151
|
Chen K, Xu M, Lu F, He Y. Development of Matrix Metalloproteinases-Mediated Extracellular Matrix Remodeling in Regenerative Medicine: A Mini Review. Tissue Eng Regen Med 2023; 20:661-670. [PMID: 37160567 PMCID: PMC10352474 DOI: 10.1007/s13770-023-00536-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 05/11/2023] Open
Abstract
Extracellular matrix (ECM) components confer biomechanical properties, maintain cell phenotype and mediate tissue homeostasis. ECM remodeling is complex and plays a key role in both physiological and pathological processes. Matrix metalloproteinases (MMPs) are a group of enzymes responsible for ECM degradation and have been accepted as a key regulator in ECM remodeling. In this mini-review, we summarize MMPs categories, functions and the targeted substrates. We then discuss current understanding of the role of MMPs-mediated events, including inflammation reaction, angiogenesis, cellular activities, etc., in ECM remodeling in the context of regenerative medicine.
Collapse
Affiliation(s)
- Kaiqi Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Mimi Xu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Yunfan He
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
152
|
Stassen RHMJ, van den Akker GGH, Surtel DAM, Housmans BAC, Cremers A, Caron MMJ, Smagul A, Peffers MJ, van Rhijn LW, Welting TJM. Unravelling the Basic Calcium Phosphate crystal-dependent chondrocyte protein secretome; a role for TGF-β signaling. Osteoarthritis Cartilage 2023; 31:1035-1046. [PMID: 37075856 DOI: 10.1016/j.joca.2023.02.079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 04/21/2023]
Abstract
OBJECTIVE Basic Calcium Phosphate (BCP) crystals play an active role in the progression of osteoarthritis (OA). However, the cellular consequences remain largely unknown. Therefore, we characterized for the first time the changes in the protein secretome of human OA articular chondrocytes as a result of BCP stimulation using two unbiased proteomic analysis methods. METHOD Isolated human OA articular chondrocytes were stimulated with BCP crystals and examined by Quantitative Reverse Transcription PCR (RT-qPCR) and enzyme-linked immune sorbent assay (ELISA) after twenty-four and forty-eight hours. Forty-eight hours conditioned media were analyzed by label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) and an antibody array. The activity of BCP dependent Transforming Growth Factor Beta (TGF-β) signaling was analyzed by RT-qPCR and luciferase reporter assays. The molecular consequences regarding BCP-dependent TGF-β signaling on BCP-dependent Interleukin 6 (IL-6) were investigated using specific pathway inhibitors. RESULTS Synthesized BCP crystals induced IL-6 expression and secretion upon stimulation of human articular chondrocytes. Concomitant induction of catabolic gene expression was observed. Analysis of conditioned media revealed a complex and diverse response with a large number of proteins involved in TGF-β signaling, both in activation of latent TGF-β and TGF-β superfamily members, which were increased compared to non-stimulated OA chondrocytes. Activity of this BCP driven TGF-β signaling was confirmed by increased activity of expression of TGF-β target genes and luciferase reporters. Inhibition of BCP driven TGF-β signaling resulted in decreased IL-6 expression and secretion with a moderate effect on catabolic gene expression. CONCLUSION BCP crystal stimulation resulted in a complex and diverse chondrocyte protein secretome response. An important role for BCP-dependent TGF-β signaling was identified in development of a pro-inflammatory environment.
Collapse
Affiliation(s)
- R H M J Stassen
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - G G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - D A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - B A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - A Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - M M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands
| | - A Smagul
- Department of Musculoskeletal Biology, Life Course and Medical Sciences, University of Liverpool, UK
| | - M J Peffers
- Department of Musculoskeletal Biology, Life Course and Medical Sciences, University of Liverpool, UK
| | - L W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - T J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, The Netherlands; Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center +, Maastricht, The Netherlands.
| |
Collapse
|
153
|
Islam MT, Sarkar C, Hossain R, Bhuia MS, Mardare I, Kulbayeva M, Ydyrys A, Calina D, Habtemariam S, Kieliszek M, Sharifi-Rad J, Cho WC. Therapeutic strategies for rheumatic diseases and disorders: targeting redox imbalance and oxidative stress. Biomed Pharmacother 2023; 164:114900. [PMID: 37216707 DOI: 10.1016/j.biopha.2023.114900] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023] Open
Abstract
Rheumatic diseases and disorders (RDDs) are a group of chronic autoimmune diseases that are collectively called "multicausal diseases". They have resulted from predisposing genetic profiles and exposure to a range of environmental, occupational and lifestyle risk factors. Other causative factors include bacterial and viral attacks, sexual habits, trauma, etc. In addition, numerous studies reported that redox imbalance is one of the most serious consequences of RDDs. For example, rheumatoid arthritis (RA) as a classic example of chronic RDDs is linked to oxidative stress. This paper summarizes the contributions of redox imbalance to RDDs. The findings suggest that establishing direct or indirect therapeutic strategies for RDDs requires a more in-depth understanding of the redox dysregulation in these diseases. For example, the recent awareness of the roles of peroxiredoxins (Prdxs, e.g. Prdx2, Prdx3) in RDDs provided one potential route of therapeutic intervention of these pathologies. Changes in stressful lifestyles and dietary habits may also provide additional benefits in the management of RDDs. Future studies should be directed to explore molecular interactions in redox regulations associated with RDDS and potential therapeutic interventions.
Collapse
Affiliation(s)
- Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| | - Chandan Sarkar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Rajib Hossain
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon, South Korea
| | - Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Ileana Mardare
- Department of Public Health and Management, Carol Davila University of Medicine and Pharmacy of Bucharest, Bucharest, Romania
| | - Marzhan Kulbayeva
- Department of Biophysics, Biomedicine and Neuroscience, Al-Farabi Kazakh National University, Al-Farabi Av. 71, 050040 Almaty, Kazakhstan
| | - Alibek Ydyrys
- Biomedical Research Centre, Al-Farabi Kazakh National University, Al-Farabi ave. 71, 050040 Almaty, Kazakhstan
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| | - Marek Kieliszek
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159C, 02-776 Warsaw, Poland.
| | | | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
154
|
Kuppa SS, Kim HK, Kang JY, Lee SC, Yang HY, Sankaranarayanan J, Seon JK. Polynucleotides Suppress Inflammation and Stimulate Matrix Synthesis in an In Vitro Cell-Based Osteoarthritis Model. Int J Mol Sci 2023; 24:12282. [PMID: 37569659 PMCID: PMC10418450 DOI: 10.3390/ijms241512282] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/19/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Osteoarthritis (OA) is characterized by degeneration of the joint cartilage, inflammation, and a change in the chondrocyte phenotype. Inflammation also promotes cell hypertrophy in human articular chondrocytes (HC-a) by activating the NF-κB pathway. Chondrocyte hypertrophy and inflammation promote extracellular matrix degradation (ECM). Chondrocytes depend on Smad signaling to control and regulate cell hypertrophy as well as to maintain the ECM. The involvement of these two pathways is crucial for preserving the homeostasis of articular cartilage. In recent years, Polynucleotides Highly Purified Technology (PN-HPT) has emerged as a promising area of research for the treatment of OA. PN-HPT involves the use of polynucleotide-based agents with controlled natural origins and high purification levels. In this study, we focused on evaluating the efficacy of a specific polynucleotide sodium agent, known as CONJURAN, which is derived from fish sperm. Polynucleotides (PN), which are physiologically present in the matrix and function as water-soluble nucleic acids with a gel-like property, have been used to treat patients with OA. However, the specific mechanisms underlying the effect remain unclear. Therefore, we investigated the effect of PN in an OA cell model in which HC-a cells were stimulated with interleukin-1β (IL-1β) with or without PN treatment. The CCK-8 assay was used to assess the cytotoxic effects of PN. Furthermore, the enzyme-linked immunosorbent assay was utilized to detect MMP13 levels, and the nitric oxide assay was utilized to determine the effect of PN on inflammation. The anti-inflammatory effects of PN and related mechanisms were investigated using quantitative PCR, Western blot analysis, and immunofluorescence to examine and analyze relative markers. PN inhibited IL-1β induced destruction of genes and proteins by downregulating the expression of MMP3, MMP13, iNOS, and COX-2 while increasing the expression of aggrecan (ACAN) and collagen II (COL2A1). This study demonstrates, for the first time, that PN exerted anti-inflammatory effects by partially inhibiting the NF-κB pathway and increasing the Smad2/3 pathway. Based on our findings, PN can potentially serve as a treatment for OA.
Collapse
Affiliation(s)
- Sree Samanvitha Kuppa
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Republic of Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42, Jebong-ro, Dong-gu, Gwangju 501-757, Republic of Korea
| | - Hyung-Keun Kim
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Republic of Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42, Jebong-ro, Dong-gu, Gwangju 501-757, Republic of Korea
| | - Ju-Yeon Kang
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Republic of Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42, Jebong-ro, Dong-gu, Gwangju 501-757, Republic of Korea
| | - Seok-Cheol Lee
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Republic of Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42, Jebong-ro, Dong-gu, Gwangju 501-757, Republic of Korea
| | - Hong-Yeol Yang
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Republic of Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42, Jebong-ro, Dong-gu, Gwangju 501-757, Republic of Korea
| | - Jaishree Sankaranarayanan
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Republic of Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42, Jebong-ro, Dong-gu, Gwangju 501-757, Republic of Korea
| | - Jong-Keun Seon
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
- Department of Orthopaedics Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519-763, Republic of Korea
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42, Jebong-ro, Dong-gu, Gwangju 501-757, Republic of Korea
| |
Collapse
|
155
|
Upadhyay U, Kolla S, Chelluri LK. Extracellular matrix composition analysis of human articular cartilage for the development of organ-on-a-chip. Biochem Biophys Res Commun 2023; 667:81-88. [PMID: 37209566 DOI: 10.1016/j.bbrc.2023.04.117] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 04/18/2023] [Accepted: 04/29/2023] [Indexed: 05/22/2023]
Abstract
INTRODUCTION Articular cartilage has a complex extracellular matrix (ECM) that provides it a defined architecture for its load-bearing properties. The complete understanding of ECM components is imperative for developing biomimetic organ-on-a-chip tissue construct. OBJECTIVE This study aimed to decellularize and characterize the ECM for its protein profiling to generate a niche for enhanced chondrocyte proliferation. METHODS Articular cartilage scrapings were subjected to mechanical and collagenase digestion, followed by sodium dodecyl sulfate (SDS) treatment for 8 h and 16 h. The de-cellularization efficiency was confirmed by hematoxylin & eosin, alcian blue, masson's trichrome staining, and scanning electron microscopy (SEM). The ECM protein profile was quantified by liquid chromatography tandem mass spectrometry (LC-MS/MS) using a bottom-up approach. RESULTS Histological characterization revealed void lacunae that lacked staining for cellular components. The ECM, sulfated glycosaminoglycan content, and collagen fibers were preserved after 8 h and 16 h of de-cellularization. The SEM ultrastructure images showed that few chondrocytes adhered to the ECM after 8 h and cell-free ECM after 16 h of de-cellularization. LC-MS/MS analysis identified 66 proteins with heterotypic collagen types COL1A1-COL6A1, COL14A1, COL22A1 and COL25A1 showed moderate fold change and expression levels, while COL18A1, COL26A1, chondroitin sulfate, matrix metalloproteinase-9 (MMP9), fibronectin, platelet glycoprotein 1 beta alpha (GP1BA), vimentin, bone morphogenetic protein 6 (BMP6), fibroblast growth factor 4 (FGF4) and growth hormone receptor (GHR) showed maximum fold change and expression levels. CONCLUSIONS The standardized de-cellularization process could preserve majority of ECM components, providing structural integrity and architecture to the ECM. The Identified proteins quantified for their expression levels provided insight into engineering the ECM composition for developing cartilage-on-a-chip.
Collapse
Affiliation(s)
- Upasna Upadhyay
- Stem Cell Unit, Global Medical Education and Research Foundation, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India.
| | - Saketh Kolla
- Department of Orthopedics, Gleneagles Global Hospitals, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India.
| | - Lakshmi Kiran Chelluri
- Stem Cell Unit, Global Medical Education and Research Foundation, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India; Advanced Diagnostics & Therapeutics, Gleneagles Global Hospitals, Hyderabad, 500004, India.
| |
Collapse
|
156
|
Chen W, Liu W, Jiang T, Liu L, He Q, Lin T, Zhang J, Huo L, Xu X, Wang H, Liang D, Liu W. Tongbi Huoluo Decoction alleviates cartilage degeneration in knee osteoarthritis by inhibiting degradation of extracellular matrix. Chin Med 2023; 18:91. [PMID: 37507774 PMCID: PMC10385923 DOI: 10.1186/s13020-023-00802-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is an age-related degenerative disease characterized by abrasion of articular cartilage. Tongbi Huoluo Decoction (TBHLD) has been transformed from the famous traditional Chinese medicine Duhuo Jisheng Decoction, which can effectively alleviate pain symptoms in KOA. However, the active components and mechanisms of TBHLD in treating KOA have not yet been elucidated. The purpose of the study was to demonstrate the molecular mechanism of TBHLD in treating KOA. METHODS The components and targets of TBHLD and KOA were collected from multiple databases, and the protein to protein interaction (PPI) network was constructed. Next, we performed topological calculation and enrichment analysis. Besides, we performed virtual screening for molecular docking and molecular dynamics simulation (MDS). Furthermore, the vitro and vivo experiments were performed to evaluate the validity and mechanism of TBHLD. RESULTS 206 active components and 187 potential targets were screened from Tongbi Huoluo Decoction. A total of 50 intersecting genes were identified between TBHLD and KOA, 20 core targets were calculated by network topology analysis. The core targets were enriched in the ECM interaction pathways. The results of virtual screening for molecular docking and MDS showed that the active components of TBHLD had steady binding conformations with core genes. Moreover, we identified 32 differential serum components in TBHLD-containing serum using LC-MS, including 22 upregulated and 10 downregulated serum components. TBHLD improved the proliferation activity of OA chondrocytes, decreased the expression of Col1a1, Col1a2, Mmp2, Mmp13 in OA chondrocytes, ameliorated the cartilage lesions and restored the cartilage abrasion. CONCLUSION TBHLD inhibited degradation of cartilage ECM by regulating the expression of type I collagens and Mmps to ameliorate cartilage degeneration in KOA.
Collapse
Affiliation(s)
- Weijian Chen
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510095, China
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China
| | - Weinian Liu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510045, Guangdong, China
| | - Tao Jiang
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China
| | - Lingyun Liu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Qi He
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Tianye Lin
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Jiayuan Zhang
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510095, China
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China
| | - Liwei Huo
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510045, Guangdong, China
| | - Xuemeng Xu
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510095, China
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China
| | - Haibin Wang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
- Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| | - Du Liang
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510045, Guangdong, China.
| | - Wengang Liu
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510095, China.
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China.
| |
Collapse
|
157
|
Zhao F, Bai Y, Xiang X, Pang X. The role of fibromodulin in inflammatory responses and diseases associated with inflammation. Front Immunol 2023; 14:1191787. [PMID: 37483637 PMCID: PMC10360182 DOI: 10.3389/fimmu.2023.1191787] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Inflammation is an immune response that the host organism eliminates threats from foreign objects or endogenous signals. It plays a key role in the progression, prognosis as well as therapy of diseases. Chronic inflammatory diseases have been regarded as the main cause of death worldwide at present, which greatly affect a vast number of individuals, producing economic and social burdens. Thus, developing drugs targeting inflammation has become necessary and attractive in the world. Currently, accumulating evidence suggests that small leucine-rich proteoglycans (SLRPs) exhibit essential roles in various inflammatory responses by acting as an anti-inflammatory or pro-inflammatory role in different scenarios of diseases. Of particular interest was a well-studied member, termed fibromodulin (FMOD), which has been largely explored in the role of inflammatory responses in inflammatory-related diseases. In this review, particular focus is given to the role of FMOD in inflammatory response including the relationship of FMOD with the complement system and immune cells, as well as the role of FMOD in the diseases associated with inflammation, such as skin wounding healing, osteoarthritis (OA), tendinopathy, atherosclerosis, and heart failure (HF). By conducting this review, we intend to gain insight into the role of FMOD in inflammation, which may open the way for the development of new anti-inflammation drugs in the scenarios of different inflammatory-related diseases.
Collapse
Affiliation(s)
- Feng Zhao
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Bai
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xuerong Xiang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoxiao Pang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
158
|
Guillán-Fresco M, Franco-Trepat E, Alonso-Pérez A, Jorge-Mora A, López-López V, Pazos-Pérez A, Piñeiro-Ramil M, Gómez R. Formononetin, a Beer Polyphenol with Catabolic Effects on Chondrocytes. Nutrients 2023; 15:2959. [PMID: 37447284 DOI: 10.3390/nu15132959] [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: 06/02/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Beer consumption has been identified as a risk factor for osteoarthritis (OA), a rheumatic disease characterised by cartilage degradation, joint inflammation, and eventual joint failure. One of the main isoflavonoids in beer is formononetin (FNT), an estrogenic compound also found in multiple plants and herbs. In this study, we aimed to investigate the effect of FNT on chondrocyte viability, inflammation, and metabolism. Cells were treated with FNT with or without IL-1β for 48 h and during 7 days of differentiation. Cell viability was determined via MTT assay. Nitrite accumulation was determined by Griess reaction. The expression of genes involved in inflammation and metabolism was determined by RT-PCR. The results revealed that a low concentration of FNT had no deleterious effect on cell viability and decreased the expression of inflammation-related genes. However, our results suggest that FNT overexposure negatively impacts on chondrocytes by promoting catabolic responses. Finally, these effects were not mediated by estrogen receptors (ERs) or aryl hydrocarbon receptor (AhR). In conclusion, factors that favour FNT accumulation, such as long exposure times or metabolic disorders, can promote chondrocyte catabolism. These data may partially explain why beer consumption increases the risk of OA.
Collapse
Affiliation(s)
- María Guillán-Fresco
- Musculoskeletal Pathology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Clinical Hospital SERGAS, 15706 Santiago de Compostela, Spain
| | - Eloi Franco-Trepat
- Musculoskeletal Pathology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Clinical Hospital SERGAS, 15706 Santiago de Compostela, Spain
| | - Ana Alonso-Pérez
- Musculoskeletal Pathology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Clinical Hospital SERGAS, 15706 Santiago de Compostela, Spain
| | - Alberto Jorge-Mora
- Musculoskeletal Pathology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Clinical Hospital SERGAS, 15706 Santiago de Compostela, Spain
| | - Verónica López-López
- Musculoskeletal Pathology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Clinical Hospital SERGAS, 15706 Santiago de Compostela, Spain
| | - Andrés Pazos-Pérez
- Musculoskeletal Pathology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Clinical Hospital SERGAS, 15706 Santiago de Compostela, Spain
| | - María Piñeiro-Ramil
- Musculoskeletal Pathology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Clinical Hospital SERGAS, 15706 Santiago de Compostela, Spain
| | - Rodolfo Gómez
- Musculoskeletal Pathology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago University Clinical Hospital SERGAS, 15706 Santiago de Compostela, Spain
| |
Collapse
|
159
|
Shigley C, Trivedi J, Meghani O, Owens BD, Jayasuriya CT. Suppressing Chondrocyte Hypertrophy to Build Better Cartilage. Bioengineering (Basel) 2023; 10:741. [PMID: 37370672 DOI: 10.3390/bioengineering10060741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Current clinical strategies for restoring cartilage defects do not adequately consider taking the necessary steps to prevent the formation of hypertrophic tissue at injury sites. Chondrocyte hypertrophy inevitably causes both macroscopic and microscopic level changes in cartilage, resulting in adverse long-term outcomes following attempted restoration. Repairing/restoring articular cartilage while minimizing the risk of hypertrophic neo tissue formation represents an unmet clinical challenge. Previous investigations have extensively identified and characterized the biological mechanisms that regulate cartilage hypertrophy with preclinical studies now beginning to leverage this knowledge to help build better cartilage. In this comprehensive article, we will provide a summary of these biological mechanisms and systematically review the most cutting-edge strategies for circumventing this pathological hallmark of osteoarthritis.
Collapse
Affiliation(s)
- Christian Shigley
- The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Jay Trivedi
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Ozair Meghani
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Brett D Owens
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Sports Surgery, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
160
|
Fan M, Tong P, Yan L, Li T, Ren J, Huang J, Du W, Zhou L, Shan L. Detrimental alteration of mesenchymal stem cells by an articular inflammatory microenvironment results in deterioration of osteoarthritis. BMC Med 2023; 21:215. [PMID: 37337188 PMCID: PMC10280917 DOI: 10.1186/s12916-023-02923-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Articular injection of mesenchymal stem cells (MSCs) has been applied to treat knee osteoarthritis (kOA), but its clinical outcomes are controversial. This study investigated whether an articular inflammatory microenvironment (AIM) impacts MSC-based therapy in a rat model of kOA. METHODS The biological change of MSCs and the functional change of MSCs on chondrocytes were evaluated under AIM. The key mediator and mechanism for the AIM impact on MSC therapy were explored via gain- and loss-of-function approaches. RESULTS The results showed that MSCs exerted potent anti-kOA effects in vivo and in vitro, but that this therapy become chondrodestructive if a chronic AIM was present. Mechanistically, the overexpression of MMP13 in the injected MSCs via a MAPKs-AP1 signaling axis was revealed as the underlying mechanism for the detriment outcome. CONCLUSIONS This study thus clarifies recent clinical findings while also suggesting a means to overcome any detrimental effects of MSC-based therapy while improving its efficacy.
Collapse
Affiliation(s)
- Mengqiang Fan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yan
- Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China
| | - Ting Li
- Department of Plastic & Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jiadan Ren
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiefeng Huang
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenxi Du
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China.
- Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China.
| |
Collapse
|
161
|
Wei Q, Zhu X, Wang L, Zhang W, Yang X, Wei W. Extracellular matrix in synovium development, homeostasis and arthritis disease. Int Immunopharmacol 2023; 121:110453. [PMID: 37331300 DOI: 10.1016/j.intimp.2023.110453] [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: 03/14/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 06/20/2023]
Abstract
Extracellular matrix (ECM) is a three-dimensional network entity composed of extracellular macromolecules. ECM in synovium not only supports the structural integrity of synovium, but also plays a crucial role in regulating homeostasis and damage repair response in synovium. Obvious disorders in the composition, behavior and function of synovial ECM will lead to the occurrence and development of arthritis diseases such as rheumatoid arthritis (RA), osteoarthritis (OA) and psoriatic arthritis (PsA). Based on the importance of synovial ECM, targeted regulation of the composition and structure of ECM is considered to be an effective measure for the treatment of arthritis disease. This paper reviews the current research status of synovial ECM biology, discusses the role and mechanism of synovial ECM in physiological status and arthritis disease, and summarizes the current strategies for targeting synovial ECM to provide information for the pathogenesis, diagnosis and treatment of arthritis disease.
Collapse
Affiliation(s)
- Qi Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Xuemin Zhu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Luping Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wankang Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Xuezhi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
162
|
Shih PC, Lee YH, Tsou HK, Cheng-Chung Wei J. Recent targets of osteoarthritis research. Best Pract Res Clin Rheumatol 2023; 37:101851. [PMID: 37422344 DOI: 10.1016/j.berh.2023.101851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/14/2023] [Indexed: 07/10/2023]
Abstract
Osteoarthritis is one of the most common diseases and poses a significant medical burden worldwide. Currently, the diagnosis and treatment of osteoarthritis primarily rely on clinical symptoms and changes observed in radiographs or other image modalities. However, identification based on reliable biomarkers would greatly improve early diagnosis, help with precise monitoring of disease progression, and provide aid for accurate treatment. In recent years, several biomarkers for osteoarthritis have been identified, including image modalities and biochemical biomarkers such as collagen degradation products, pro- or anti-inflammatory cytokines, micro RNAs, long non-coding RNAs, and circular RNAs. These biomarkers offer new insights in the pathogenesis of osteoarthritis and provide potential targets for further research. This article reviews the evolution of osteoarthritis biomarkers from the perspective of pathogenesis and emphasizes the importance of continued research to improve the diagnosis, treatment, and management of osteoarthritis.
Collapse
Affiliation(s)
- Po-Cheng Shih
- Department of Allergy, Immunology & Rheumatology, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yung-Heng Lee
- Department of Orthopedics, Cishan Hospital, Ministry of Health and Welfare, Kaohsiung, Taiwan; Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Senior Services Industry Management, Minghsin University of Science and Technology, Hsinchu, Taiwan; Department of Recreation and Sport Management, Shu-Te University, Kaohsiung, Taiwan
| | - Hsi-Kai Tsou
- Functional Neurosurgery Division, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Houlong, Miaoli County, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Division of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
163
|
Jiang Q, Zhang S. Stimulus-Responsive Drug Delivery Nanoplatforms for Osteoarthritis Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206929. [PMID: 36905239 DOI: 10.1002/smll.202206929] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/16/2023] [Indexed: 06/08/2023]
Abstract
Osteoarthritis (OA) is one of the most prevalent age-related degenerative diseases. With an increasingly aging global population, greater numbers of OA patients are providing clear economic and societal burdens. Surgical and pharmacological treatments are the most common and conventional therapeutic strategies for OA, but often fall considerably short of desired or optimal outcomes. With the development of stimulus-responsive nanoplatforms has come the potential for improved therapeutic strategies for OA. Enhanced control, longer retention time, higher loading rates, and increased sensitivity are among the potential benefits. This review summarizes the advanced application of stimulus-responsive drug delivery nanoplatforms for OA, categorized by either those that depend on endogenous stimulus (reactive oxygen species, pH, enzyme, and temperature), or those that depend on exogenous stimulus (near-infrared ray, ultrasound, magnetic fields). The opportunities, restrictions, and limitations related to these various drug delivery systems, or their combinations, are discussed in areas such as multi-functionality, image guidance, and multi-stimulus response. The remaining constraints and potential solutions that are represented by the clinical application of stimulus-responsive drug delivery nanoplatforms are finally summarized.
Collapse
Affiliation(s)
- Qi Jiang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| |
Collapse
|
164
|
Poudel SB, Ruff RR, Yildirim G, Dixit M, Michot B, Gibbs JL, Ortiz SD, Kopchick JJ, Kirsch T, Yakar S. Excess Growth Hormone Triggers Inflammation-Associated Arthropathy, Subchondral Bone Loss, and Arthralgia. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:829-842. [PMID: 36870529 PMCID: PMC10284029 DOI: 10.1016/j.ajpath.2023.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 03/06/2023]
Abstract
Growth hormone (GH) is a key mediator of skeletal growth. In humans, excess GH secretion due to pituitary adenoma, seen in patients with acromegaly, results in severe arthropathies. This study investigated the effects of long-term excess GH on the knee joint tissues. One year-old wild-type (WT) and bovine GH (bGH) transgenic mice were used as a model for excess GH. bGH mice showed increased sensitivity to mechanical and thermal stimuli, compared with WT mice. Micro-computed tomography analyses of the distal femur subchondral bone revealed significant reductions in trabecular thickness and significantly reduced bone mineral density of the tibial subchondral bone-plate associated with increased osteoclast activity in both male and female bGH compared with WT mice. bGH mice showed severe loss of matrix from the articular cartilage, osteophytosis, synovitis, and ectopic chondrogenesis. Articular cartilage loss in the bGH mice was associated with elevated markers of inflammation and chondrocyte hypertrophy. Finally, hyperplasia of synovial cells was associated with increased expression of Ki-67 and diminished p53 levels in the synovium of bGH mice. Unlike the low-grade inflammation seen in primary osteoarthritis, arthropathy caused by excess GH affects all joint tissues and triggers severe inflammatory response. Data from this study suggest that treatment of acromegalic arthropathy should involve inhibition of ectopic chondrogenesis and chondrocyte hypertrophy.
Collapse
Affiliation(s)
- Sher B Poudel
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Ryan R Ruff
- Department of Epidemiology and Health Promotion, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Gozde Yildirim
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Manisha Dixit
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Benoit Michot
- Department of Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Jennifer L Gibbs
- Department of Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Silvana D Ortiz
- Department of Biomedical Sciences, Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - John J Kopchick
- Department of Biomedical Sciences, Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - Thorsten Kirsch
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, New York; Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, New York
| | - Shoshana Yakar
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York.
| |
Collapse
|
165
|
Scuruchi M, Aliquò F, Avenoso A, Mandraffino G, Vermiglio G, Minuti A, Campo S, Campo GM, D’Ascola A. Endocan Knockdown Down-Regulates the Expression of Angiogenesis-Associated Genes in Il-1ß Activated Chondrocytes. Biomolecules 2023; 13:851. [PMID: 37238720 PMCID: PMC10216645 DOI: 10.3390/biom13050851] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Endocan is a small soluble proteoglycan (PG) known to be involved in inflammation and angiogenesis. Increased endocan expression was found in the synovia of arthritic patients and chondrocytes stimulated with IL-1ß. Considering these findings, we aimed to investigate the effects of endocan knockdown on the modulation of pro-angiogenic molecules expression in a model of IL-1ß-induced inflammation in human articular chondrocytes. Endocan, VEGF-A, MMP-9, MMP-13, and VEGFR-2 expression was measured in both normal and endocan knockdown chondrocytes stimulated with IL-1ß. VEGFR-2 and NF-kB activation were also measured. Results have shown that endocan, VEGF-A, VEGFR-2, MMP-9, and MMP-13 were significantly up-regulated during IL-1ß-induced inflammation; interestingly, the expression of such pro-angiogenic molecules and NF-kB activation were significantly reduced by endocan knockdown. These data support the hypothesis that endocan released by activated chondrocytes may be involved in the mechanisms that stimulate cell migration and invasion, as well as angiogenesis, in the pannus of arthritic joints.
Collapse
Affiliation(s)
- Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Federica Aliquò
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98122 Messina, Italy
| | - Angela Avenoso
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98122 Messina, Italy
| | - Giuseppe Mandraffino
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Giovanna Vermiglio
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98122 Messina, Italy
| | - Aurelio Minuti
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98122 Messina, Italy
| | - Salvatore Campo
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98122 Messina, Italy
| | - Giuseppe Maurizio Campo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Angela D’Ascola
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| |
Collapse
|
166
|
Grillet B, Pereira RVS, Van Damme J, Abu El-Asrar A, Proost P, Opdenakker G. Matrix metalloproteinases in arthritis: towards precision medicine. Nat Rev Rheumatol 2023; 19:363-377. [PMID: 37161083 DOI: 10.1038/s41584-023-00966-w] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2023] [Indexed: 05/11/2023]
Abstract
Proteolysis of structural molecules of the extracellular matrix (ECM) is an irreversible post-translational modification in all arthropathies. Common joint disorders, including osteoarthritis and rheumatoid arthritis, have been associated with increased levels of matrix remodelling enzymes, including matrix metalloproteinases (MMPs). MMPs, in concert with other host proteinases and glycanases, destroy proteoglycans, collagens and other ECM molecules. MMPs may also control joint remodelling indirectly by signalling through cell-surface receptors or by proteolysis of cytokines and receptor molecules. After synthesis as pro-forms, MMPs can be activated by various types of post-translational modifications, including proteolysis. Once activated, MMPs are controlled by general and specific tissue inhibitors of metalloproteinases (TIMPs). In rheumatoid arthritis, proteolysis of the ECM results in so-called remnant epitopes that enhance and perpetuate autoimmune processes in susceptible hosts. In osteoarthritis, the considerable production of MMP-13 by chondrocytes, often concurrent with mechanical overload, is a key event. Hence, information about the regulation, timing, localization and activities of MMPs in specific disease phases and arthritic entities will help to develop better diagnostics. Insights into beneficial and detrimental effects of MMPs on joint tissue inflammation are also necessary to plan and execute (pre)clinical studies for better therapy and precision medicine with MMP inhibitors. With the advances in proteomics and single-cell transcriptomics, two critical points need attention: neglected neutrophil MMP biology, and the analysis of net proteolytic activities as the result of balances between MMPs and their inhibitors.
Collapse
Affiliation(s)
- Bernard Grillet
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Rafaela Vaz Sousa Pereira
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Ahmed Abu El-Asrar
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Ophthalmology, King Saud University, Riyadh, Saudi Arabia
| | - Paul Proost
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.
- Department of Ophthalmology, King Saud University, Riyadh, Saudi Arabia.
- University Hospitals Gasthuisberg, UZ Leuven, KU Leuven, Leuven, Belgium.
| |
Collapse
|
167
|
Shanmugasundaram S, Solanki K, Saseendar S, Chavada VK, D’Ambrosi R. Role of Doxycycline as an Osteoarthritis Disease-Modifying Drug. J Clin Med 2023; 12:2927. [PMID: 37109263 PMCID: PMC10145123 DOI: 10.3390/jcm12082927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Doxycycline is a drug that has been proposed to modify osteoarthritis (OA) progression, in addition to its role as an antibiotic. However, available evidence thus far comprises sporadic reports, with no consensus on its benefits. Hence, this review attempts to analyze the evidence available thus far on the role of doxycycline as a disease-modifying osteoarthritis drug (DMOAD) in knee osteoarthritis. The earliest evidence of doxycycline in OA appeared in 1991 when doxycycline was found to inhibit the type XI collagenolytic activity of extracts from the human osteoarthritic cartilage, and gelatinase and tetracycline were found to inhibit this metalloproteinase activity in articular cartilage in vivo, which could modify cartilage breakdown in osteoarthritis. Apart from the inhibition of cartilage damage by metalloproteinases (MMPs) and other cartilage-related mechanisms, doxycycline also affects the bone and interferes with many enzyme systems. The most significant finding after reviewing various studies was that doxycycline has a definitive role in structural changes in osteoarthritis progression and radiological joint space width, but its role in the improvement of clinical outcomes as a DMOAD has not been established. However, there is much of a gap and lack of evidence in this regard. Doxycycline, as an MMP inhibitor, has theoretical advantages for clinical outcomes, but the present studies reveal only beneficial structural changes in osteoarthritis and very minimal or nonexistent advantages in clinical outcomes. Current evidence does not favor the regular use of doxycycline for the treatment of osteoarthritis as an individual treatment option or in combination with others. However, multicenter large cohort studies are warranted to determine the long-term benefits of doxycycline.
Collapse
Affiliation(s)
| | | | | | - Vijay K. Chavada
- Department of Community Medicine, Indira Gandhi Medical College and Research Institute, Puducherry 605009, India
| | - Riccardo D’Ambrosi
- IRCCS Ospedale Galeazzi—Sant’Ambrogio, 20161 Milan, Italy
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
168
|
Liu D, Li X, Zhang L, Hu B, Hu S, Zhang X, Hu J. Small molecule inhibitors of osteoarthritis: Current development and future perspective. Front Physiol 2023; 14:1156913. [PMID: 37089415 PMCID: PMC10119395 DOI: 10.3389/fphys.2023.1156913] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
Osteoarthritis (OA) is one of the common degenerative joint diseases in clinic. It mainly damages articular cartilage, causing pain, swelling and stiffness around joints, and is the main cause of disability of the elderly. Due to the unclear pathogenesis of osteoarthritis and the poor self-healing ability of articular cartilage, the treatment options for this disease are limited. At present, NSAIDs, Glucocorticoid and Duloxetine are the most commonly used treatment choice for osteoarthritis. Although it is somewhat effective, the adverse reactions are frequent and serious. The development of safer and more effective anti-osteoarthritis drugs is essential and urgent. This review summarizes recent advances in the pharmacological treatment of OA, focusing on small molecule inhibitors targeting cartilage remodeling in osteoarthritis as well as the research idea of reducing adverse effects by optimizing the dosage form of traditional drugs for the treatment of osteoarthritis. It should provide a reference for exploration of new potential treatment options.
Collapse
Affiliation(s)
- Dan Liu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Xingxing Li
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Lin Zhang
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Bin Hu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Sang Hu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiao Zhang
- Institute of Pathology, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing, China
| | - Jing Hu
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
169
|
Zhang W, Kuss M, Yan Y, Shi W. Dynamic Alginate Hydrogel as an Antioxidative Bioink for Bioprinting. Gels 2023; 9:gels9040312. [PMID: 37102924 PMCID: PMC10137987 DOI: 10.3390/gels9040312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/28/2023] Open
Abstract
3D bioprinting holds great potential for use in tissue engineering to treat degenerative joint disorders, such as osteoarthritis. However, there is a lack of multifunctional bioinks that can not only support cell growth and differentiation, but also offer protection to cells against injuries caused by the elevated oxidative stress; this conditions is a common characteristic of the microenvironment of the osteoarthritis disease. To mitigate oxidative stress-induced cellular phenotype change and malfunction, an anti-oxidative bioink derived from an alginate dynamic hydrogel was developed in this study. The alginate dynamic hydrogel gelated quickly via the dynamic covalent bond between the phenylboronic acid modified alginate (Alg-PBA) and poly (vinyl alcohol) (PVA). It presented good self-healing and shear-thinning abilities because of the dynamic feature. The dynamic hydrogel supported long-term growth of mouse fibroblasts after stabilization with a secondary ionic crosslinking between introduced calcium ions and the carboxylate group in the alginate backbone. In addition, the dynamic hydrogel showed good printability, resulting in the fabrication of scaffolds with cylindrical and grid structures with good structural fidelity. Encapsulated mouse chondrocytes maintained high viability for at least 7 days in the bioprinted hydrogel after ionic crosslinking. Most importantly, in vitro studies implied that the bioprinted scaffold could reduce the intracellular oxidative stress for embedded chondrocytes under H2O2 exposure; it could also protect the chondrocytes from H2O2-induced downregulation of extracellular matrix (ECM) relevant anabolic genes (ACAN and COL2) and upregulation of a catabolic gene (MMP13). In summary, the results suggest that the dynamic alginate hydrogel can be applied as a versatile bioink for the fabrication of 3D bioprinted scaffolds with an innate antioxidative ability; this technique is expected to improve the regenerative efficacy of cartilage tissues for the treatment of joint disorders.
Collapse
Affiliation(s)
- Wenhai Zhang
- Orthopedic Department, Tianjin Hospital, Tianjin 300211, China
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Yan
- Healthcare Security Office, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
170
|
Kamar SS, ShamsEldeen AM, Hosny SA, El-Shafei AA, Rashid LA, Hassanein RT, Hassan RM. Comparing Effectiveness of Hyaluronic Acid-Chitosan Nanoparticles Encapsulation Versus Hyaluronic Acid Monotherapy in Osteoarthritis Rat Model: Microarray Screening for miR-140. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:686-697. [PMID: 37749722 DOI: 10.1093/micmic/ozac048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/02/2022] [Accepted: 12/04/2022] [Indexed: 09/27/2023]
Abstract
Osteoarthritis is a debilitating, progressive joint disease linked to lower quality of life and higher health care costs. This study compared hyaluronic acid-chitosan nanoparticle encapsulation to hyaluronic-acid monotherapy in a rat model of knee osteoarthritis. Four groups of 40 adult male albino rats were designed. Group (Gp) I: control; Gp II (osteoarthritis model): intra-articular injection of monoiodoacetate; Gp III (hyaluronic acid-treated): intra-articular injections of hyaluronic-acid on days 14 and 21 after monoiodoacetate injection; and Gp IV (hyaluronic acid-chitosan nanoparticle-treated): intra-articular injections of hyaluronic acid-chitosan nanoparticle on days 14 and 21 after monoiodoacetate injection. After 28 days, knee joints were examined using H&E, Safranin O, and immunohistochemistry for nuclear factor kappa beta (NF-κB), inducible nitric oxide synthase (iNOS), and matrix metalloproteinase (MMP)-13. Quantification for gene expression of collagen-II, aggrecan, and micro-RNA-140; ELISA for interleukin (IL)-1β and IL-8; and western blotting for IKBα and NF-κB was estimated. Osteoarthritis-knee joints showed a severe cartilage damage and synovial inflammation with increased NF-κB, iNOS, and MMP-13 immunostaining, decreased miR-140, collagen II, and aggrecan levels, and increased inflammatory markers' gene expressions. The hyaluronic acid-chitosan nanoparticle significantly improved knee joint structure and reduced inflammatory cytokines compared to hyaluronic acid monotherapy. Intra-articular injection of hyaluronic acid-chitosan nanoparticle encapsulation revealed a significant improvement in the knee joint structure compared to hyaluronic-acid in a rat model of osteoarthritis.
Collapse
Affiliation(s)
- Samaa Samir Kamar
- Histology Department, Kasr Al-ainy Faculty of Medicine, Cairo University, Cairo 11559, Egypt
| | | | - Sara Adel Hosny
- Histology Department, Kasr Al-ainy Faculty of Medicine, Cairo University, Cairo 11559, Egypt
| | - Asmaa Ahmed El-Shafei
- Histology Department, Kasr Al-ainy Faculty of Medicine, Cairo University, Cairo 11559, Egypt
| | - Laila Ahmad Rashid
- Biochemistry Department, Kasr Al-ainy Faculty of Medicine, Cairo University, Cairo 11559, Egypt
| | - Radwa Taha Hassanein
- Biochemistry Department, Kasr Al-ainy Faculty of Medicine, Cairo University, Cairo 11559, Egypt
| | - Rokia Mohamad Hassan
- Histology Department, Kasr Al-ainy Faculty of Medicine, Cairo University, Cairo 11559, Egypt
| |
Collapse
|
171
|
Liu S, Zhang C, Zhou Y, Zhang F, Duan X, Liu Y, Zhao X, Liu J, Shuai X, Wang J, Cao Z. MRI-visible mesoporous polydopamine nanoparticles with enhanced antioxidant capacity for osteoarthritis therapy. Biomaterials 2023; 295:122030. [PMID: 36758340 DOI: 10.1016/j.biomaterials.2023.122030] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 01/21/2023] [Accepted: 01/26/2023] [Indexed: 02/01/2023]
Abstract
Since the progression of osteoarthritis (OA) is closely associated with synovitis and cartilage destruction, the inhibition of inflammatory responses in synovial macrophages and reactive oxygen species (ROS) induced apoptosis in chondrocytes is crucial for OA amelioration. However, most of the current anti-inflammatory and antioxidant drugs are small molecules apt to be eliminated in vivo. Herein, mesoporous polydopamine nanoparticles (DAMM NPs) doped with arginine and manganese (Mn) ions were prepared to load dexamethasone (DEX) for OA intervention. A series of in vitro studies showed that the sustained release of DEX from DAMM NPs suppressed synovial inflammation and simultaneously inhibited toll-like receptor 3 (TLR-3) production in chondrocytes, contributing to prevention of chondrocyte apoptosis through the inflammatory factor-dependent TLR-3/NF-κB signaling pathway via modulation of macrophage-chondrocyte crosstalk. In addition, DAMM NPs exerted a predominant role in removal of ROS generated in chondrocytes. Therefore, the DEX-loaded DAMM NPs significantly attenuated OA development in mice model. Importantly, the T1-T2 magnetic contrast capabilities of DAMM NPs allowed an MRI-trackable delivery, manifesting a distinct feature widely regarded to boost the potential of nanomedicines for clinical applications. Together, our developed antioxidant-enhanced DAMM NPs with MRI-visible signals may serve as a novel multifunctional nanocarriers for prevention of OA progression.
Collapse
Affiliation(s)
- Sitong Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Chen Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yuanyuan Zhou
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Fang Zhang
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Xiaohui Duan
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Yang Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Xibang Zhao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jiali Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| | - Zhong Cao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| |
Collapse
|
172
|
Gu Z, Wang J, Fu Y, Pan H, He H, Gan Q, Liu C. Smart Biomaterials for Articular Cartilage Repair and Regeneration. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202212561] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Indexed: 01/06/2025]
Abstract
AbstractArticular cartilage defects bring about disability and worldwide socioeconomic loss, therefore, articular cartilage repair and regeneration is recognized as a global issue. However, due to its avascular and nearly acellular characteristic, cartilage tissue regeneration ability is limited to some extent. Despite the availability of various treatment methods, including palliative drugs and surgical regenerative therapy, articular cartilage repair and regeneration still face major challenges due to the lack of appropriate methods and materials. Smart biomaterials can regulate cell behavior and provide excellent tissue repair and regeneration microenvironment, thus inducing articular cartilage repair and regeneration. This process is adjusted by controlling drug/bioactive factors release via responding to exogenous/endogenous stimuli, tailoring materials’ structure and function similar to native cartilage or providing physiochemical and physical signaling factors. Herein, smart biomaterials, recently applied in articular cartilage repair and regeneration, are elaborated from two aspects: smart drug release system and smart scaffolds. Furthermore, articular cartilage and its defects and advanced manufacturing techniques of smart biomaterials are discussed in brief. Finally, perspectives for smart biomaterials used in articular cartilage repair and regeneration are presented and the clinical translation of smart biomaterials is emphasized.
Collapse
Affiliation(s)
- Zhanghao Gu
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Jiayi Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yu Fu
- School of Aerospace Engineering and Applied Mechanics Tongji University Zhangwu Road 100 Shanghai 200092 P. R. China
| | - Hao Pan
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Hongyan He
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Qi Gan
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of the Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry East China University of Science and Technology Shanghai 200237 P. R. China
| |
Collapse
|
173
|
Zeng J, Sun P, Zhao Y, Fang X, Wu Z, Qi X. Bone Mesenchymal Stem Cell-derived Exosomes Involved Co-delivery and Synergism Effect with Icariin via Mussel-inspired Multifunctional Hydrogel for Cartilage Protection. Asian J Pharm Sci 2023. [DOI: 10.1016/j.ajps.2023.100799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
|
174
|
Gerami MH, Khorram R, Rasoolzadegan S, Mardpour S, Nakhaei P, Hashemi S, Al-Naqeeb BZT, Aminian A, Samimi S. Emerging role of mesenchymal stem/stromal cells (MSCs) and MSCs-derived exosomes in bone- and joint-associated musculoskeletal disorders: a new frontier. Eur J Med Res 2023; 28:86. [PMID: 36803566 PMCID: PMC9939872 DOI: 10.1186/s40001-023-01034-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 01/26/2023] [Indexed: 02/22/2023] Open
Abstract
Exosomes are membranous vesicles with a 30 to 150 nm diameter secreted by mesenchymal stem/stromal cells (MSCs) and other cells, such as immune cells and cancer cells. Exosomes convey proteins, bioactive lipids, and genetic components to recipient cells, such as microRNAs (miRNAs). Consequently, they have been implicated in regulating intercellular communication mediators under physiological and pathological circumstances. Exosomes therapy as a cell-free approach bypasses many concerns regarding the therapeutic application of stem/stromal cells, including undesirable proliferation, heterogeneity, and immunogenic effects. Indeed, exosomes have become a promising strategy to treat human diseases, particularly bone- and joint-associated musculoskeletal disorders, because of their characteristics, such as potentiated stability in circulation, biocompatibility, low immunogenicity, and toxicity. In this light, a diversity of studies have indicated that inhibiting inflammation, inducing angiogenesis, provoking osteoblast and chondrocyte proliferation and migration, and negative regulation of matrix-degrading enzymes result in bone and cartilage recovery upon administration of MSCs-derived exosomes. Notwithstanding, insufficient quantity of isolated exosomes, lack of reliable potency test, and exosomes heterogeneity hurdle their application in clinics. Herein, we will deliver an outline respecting the advantages of MSCs-derived exosomes-based therapy in common bone- and joint-associated musculoskeletal disorders. Moreover, we will have a glimpse the underlying mechanism behind the MSCs-elicited therapeutic merits in these conditions.
Collapse
Affiliation(s)
- Mohammad Hadi Gerami
- grid.412571.40000 0000 8819 4698Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roya Khorram
- grid.412571.40000 0000 8819 4698Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheil Rasoolzadegan
- grid.411600.2Department of Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Mardpour
- grid.411705.60000 0001 0166 0922Department of Radiology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Pooria Nakhaei
- grid.411705.60000 0001 0166 0922Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheyla Hashemi
- grid.411036.10000 0001 1498 685XObstetrician, Gynaecology & Infertility Department, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Amir Aminian
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Sahar Samimi
- Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
175
|
Zhang RD, Chen C, Wang P, Fang Y, Jiang LQ, Fang X, Zhao Y, Ni J, Wang DG, Pan HF. Air pollution exposure and auto-inflammatory and autoimmune diseases of the musculoskeletal system: a review of epidemiologic and mechanistic evidence. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2023:10.1007/s10653-023-01495-x. [PMID: 36735155 DOI: 10.1007/s10653-023-01495-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Auto-inflammatory and autoimmune diseases of the musculoskeletal system can be perceived as a spectrum of rheumatic diseases, with the joints and connective tissues are eroded severely that progressively develop chronic inflammation and lesion. A wide range of risk factors represented by genetic and environmental factors have been uncovered by population-based surveys and experimental studies. Lately, the exposure to air pollution has been found to be potentially involved in the mechanisms of occurrence or development of such diseases, principally manifest in oxidative stress, local and systemic inflammation, and epigenetic modifications, as well as the mitochondrial dysfunction, which has been reported to participate in the intermediate links. The lungs might serve as a starting area of air pollutants, which would cause oxidative stress-induced bronchial-associated lymphoid tissue (iBALT) to further to influence T, B cells, and the secretion of pro-inflammatory cytokines. The binding of aromatic hydrocarbon receptor (AhR) to the corresponding contaminant ligands tends to regulate the reaction of Th17 and Tregs. Furthermore, air pollution components might spur on immune and inflammatory responses by damaging mitochondria that could interact with and exacerbate oxidative stress and pro-inflammatory cytokines. In this review, we focused on the association between air pollution and typical auto-inflammatory and autoimmune diseases of the musculoskeletal system, mainly including osteoarthritis (OA), rheumatoid arthritis (RA), spondyloarthritis (SpA) and juvenile idiopathic arthritis (JIA), and aim to collate the mechanisms involved and the potential channels. A complete summary and in-depth understanding of the autoimmune and inflammatory effects of air pollution exposure should hopefully contribute new perspectives on how to formulate better public health policies to alleviate the adverse health effects of air pollutants.
Collapse
Affiliation(s)
- Ruo-Di Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Cong Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Peng Wang
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
- Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Yang Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Xi Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Yan Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - De-Guang Wang
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
- Department of Nephrology, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China.
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
176
|
Paramakrishnan N, Chavan L, Lim KG, Paramaswaran Y, Muthuraman A. Reversal of Neuralgia Effect of Beta Carotene in Streptozotocin-Associated Diabetic Neuropathic Pain in Female Zebrafish via Matrix Metalloprotease-13 Inhibition. Pharmaceuticals (Basel) 2023; 16:157. [PMID: 37259308 PMCID: PMC9959792 DOI: 10.3390/ph16020157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 08/26/2023] Open
Abstract
Beta carotene is a natural anti-oxidant agent, and it inhibits the matrix metalloprotease (MMP) activity. Diabetic neuropathic pain (DNP) is produced by cellular oxidative stress. The role of the beta carotene effect in diabetic neuropathic pain is not explored yet. The present study is designed for the evaluation of the palm oil mill effluent-derived beta carotene (PBC) effect in DNP in zebrafish. The DNP was induced by the intraperitoneal administration of streptozotocin (STZ). Blood glucose levels of above 15 mM were considered to be diabetic conditions. The zebrafish were exposed to test compound PBC (25, 50, and 100 µM), pregabalin (PG: 10 μM), and an MMP-13 inhibitor (CL-82198; 10 μM) for 10 consecutive days from day 11. The neuralgic behavioral parameters, i.e., temperature test, acetic acid test, and fin clip test were assessed on day 0 and the 7th, 14th, and 21st days. On the 22nd day, the blood glucose and MMP-13 levels and brain thiobarbituric acid reactive substances (TBARS), reduced glutathione (GSH), and MMP-13 activity levels were estimated. The treatment of PBC ameliorated the DNP-associated behavioral and biochemical changes. The results are similar to those of PG and CL-82198 treatments. Hence, the PBC possesses a potentially ameliorative effect against DNP due to its potential anti-oxidant, anti-lipid peroxidation, and MMP-13 inhibitory actions.
Collapse
Affiliation(s)
| | - Laxmikant Chavan
- Faculty of Medicine, AIMST University, Semeling, Bedong 08100, Kedah, Malaysia
| | - Khian Giap Lim
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, Bedong 08100, Kedah, Malaysia
| | - Yamunna Paramaswaran
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, Bedong 08100, Kedah, Malaysia
| | - Arunachalam Muthuraman
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, Bedong 08100, Kedah, Malaysia
| |
Collapse
|
177
|
The Role of Membrane-Type 1 Matrix Metalloproteinase-Substrate Interactions in Pathogenesis. Int J Mol Sci 2023; 24:ijms24032183. [PMID: 36768503 PMCID: PMC9917210 DOI: 10.3390/ijms24032183] [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: 12/31/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023] Open
Abstract
A protease is an enzyme with a proteolytic activity that facilitates the digestion of its substrates. Membrane-type I matrix metalloproteinase (MT1-MMP), a member of the broader matrix metalloproteinases (MMP) family, is involved in the regulation of diverse cellular activities. MT1-MMP is a very well-known enzyme as an activator of pro-MMP-2 and two collagenases, MMP-8 and MMP-13, all of which are essential for cell migration. As an anchored membrane enzyme, MT1-MMP has the ability to interact with a diverse group of molecules, including proteins that are not part of the extracellular matrix (ECM). Therefore, MT1-MMP can regulate various cellular activities not only by changing the extra-cellular environment but also by regulating cell signaling. The presence of both intracellular and extra-cellular portions of MT1-MMP can allow it to interact with proteins on both sides of the cell membrane. Here, we reviewed the MT1-MMP substrates involved in disease pathogenesis.
Collapse
|
178
|
Mulberroside A alleviates osteoarthritis via restoring impaired autophagy and suppressing MAPK/NF-κB/PI3K-AKT-mTOR signaling pathways. iScience 2023; 26:105936. [PMID: 36698724 PMCID: PMC9868682 DOI: 10.1016/j.isci.2023.105936] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/11/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023] Open
Abstract
Osteoarthritis (OA) is a trauma-/age-related degenerative disease characterized by chronic inflammation as one of its pathogenic mechanisms. Mulberroside A (MA), a natural bioactive withanolide, demonstrates anti-inflammatory properties in various diseases; however, little is known about the effect of MA on OA. We aim to examine the role of MA on OA and to identify the potential mechanisms through which it protects articular cartilage. In vitro, MA improved inflammatory response, anabolism, and catabolism in IL-1β-induced OA chondrocytes. The chondroprotective effects of MA were attributed to suppressing the MAPK, NF-κB, and PI3K-AKT-mTOR signaling pathways, as well as promoting the autophagy process. In vivo, intra-articular injection of MA reduced the cartilage destruction and reversed the change of anabolic and catabolic-related proteins in destabilized medial meniscus (DMM)-induced OA models. Thus, the study indicates that MA exhibits a chondroprotective effect and might be a promising agent for OA treatment.
Collapse
|
179
|
罗 媚, 杜 信, 周 学. [Developments in Research on the Relationship Between Matrix Metalloproteinases and Osteoarthritis]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:77-82. [PMID: 36647647 PMCID: PMC10409029 DOI: 10.12182/20230160110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Indexed: 01/18/2023]
Abstract
Matrix metalloproteinases (MMPs) acquired their names because they depend on metal ions such as Ca 2+ and Zn 2+ as their cofactors. Members of this family of proteins share a similar structure consisting of five functionally distinct structural domains. MMPs, including MMP-1, MMP-3, MMP-9, and MMP-13, are key substances that promote cartilage matrix degradation and play an important role in the occurrence and progression of osteoarthritis (OA). MMPs boost the development of OA through the degradation of extracellular matrix proteins of chondrocytes, the promotion of inflammation, and other mechanisms, and are hence attracting extensive and increasing attention from the medical community. OA is a common degenerative disease that occurs in the joints and is associated with aging, metabolism, infections, genetics, exercise, and other predisposing factors. The pathological changes it causes can lead to a series of clinical symptoms such as joint pain, morning stiffness, and restricted joint movement, severely affecting patients' quality of life. The pathogenic mechanism of this highly prevalent disease is still unclear. At present, there is no effective treatment available for disease improvement. In the future, selective inhibition of MMPs, the key enzymes, may become an effective therapeutic approach. Focusing on the pathogenic effects of MMPs in OA, we herein reviewed the latest findings on the role of MMPs in the occurrence and progression of OA.
Collapse
Affiliation(s)
- 媚 罗
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓病科 (成都 610041)State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 信眉 杜
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓病科 (成都 610041)State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 学东 周
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 牙体牙髓病科 (成都 610041)State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
180
|
Ashruf OS, Ansari MY. Natural Compounds: Potential Therapeutics for the Inhibition of Cartilage Matrix Degradation in Osteoarthritis. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010102. [PMID: 36676051 PMCID: PMC9866583 DOI: 10.3390/life13010102] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/24/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease characterized by enzymatic degradation of the cartilage extracellular matrix (ECM) causing joint pain and disability. There is no disease-modifying drug available for the treatment of OA. An ideal drug is expected to stop cartilage ECM degradation and restore the degenerated ECM. The ECM primarily contains type II collagen and aggrecan but also has minor quantities of other collagen fibers and proteoglycans. In OA joints, the components of the cartilage ECM are degraded by matrix-degrading proteases and hydrolases which are produced by chondrocytes and synoviocytes. Matrix metalloproteinase-13 (MMP-13) and a disintegrin and metalloproteinase with thrombospondin motifs 4 and 5 (ADAMTS5) are the major collagenase and aggrecanase, respectively, which are highly expressed in OA cartilage and promote cartilage ECM degradation. Current studies using various in vitro and in vivo approaches show that natural compounds inhibit the expression and activity of MMP-13, ADAMTS4, and ADAMTS5 and increase the expression of ECM components. In this review, we have summarized recent advancements in OA research with a focus on natural compounds as potential therapeutics for the treatment of OA with emphasis on the prevention of cartilage ECM degradation and improvement of joint health.
Collapse
Affiliation(s)
- Omer S. Ashruf
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, 4209, State Route 44, Rootstown, OH 44272, USA
- College of Medicine, Northeast Ohio Medical University, 4209, State Route 44, Rootstown, OH 44272, USA
| | - Mohammad Yunus Ansari
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, 4209, State Route 44, Rootstown, OH 44272, USA
- Musculoskeletal Research Focus Area, Northeast Ohio Medical University, 4209, State Route 44, Rootstown, OH 44272, USA
- Correspondence:
| |
Collapse
|
181
|
Wu D, Luo Y, Li T, Zhao X, Lv T, Fang G, Ou P, Li H, Luo X, Huang A, Pang Y. Systemic complications of rheumatoid arthritis: Focus on pathogenesis and treatment. Front Immunol 2022; 13:1051082. [PMID: 36618407 PMCID: PMC9817137 DOI: 10.3389/fimmu.2022.1051082] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
As a systemic autoimmune disease, rheumatoid arthritis (RA) usually causes damage not only to joints, but also to other tissues and organs including the heart, kidneys, lungs, digestive system, eyes, skin, and nervous system. Excessive complications are closely related to the prognosis of RA patients and even lead to increased mortality. This article summarizes the serious complications of RA, focusing on its incidence, pathogenesis, clinical features, and treatment methods, aiming to provide a reference for clinicians to better manage the complications of RA.
Collapse
Affiliation(s)
- Di Wu
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yehao Luo
- School of Second Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Tong Li
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xinyi Zhao
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Ting Lv
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Gang Fang
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Peiqi Ou
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Hongyi Li
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xiaofan Luo
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - An Huang
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China,*Correspondence: An Huang, ; Yuzhou Pang,
| | - Yuzhou Pang
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, Guangxi, China,*Correspondence: An Huang, ; Yuzhou Pang,
| |
Collapse
|
182
|
Abshirini M, Coad J, Wolber FM, von Hurst P, Miller MR, Tian HS, Kruger MC. Effects of Greenshell™ mussel intervention on biomarkers of cartilage metabolism, inflammatory markers and joint symptoms in overweight/obese postmenopausal women: A randomized, double-blind, and placebo-controlled trial. Front Med (Lausanne) 2022; 9:1063336. [PMID: 36544504 PMCID: PMC9760926 DOI: 10.3389/fmed.2022.1063336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Objective To investigate the effect of whole greenshell mussel (GSM) powder on biomarkers of cartilage metabolism, inflammatory cytokines, and joint symptoms in postmenopausal women with overweight/obesity and joint discomfort. Design Fifty-five postmenopausal women with overweight/obesity were randomly assigned to receive 3 g/day whole GSM powder or placebo for 12 weeks. Cartilage turnover biomarkers urinary C-telopeptide of type II collagen (CTX-II) and serum cartilage oligomeric matrix protein (COMP) were measured at baseline, week 6 and 12. Plasma cytokines were measured at baseline and week 12. Joint pain and knee-related problems were assessed at baseline and week 12 using a 100 mm Visual Analogue Scale (VAS) and the Knee injury and Osteoarthritis Outcome Score (KOOS) questionnaire, respectively. Results Forty-nine participants completed the study (GSM n = 25, placebo n = 24). After 12 weeks, urinary CTX-II showed no significant change over time or between the groups (interaction effect P = 0.1). However, in women with symptomatic knees, a significant difference was noted between the group (treatment effect P = 0.04), as it was lower in the GSM group compared to placebo group at week 6 (P = 0.04) and week 12 (P = 0.03). Serum COMP and plasma cytokines were not affected. GSM supplementation showed greater reduction in the VAS pain score than placebo (-13.2 ± 20.3 vs. -2.9 ± 15.9; P = 0.04). No significant change in KOOS domains between the two groups was observed. Conclusion Oral supplementation of whole GSM powder at 3 g/day may slow down the degradation of type II collagen in postmenopausal women with symptomatic knees. GSM treatment conferred clinical benefit on overall joint pain. No significant effect was noted for inflammatory cytokines, suggesting that GSM may act within the joint microenvironment rather than at the systemic level. Clinical trial registration [www.australianclinicaltrials.gov.au/clinical-trialregistries], identifier [ACTRN12620000413921p].
Collapse
Affiliation(s)
- Maryam Abshirini
- School of Health Sciences, College of Health, Massey University, Palmerston North, New Zealand
| | - Jane Coad
- School of Food and Advanced Technology, College of Sciences, Massey University, Palmerston North, New Zealand
| | - Frances M. Wolber
- School of Food and Advanced Technology, College of Sciences, Massey University, Palmerston North, New Zealand,Centre for Metabolic Health Research, Massey University, Palmerston North, New Zealand
| | - Pamela von Hurst
- School of Sport, Exercise and Nutrition, College of Health, Massey University, Auckland, New Zealand
| | | | | | - Marlena C. Kruger
- School of Health Sciences, College of Health, Massey University, Palmerston North, New Zealand,*Correspondence: Marlena C. Kruger,
| |
Collapse
|
183
|
Manning JC, Baldoneschi V, Romero-Hernández LL, Pichler KM, GarcÍa Caballero G, André S, Kutzner TJ, Ludwig AK, Zullo V, Richichi B, Windhager R, Kaltner H, Toegel S, Gabius HJ, Murphy PV, Nativi C. Targeting osteoarthritis-associated galectins and an induced effector class by a ditopic bifunctional reagent: Impact of its glycan part on binding measured in the tissue context. Bioorg Med Chem 2022; 75:117068. [PMID: 36327696 DOI: 10.1016/j.bmc.2022.117068] [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: 02/24/2022] [Revised: 09/07/2022] [Accepted: 10/12/2022] [Indexed: 11/19/2022]
Abstract
Pairing glycans with tissue lectins controls multiple effector pathways in (patho)physiology. A clinically relevant example is the prodegradative activity of galectins-1 and -3 (Gal-1 and -3) in the progression of osteoarthritis (OA) via matrix metalloproteinases (MMPs), especially MMP-13. The design of heterobifunctional inhibitors that can block galectin binding and MMPs both directly and by preventing their galectin-dependent induction selectively offers a perspective to dissect the roles of lectins and proteolytic enzymes. We describe the synthesis of such a reagent with a bivalent galectin ligand connected to an MMP inhibitor and of two tetravalent glycoclusters with a subtle change in headgroup presentation for further elucidation of influence on ligand binding. Testing was performed on clinical material with mixtures of galectins as occurring in vivo, using sections of fixed tissue. Two-colour fluorescence microscopy monitored binding to the cellular glycome after optimization of experimental parameters. In the presence of the inhibitor, galectin binding to OA specimens was significantly reduced. These results open the perspective to examine the inhibitory capacity of custom-made ditopic compounds on binding of lectins in mixtures using sections of clinical material with known impact of galectins and MMPs on disease progression.
Collapse
Affiliation(s)
- Joachim C Manning
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Lena-Christ-Str. 48, 82152 Planegg, Germany
| | - Veronica Baldoneschi
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia, 3-13, Sesto Fiorentino, Florence 50019, Italy
| | - Laura L Romero-Hernández
- School of Biological and Chemical Sciences, University of Galway, University Road, Galway H91 TK33, Ireland
| | - Katharina M Pichler
- Karl Chiari Lab for Orthopedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Gabriel GarcÍa Caballero
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Lena-Christ-Str. 48, 82152 Planegg, Germany
| | - Sabine André
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Lena-Christ-Str. 48, 82152 Planegg, Germany
| | - Tanja J Kutzner
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Lena-Christ-Str. 48, 82152 Planegg, Germany
| | - Anna-Kristin Ludwig
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Lena-Christ-Str. 48, 82152 Planegg, Germany
| | - Valerio Zullo
- Dipartimento di Chimica e Chimica Industriale, University of Pisa, Via Moruzzi 13, Pisa 56124, Italy
| | - Barbara Richichi
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia, 3-13, Sesto Fiorentino, Florence 50019, Italy
| | - Reinhard Windhager
- Karl Chiari Lab for Orthopedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Herbert Kaltner
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Lena-Christ-Str. 48, 82152 Planegg, Germany
| | - Stefan Toegel
- Karl Chiari Lab for Orthopedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, 1090 Vienna, Austria
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Lena-Christ-Str. 48, 82152 Planegg, Germany
| | - Paul V Murphy
- School of Biological and Chemical Sciences, University of Galway, University Road, Galway H91 TK33, Ireland; SSPC - Science Foundation Ireland Research Centre for Pharmaceuticals, CÚRAM - Science Foundation Ireland Research Centre for Medical Devices, School of Biological and Chemical Sciences, National University of Ireland Galway, University Road, Galway H91 TK33, Ireland.
| | - Cristina Nativi
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia, 3-13, Sesto Fiorentino, Florence 50019, Italy; CeRM, University of Florence, via L. Sacconi, 6, Sesto Fiorentino, Florence 50019, Italy
| |
Collapse
|
184
|
Wang CC, Lu JW, Chiang KH, Cheng YS, Chu YH, Peng YJ, Cheng CH, Chang CY, Chuu JJ. Anti-Inflammatory and Chondro-Protective Effects of Acidic Polysaccharide from Enteromorpha Prolifera in Experimental Models of Osteoarthritis In-Vitro and In-Vivo. Cartilage 2022; 13:157-170. [PMID: 36250247 PMCID: PMC9924978 DOI: 10.1177/19476035221129573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE Osteoarthritis (OA) progression has been shown to increase the expression of inflammatory cytokines in joints, leading to the destruction of cartilage matrix. Interleukin (IL)-1β is a potent inflammatory cytokine associated with osteoarthritic synovial fluid. The protective effects of polysaccharides from Enteromorpha prolifera against acute hepatic injury was reported. DESIGN In this study, we examined the effects of Enteromorpha polysaccharide extracts (EPEs) in the treatment of OA. The effects of the EPEs were assessed using an IL-1β-stimulated SW1353 and SW982 cells. The expression levels of specific mRNA and proteins were evaluated using semi-quantitative reverse transcription polymerase chain reaction (sqRT-PCR) and western immunoblotting. An OA animal study involving C57BL/6J mice was also conducted to assess the effects on tactile sensitivity and anterior cruciate ligament transection (ACLT). RESULTS Acidic polysaccharide extract (APE) was shown to significantly reduce cytokine and chemokine mRNA levels in IL-1β-stimulated SW1353 and SW982 cells and attenuate the expression of proinflammatory cytokines and p38/AP-1 in SW1353 cells. APE was also shown to minimize the effect of osteolytic lesions in the knee joints of ACLT-induced osteoarthritic mice. CONCLUSIONS APE is a potent inhibitor of joint degeneration associated with OA.
Collapse
Affiliation(s)
- Chih-Chien Wang
- Department of Orthopedics, Tri-Service
General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jeng-Wei Lu
- Antimicrobial Resistance
Interdisciplinary Research Group, Singapore-MIT Alliance for Research and
Technology, Singapore
| | - Kuang-Hsing Chiang
- Taipei Heart Institute, Taipei Medical
University, Taipei, Taiwan,Division of Cardiology and
Cardiovascular Research Center, Taipei Medical University Hospital, Taipei,
Taiwan,Department of Internal Medicine, School
of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,Graduate Institute of Biomedical
Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Yu-Shuan Cheng
- Department of Biotechnology and Food
Technology, College of Engineering, Southern Taiwan University of Science and
Technology, Tainan, Taiwan
| | - You-Hsiang Chu
- Graduate Institute of Life Sciences,
National Defense Medical Center, Taipei, Taiwan
| | - Yi-Jen Peng
- Department of Pathology, Tri-Service
General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Hui Cheng
- Department of Biotechnology and Food
Technology, College of Engineering, Southern Taiwan University of Science and
Technology, Tainan, Taiwan
| | - Chia-Yu Chang
- Department of Neurology, Chi Mei
Medical Center, Tainan, Taiwan,Center for General Education,
Southern Taiwan University of Science and Technology, Tainan, Taiwan,Chia-Yu Chang, Department of Neurology,
Chi-Mei Medical Center, No. 901, Zhonghua Rd., Yongkang Dist., Tainan 71004,
Taiwan.
| | - Jiunn-Jye Chuu
- Department of Biotechnology and Food
Technology, College of Engineering, Southern Taiwan University of Science and
Technology, Tainan, Taiwan,Pharmacy Department, Wei Gong
Memorial Hospital, Miaoli, Taiwan,Chia-Yu Chang, Department of Neurology,
Chi-Mei Medical Center, No. 901, Zhonghua Rd., Yongkang Dist., Tainan 71004,
Taiwan.
| |
Collapse
|
185
|
Mostofa F, Yasid NA, Shamsi S, Ahmad SA, Mohd-Yusoff NF, Abas F, Ahmad S. In Silico Study and Effects of BDMC33 on TNBS-Induced BMP Gene Expressions in Zebrafish Gut Inflammation-Associated Arthritis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238304. [PMID: 36500396 PMCID: PMC9740523 DOI: 10.3390/molecules27238304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022]
Abstract
The bone morphogenic protein (BMP) family is a member of the TGF-beta superfamily and plays a crucial role during the onset of gut inflammation and arthritis diseases. Recent studies have reported a connection with the gut-joint axis; however, the genetic players are still less explored. Meanwhile, BDMC33 is a newly synthesized anti-inflammatory drug candidate. Therefore, in our present study, we analysed the genome-wide features of the BMP family as well as the role of BMP members in gut-associated arthritis in an inflammatory state and the ability of BDMC33 to attenuate this inflammation. Firstly, genome-wide analyses were performed on the BMP family in the zebrafish genome, employing several in silico techniques. Afterwards, the effects of curcumin analogues on BMP gene expression in zebrafish larvae induced with TNBS (0.78 mg/mL) were determined using real time-qPCR. A total of 38 identified BMP proteins were revealed to be clustered in five major clades and contain TGF beta and TGF beta pro peptide domains. Furthermore, BDMC33 suppressed the expression of four selected BMP genes in the TNBS-induced larvae, where the highest gene suppression was in the BMP2a gene (an eight-fold decrement), followed by BMP7b (four-fold decrement), BMP4 (four-fold decrement), and BMP6 (three-fold decrement). Therefore, this study reveals the role of BMPs in gut-associated arthritis and proves the ability of BDMC33 to act as a potential anti-inflammatory drug for suppressing TNBS-induced BMP genes in zebrafish larvae.
Collapse
Affiliation(s)
- Farhana Mostofa
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Nur Adeela Yasid
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Suhaili Shamsi
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Siti Aqlima Ahmad
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Nur Fatihah Mohd-Yusoff
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Bimolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Faridah Abas
- Department of Food Science, Faculty of Food Science & Technology, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Syahida Ahmad
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Correspondence: ; Tel.: +603-97696724
| |
Collapse
|
186
|
Sohn HS, Choi JW, Jhun J, Kwon SP, Jung M, Yong S, Na HS, Kim JH, Cho ML, Kim BS. Tolerogenic nanoparticles induce type II collagen-specific regulatory T cells and ameliorate osteoarthritis. SCIENCE ADVANCES 2022; 8:eabo5284. [PMID: 36427299 PMCID: PMC9699678 DOI: 10.1126/sciadv.abo5284] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Local inflammation in the joint is considered to contribute to osteoarthritis (OA) progression. Here, we describe an immunomodulating nanoparticle for OA treatment. Intradermal injection of lipid nanoparticles (LNPs) loaded with type II collagen (Col II) and rapamycin (LNP-Col II-R) into OA mice effectively induced Col II-specific anti-inflammatory regulatory T cells, substantially increased anti-inflammatory cytokine expression, and reduced inflammatory immune cells and proinflammatory cytokine expression in the joints. Consequently, LNP-Col II-R injection inhibited chondrocyte apoptosis and cartilage matrix degradation and relieved pain, while injection of LNPs loaded with a control peptide and rapamycin did not induce these events. Adoptive transfer of CD4+CD25+ T cells isolated from LNP-Col II-R-injected mice suggested that Tregs induced by LNP-Col II-R injection were likely responsible for the therapeutic effects. Collectively, this study suggests nanoparticle-mediated immunomodulation in the joint as a simple and effective treatment for OA.
Collapse
Affiliation(s)
- Hee Su Sohn
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong Won Choi
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - JooYeon Jhun
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sangmin Yong
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun Sik Na
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin-Hong Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
- Corresponding author. (M.-L.C.), (B.-S.K.)
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
- Corresponding author. (M.-L.C.), (B.-S.K.)
| |
Collapse
|
187
|
Wu X, Xu P, Shi X, Shang J, Chen X, Guo A, Wang F, Yin Z. Intra-articular injection of clioquinol ameliorates osteoarthritis in a rabbit model. Front Med (Lausanne) 2022; 9:1028575. [DOI: 10.3389/fmed.2022.1028575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Osteoarthritis (OA) is characterized by the degeneration of articular cartilage. Decreased autophagy is tightly associated with chondrocyte death, which contributes to the progression of OA. Thus, pharmacological activation of autophagy may be a promising therapeutic approach for OA. Here, we discovered that clioquinol, an antibiotic, significantly induces autophagy in OA chondrocytes from human tissue and rabbit model. Meanwhile, clioquinol can also augment the expression of extracellular matrix (ECM) components and suppress inflammatory mediators to improve OA microenvironment. Intra-articular injection of clioquinol can greatly prevent or slow down the development of this disease in a trauma-induced rabbit model of osteoarthritis. Such protective effect induced by clioquinol was at least in part explained by decreasing chondrocyte apoptosis and increasing autophagy. This study reveals the therapeutic potential of clioquinol in OA treatment.
Collapse
|
188
|
Stöckl S, Reichart J, Zborilova M, Johnstone B, Grässel S. Semaphorin 3A-Neuropilin-1 Signaling Modulates MMP13 Expression in Human Osteoarthritic Chondrocytes. Int J Mol Sci 2022; 23:ijms232214180. [PMID: 36430655 PMCID: PMC9699590 DOI: 10.3390/ijms232214180] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022] Open
Abstract
Osteoarthritis (OA) is a complex disorder of diarthrodial joints caused by multiple risk factors and is characterized by articular cartilage destruction as well as changes in other articular tissues. Semaphorin 3A (Sema3A), known to be a chemo-repellent for sensory nerve fibers, has recently been implicated in cartilage OA pathophysiology. We demonstrated that the expression of SEMA3A and its receptor neuropilin-1 (NRP1) are synchronously upregulated in chondrocytes isolated from knee cartilage of OA patients compared to non-OA control chondrocytes. In addition, we observed that during in vitro passaging of OA chondrocytes, the Nrp-1 level increases, whereas the Sema3A level decreases. In this study, we aimed to uncover how Sema3A-Nrp-1 signaling affects metabolism and viability of OA chondrocytes via siRNA-mediated inhibition of Nrp-1 expression. We observed a decreased proliferation rate and an increase in adhesion and senescence after Nrp-1 silencing. Moreover, MMP13 gene expression was reduced by approximately 75% in NRP1 knockdown OA chondrocytes, whereas MMP13 expression was induced by Sema3A treatment in control (nt siRNA) OA chondrocytes, accompanied by an impaired AKT phosphorylation. These findings suggest a potential catabolic function of Sema3A signaling in OA chondrocytes by inducing MMP13 expression and by compromising pro-survival AKT activation. We propose that targeting the Sema3A-Nrp-1 signaling axis might be an opportunity to interfere with OA pathogenesis and progression.
Collapse
Affiliation(s)
- Sabine Stöckl
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, 93053 Regensburg, Germany
| | - Johanna Reichart
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, 93053 Regensburg, Germany
| | - Magdalena Zborilova
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, 93053 Regensburg, Germany
| | - Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, OR 97239, USA
| | - Susanne Grässel
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, 93053 Regensburg, Germany
- Correspondence:
| |
Collapse
|
189
|
Chondrogenic Potential of Human Umbilical Cord Mesenchymal Stem Cells Cultured with Exosome-Depleted Fetal Bovine Serum in an Osteoarthritis Mouse Model. Biomedicines 2022; 10:biomedicines10112773. [PMID: 36359292 PMCID: PMC9687487 DOI: 10.3390/biomedicines10112773] [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: 09/16/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Osteoarthritis (OA) is characterized by the loss of articular cartilage and is also an age-related disease. Recently, stem cell therapy for cartilage repair has emerged. The stem cells need to be cultured with a fetal bovine serum (FBS)-supplemented medium. The effect of FBS-containing exosomes on the differentiation of human umbilical cord mesenchymal stem cells (HUCMSCs) is unknown. The morphology, proliferation, surface marker expressions, and trilineage differentiation ability of two groups of HUCMSCs, cultured with conventional (FBS) and exosome-depleted FBS (Exo(-)FBS), were evaluated. In a mouse OA model after two groups of HUCMSCs transplantation, the rotarod activity, histology, and immunohistochemistry (type II collagen, aggrecan, IL-1β, and MMP13) of the cartilage were evaluated. The Exo(-)FBS-cultured HUCMSCs, like FBS-cultured HUCMSCs, displayed classic MSC characteristics, including spindle-shaped morphology, surface marker expression (positive for CD44, CD73, CD90, CD105, and HLA-ABC and negative for CD34, CD45, and HLA-DR), and trilineage differentiation (chondrogenesis, osteogenesis, and adipogenesis). The Exo(-)FBS-cultured HUCMSCs proliferated significantly slower than those of the FBS-cultured HUCMSCs (p < 0.01). The trilineage gene expression of PPAR-γ, FABP4, APAL, type II collagen, aggrecan, and SOX9 was significantly increased in the Exo(-)FBS-cultured HUCMSCs than in the FBS-cultured HUCMSCs and undifferentiated controls. The Exo(-)FBS- and FBS-cultured HUCMSCs-transplanted mice showed a better rotarod activity than in the control OA mice (n = 3 in each group). A significant histological improvement in hyaline cartilage destruction after the transplantation of both types of FBS-cultured HUCMSCs was noted when compared with the OA knees. The Exo(-)FBS-cultured HUCMSCs-transplanted knees showed a higher International Cartilage Repair Society histological score (p < 0.05), staining intensity of type II collagen (p < 0.01), and aggrecan (p < 0.01) than in the control knees. Moreover, both types of the FBS-cultured HUCMSCs-transplanted knees significantly decreased the expression of MMP13 and IL-1β compared to that in the OA knees (p < 0.01). The Exo(-)FBS-cultured HUCMSCs harbor chondrogenic potential and attenuated cartilage destruction in a mouse OA model. Our study provides a basis for future clinical trials using Exo(-)FBS-cultured stem cells to treat OA.
Collapse
|
190
|
Ma T, Chen H, Ruan H, Lv L, Yu Y, Jia L, Zhao J, Li X, Zang Y, Xu X, Zhang J, Gao L. Natural product, bilobalide, improves joint health in rabbits with osteoarthritis by anti-matrix degradation and antioxidant activities. Front Vet Sci 2022; 9:1034623. [PMID: 36337189 PMCID: PMC9631767 DOI: 10.3389/fvets.2022.1034623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/07/2022] [Indexed: 05/01/2024] Open
Abstract
Osteoarthritis (OA) is a common chronic musculoskeletal disease reported in veterinary clinics that severely reduces the quality of life of animals. The natural product, bilobalide, has positive effects on chondroprotection but its exact mechanism of action is unclear. This study aimed to investigate the antioxidant and anti-matrix degradation activities of bilobalide in a rabbit model of OA and its protective effects on joints. We also investigated the possible mechanisms underlying these effects. The rabbit OA model was established by intra-articular injection of 4% papain. Thirty healthy male New Zealand rabbits were randomly divided into control, untreated OA, Cel (100 mg/kg celecoxib intervention as a positive control), BB-L and BB-H (40 mg /kg and 80 mg /kg bilobalide gavage treatment, respectively) groups. Two weeks after surgical induction, bilobalide or celecoxib was administered by gavage daily for 8 weeks. After 8 weeks of bilobalide intervention, cartilage macroscopic observation and histopathological images showed alleviation of cartilage damage after bilobalide treatment, and the Osteoarthritis Research Society International (OARSI) score was significantly lower than that in the OA group. Bilobalide reduced the expression of metalloproteinase 3 (MMP-3) and MMP-13 in cartilage tissue of OA rabbits and reversed the levels of serum C-telopeptides of type II collagen (CTX-II), cartilage oligomeric matrix protein (COMP), interleukin 1(IL-1), and tumor necrosis factor (TNF-α). Bilobalide (80 mg/kg) could improve the biomechanical properties and microstructural changes in subchondral bone in the early stage of OA in rabbits, thereby delaying subchondral bone damage. Mechanistically, bilobalide exerted antioxidant and anti-matrix degradation effects by upregulating the oxidative stress signaling Nrf2/HO-1 pathway and inhibiting cartilage degeneration in rabbit OA. We thus speculate that bilobalide supplements recovery from OA damage.
Collapse
Affiliation(s)
- Tianwen Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hong Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hongri Ruan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Liangyu Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yue Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Lina Jia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jinghua Zhao
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Xin Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuxin Zang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinyu Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jiantao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Li Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| |
Collapse
|
191
|
Lei Y, Wang X, Liao J, Shen J, Li Y, Cai Z, Hu N, Luo X, Cui W, Huang W. Shear-responsive boundary-lubricated hydrogels attenuate osteoarthritis. Bioact Mater 2022; 16:472-484. [PMID: 35415286 PMCID: PMC8967971 DOI: 10.1016/j.bioactmat.2022.02.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/20/2022] [Accepted: 02/12/2022] [Indexed: 12/15/2022] Open
Abstract
Lipid-based boundary layers formed on liposome-containing hydrogels can facilitate lubrication. However, these boundary layers can be damaged by shear, resulting in decreased lubrication. Here, a shear-responsive boundary-lubricated drug-loaded hydrogel is created by incorporating celecoxib (CLX)-loaded liposomes within dynamic covalent bond-based hyaluronic acid (HA) hydrogels (CLX@Lipo@HA-gel). The dynamic cross-linked network enables the hydrogel to get restructured in response to shear, and the HA matrix allows the accumulation of internal liposome microreservoirs on the sliding surfaces, which results in the formation of boundary layers to provide stable lubrication. Moreover, hydration shells formed surrounding the hydrogel can retard the degradation process, thus helping in sustaining lubrication. Furthermore, in vitro and in vivo experiments found that CLX@Lipo@HA-gels can maintain anabolic-catabolic balance, alleviate cartilage wear, and attenuate osteoarthritis progression by delivering CLX and shear-responsive boundary lubrication. Overall, CLX@Lipo@HA-gels can serve as shear-responsive boundary lubricants and drug-delivery vehicles to alleviate friction-related diseases like osteoarthritis.
Collapse
Affiliation(s)
- Yiting Lei
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, PR China
| | - Xingkuan Wang
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, No. 1 the South of Maoyuan Road, Nanchong, Sichuan, 637000, PR China
| | - Junyi Liao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, PR China
| | - Jieliang Shen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, PR China
| | - Yuling Li
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, No. 1 the South of Maoyuan Road, Nanchong, Sichuan, 637000, PR China
| | - Zhengwei Cai
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Ning Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, PR China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopedic Laboratory of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, 400016, PR China
| |
Collapse
|
192
|
Su Z, Zong Z, Deng J, Huang J, Liu G, Wei B, Cui L, Li G, Zhong H, Lin S. Lipid Metabolism in Cartilage Development, Degeneration, and Regeneration. Nutrients 2022; 14:3984. [PMID: 36235637 PMCID: PMC9570753 DOI: 10.3390/nu14193984] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Lipids affect cartilage growth, injury, and regeneration in diverse ways. Diet and metabolism have become increasingly important as the prevalence of obesity has risen. Proper lipid supplementation in the diet contributes to the preservation of cartilage function, whereas excessive lipid buildup is detrimental to cartilage. Lipid metabolic pathways can generate proinflammatory substances that are crucial to the development and management of osteoarthritis (OA). Lipid metabolism is a complicated metabolic process involving several regulatory systems, and lipid metabolites influence different features of cartilage. In this review, we examine the current knowledge about cartilage growth, degeneration, and regeneration processes, as well as the most recent research on the significance of lipids and their metabolism in cartilage, including the extracellular matrix and chondrocytes. An in-depth examination of the involvement of lipid metabolism in cartilage metabolism will provide insight into cartilage metabolism and lead to the development of new treatment techniques for metabolic cartilage damage.
Collapse
Affiliation(s)
- Zhanpeng Su
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524013, China
| | - Zhixian Zong
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Jinxia Deng
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524013, China
| | - Jianping Huang
- Department of Prosthodontics, Yonsei University College of Dentistry, Seoul 03722, Korea
| | - Guihua Liu
- Institute of Orthopaedics, Huizhou Municipal Central Hospital, Huizhou 516001, China
| | - Bo Wei
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524013, China
| | - Liao Cui
- Department of Pharmacology, Marine Biomedical Research Institute, Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical Unversity, Zhanjiang 524023, China
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Huan Zhong
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524013, China
| | - Sien Lin
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524013, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| |
Collapse
|
193
|
Liu XH, Ding JY, Zhu ZH, Wu XC, Song YJ, Xu XL, Ding DF. Recent advances in enzyme-related biomaterials for arthritis treatment. Front Chem 2022; 10:988051. [PMID: 36051622 PMCID: PMC9424673 DOI: 10.3389/fchem.2022.988051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/21/2022] [Indexed: 12/27/2022] Open
Abstract
Arthritis is a group of highly prevalent joint disorders, and osteoarthritis (OA) and rheumatoid arthritis are the two most common types. The high prevalence of arthritis causes severe burdens on individuals, society and the economy. Currently, the primary treatment of arthritis is to relieve symptoms, but the development of arthritis cannot be effectively prevented. Studies have revealed that the disrupted balance of enzymes determines the pathological changes in arthritis. In particular, the increased levels of matrix metalloproteinases and the decreased expression of endogenous antioxidant enzymes promote the progression of arthritis. New therapeutic strategies have been developed based on the expression characteristics of these enzymes. Biomaterials have been designed that are responsive when the destructive enzymes MMPs are increased or have the activities of the antioxidant enzymes that play a protective role in arthritis. Here, we summarize recent studies on biomaterials associated with MMPs and antioxidant enzymes involved in the pathological process of arthritis. These enzyme-related biomaterials have been shown to be beneficial for arthritis treatment, but there are still some problems that need to be solved to improve efficacy, especially penetrating the deeper layer of articular cartilage and targeting osteoclasts in subchondral bone. In conclusion, enzyme-related nano-therapy is challenging and promising for arthritis treatment.
Collapse
Affiliation(s)
- Xin-Hao Liu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia-Ying Ding
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhi-Heng Zhu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xi-Chen Wu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Jia Song
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- *Correspondence: Xiao-Ling Xu, ; Dao-Fang Ding,
| | - Dao-Fang Ding
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Xiao-Ling Xu, ; Dao-Fang Ding,
| |
Collapse
|
194
|
Celecoxib nanocrystal-loaded dissolving microneedles with highly efficient for osteoarthritis treatment. Int J Pharm 2022; 625:122108. [PMID: 35970280 DOI: 10.1016/j.ijpharm.2022.122108] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/27/2022] [Accepted: 08/09/2022] [Indexed: 11/20/2022]
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease that has a significant impact on patients' lives. Celecoxib (CXB) is now primarily used to treat OA with oral dosing. CXB's limited water solubility, on the other hand, restricts its therapeutic application. We developed a delivery system of dissolving microneedles (DMNs) loaded with CXB-nanocrystals (CXB-NCs) for the treatment of OA. Oral administration's inefficiency and injectable administration's poor compliance might be solved using DMNs. Furthermore, carrier-free NCs may dramatically increase the dissolution of drugs with poorly water-solubility, as well as the drug load of DMNs. Antisolvent precipitation was used to make CXB-NCs. CXB-NC@DMNs were prepared by mixing CXB-NCs with hyaluronic acid (HA) that had high mechanical qualities and could permeate the skin efficiently in vitro. The therapeutic effect of oral CXB-NCs was substantially better than that of the same dose of oral CXB in an in vivo pharmacodynamic trial, demonstrating that the preparation of CXB into NCs might greatly increase CXB bioavailability. Furthermore, we discovered that DMNs loaded with low-dose CXB-NCs had similar or even better efficacy than the oral CXB-NCs group. The findings suggested that CXB-NC@DMNs may be a very efficient and promising drug delivery strategy in the treatment of OA.
Collapse
|
195
|
Damayanti MM, Rachmawati M. Pre-Clinical Study: Immunohistochemical evaluation of matrix metalloproteinase-13 on rabbit (Oryctolagus cuniculus) socket healing after application of platelet-rich fibrin with and without hydroxyapatite. F1000Res 2022; 11:29. [PMID: 36101858 PMCID: PMC9440371 DOI: 10.12688/f1000research.74094.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Tissue engineering technology has been used globally and proven to accelerate wound healing. This study aimed to analyse the effect of adding hydroxyapatite (HA) as a scaffold to platelet-rich fibrin (PRF) as a growth factor in accelerating the wound healing process as seen from the expression of matrix metalloproteinase-13 (MMP-13). Methods: This research is an animal experiment conducted on 18 rabbits (Oryctolagus cuniculus). Rabbits were randomly divided into the following three groups of treatment: (G1) the application of PRF group, (G2) the application of PRF+HA group and (C) the control group without any application. Furthermore, each treatment group was split randomly into three groups of observation time. Periodontal tissue biopsy was performed to analyse the histopathological features that were examined on the basis of the level of MMP-13 immunoexpression. Results: MMP-13 immunoexpression in the PRF+HA group showed better histoscore results, indicating a substantial reduction in MMP-13 values compared with other groups. The healing process was shown to increase with increasing observation time (p<0.05), and the PRF+HA group outperformed the PRF and control groups. On day 3, MMP-13 exhibited a dark brown colour of Immunohistochemistry (IHC), which indicated an increase in the expression value of MMP-13 in the early stages of healing, namely, inflammation. On day 14, light brown IHC was seen, especially in group 2, as a reference that the remodeling process had begun. Conclusions: This study indicates that the administration of PRF and HA was capable of reducing the MMP-13 expression that significantly accelerates the socket healing process. Hydroxyapatite is an alloplastic material that has inherent bioactive properties that support osteoconduction, can bind MMPs, and showed faster healing results based on the observation time as documented by immunohistochemistry.
Collapse
Affiliation(s)
- Meta Maulida Damayanti
- Pathology Anatomy, Universitas Islam Bandung, Unisba, Bandung, West Java, 40116, Indonesia
| | - Meike Rachmawati
- Pathology Anatomy, Universitas Islam Bandung, Unisba, Bandung, West Java, 40116, Indonesia
| |
Collapse
|
196
|
Haartmans MJ, Timur UT, Emanuel KS, Caron MM, Jeuken RM, Welting TJ, van Osch GJ, Heeren RM, Cillero-Pastor B, Emans PJ. Evaluation of the Anti-Inflammatory and Chondroprotective Effect of Celecoxib on Cartilage Ex Vivo and in a Rat Osteoarthritis Model. Cartilage 2022; 13:19476035221115541. [PMID: 35932105 PMCID: PMC9364198 DOI: 10.1177/19476035221115541] [Citation(s) in RCA: 5] [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: 11/15/2022] Open
Abstract
OBJECTIVE The potential chondroprotective effect of celecoxib, a nonsteroidal anti-inflammatory drug and selective cyclooxygenase-2 inhibitor used to reduce pain and inflammation in knee osteoarthritis patients, is disputed. This study aimed at investigating the chondroprotective effects of celecoxib on (1) human articular cartilage explants and (2) in an in vivo osteoarthritis rat model. DESIGN Articular cartilage explants from 16 osteoarthritis patients were cultured for 24 hours with celecoxib or vehicle. Secreted prostaglandins (prostaglandin E2, prostaglandin F2α, prostaglandin D2) and thromboxane B2 (TXB2) concentrations were determined in medium by ELISA, and protein regulation was measured with label-free proteomics. Cartilage samples from 7 of these patients were analyzed for gene expression using real-time quantitative polymerase chain reaction. To investigate the chondroprotective effect of celecoxib in vivo, 14 rats received an intra-articular injection of celecoxib or 0.9% NaCl after osteoarthritis induction by anterior cruciate ligament transection and partial medial meniscectomy (ACLT/pMMx model). Histopathological scoring was used to evaluate osteoarthritis severity 12 weeks after injection. RESULTS Secretion of prostaglandins, target of Nesh-SH3 (ABI3BP), and osteonectin proteins decreased, whereas tissue inhibitor of metalloproteinase 2 (TIMP-2) increased significantly after celecoxib treatment in the human (ex vivo) explant culture. Gene expression of a disintegrin and metalloproteinase with thrombospondin motifs 4 and 5 (ADAMTS4/5) and metalloproteinase 13 (MMP13) was significantly reduced after celecoxib treatment in human cartilage explants. Cartilage degeneration was reduced significantly in an in vivo osteoarthritis knee rat model. CONCLUSIONS Our data demonstrated that celecoxib acts chondroprotective on cartilage ex vivo and a single intra-articular bolus injection has a chondroprotective effect in vivo.
Collapse
Affiliation(s)
- Mirella J.J. Haartmans
- Laboratory for Experimental
Orthopedics, Department of Orthopaedic Surgery, Maastricht University, Maastricht,
The Netherlands,Maastricht MultiModal Molecular Imaging
Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University,
Maastricht, The Netherlands
| | - Ufuk Tan Timur
- Laboratory for Experimental
Orthopedics, Department of Orthopaedic Surgery, Maastricht University, Maastricht,
The Netherlands
| | - Kaj S. Emanuel
- Laboratory for Experimental
Orthopedics, Department of Orthopaedic Surgery, Maastricht University, Maastricht,
The Netherlands,Department of Orthopaedic Surgery,
Amsterdam Movement Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The
Netherlands
| | - Marjolein M.J. Caron
- Laboratory for Experimental
Orthopedics, Department of Orthopaedic Surgery, Maastricht University, Maastricht,
The Netherlands
| | - Ralph M. Jeuken
- Laboratory for Experimental
Orthopedics, Department of Orthopaedic Surgery, Maastricht University, Maastricht,
The Netherlands
| | - Tim J.M. Welting
- Laboratory for Experimental
Orthopedics, Department of Orthopaedic Surgery, Maastricht University, Maastricht,
The Netherlands
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports
Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The
Netherlands,Department of Otorhinolaryngology,
Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ron M.A. Heeren
- Maastricht MultiModal Molecular Imaging
Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University,
Maastricht, The Netherlands
| | - Berta Cillero-Pastor
- Maastricht MultiModal Molecular Imaging
Institute (M4i), Division of Imaging Mass Spectrometry, Maastricht University,
Maastricht, The Netherlands,MERLN Institute for Technology-Inspired
Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering,
Maastricht University, Maastricht, The Netherlands,Dr. Berta Cillero-Pastor, Maastricht
MultiModal Molecular Imaging Institute (M4i), Division of Imaging Mass
Spectrometry, Maastricht University, Maastricht, The Netherlands; MERLN
Institute for Technology-Inspired Regenerative Medicine, Department of Cell
Biology-Inspired Tissue Engineering, Maastricht University, Universiteitssingel
40, 6229 ER, Maastricht, PO Box 616, Maastricht, 6200 MD, The Netherlands.
| | - Pieter J. Emans
- Laboratory for Experimental
Orthopedics, Joint Preserving Clinic, Department of Orthopaedic Surgery, Maastricht
University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
197
|
Intermittent Hydrostatic Pressure Promotes Cartilage Repair in an Inflammatory Environment through Hippo-YAP Signaling In Vitro and In Vivo. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3215461. [PMID: 35968240 PMCID: PMC9371873 DOI: 10.1155/2022/3215461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022]
Abstract
The study of chondrogenic progenitor cells (CPCs) as seed cells has become a new focus of cartilage regeneration. The inflammatory environment of osteoarthritis (OA) inhibits the repair ability of CPCs. But the OA patients' CPCs showed an excellent regeneration ability with intermittent hydrostatic pressure (IHP). However, the mechanism is unclear. We compared the expression of the Hippo signaling effect factor YAP between OA and normal cartilages. Then, the relationship between the Kellgren-Lawrence (K-L) score of OA and the rate of YAP-positive cells was analyzed. The changes of CPCs after IHP and IL-1β applications were observed. The OA model was established by cutting the anterior cruciate ligament of rats. The knee joint of the OA rats was distracted by hinged external fixator to create suitable IHP, named as the IHP group. The IHP group plus intra-articular injection of Verteporfin (VP) was named as the IHP+VP group, and the untreated rat group was named as the CON group. Four and 8 weeks after the operation, the reparative effect was evaluated by MASSON staining and immunohistochemical staining. Lower levels of YAP1 and higher expressions of p-YAP1 were found in the OA group as compared to the normal group. IHP inhibited the Hippo signaling in an inflammatory environment and promoted the proliferation of CPCs. The cartilage deterioration in the CON group progressed more significantly than that in the IHP+VP group. The best reparative effect was observed in the IHP group with increased expression of YAP1 and decreased p-YAP1. These results hint that mechanical stress can activate CPCs and promote cartilage repair in an inflammatory environment through inhibiting Hippo signaling.
Collapse
|
198
|
Cheleschi S, Tenti S, Lorenzini S, Seccafico I, Barbagli S, Frati E, Fioravanti A. Synovial Fluid Regulates the Gene Expression of a Pattern of microRNA via the NF-κB Pathway: An In Vitro Study on Human Osteoarthritic Chondrocytes. Int J Mol Sci 2022; 23:ijms23158334. [PMID: 35955467 PMCID: PMC9369022 DOI: 10.3390/ijms23158334] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Synovial fluid (SF) represents the primary source of nutrients of articular cartilage and is implicated in maintaining cartilage metabolism. We investigated the effects of SF, from patients with osteoarthritis (OA), rheumatoid arthritis (RA), and controls, on a pattern of microRNA (miRNA) in human OA chondrocytes. Cells were stimulated with 50% or 100% SF for 24 h and 48 h. Apoptosis and superoxide anion production were detected by cytometry; miRNA (34a, 146a, 155, 181a), cytokines, metalloproteinases (MMPs), type II collagen (Col2a1), antioxidant enzymes, B-cell lymphoma (BCL)2, and nuclear factor (NF)-κB by real-time PCR. The implication of the NF-κB pathway was assessed by the use of NF-κB inhibitor (BAY-11-7082). RA and OA SF up-regulated miR-34a, -146a, -155, -181a, interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, MMP-1, MMP-13, and ADAMTs-5 gene expression, while it down-regulated Col2a1. Pathological SF also induced apoptosis, reduced viability, and decreased BCL2 mRNA, whereas it increased superoxide anions, the expression of antioxidant enzymes, p65 and p50 NF-κB. Opposite and positive results were obtained with 100% control SF. Pre-incubation with BAY-11-7082 counteracted SF effects on miRNA. We highlight the role of the SF microenvironment in regulating some miRNA involved in inflammation and cartilage degradation during OA and RA, via the NF-κB pathway.
Collapse
|
199
|
Jiang H, Zhang Y, Hu G, Shang X, Ming J, Deng M, Li Y, Ma Y, Liu S, Zhou Y. Innate/Inflammatory Bioregulation of Surfactant Protein D Alleviates Rat Osteoarthritis by Inhibiting Toll-Like Receptor 4 Signaling. Front Immunol 2022; 13:913901. [PMID: 35865531 PMCID: PMC9294227 DOI: 10.3389/fimmu.2022.913901] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a deteriorating disease of cartilage tissues mainly characterized as low-grade inflammation of the joint. Innate immune molecule surfactant protein D (SP-D) is a member of collectin family of collagenous Ca2+-dependent defense lectins and plays a vital role in the inflammatory and innate immune responses. The present study investigated the SP-D-mediated innate/inflammatory bioregulation in OA and explored the underlying molecular mechanism. Transcriptome analysis revealed that SP-D regulated genes were strongly enriched in the inflammatory response, immune response, cellular response to lipopolysaccharide (LPS), PI3K-Akt signaling, Toll-like receptor (TLR) signaling, and extracellular matrix (ECM)-receptor interaction pathways. Knockdown of the SP-D gene by the recombinant adeno-associated virus promoted the macrophage specific markers of CD68, F4/80 and TLR4 in the articular cartilage in vivo. SP-D alleviated the infiltration of synovial macrophages and neutrophils, and inhibited TLR4, TNF-α and the phosphorylation of PI3K, Akt and NF-κB p65 in cartilage. SP-D suppressed cartilage degeneration, inflammatory and immune responses in the rat OA model, whilst TAK-242 strengthened this improvement. In in vitro conditions, SP-D pre-treatment inhibited LPS-induced overproduction of inflammation-correlated cytokines such as IL-1β and TNF-α, and suppressed the overexpression of TLR4, MD-2 and NLRP3. SP-D prevented the LPS-induced degradation of ECM by down-regulating MMP-13 and up-regulating collagen II. Blocking of TLR4 by TAK-242 further enhanced these manifestations. We also demonstrated that SP-D binds to the TLR4/MD-2 complex to suppress TLR4-mediated PI3K/Akt and NF-κB signaling activation in chondrocytes. Taken together, these findings indicate that SP-D has chondroprotective properties dependent on TLR4-mediated PI3K/Akt and NF-κB signaling and that SP-D has an optimal bioregulatory effect on the inflammatory and innate responses in OA.
Collapse
Affiliation(s)
- Huanyu Jiang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yubiao Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Geliang Hu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaobin Shang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jianghua Ming
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ming Deng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yaming Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yonggang Ma
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiqing Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Yan Zhou,
| |
Collapse
|
200
|
Assirelli E, Caravaggi P, Mazzotti A, Ursini F, Leardini A, Belvedere C, Neri S. Location-Dependent Human Osteoarthritis Cartilage Response to Realistic Cyclic Loading: Ex-Vivo Analysis on Different Knee Compartments. Front Bioeng Biotechnol 2022; 10:862254. [PMID: 35782520 PMCID: PMC9240619 DOI: 10.3389/fbioe.2022.862254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Osteoarthritis (OA) is a multifactorial musculoskeletal disorder affecting mostly weight-bearing joints. Chondrocyte response to load is modulated by inflammatory mediators and factors involved in extracellular cartilage matrix (ECM) maintenance, but regulatory mechanisms are not fully clarified yet. By using a recently proposed experimental model combining biomechanical data with cartilage molecular information, basally and following ex-vivo load application, we aimed at improving the understanding of human cartilage response to cyclic mechanical compressive stimuli by including cartilage original anatomical position and OA degree as independent factors. Methods: 19 mono-compartmental Knee OA patients undergoing total knee replacement were recruited. Cartilage explants from four different femoral condyles zones and with different degeneration levels were collected. The response of cartilage samples, pooled according to OA score and anatomical position was tested ex-vivo in a bioreactor. Mechanical stimulation was obtained via a 3-MPa 1-Hz sinusoidal compressive load for 45-min to replicate average knee loading during normal walking. Samples were analysed for chondrocyte gene expression and ECM factor release. Results: Non parametric univariate and multivariate (generalized linear mixed model) analysis was performed to evaluate the effect of compression and IL-1β stimulation in relationship to the anatomical position, local disease severity and clinical parameters with a level of significance set at 0.05. We observed an anti-inflammatory effect of compression inducing a significant downmodulation of IL-6 and IL-8 levels correlated to the anatomical regions, but not to OA score. Moreover, ADAMTS5, PIICP, COMP and CS were upregulated by compression, whereas COL-2CAV was downmodulated, all in relationship to the anatomical position and to the OA degree. Conclusion: While unconfined compression testing may not be fully representative of the in-vivo biomechanical situation, this study demonstrates the importance to consider the original cartilage anatomical position for a reliable biomolecular analysis of knee OA metabolism following mechanical stimulation.
Collapse
Affiliation(s)
- Elisa Assirelli
- Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Paolo Caravaggi
- Laboratory of Movement Analysis and Functional Evaluation of Prosthesis, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Antonio Mazzotti
- I Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesco Ursini
- Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Biomedical and Neuromotor Science, IRCCS Istituto Ortopedico Rizzoli, University of Bologna, Bologna, Italy
| | - Alberto Leardini
- Laboratory of Movement Analysis and Functional Evaluation of Prosthesis, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Claudio Belvedere
- Laboratory of Movement Analysis and Functional Evaluation of Prosthesis, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Simona Neri
- Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|