil.
| |
Collapse
|
19
|
Abstract
Osteochondritis dissecans (OCD) is a chronic disease of the articular cartilage characterized by focal lesions of subchondral bone and overlaying cartilage. Through the growing number of reports describing the high prevalence of OCD in some families, the subcategory termed familial OCD (FOCD) was established. With the development of genetic approaches such as genome-wide association studies and sequencing, aggrecan (ACAN) has been identified as one of the genes of interest associated with FOCD. Aggrecan is a crucial protein for the preservation and function of cartilage. However, due to FOCD being characterized relatively recently, there is a paucity of literature on the subject. The purpose of this review is to explore the relationship between ACAN mutations and familial OCD as well as to explore current treatment options and avenues for future research. In vitro and animal studies have shown the importance of ACAN in the preservation of cartilage. However, the only human ACAN mutation related to OCD ever identified is a V2303M mutation in the G3 domain. Multiple treatments have been superficially explored, and some options such as growth hormone (GH) and gonadotrophin-releasing hormone agonists (GnRHa) show potential. Thus, further research on FOCD in needed to identify other ACAN mutations and determine optimal treatment modalities for this patient population.
Collapse
|
20
|
Sugitani K, Egorova D, Mizumoto S, Nishio S, Yamada S, Kitagawa H, Oshima K, Nadano D, Matsuda T, Miyata S. Hyaluronan degradation and release of a hyaluronan-aggrecan complex from perineuronal nets in the aged mouse brain. Biochim Biophys Acta Gen Subj 2020; 1865:129804. [PMID: 33253804 DOI: 10.1016/j.bbagen.2020.129804] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Perineuronal nets (PNNs) are insoluble aggregates of extracellular matrix molecules in the brain that consist of hyaluronan (HA) and chondroitin sulfate proteoglycans (CSPGs). PNNs promote the acquisition and storage of memories by stabilizing the formation of synapses in the adult brain. Although the deterioration of PNNs has been suggested to contribute to the age-dependent decline in brain function, the molecular mechanisms underlying age-related changes in PNNs remain unclear. METHODS The amount and solubility of PNN components were investigated by sequential extraction followed by a disaccharide analysis and immunoblotting. We examined the interaction between HA and aggrecan, a major HA-binding CSPG, by combining mass spectrometry and pull-down assays. RESULTS The solubility and amount of HA increased in the brain with age. Among several CSPGs, the solubility of aggrecan was selectively elevated during aging. In contrast to alternations in biochemical properties, the expression of PNN components at the transcript level was not markedly changed by aging. The increased solubility of aggrecan was not due to the loss of HA-binding properties. Our results indicated that the degradation of high-molecular-mass HA induced the release of the HA-aggrecan complex from PNNs in the aged brain. CONCLUSION The present study revealed a novel mechanism underlying the age-related deterioration of PNNs in the brain.
Collapse
Affiliation(s)
- Kei Sugitani
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Diana Egorova
- Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan
| | - Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-Ku, Nagoya 468-8503, Japan
| | - Shunsuke Nishio
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-Ku, Nagoya 468-8503, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada-Ku, Kobe 658-8558, Japan
| | - Kenzi Oshima
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Daita Nadano
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Tsukasa Matsuda
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Food and Agricultural Sciences, Fukushima University, Kanayagawa 1, Fukushima 960-1296, Japan
| | - Shinji Miyata
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan.
| |
Collapse
|
21
|
Matrix Metalloproteinases MMP-2 and MMP-9, Their Inhibitors TIMP-1 and TIMP-2, Vascular Endothelial Growth Factor and sVEGFR-2 as Predictive Markers of Ischemic Retinopathy in Patients with Systemic Sclerosis-Case Series Report. Int J Mol Sci 2020; 21:ijms21228703. [PMID: 33218057 PMCID: PMC7698901 DOI: 10.3390/ijms21228703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/27/2020] [Accepted: 11/14/2020] [Indexed: 02/06/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune connective tissue disorder associated with multiple organ involvement. The aim of the study was to present two SSc patients who were diagnosed with ischemic retinopathy in both eyes. As a background to our case study, we decided to investigate the imbalance of angiogenesis factors in 25 SSc patients in relation to 25 healthy controls. Assays of matrix metalloproteinases-2 and -9 (MMP-2, MMP-9), tissue inhibitor of metalloproteinases-1 (TIMP-1) and -2 (TIMP-2), vascular endothelial growth factor (VEGF), and soluble VEGF receptor-2 (sVEGFR-2) in blood serum and tears were performed. A significantly increased levels of MMP-9 in serum and tears, (p = 0.0375 and p < 0.001, respectively) as well as VEGF/sVEGFR-2 ratio in tears (p < 0.001) were found in the whole SSc patients group compared with controls, while reduced levels of these parameters in patients with ischemic sclerodermic retinopathy were noted. We also observed decreased level MMP-2 in tears and increased levels of TIMP-2 in blood serum and tears of SSc patients with retinal ischemic changes. MMP-9, MMP-2, TIMP-2, and VEGF/sVEGFR-2 may play a crucial role in ischemic retinal degeneration or retinal reorganization in SSc.
Collapse
|
22
|
The Anti-ADAMTS-5 Nanobody ® M6495 Protects Cartilage Degradation Ex Vivo. Int J Mol Sci 2020; 21:ijms21175992. [PMID: 32825512 PMCID: PMC7503673 DOI: 10.3390/ijms21175992] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/28/2020] [Accepted: 08/02/2020] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is associated with cartilage breakdown, brought about by ADAMTS-5 mediated aggrecan degradation followed by MMP-derived aggrecan and type II collagen degradation. We investigated a novel anti-ADAMTS-5 inhibiting Nanobody® (M6495) on cartilage turnover ex vivo. Bovine cartilage (BEX, n = 4), human osteoarthritic - (HEX, n = 8) and healthy—cartilage (hHEX, n = 1) explants and bovine synovium and cartilage were cultured up to 21 days in medium alone (w/o), with pro-inflammatory cytokines (oncostatin M (10 ng/mL) + TNFα (20 ng/mL) (O + T), IL-1α (10 ng/mL) or oncostatin M (50 ng/mL) + IL-1β (10 ng/mL)) with or without M6495 (1000−0.46 nM). Cartilage turnover was assessed in conditioned medium by GAG (glycosaminoglycan) and biomarkers of ADAMTS-5 driven aggrecan degradation (huARGS and exAGNxI) and type II collagen degradation (C2M) and formation (PRO-C2). HuARGS, exAGNxI and GAG peaked within the first culture week in pro-inflammatory stimulated explants. C2M peaked from day 14 by O + T and day 21 in co-culture experiments. M6495 dose dependently decreased huARGS, exAGNxI and GAG after pro-inflammatory stimulation. In HEX C2M was dose-dependently reduced by M6495. M6495 showed no effect on PRO-C2. M6495 showed cartilage protective effects by dose-dependently inhibiting ADAMTS-5 mediated cartilage degradation and inhibiting overall cartilage deterioration in ex vivo cartilage cultures.
Collapse
|
23
|
Velloso Alvarez A, Boone LH, Pondugula SR, Caldwell F, Wooldridge AA. Effects of Autologous Conditioned Serum, Autologous Protein Solution, and Triamcinolone on Inflammatory and Catabolic Gene Expression in Equine Cartilage and Synovial Explants Treated With IL-1β in Co-culture. Front Vet Sci 2020; 7:323. [PMID: 32671108 PMCID: PMC7332692 DOI: 10.3389/fvets.2020.00323] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/11/2020] [Indexed: 01/13/2023] Open
Abstract
Autologous conditioned serum (ACS) and autologous protein solution (APS) are newer therapeutic options for osteoarthritis (OA). Co-culture of cartilage and synovium stimulated with IL-1β produces a similar physiologic response to tissues from naturally-ocurring OA. The study objective was to investigate the effects of ACS, APS, and triamcinolone (TA) on inflammatory and catabolic gene expression of inflamed joint tissues in co-culture. Blood was collected and processed for ACS and APS from six horses. Cartilage and synovial explants were harvested from the stifle, placed in co-culture, and treated as: (1) unstimulated control (2) stimulated control (3) ACS at 25% v/v (4) ACS at 50% v/v (5) APS at 25% v/v (6) APS at 50% v/v, (7) TA (10−6 M). Treatment groups 2–7 were stimulated with IL-1β (10 ng/ml). Cultures were maintained for 96 hours, and then both media and explants were harvested for measurement of gene expression and protein. IL-1β stimulation significantly increased IL-1β (p = 0.029), IL-8 (p = 0.011) and MMP-3 (p = 0.043) expression in synovium and IL-1β (p = 0.003) and TNF-α (p = 0.001) expression in cartilage. Treatment with 50% ACS and APS v/v downregulated IL-1β expression in cartilage more than TA treatment (p = 0.001 and p = 0.0004) and APS downregulated MMP-1 expression in synovial membrane (p = 0.025). Treatment with ACS and APS caused a trend in upregulation of IL-10 expression in synovium and type II collagen and aggrecan expression in cartilage. PGE2 media concentrations were significantly reduced following treatment with APS (13.7-fold decrease, p = 0.0001) and ACS (4.13-fold decrease, p = 0.024); while TA did not reduce PGE2 significantly (2.3-fold decreased p = 0.406). As disease-modifying therapies, ACS and APS modified the cellular response from synovial membrane and articular cartilage. ACS and APS may offer an improved strategy to improve clinical signs of horses with naturally occurring OA, compared to TA treatment.
Collapse
Affiliation(s)
- Ana Velloso Alvarez
- Department of Clinical Sciences, Auburn University, Auburn, AL, United States
| | - Lindsey H Boone
- Department of Clinical Sciences, Auburn University, Auburn, AL, United States
| | | | - Fred Caldwell
- Department of Clinical Sciences, Auburn University, Auburn, AL, United States
| | - Anne A Wooldridge
- Department of Clinical Sciences, Auburn University, Auburn, AL, United States
| |
Collapse
|
24
|
Onitsuka K, Murata K, Kokubun T, Fujiwara S, Nakajima A, Morishita Y, Kanemura N. Effects of Controlling Abnormal Joint Movement on Expression of MMP13 and TIMP-1 in Osteoarthritis. Cartilage 2020; 11:98-107. [PMID: 29938527 PMCID: PMC6921957 DOI: 10.1177/1947603518783449] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Abnormal joint movement is associated with osteoarthritis (OA). Previous studies using the controlling abnormal joint movement (CAJM) model of OA reported delayed cartilage degeneration; however, none of them focused on gait performance and the localization of matrix metalloproteinase 13 (MMP13) and tissue inhibitor of metalloproteinase-1 (TIMP-1) in chondrocytes. Therefore, we aimed to investigate the effect of controlling abnormal joint movement on gait performance and the localization of MMP13 and TIMP-1, using kinematic and histological analyses. DESIGN Rats were assigned to 2 groups: anterior cruciate ligament transection (ACL-T) group and CAJM group (n = 5/group); contralateral hind limbs of ACL-T rats were designated as intact. After 1, 2, and 4 weeks, step length was analyzed, and after 2, 4, and 8 weeks, Safranin O-Fast Green staining and immunohistochemical staining for MMP13 and TIMP-1 were performed. RESULTS Step length did not differ significantly between the groups. However, degeneration of articular cartilage was higher in the ACL-T group than in the intact group (P < 0.05). There was no significant difference in the CAJM group at all time points. Immunohistochemical analysis of the MMP13/TIMP-1 relationship revealed a significant increase in the expression ratio of MMP13 after 4 weeks in the ACL-T group compared to the CAJM group (P < 0.05). CONCLUSIONS Controlling abnormal joint movement may reduce mechanical stress owing to kinematic elements of small articulation including joint instability and delayed cartilage degeneration, despite the lack of kinematic change in step length.
Collapse
Affiliation(s)
- Katsuya Onitsuka
- Department of Rehabilitation, Tokyo
Women’s Medical University Yachiyo Medical Center, Chiba, Japan
| | - Kenji Murata
- Department of Physical Therapy, School
of Health and Social Services, Saitama Prefectural University, Saitama, Japan
| | - Takanori Kokubun
- Department of Physical Therapy, School
of Health and Social Services, Saitama Prefectural University, Saitama, Japan
| | - Shuhei Fujiwara
- Graduate Course of Health and Social
Services, Graduate School of Saitama Prefectural University, Saitama, Japan
| | - Aya Nakajima
- Graduate Course of Health and Social
Services, Graduate School of Saitama Prefectural University, Saitama, Japan
| | - Yuri Morishita
- Graduate Course of Health and Social
Services, Graduate School of Saitama Prefectural University, Saitama, Japan
| | - Naohiko Kanemura
- Department of Physical Therapy, School
of Health and Social Services, Saitama Prefectural University, Saitama, Japan,Naohiko Kanemura, Department of Physical
Therapy, School of Health and Social Services, Saitama Prefectural University,
820 Sannomiya, Saitama 343-8540, Japan.
| |
Collapse
|
25
|
Camargo Garbin L, McIlwraith CW, Frisbie DD. Evaluation of allogeneic freeze-dried platelet lysate in cartilage exposed to interleukin 1-β in vitro. BMC Vet Res 2019; 15:386. [PMID: 31675958 PMCID: PMC6824121 DOI: 10.1186/s12917-019-2118-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 09/27/2019] [Indexed: 11/24/2022] Open
Abstract
Background Platelet-rich plasma (PRP) as well as other platelet-derived products have been used as a potential disease-modifying treatment for musculoskeletal diseases, such as osteoarthritis (OA). The restorative properties of such products rely mainly on the high concentrations of growth factors, demonstrating encouraging results experimentally and clinically. Yet, the autologous blood-derived nature of the PRP product lead to limitations that precludes it’s widespread use. The main limitations for PRP use are; product variability, the need for minimum laboratory settings in most cases, and the need for storage at low temperatures to preserve its properties. Based on these limitations, the objective of this study was to investigate an allogeneic off-the-shelf platelet lysate (PL) in cartilage exposed to interleukin 1β (IL-1β). For this purpose, blood and cartilage were harvested from eight skeletally mature and healthy horses. Blood was processed into PL aliquots and divided into three groups (Frozen, Freeze-dried and Filtered freeze-dried), used in autologous and allogeneic conditions and in three different concentrations (1.5, 3 and 6-fold). Different PL preparations were then applied in cartilage culture with interleukin-1 beta and cultured for 10 days. Cartilage and media samples were collected and analyzed for total GAG and 35SO4-labeled GAG content. Results No significant differences between the controls and PL groups in cartilage and media were demonstrated. The effects of PL on cartilage matrix were concentration dependent and intermediate concentrations (3-fold) in PL showed increased 35SO4-labelled GAG in cartilage. Conclusion In conclusion, the allogeneic freeze-dried PL presented equivalent effects compared to frozen autologous PL. Intermediate platelet concentration on average demonstrated improved results, demonstrating less GAG loss compared to other concentrations.
Collapse
Affiliation(s)
- Livia Camargo Garbin
- Department of Clinical Veterinary Sciences, School of Veterinary Medicine, Faculty of Medical Sciences, University of West Indies, St. Augustine Campus, St. Augustine, Trinidad and Tobago.
| | - C Wayne McIlwraith
- C.Wayne McIlwraith Translational Medicine Institute, Orthopaedic Research Center, Colorado State University, 2350 Gillette Drive, Fort Collins, CO, 80523, USA
| | - David D Frisbie
- C.Wayne McIlwraith Translational Medicine Institute, Orthopaedic Research Center, Colorado State University, 2350 Gillette Drive, Fort Collins, CO, 80523, USA
| |
Collapse
|
26
|
Yan L, Zhou L, Xie D, Du W, Chen F, Yuan Q, Tong P, Shan L, Efferth T. Chondroprotective effects of platelet lysate towards monoiodoacetate-induced arthritis by suppression of TNF-α-induced activation of NF-ĸB pathway in chondrocytes. Aging (Albany NY) 2019; 11:2797-2811. [PMID: 31089001 PMCID: PMC6535074 DOI: 10.18632/aging.101952] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 05/02/2019] [Indexed: 12/14/2022]
Abstract
Platelet lysate (PL) contains a cocktail of growth factors that actively participates in cartilage repair. This study was designed to determine the effect and mechanism of PL on osteoarthritis (OA). An arthritis model was established to mimic human OA by intra-articular injection of monoiodoacetate (MIA) to Sprague Dawley (SD) rats. The model was weekly treated with PL by intra-articular injection. Thermal withdrawal latency, mechanical withdrawal threshold, and treadmill gait were tested for pain behavior observation. Histopathological and immunohistochemical analyses were conducted for evaluating cartilage degradation. Real time PCRs and Western blots were conducted to elucidate the mechanism of PL on primary chondrocytes. Results showed that, in vivo, PL significantly attenuated pain symptoms and exerted chondrocyte-protective and extracellular matrix (ECM)-modifying effect on the arthritic cartilage in a dose-dependent manner. The in situ expressions of type II Collagen (Col2) and matrix metalloproteinase 13 (Mmp13) in the arthritic cartilage was abnormal and was restored by PL. In vitro, PL significantly restored tumor necrosis factor α (TNF-α)-suppressed anabolic gene expression (Col2 and aggrecan) and TNF-α-increased catabolic gene expression (Col10, Mmp13, Adamts5, and Adamts9) in chondrocytes. The effects were mediated by TNF-α downstream signaling, including inhibition of NF-κB and c-Jun activities. This study provides certain knowledge of anti-OA effect and TNF signaling-related mechanism of PL, placing it as a promising and alternative option for OA therapy in the future.
Collapse
Affiliation(s)
- Li Yan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
- Center for Stem Cell Translational Research, Zhejiang Chinese Medical University, Hangzhou, China
- Equal contribution
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
- Center for Stem Cell Translational Research, Zhejiang Chinese Medical University, Hangzhou, China
- Equal contribution
| | - Danting Xie
- Center for Stem Cell Translational Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenxi Du
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangming Chen
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Yuan
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
- Center for Stem Cell Translational Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
27
|
Aterido A, Cañete JD, Tornero J, Ferrándiz C, Pinto JA, Gratacós J, Queiró R, Montilla C, Torre-Alonso JC, Pérez-Venegas JJ, Fernández Nebro A, Muñoz-Fernández S, González CM, Roig D, Zarco P, Erra A, Rodríguez J, Castañeda S, Rubio E, Salvador G, Díaz-Torné C, Blanco R, Willisch Domínguez A, Mosquera JA, Vela P, Sánchez-Fernández SA, Corominas H, Ramírez J, de la Cueva P, Fonseca E, Fernández E, Puig L, Dauden E, Sánchez-Carazo JL, López-Estebaranz JL, Moreno D, Vanaclocha F, Herrera E, Blanco F, Fernández-Gutiérrez B, González A, Pérez-García C, Alperi-López M, Olivé Marques A, Martínez-Taboada V, González-Álvaro I, Sanmartí R, Tomás Roura C, García-Montero AC, Bonàs-Guarch S, Mercader JM, Torrents D, Codó L, Gelpí JL, López-Corbeto M, Pluma A, López-Lasanta M, Tortosa R, Palau N, Absher D, Myers R, Marsal S, Julià A. Genetic variation at the glycosaminoglycan metabolism pathway contributes to the risk of psoriatic arthritis but not psoriasis. Ann Rheum Dis 2019; 78:e214158. [PMID: 30552173 DOI: 10.1136/annrheumdis-2018-214158] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Psoriatic arthritis (PsA) is a chronic inflammatory arthritis affecting up to 30% of patients with psoriasis (Ps). To date, most of the known risk loci for PsA are shared with Ps, and identifying disease-specific variation has proven very challenging. The objective of the present study was to identify genetic variation specific for PsA. METHODS We performed a genome-wide association study in a cohort of 835 patients with PsA and 1558 controls from Spain. Genetic association was tested at the single marker level and at the pathway level. Meta-analysis was performed with a case-control cohort of 2847 individuals from North America. To confirm the specificity of the genetic associations with PsA, we tested the associated variation using a purely cutaneous psoriasis cohort (PsC, n=614) and a rheumatoid arthritis cohort (RA, n=1191). Using network and drug-repurposing analyses, we further investigated the potential of the PsA-specific associations to guide the development of new drugs in PsA. RESULTS We identified a new PsA risk single-nucleotide polymorphism at B3GNT2 locus (p=1.10e-08). At the pathway level, we found 14 genetic pathways significantly associated with PsA (pFDR<0.05). From these, the glycosaminoglycan (GAG) metabolism pathway was confirmed to be disease-specific after comparing the PsA cohort with the cohorts of patients with PsC and RA. Finally, we identified candidate drug targets in the GAG metabolism pathway as well as new PsA indications for approved drugs. CONCLUSION These findings provide insights into the biological mechanisms that are specific for PsA and could contribute to develop more effective therapies.
Collapse
Affiliation(s)
- Adrià Aterido
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Juan D Cañete
- Rheumatology Department, Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | - Jesús Tornero
- Rheumatology Department, Hospital Universitario Guadalajara, Guadalajara, Spain
| | - Carlos Ferrándiz
- Dermatology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - José Antonio Pinto
- Rheumatology Department, Complejo Hospitalario Juan Canalejo, A Coruña, Spain
| | - Jordi Gratacós
- Rheumatology Department, Hospital Parc Taulí, Sabadell, Spain
| | - Rubén Queiró
- Rheumatology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Carlos Montilla
- Rheumatology Department, Hospital Virgen de la Vega, Salamanca, Spain
| | | | | | - Antonio Fernández Nebro
- Rheumatology Department, Instituto de Investigación Biomédica de Málaga, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Santiago Muñoz-Fernández
- Rheumatology Department, Hospital Universitario Infanta Sofía, Universidad Europea, Madrid, Spain
| | - Carlos M González
- Rheumatology Department, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Daniel Roig
- Rheumatology Department, Hospital Moisès Broggi, Barcelona, Spain
| | - Pedro Zarco
- Rheumatology Department, Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | - Alba Erra
- Rheumatology Department, Hospital Sant Rafael, Barcelona, Spain
| | - Jesús Rodríguez
- Rheumatology Department, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Santos Castañeda
- Rheumatology Department, Hospital Universitario La Princesa, IIS La Princesa, Madrid, Spain
| | - Esteban Rubio
- Rheumatology Department, Centro de Salud Virgen de los Reyes, Sevilla, Spain
| | - Georgina Salvador
- Rheumatology Department, Hospital Universitario Mútua de Terrassa, Terrassa, Spain
| | - Cesar Díaz-Torné
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ricardo Blanco
- Rheumatology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | - José Antonio Mosquera
- Rheumatology Department, Complejo Hospitalario Hospital Provincial de Pontevedra, Pontevedra, Spain
| | - Paloma Vela
- Rheumatology Department, Hospital General Universitario de Alicante, Alicante, Spain
| | | | - Héctor Corominas
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Rheumatology Department, Hospital Dos de Maig, Barcelona, Spain
| | - Julio Ramírez
- Rheumatology Department, Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | - Pablo de la Cueva
- Dermatology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Eduardo Fonseca
- Dermatology Department, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Emilia Fernández
- Dermatology Department, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Lluis Puig
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Esteban Dauden
- Dermatology Department, Hospital Universitario La Princesa, IIS La Princesa, Madrid, Spain
| | | | | | - David Moreno
- Dermatology Department, Hospital Virgen Macarena, Sevilla, Spain
| | | | - Enrique Herrera
- Dermatology Department, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Francisco Blanco
- Rheumatology Department, INIBIC-Hospital Universitario A Coruña, A Coruña, Spain
| | | | - Antonio González
- Instituto de Investigación Sanitaria Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | | | | | | | | | | | - Raimon Sanmartí
- Rheumatology Department, Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | | | | | - Sílvia Bonàs-Guarch
- Barcelona Supercomputing Centre (BSC), Joint BSC-CRG-IRB Research Program in Computational Biology, Barcelona, Spain
| | - Josep Maria Mercader
- Barcelona Supercomputing Centre (BSC), Joint BSC-CRG-IRB Research Program in Computational Biology, Barcelona, Spain
| | - David Torrents
- Barcelona Supercomputing Centre (BSC), Joint BSC-CRG-IRB Research Program in Computational Biology, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Laia Codó
- Life Sciences Department, Barcelona Supercomputing Centre, Barcelona, Spain
| | - Josep Lluís Gelpí
- Life Sciences Department, Barcelona Supercomputing Centre, Barcelona, Spain
| | | | - Andrea Pluma
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Maria López-Lasanta
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Raül Tortosa
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Nuria Palau
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Richard Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Sara Marsal
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Antonio Julià
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| |
Collapse
|
28
|
Felizatti AL, do Bomfim FRC, Bovo JL, de Aro AA, do Amaral MEC, Esquisatto MAM. Effects of low-level laser therapy on the organization of articular cartilage in an experimental microcrystalline arthritis model. Lasers Med Sci 2019; 34:1401-1412. [PMID: 30762197 DOI: 10.1007/s10103-019-02740-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022]
Abstract
The aim of this study was to evaluate the effects of low-level laser therapy using the gallium arsenide laser (λ = 830 nm) on the articular cartilage (AC) organization from knee joint in an experimental model of microcrystalline arthritis in adult male Wistar rats. Seventy-two animals were divided into three groups: A (control), B (induced arthritis), and C (induced arthritis + laser therapy). The arthritis was induced in the right knee using 2 mg of Na4P2O7 in 0.5 mL of saline solution. The treatments were daily applied in the patellar region of the right knee after 48 h of induction. On the 7th, 14th, and 21st days of treatment, the animals were euthanized and their right knees were removed and processed for structural and biochemical analysis of the AC. The chondrocytes positively labeled for the TUNEL reaction were lower in C than in B on the 14th and 21st days. The content of glycosaminoglycans and hydroxyproline in A and C was higher than B on the 21st day. The amount of tibial TNF-α in B and C was lower than in A. The amount of tibial BMP-7 in B and C was higher than in A. The femoral MMP-13 was lower in B and C than for A. The tibial TGF-β for C was higher than the others. The femoral ADAMT-S4 content of A and C presented similar and inferior data to B on the 21st day. The AsGa-830 nm therapy preserved the content of glycosaminoglycans, reduced the cellular changes and the inflammatory process compared to the untreated group.
Collapse
Affiliation(s)
- Airton Luiz Felizatti
- Programa de Pós-graduação em Ciências Biomédicas, Centro Universitário da Fundação Hermínio Ometto - FHO, Av. Dr. Maximiliano Baruto, 500 Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Fernando Russo Costa do Bomfim
- Programa de Pós-graduação em Ciências Biomédicas, Centro Universitário da Fundação Hermínio Ometto - FHO, Av. Dr. Maximiliano Baruto, 500 Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Julia Leme Bovo
- Programa de Pós-graduação em Ciências Biomédicas, Centro Universitário da Fundação Hermínio Ometto - FHO, Av. Dr. Maximiliano Baruto, 500 Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Andrea Aparecida de Aro
- Programa de Pós-graduação em Ciências Biomédicas, Centro Universitário da Fundação Hermínio Ometto - FHO, Av. Dr. Maximiliano Baruto, 500 Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Maria Esméria Corezzola do Amaral
- Programa de Pós-graduação em Ciências Biomédicas, Centro Universitário da Fundação Hermínio Ometto - FHO, Av. Dr. Maximiliano Baruto, 500 Jd. Universitário, Araras, SP, 13607-339, Brazil
| | - Marcelo Augusto Marretto Esquisatto
- Programa de Pós-graduação em Ciências Biomédicas, Centro Universitário da Fundação Hermínio Ometto - FHO, Av. Dr. Maximiliano Baruto, 500 Jd. Universitário, Araras, SP, 13607-339, Brazil.
| |
Collapse
|
29
|
Zhou X, Jiang L, Zhang Y, Zhang J, Zhou D, Wu L, Huang Y, Xu N. Genetic variation of aggrecanase-2 (ADAMTS5) in susceptibility to osteoarthritis. ACTA ACUST UNITED AC 2019; 52:e8109. [PMID: 30652828 PMCID: PMC6328970 DOI: 10.1590/1414-431x20188109] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/07/2018] [Indexed: 01/06/2023]
Abstract
Aggrecanase-2 (ADAMTS5) gene is responsible for aggrecan degradation that may contribute to cartilage destruction in a mouse osteoarthritis (OA) model. We aimed to investigate the effects of ADAMTS5 gene polymorphisms on OA risk in a Chinese population. A total of 300 OA patients and 300 controls were recruited and their genotypes for ADAMTS5 gene rs226794 and rs2830585 polymorphisms were determined using a custom-by-design 48-Plex single nucleotide polymorphism Scan™ kit. ADAMTS5-associated genes were identified by co-expression analysis and their functions were investigated by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. Bioinformatics analysis showed that ADAMTS5 was significantly related to the components, structural constituent, and organization of the extracellular matrix. The rs2830585 polymorphism, but not rs226794 polymorphism, was significantly associated with an increased risk of knee OA. Stratified analysis further confirmed this significant association in patients at age ≥55 years. In conclusion, the ADAMTS5 rs2830585 polymorphism may be involved in the development of knee OA by destroying the extracellular matrix, but this finding should be further confirmed by larger studies.
Collapse
Affiliation(s)
- Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China.,Department of Orthopedic Trauma, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Lifeng Jiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Zhang
- Department of Orthopedic Trauma, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Junjie Zhang
- Department of Orthopedic Trauma, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Dong Zhou
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Lidong Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Huang
- Department of Orthopedic Trauma, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Nanwei Xu
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
30
|
van Geffen EW, van Caam APM, Schreurs W, van de Loo FA, van Lent PLEM, Koenders MI, Thudium CS, Bay-Jensen AC, Blaney Davidson EN, van der Kraan PM. IL-37 diminishes proteoglycan loss in human OA cartilage: donor-specific link between IL-37 and MMP-3. Osteoarthritis Cartilage 2019; 27:148-157. [PMID: 30201492 DOI: 10.1016/j.joca.2018.08.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 07/23/2018] [Accepted: 08/29/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE A hallmark of osteoarthritis (OA) is degradation of articular cartilage proteoglycans. In isolated human OA chondrocytes, the anti-inflammatory cytokine Interleukin-37 (IL-37) lowers the expression of the proteolytic MMP and ADAMTS enzymes, which mediate this degradation. Therefore, we investigated if IL-37 protects against proteoglycan loss in freshly obtained human OA explants. MATERIAL AND METHODS Human OA cartilage explants were incubated with IL-37. Release of sulphated proteoglycans (sGAGs) was measured with the dimethylmethylene-blue assay. Production and degradation of newly synthesized proteoglycans was measured using 35S-sulphate. Proteoglycan and proteolytic enzyme expression were analyzed by qPCR and Western Blot. Proteolytic activity was determined by measuring MMP- and ADAMTS-generated aggrecan neo-epitopes with ELISA and by using MMP-3-, MMP-13- or ADAMTS-5-inhibitors. RESULTS Over time, a linear release of sGAGs from OA cartilage was measured. IL-37 reduced this release by 87 μg/ml (24%) 95%CI [21.04-141.4]. IL-37 did not affect 35S-sulphate incorporation or proteoglycan gene expression. In contrast, IL-37 reduced loss of 35S-sulphate labeled GAGs and reduced MMP-3 protein expression, indicating that IL-37 inhibits proteoglycan degradation. Remarkably, we observed two groups of patients; one group in which MMP-3-inhibition lowered sGAG release, and one group in which ADAMTS5-inhibition had this effect. Remarkably, IL-37 was only functional in the group of patients that responded to MMP-3-inhibition. CONCLUSION We identified a relationship between IL-37 and reduced sGAG loss in OA cartilage. Most likely, this effect is mediated by inhibition of MMP-3 expression. These results suggest that IL-37 could be applied as therapy in a subgroup of OA patients, in which cartilage degradation is mediated by MMP-3.
Collapse
Affiliation(s)
- E W van Geffen
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A P M van Caam
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W Schreurs
- Department of Orthopaedics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - F A van de Loo
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P L E M van Lent
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M I Koenders
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - C S Thudium
- Department of Rheumatology, Nordic Bioscience, Copenhagen, Denmark
| | - A C Bay-Jensen
- Department of Rheumatology, Nordic Bioscience, Copenhagen, Denmark
| | - E N Blaney Davidson
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P M van der Kraan
- Department of Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
31
|
Association of ADAMTS4 and ADAMTS5 polymorphisms with musculoskeletal degenerative diseases: a systematic review and meta-analysis. Biosci Rep 2018; 38:BSR20181619. [PMID: 30369484 PMCID: PMC6265622 DOI: 10.1042/bsr20181619] [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: 09/13/2018] [Revised: 10/15/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
Objective: This meta-analysis and systematic review was performed with the aim of investigating the association between a disintegrin and metalloproteinase with thrombospondin motif (ADAMTS)4, AMDMTS5 polymorphisms and risk of musculoskeletal degenerative diseases. Methods: PubMed, EMBASE, ISI Web of Science, Wanfang and CNKI were searched from their inception until May 2018 to identify eligible studies. Data from individual studies were extracted using a standardized data collection sheet. The estimate of association between ADAMTS4, AMDMTS5 polymorphisms and risk of musculoskeletal degenerative diseases was expressed as odds ratio (OR) along with its related 95% confidence interval (95%CI) under an allelic model of inheritance. Meta-analysis was conducted using RevMan 5.3 software. Subgroup-analyses by ethnicity and type of diseases were performed. Results: Eight studies including ten cohorts were included in this meta-analysis. The meta-analyses results based on seven studies showed that rs226794 in ADAMTS5 gene was not associated with risk of musculoskeletal degenerative diseases (A vs. G: OR 1.07; 95%CI 0.97–1.19; P=0.16). Rs2830585 in ADAMTS5 was significantly associated with musculoskeletal degenerative diseases in only Asians (OR 1.41, 95%CI 1.18–1.68; P=0.0001), but not in Caucasians. Since only two of the collected studies referred to ADAMTS4, we did not perform meta-analysis for these comparisons. Conclusion: Taken together, rs226794 and rs2830585 in ADAMTS5 gene were not associated with musculoskeletal degenerative diseases in overall population, but there seemed to be an ethnicity-dependent effect of rs2830585 in the risk of musculoskeletal degenerative diseases. Insufficient evidence was found to support the association of other single nucleotide polymorphisms and musculoskeletal degenerative diseases.
Collapse
|
32
|
Yan B, Zhou L, Wang C, Wang R, Yan L, Yu L, Liu F, Du W, Yu G, Yuan Q, Tong P, Shan L, Efferth T. Intra-Articular Injection of Fructus Ligustri Lucidi Extract Attenuates Pain Behavior and Cartilage Degeneration in Mono-Iodoacetate Induced Osteoarthritic Rats. Front Pharmacol 2018; 9:1360. [PMID: 30532708 PMCID: PMC6265644 DOI: 10.3389/fphar.2018.01360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/05/2018] [Indexed: 12/21/2022] Open
Abstract
Fructus Ligustri Lucidi (FLL) has been widely used as a traditional Chinese medicine (TCM) for treating soreness and weakness of waist and knees. It has potential for treating OA owing to its kidney-tonifying activity with bone-strengthening effects, but there is so far no report of its anti-OA effect. This study established a rat OA model by intra-articular (IA) injection of mono-iodoacetate (1.5 mg) and weekly treated by IA administration of FLL at 100 μg/mL for 4 weeks. Thermal withdrawal latency, mechanical withdrawal threshold, and spontaneous activity were tested for evaluation of pain behavior, and histopathological (HE, SO, and ABH staining) and immunohistochemical (Col2, Col10, and MMP13) analyses were conducted for observation of cartilage degradation. In vitro effect of FLL on chondrocytes was evaluated by MTT assay and qPCR analysis. Moreover, HPLC analysis was performed to determine its chemoprofile. The pain behavioral data showed that FLL attenuated joint pain hypersensitivity by increasing thresholds of mechanical allodynia and thermal hyperalgesia as well as spontaneous activity. The histopathological result showed that FLL reversed OA cartilage degradation by protecting chondrocytes and extracellular matrix in cartilage, and the immunohistochemical analysis revealed its molecular actions on protein expressions of MMP13, Col2, and Col10 in cartilage. The MTT assay showed its proliferative effects on chondrocytes, and qPCR assay clarified its mechanism associated with gene expressions of Mmp13, Col2, Col10, Adamts5, Aggrecan, and Runx2 in TNF-α treated chondrocytes. Our results revealed an anti-OA effect of FLL on pain behavior and cartilage degradation in OA rats and clarified a molecular mechanism in association with the suppression of chondrocyte hypertrophy and catabolism. IA FLL can be regarded as novel and promising option for OA therapy.
Collapse
Affiliation(s)
- Bo Yan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Caiwei Wang
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rongrong Wang
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lingying Yu
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fucun Liu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wenxi Du
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guangping Yu
- Xianju Traditional Chinese Medicine, Taizhou, China
| | - Qiang Yuan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
33
|
Kennedy KC, Martinez SA, Martinez SE, Tucker RL, Davies NM. Effects of low-level laser therapy on bone healing and signs of pain in dogs following tibial plateau leveling osteotomy. Am J Vet Res 2018; 79:893-904. [PMID: 30058855 DOI: 10.2460/ajvr.79.8.893] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To assess the effect of low-level laser therapy (LLLT) on markers of synovial inflammation and signs of pain, function, bone healing, and osteoarthritis following tibial plateau leveling osteotomy (TPLO) in dogs with spontaneous cranial cruciate ligament rupture (CCLR). ANIMALS 12 client-owned dogs with unilateral CCLR. PROCEDURES All dogs were instrumented with an accelerometer for 2 weeks before and 8 weeks after TPLO. Dogs were randomly assigned to receive LLLT (radiant exposure, 1.5 to 2.25 J/cm2; n = 6) or a control (red light; 6) treatment immediately before and at predetermined times for 8 weeks after TPLO. Owners completed a Canine Brief Pain Inventory weekly for 8 weeks after surgery. Each dog underwent a recheck appointment, which included physical and orthopedic examinations, force plate analysis, radiography and synoviocentesis of the affected joint, and evaluation of lameness and signs of pain, at 2, 4, and 8 weeks after surgery. Select markers of inflammation were quantified in synovial fluid samples. Variables were compared between the 2 groups. RESULTS For the control group, mean ground reaction forces were greater at 2 and 4 weeks after TPLO and owner-assigned pain scores were lower during weeks 1 through 5 after TPLO, compared with corresponding values for the LLLT group. CONCLUSIONS AND CLINICAL RELEVANCE Results suggested that the LLLT protocol used had no beneficial effects on signs of pain or pelvic limb function following TPLO. Further research is necessary to evaluate the effects of LLLT and to determine the optimum LLLT protocol for dogs with CCLR.
Collapse
|
34
|
Cooke ME, Lawless BM, Jones SW, Grover LM. Matrix degradation in osteoarthritis primes the superficial region of cartilage for mechanical damage. Acta Biomater 2018; 78:320-328. [PMID: 30059801 DOI: 10.1016/j.actbio.2018.07.037] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/23/2018] [Accepted: 07/17/2018] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) is a degenerative disease that affects 25% of the world's population over fifty years of age. It is a chronic disease of the synovial joints, primarily the hip and knee. The main pathologies are degradation of the articular cartilage and changes to the subchondral bone, as a result of both mechanical wear and a locally elevated inflammatory state. This study compares the viscoelastic properties of cartilage that represents the biochemical changes in OA and age-matched healthy tissue. Further, the mechanical damage induced by this compressive loading cycle was characterised and the mechanism for it was investigated. The storage modulus of OA cartilage was shown to be significantly lower than that of healthy cartilage whilst having a higher capacity to hold water. Following mechanical testing, there was a significant increase in the surface roughness of OA cartilage. This change in surface structure occurred following a reduction in sulphated glycosaminoglycan content of the superficial region in OA, as seen by alcian blue staining and quantified by micro X-ray fluorescence. These findings are important in understanding how the chemical changes to cartilage matrix in OA influence its dynamic mechanical properties and structural integrity. STATEMENT OF SIGNIFICANCE Cartilage has a very specialised tissue structure which acts to resist compressive loading. In osteoarthritis (OA), there is both mechanically- and chemically-induced damage to cartilage, resulting in severe degradation of the tissue. In this study we have undertaken a detailed mechanical and chemical analysis of macroscopically undamaged OA and healthy cartilage tissue. We have demonstrated, for the first time in human tissue, that the mechanical degradation of the tissue is attributed to a chemical change across the structure. In macroscopically undamaged OA tissue, there is a reduction in the elastic response of cartilage tissue and an associated destabilisation of the matrix that leaves it susceptible to damage. Understanding this allows us to better understand the progression of OA to design better therapeutic interventions.
Collapse
|
35
|
Sauchinone inhibits IL-1β induced catabolism and hypertrophy in mouse chondrocytes to attenuate osteoarthritis via Nrf2/HO-1 and NF-κB pathways. Int Immunopharmacol 2018; 62:181-190. [DOI: 10.1016/j.intimp.2018.06.041] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/19/2018] [Accepted: 06/27/2018] [Indexed: 12/29/2022]
|
36
|
Amar S, Smith L, Fields GB. Matrix metalloproteinase collagenolysis in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2017; 1864:1940-1951. [PMID: 28456643 PMCID: PMC5605394 DOI: 10.1016/j.bbamcr.2017.04.015] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 01/08/2023]
Abstract
The proteolytic processing of collagen (collagenolysis) is critical in development and homeostasis, but also contributes to numerous pathologies. Mammalian interstitial collagenolytic enzymes include members of the matrix metalloproteinase (MMP) family and cathepsin K. While MMPs have long been recognized for their ability to catalyze the hydrolysis of collagen, the roles of individual MMPs in physiological and pathological collagenolysis are less defined. The use of knockout and mutant animal models, which reflect human diseases, has revealed distinct collagenolytic roles for MT1-MMP and MMP-13. A better understanding of temporal and spatial collagen processing, along with the knowledge of the specific MMP involved, will ultimately lead to more effective treatments for cancer, arthritis, cardiovascular conditions, and infectious diseases. This article is part of a Special Issue entitled: Matrix Metalloproteinases edited by Rafael Fridman.
Collapse
Affiliation(s)
- Sabrina Amar
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL 33458, USA.
| | - Lyndsay Smith
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL 33458, USA.
| | - Gregg B Fields
- Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL 33458, USA; Department of Chemistry, The Scripps Research Institute/Scripps Florida, Jupiter, FL 33458, USA.
| |
Collapse
|
37
|
Doyran B, Tong W, Li Q, Jia H, Zhang X, Chen C, Enomoto-Iwamoto M, Lu XL, Qin L, Han L. Nanoindentation modulus of murine cartilage: a sensitive indicator of the initiation and progression of post-traumatic osteoarthritis. Osteoarthritis Cartilage 2017; 25:108-117. [PMID: 27568574 PMCID: PMC5182132 DOI: 10.1016/j.joca.2016.08.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/30/2016] [Accepted: 08/17/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study aims to demonstrate that cartilage nanoindentation modulus is a highly sensitive indicator of the onset and spatiotemporal progression of post-traumatic osteoarthritis (PTOA) in murine models. DESIGN Destabilization of the medial meniscus (DMM) surgery was performed on the right knees of 12-week old male, wild-type C57BL/6 mice, with Sham control on contralateral left knees. Atomic force microscopy (AFM)-based nanoindentation was applied to quantify the nanoindentation modulus, Eind, of femoral condyle cartilage at 3 days to 12 weeks after surgery. The modulus changes were compared against the timeline of histological OA signs. Meanwhile, at 8 weeks after surgery, changes in meniscus, synovium and subchondral bone were evaluated to reveal the spatial progression of PTOA. RESULTS The modulus of medial condyle cartilage was significantly reduced at 1 week after DMM, preceding the histological OA signs, which only became detectable at 4-8 weeks after. This reduction is likely due to concomitantly elevated proteolytic activities, as blocking enzymatic activities in mice can attenuate this modulus reduction. In later OA, lateral condyle cartilage and medial meniscus also started to be weakened, illustrating the whole-organ nature of PTOA. CONCLUSIONS This study underscores the high sensitivity of nanoindentation in examining the initiation, attenuation and progression of PTOA in murine models. Meanwhile, modulus changes highlight concomitant changes in lateral cartilage and meniscus during the advancement of OA.
Collapse
Affiliation(s)
- B Doyran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - W Tong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Q Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - H Jia
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Orthopaedic Surgery, School of Medicine, ShiHeZi University, ShiHeZi, Xinjiang 832000, PR China
| | - X Zhang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - C Chen
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - M Enomoto-Iwamoto
- Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - X L Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - L Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - L Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
38
|
Silva SV, Lima MA, Cella N, Jaeger RG, Freitas VM. ADAMTS-1 Is Found in the Nuclei of Normal and Tumoral Breast Cells. PLoS One 2016; 11:e0165061. [PMID: 27764205 PMCID: PMC5072708 DOI: 10.1371/journal.pone.0165061] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 10/05/2016] [Indexed: 12/13/2022] Open
Abstract
Proteins secreted in the extracellular matrix microenvironment (ECM) by tumor cells are involved in cell adhesion, motility, intercellular communication and invasion. The tumor microenvironment is expansively modified and remodeled by proteases, resulting in important changes in both cell-cell and cell-ECM interactions and in the generation of new signals from the cell surface. Metalloproteinases belonging to the ADAMTS (a disintegrin and metalloprotease with thrombospondin motifs) family have been implicated in tissue remodeling events observed in cancer development, growth and progression. Here we investigated the subcellular localization of ADAMTS-1 in normal-like (MCF10-A) and tumoral (MCF7 and MDA-MB-231) human breast cells. ADAMTS-1 is a secreted protease found in the extracellular matrix. However, in this study we show for the first time that ADAMTS-1 is also present in the nuclei and nucleoli of the three mammary cell lines studied here. Our findings indicate that ADAMTS-1 has proteolytic functions in the nucleus through its interaction with aggrecan substrate.
Collapse
Affiliation(s)
- Suély V. Silva
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Maíra A. Lima
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Nathalie Cella
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Ruy G. Jaeger
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Vanessa M Freitas
- Department of Cell and Developmental Biology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
39
|
Sirri F, Maiorano G, Tavaniello S, Chen J, Petracci M, Meluzzi A. Effect of different levels of dietary zinc, manganese, and copper from organic or inorganic sources on performance, bacterial chondronecrosis, intramuscular collagen characteristics, and occurrence of meat quality defects of broiler chickens. Poult Sci 2016; 95:1813-24. [DOI: 10.3382/ps/pew064] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2016] [Indexed: 11/20/2022] Open
|
40
|
Azadi M, Nia HT, Gauci SJ, Ortiz C, Fosang AJ, Grodzinsky AJ. Wide bandwidth nanomechanical assessment of murine cartilage reveals protection of aggrecan knock-in mice from joint-overuse. J Biomech 2016; 49:1634-1640. [PMID: 27086115 DOI: 10.1016/j.jbiomech.2016.03.055] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 03/04/2016] [Accepted: 03/28/2016] [Indexed: 11/28/2022]
Abstract
Aggrecan loss in human and animal cartilage precedes clinical symptoms of osteoarthritis, suggesting that aggrecan loss is an initiating step in cartilage pathology. Characterizing early stages of cartilage degeneration caused by aging and overuse is important in the search for therapeutics. In this study, atomic force microscopy (AFM)-based force-displacement micromechanics, AFM-based wide bandwidth nanomechanics (nanodynamic), and histologic assessments were used to study changes in distal femur cartilage of wildtype mice and mice in which the aggrecan interglobular domain was mutated to make the cartilage aggrecanase-resistant. Half the animals were subjected to voluntary running-wheel exercise of varying durations. Wildtype mice at three selected age groups were compared. While histological assessment was not sensitive enough to capture any statistically significant changes in these relatively young populations of mice, micromechanical assessment captured changes in the quasi-equilibrium structural-elastic behavior of the cartilage matrix. Additionally, nanodynamic assessment captured changes in the fluid-solid poroelastic behavior and the high frequency stiffness of the tissue, which proved to be the most sensitive assessment of changes in cartilage associated with aging and joint-overuse. In wildtype mice, aging caused softening of the cartilage tissue at the microscale and at the nanoscale. Softening with increased animal age was found at high loading rates (frequencies), suggesting an increase in hydraulic permeability, with implications for loss of function pertinent to running and impact-injury. Running caused substantial changes in fluid-solid interactions in aggrecanase-resistant mice, suggestive of tissue degradation. However, higher nanodynamic stiffness magnitude and lower hydraulic permeability was observed in running aggrecanase-resistant mice compared to running wildtype controls at the same age, thereby suggesting protection from joint-overuse.
Collapse
Affiliation(s)
- Mojtaba Azadi
- School of Engineering, College of Science and Engineering, San Francisco State University, San Francisco, CA 94132, United States; Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States.
| | - Hadi Tavakoli Nia
- Massachusetts General Hospital, Harvard Medical School, Cambridge, MA 02139, United States
| | - Stephanie J Gauci
- University of Melbourne Department of Pediatrics & Murdoch Children׳s Research Institute, Parkville, Australia
| | - Christine Ortiz
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Amanda J Fosang
- University of Melbourne Department of Pediatrics & Murdoch Children׳s Research Institute, Parkville, Australia
| | - Alan J Grodzinsky
- Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| |
Collapse
|
41
|
Single-cell differences in matrix gene expression do not predict matrix deposition. Nat Commun 2016; 7:10865. [PMID: 26936319 PMCID: PMC4782061 DOI: 10.1038/ncomms10865] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/28/2016] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) display substantial cell-to-cell heterogeneity, complicating their use in regenerative medicine. However, conventional bulk assays mask this variability. Here we show that both chondrocytes and chondrogenically induced MSCs exhibit substantial mRNA expression heterogeneity. Single-molecule RNA FISH to measure mRNA expression of differentiation markers in single cells reveals that sister cell pairs have high levels of mRNA variability, suggesting that marker expression is not heritable. Surprisingly, this variability does not correlate with cell-to-cell differences in cartilage-like matrix production. Transcriptome-wide analysis suggests that no combination of markers can predict functional potential. De-differentiating chondrocytes also show a disconnect between mRNA expression of the cartilage marker aggrecan and cartilage-like matrix accumulation. Altogether, these quantitative analyses suggest that sorting subpopulations based on these markers would only marginally enrich the progenitor population for ‘superior' MSCs. Our results suggest that instantaneous mRNA abundance of canonical markers is tenuously linked to the chondrogenic phenotype at the single-cell level. Regenerative tissue engineering with mesenchymal stem cells is hampered by bulk methods of assessing differentiation status and a general assumption that expression of individual markers of stem cell differentiation correlate with functional capacity. Here the authors debunk this assumption by applying single-cell techniques to disassociate aggrecan mRNA abundance and matrix deposition.
Collapse
|
42
|
Hypoxia-inducible factor-2α induces expression of type X collagen and matrix metalloproteinases 13 in osteoarthritic meniscal cells. Inflamm Res 2016; 65:439-48. [PMID: 26892680 DOI: 10.1007/s00011-016-0926-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 02/03/2016] [Accepted: 02/09/2016] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES To evaluate whether Hypoxia-inducible factor-2α (HIF-2α) regulates expression of endochondral ossification-related molecules in human OA meniscus. METHODS Expressions of HIF-2α, type X collagen (COL10), matrix metalloproteinase (MMP)-13, and vascular endothelial growth factor (VEGF) in non-OA and OA menisci were analyzed by real-time RT-PCR and immunohistochemistry (IHC). Meniscal cells from OA patients were treated with interleukin-1β (IL-1β) and gene expression was analyzed. After knockdown of HIF-2α in OA meniscal cells, COL10 and MMP-13 expression were analyzed by RT-PCR, western blotting, immunofluorescence and ELISA. RESULT Histological analysis demonstrated weak staining of the superficial layer and large round cells in OA meniscus. RT-PCR analysis showed that HIF-2α, COL10, MMP-13, and VEGF mRNA expressions were higher in OA than non-OA meniscal cells. IHC showed a coordinated staining pattern of HIF-2α, COL10, and MMP-13 in OA meniscus. IL-1β treatment increased HIF-2α, COL10, and MMP-13 expressions in OA meniscal cells, and knockdown of HIF-2α suppressed IL-1β-mediated increase in COL10 and MMP-13 expression. CONCLUSIONS These results suggested that HIF-2α may cause meniscal matrix degradation by transactivation of MMP-13. HIF-2α may be a therapeutic target for modulating matrix degradation in both articular cartilage and meniscus during knee OA progression.
Collapse
|
43
|
Bascoul-Colombo C, Garaiova I, Plummer SF, Harwood JL, Caterson B, Hughes CE. Glucosamine Hydrochloride but Not Chondroitin Sulfate Prevents Cartilage Degradation and Inflammation Induced by Interleukin-1α in Bovine Cartilage Explants. Cartilage 2016; 7:70-81. [PMID: 26958319 PMCID: PMC4749751 DOI: 10.1177/1947603515603762] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE Glucosamine hydrochloride (GH) and chondroitin sulfate (CS) are commonly used for the treatment of osteoarthritis (OA). The aim of this study was to assess their effects, alone and in combination, on preventing aggrecan degradation and inflammation in an in vitro model of OA. DESIGN To test the effects of GH and/or CS as a preventative treatment, cartilage explants were pretreated with the compound(s) using concentrations that showed no detrimental effect on chondrocyte viability. Interleukin-1α (IL-1α) was added to induce cartilage degradation, supernatant and explants were analyzed for proteoglycan degradation products, aggrecanase mRNA expression and activity, and for the release of inflammatory markers. RESULTS Following treatment with IL-1α, 2 mg/mL dose of GH pretreatment was associated with a reduction of glycosaminoglycan release, reduced generation of the pathological interglobular domain aggrecan catabolites, decreased mRNA levels of ADAMTS-4 and -5 and reduced activity of ADAMTS-4. In contrast, CS alone did not have a significant effect on IL-1α-induced cartilage degradation and the addition of 0.4 mg/mL CS to 2 mg/mL GH did not further inhibit IL-1α-induced activity. Pretreatment with 2 mg/mL GH also reduced the release of inflammatory markers, prostaglandin E2 and nitric oxide induced by IL-1α while CS did not have a significant effect. CONCLUSIONS The results suggest that GH prevents cartilage degradation mediated by aggrecanases ADAMTS-4 and -5, and may also reduce inflammation. This could be part of the mechanisms by which GH is effective in maintaining joint integrity and function, and preventing or delaying early symptoms of OA.
Collapse
Affiliation(s)
- Cécile Bascoul-Colombo
- School of Biosciences, Cardiff University, Cardiff, UK,Research Department, Obsidian Research Ltd (now Cultech Ltd), Port Talbot, UK
| | - Iveta Garaiova
- Research Department, Obsidian Research Ltd (now Cultech Ltd), Port Talbot, UK
| | - Sue F. Plummer
- Research Department, Obsidian Research Ltd (now Cultech Ltd), Port Talbot, UK
| | | | | | - Clare E Hughes
- School of Biosciences, Cardiff University, Cardiff, UK,Clare E. Hughes, School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff, CF10 3AX, UK.
| |
Collapse
|
44
|
|
45
|
Abstract
The term "chondropenia" indicates the early stage of degenerative cartilage disease, and it has been identified by carefully monitoring early-stage osteoarthritis (OA). Not only is it the loss of articular cartilage volume, but it is also a rearrangement of biomechanical, ultrastructural, biochemical and molecular properties typical of healthy cartilage tissue. Diagnosing OA at an early stage or an advanced stage is valuable in terms of clinical and therapeutic outcome. In fact degenerative phenomena are supported by a complex biochemical cascade which unbalances the extracellular matrix homeostasis, closely regulated by chondrocytes. In the first stage an intense inflammatory reaction is triggered: pro-catabolic cytokines such as IL-1β and TNF-α triggering matrix metalloproteases and aggrecanase (ADAMT-4 and 5), responsible for the early loss of ultrastructural components, such as type II collagen and aggrecan. In addition nitric oxide and reactive oxygen species modulate the physiopathology of the condral matrix inducing apoptosis of chondrocytes through a mitochondria-dependent pathway. In addition, "Lonely Death": chondrocytes, are confined within a dense, avascular extracellular matrix capsule, and can trigger a genetically induced apoptosis and necrosis. The degenerative process starts from a central point and then spreads in a centrifugal manner in depth and in adjacent areas, eventually covering the whole joint; chondropenia represents a journey from the first clinically detectable time-point until it can be characterized as frank osteoarthritis. Currently, there are no instruments sensitive enough which allow a timely diagnosis of chondropenia. Innovative magnetic resonance imaging techniques, such as T2 mapping, can be effective and a sensitive diagnostic instrument for quantifying cartilage volume and proteoglycan content. However, avant-garde biophysical techniques, such as mechanical indenters, ultrasound and biochemical markers (uCTX-II), are rational and scientific tools applicable to the clinical and therapeutic management of early degenerative cartilage disease. The objective of this review on chondropenia is to present a state of the art and innovative concepts.
Collapse
|
46
|
Svala E, Löfgren M, Sihlbom C, Rüetschi U, Lindahl A, Ekman S, Skiöldebrand E. An inflammatory equine model demonstrates dynamic changes of immune response and cartilage matrix molecule degradation in vitro. Connect Tissue Res 2015; 56:315-25. [PMID: 25803623 DOI: 10.3109/03008207.2015.1027340] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The molecular aspects of inflammation were investigated in equine articular cartilage explants using quantitative proteomics. Articular cartilage explants were stimulated with interleukin (IL)-1β in vitro for 25 days, and proteins released into cell culture media were chemically labeled with isobaric mass tags and analyzed by liquid chromatography-tandem mass spectrometry. A total of 127 proteins were identified and quantified in media from explants. IL-1β-stimulation resulted in an abundance of proteins related to inflammation, including matrix metalloproteinases, acute phase proteins, complement components and IL-6. Extracellular matrix (ECM) molecules were released at different time points, and fragmentation of aggrecan and cartilage oligomeric matrix protein was observed at days 3 and 6, similar to early-stage OA in vivo. Degradation products of the collagenous network were observed at days 18 and 22, similar to late-stage OA. This model displays a longitudinal quantification of released molecules from the ECM of articular cartilage. Identification of dynamic changes of extracellular matrix molecules in the secretome of equine explants stimulated with IL-1β over time may be useful for identifying components released at different time points during the spontaneous OA process.
Collapse
Affiliation(s)
- Emilia Svala
- Section of Pathology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences , Uppsala , Sweden
| | | | | | | | | | | | | |
Collapse
|
47
|
Crascì L, Vicini P, Incerti M, Cardile V, Avondo S, Panico A. 2-Benzisothiazolylimino-5-benzylidene-4-thiazolidinones as protective agents against cartilage destruction. Bioorg Med Chem 2015; 23:1551-1556. [PMID: 25725607 DOI: 10.1016/j.bmc.2015.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 11/17/2022]
Abstract
We report the synthesis, the antioxidant and the inhibitory activity (IC50) on metalloproteinases (MMPs) 3 and 13 of 2-benzo[d]isothiazolylimino-5-benzylidene-4-thiazolidinones. Their potential as protective agents in osteoarthritis (OA) was evaluated by biological assays on chondrocytes cultures, stimulated by IL-1β. The chondroprotective capability, related both to antioxidant activity and to inhibition of MMPs, was studied in vitro, by determining nitric oxide production and glycosaminoglycans release. Moreover, selected derivatives 1h and 1g was studied for nuclear factor kappaB (NF-κB) inhibition by Western Blot analysis and for MMP-3 protein release using ELISA test. The structure-activity relationship of tested compounds demonstrates a favorable effect of the para substitution on the 5-benzilydene ring. Compound 1g shows a potent and selective activity on MMP-3 versus MMP-13. Accordingly, 1g possesses high antioxidant effect, NO lowering and GAGs restoring capability and also reduces the production of MMPs and NF-κB expression. Thus 1g can be considered as new potential chondroprotective agents.
Collapse
Affiliation(s)
- Lucia Crascì
- Department of Drug Sciences, University of Catania, V. le A. Doria 6, 95125 Catania, Italy.
| | - Paola Vicini
- Department of Pharmacy, University of Parma, Parco Area Delle Scienze 27/A, 43124 Parma, Italy
| | - Matteo Incerti
- Department of Pharmacy, University of Parma, Parco Area Delle Scienze 27/A, 43124 Parma, Italy
| | - Venera Cardile
- Department of Physiological Sciences, University of Catania, V. le A. Doria 6, 95125 Catania, Italy
| | - Sergio Avondo
- Department of Surgery, University of Catania, V. le A. Doria 6, 95125 Catania, Italy
| | - Annamaria Panico
- Department of Drug Sciences, University of Catania, V. le A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
48
|
Bolton SN, Whitehead MP, Dudhia J, Baldwin TC, Sutton R. Investigating the Postmortem Molecular Biology of Cartilage and its Potential Forensic Applications. J Forensic Sci 2015; 60:1061-7. [PMID: 25823955 DOI: 10.1111/1556-4029.12764] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 06/30/2014] [Accepted: 07/13/2014] [Indexed: 11/29/2022]
Abstract
This study investigated the postmortem molecular changes that articular cartilage undergoes following burial. Fresh pig trotters were interred in 30-cm-deep graves at two distinct locations exhibiting dissimilar soil environments for up to 42 days. Extracts of the metacarpophalangeal (MCP) and metatarsophalangeal (MTP) joint cartilage from trotters disinterred weekly over 6 weeks were analyzed by Western blot against the monoclonal antibody 2-B-6 to assess aggrecan degradation. In both soil conditions, aggrecan degradation by-products of decreasing molecular size and complexity were observed up to 21 days postmortem. Degradation products were undetected after this time and coincided with MCP/MTP joint exposure to the soil environment. These results show that cartilage proteoglycans undergo an ordered molecular breakdown, the analysis of which may have forensic applications. This model may prove useful for use as a human model and for forensic investigations concerning crimes against animals and the mortality of endangered species.
Collapse
Affiliation(s)
- Shawna N Bolton
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton, WV1 1LY, U.K
| | - Michael P Whitehead
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton, WV1 1LY, U.K
| | - Jayesh Dudhia
- Department of Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire, AL9 7TA, U.K
| | - Timothy C Baldwin
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton, WV1 1LY, U.K
| | - Raul Sutton
- Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton, WV1 1LY, U.K
| |
Collapse
|
49
|
Casagrande D, Stains JP, Murthi AM. Identification of shoulder osteoarthritis biomarkers: comparison between shoulders with and without osteoarthritis. J Shoulder Elbow Surg 2015; 24:382-90. [PMID: 25595362 PMCID: PMC4331258 DOI: 10.1016/j.jse.2014.11.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 11/12/2014] [Accepted: 11/15/2014] [Indexed: 02/01/2023]
Abstract
BACKGROUND The biologic factors associated with shoulder osteoarthritis (OA) have not been elucidated. The purpose of this study was to investigate osteoarthritic biomarkers of the shoulder. To our knowledge, this is the first study to analyze shoulder cartilage for OA-associated genes and to examine human shoulder cartilage for a possible biomarker, connexin 43 (Cx43). MATERIALS AND METHODS Cartilage from 16 osteoarthritic and 10 nonosteoarthritic humeral heads was assessed for expression of the following genes by real-time polymerase chain reaction: types I, II, and X collagen; matrix metalloproteinases (MMPs); tissue inhibitors of MMP (TIMPs); interleukins; versican; cyclooxygenase 2 (Cox-2); inducible nitric oxide synthase (iNOS); tumor necrosis factor α (TNF-α); aggrecanase 2 (ADAMTS5); and Cx43. RESULTS In osteoarthritic shoulders, Cx43, Cox-2, versican, collagen type I, ADAMTS5, MMP-3, and TNF-α expressions were significantly increased compared with controls. TIMP-3 and iNOS trended toward significance, with robust expression in osteoarthritic shoulders and low expression in nonosteoarthritic shoulders. In osteoarthritic shoulders, gene expression of Cx43, ADAMTS5, collagen type I, Cox-2, versican, and TIMP-3 showed predominance (85-, 33-, 13-, 12-, 11.5-, and 3-fold increases, respectively) relative to nonosteoarthritic controls. Spearman correlation analysis showed significant correlations between Cx43 and collagen (types I, II, and X), MMP-9, TIMP-2 and TIMP-3, versican, Cox-2, iNOS, and ADAMTS5. CONCLUSIONS Certain genes are markedly upregulated in osteoarthritic shoulders compared with nonosteoarthritic shoulders, with Cx43, Cox-2, versican, collagen type I, ADAMTS5, MMP-3, and TNF-α expression being significantly increased. These genes might be useful biomarkers for examining shoulder OA. CLINICAL RELEVANCE Identification of osteoarthritic biomarkers can help us better understand shoulder OA and build the foundation for future research on disease progression and treatments.
Collapse
|
50
|
Skaalure SC, Chu S, Bryant SJ. An enzyme-sensitive PEG hydrogel based on aggrecan catabolism for cartilage tissue engineering. Adv Healthc Mater 2015; 4:420-31. [PMID: 25296398 PMCID: PMC4516272 DOI: 10.1002/adhm.201400277] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/04/2014] [Indexed: 11/06/2022]
Abstract
A new cartilage-specific degradable hydrogel based on photoclickable thiol-ene poly(ethylene glycol) (PEG) hydrogels is presented. The hydrogel crosslinks are composed of the peptide, CRDTEGE-ARGSVIDRC, derived from the aggrecanase-cleavable site in aggrecan. This new hydrogel is evaluated for use in cartilage tissue engineering by encapsulating bovine chondrocytes from different cell sources (skeletally immature (juvenile) and mature (adult) donors and adult cells stimulated with proinflammatory lipopolysaccharide (LPS)) and culturing for 12 weeks. Regardless of cell source, a twofold decrease in compressive modulus is observed by 12 weeks, but without significant hydrogel swelling indicating limited bulk degradation. For juvenile cells, a connected matrix rich in aggrecan and collagen II, but minimal collagens I and X is observed. For adult cells, less matrix, but similar quality, is deposited. Aggrecanase activity is elevated, although without accelerating bulk hydrogel degradation. LPS further decreases matrix production, but does not affect aggrecanase activity. In contrast, matrix deposition in the nondegradable hydrogels consists of aggrecan and collagens I, II, and X, indicative of hypertrophic cartilage. Lastly, no inflammatory response in chondrocytes is observed by the aggrecanase-sensitive hydrogels. Overall, it is demonstrated that this new aggrecanase-sensitive hydrogel, which is degradable by chondrocytes and promotes a hyaline-like engineered cartilage, is promising for cartilage regeneration.
Collapse
Affiliation(s)
- Stacey C. Skaalure
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309
| | - Stanley Chu
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309
| | - Stephanie J. Bryant
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309
- Materials Science and Engineering Program, University of Colorado, Boulder, CO 80309
| |
Collapse
|