1
|
Zhang M, Jia G, Weng J, Zhu Y, Lin J, Yang Q, Fang C, Zeng H, Yuan G, Yang J, Yu F. A Novel Scaffold of Icariin/Porous Magnesium Alloy-Repaired Knee Cartilage Defect in Rat by Wnt/β-Catenin Signaling Pathway. ACS Biomater Sci Eng 2024; 10:5796-5806. [PMID: 39155687 DOI: 10.1021/acsbiomaterials.4c00713] [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] [Indexed: 08/20/2024]
Abstract
Cartilage defects caused by joint diseases are difficult to treat clinically. Tissue engineering materials provide a new means to promote the repair of cartilage defects. The purpose of this study is to design a novel scaffold of porous magnesium alloy loaded with icariin and sustained release in order to explore the effect and possible mechanism of this scaffold in repairing SD rat knee articular cartilage defect. We constructed a novel type of icariin/porous magnesium alloy scaffold, observed the structure of the scaffold by electron microscope, detected the drug release of icariin in the scaffold and the biological safety, and established an animal model of cartilage defect in the femoral intercondylar fossa of the knee joint in rats; the scaffold was placed in the defect. After 12 weeks of repair, the rat knee articular cartilage repair was evaluated by gross specimens and micro-CT, HE, safranin O-fast green, and toluidine blue staining combined with the modified Mankin's score. The protein expressions of the Wnt/β-catenin signaling pathway-related factors (β-catenin, Wnt5a, Wnt1, sFRP1) and chondrogenic differentiation-related factors (Sox9, Aggrecan, Col2α1) were detected by immunohistochemical staining. We found that the novel scaffold of icariin/porous magnesium alloy can release icariin slowly and has biosafety in rats. Compared with other groups, icariin/porous magnesium alloy can significantly promote the repair of cartilage defects and the expressions of β-catenin, Wnt5a, Wnt1, Sox9, Aggrecan, and Col2α1 (P < 0.05). This novel scaffold can promote the repair of rat knee cartilage defects, and this process may be achieved by activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Mengwei Zhang
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Gaozhi Jia
- School of Intelligent Manufacturing and Equipment, Shenzhen Institute of Information Technology, Shenzhen 518172, China
| | - Jian Weng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yuanchao Zhu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jianjin Lin
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Qi Yang
- Department of Medical Ultrasound, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Chongzhou Fang
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Hui Zeng
- Department of Orthopedics, Shenzhen Second Peoples Hospital, Shenzhen 518000, China
| | - Guangyin Yuan
- Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Fei Yu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
2
|
Cheng Q, He K, Zhu J, Li X, Wu X, Zeng C, Lei G, Wang N, Li H, Wei J. Memantine attenuates the development of osteoarthritis by blocking NMDA receptor mediated calcium overload and chondrocyte senescence. J Orthop Translat 2024; 48:204-216. [PMID: 39280634 PMCID: PMC11399475 DOI: 10.1016/j.jot.2024.08.007] [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] [Received: 07/15/2023] [Revised: 06/22/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
Background Memantine, which is an FDA-proven drug for the treatment of dementia, exerts its function by blocking the function of NMDA (N-methyl-D-aspartate) receptor, a calcium-permeable ion channel that reduces cytotoxic calcium overload. Chondrocyte senescence is a crucial cellular event that contributes to articular cartilage degeneration during osteoarthritis (OA) development. To date, the effects of memantine and its downstream NMDA receptor on chondrocyte senescence and OA have been rarely reported. Methods The protein levels of NMDA receptor and its agonistic ligand, glutamate, were compared between normal and OA chondrocytes. The quantity of intracellular calcium ions and the level of mitochondrial damage were evaluated using specific fluorescent probes and transmission electron microscopy (TEM), respectively. Chondrocyte senescence was evaluated by senescence-associated β-galactosidase (SA-β-Gal) staining and p16INK4a analysis. The function of NMDA receptor in chondrocyte senescence and OA was tested via agonists activation and gene knockdown experiments. The therapeutic effects of memantine on OA were examined both in vitro and in vivo. Additionally, to verify the findings from animal samples, a propensity score-matched cohort study was conducted using data from a United Kingdom primary care database (i.e., IQVIA Medical Research Database [IMRD]) to compare the risk of OA-related joint replacement involved in memantine initiators versus active comparators (i.e., acetylcholinesterase [AchE] initiators) in patients with dementia. Results The protein expression of NMDA receptor and the secretion of glutamate were both significantly increased in OA chondrocytes. NMDA receptor activation was found to stimulate chondrocyte calcium overload, which further led to mitochondrial fragmentation and chondrocyte senescence. Blocking the NMDA receptor with memantine and N-methyl-D-aspartate receptor subunit 1(NR1, the gene encoding NMDA receptor) knockdown resulted in reduced calcium influx, mitochondrial fragmentation, as well as cellular senescence in OA chondrocytes. Intra-articular injection of memantine in OA mice also exhibited protective effects against cartilage degeneration. Moreover, in the 1:5 propensity score-matched cohort study consisting of 6218 patients (n = 1435 in the memantine cohort; n = 4783 in the AchE cohort), the memantine initiator was associated with a lower risk of OA-related joint replacement than AchE initiators (Hazard ratio = 0.56, 95 % confidence interval: 0.34 to 0.99). Conclusion NMDA receptor plays an important role in inflammatory-induced cytotoxic calcium overload in chondrocytes, while memantine can effectively block the NMDA receptor to reduce chondrocyte senescence and retard the development of OA. The translational potential of this article As a clinically licensed drug used for the treatment of dementia, memantine has shown promising therapeutic effects on OA. Mechanistically, it functions by blocking NMDA receptor from mediating chondrocyte senescence. The protective effects of memantine against OA were verified not only by in vivo and in vitro experiments but also via a propensity score-matched human cohort study. These findings presented robust evidence for repurposing memantine for the treatment of OA.
Collapse
Affiliation(s)
- Qingmei Cheng
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Ke He
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Junyu Zhu
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiao Li
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Xuan Wu
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Zeng
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guanghua Lei
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Wang
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Li
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wei
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Health Management Center, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Mei S, Jiang F, Liu N, Feng Z, Zheng Y, Yang W, Zhang W, Cui Y, Wang W, Xie J, Zhang N. Sol-gel synthesis of magnesium oxide nanoparticles and their evaluation as a therapeutic agent for the treatment of osteoarthritis. Nanomedicine (Lond) 2024; 19:1867-1878. [PMID: 39109508 PMCID: PMC11457622 DOI: 10.1080/17435889.2024.2382421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/16/2024] [Indexed: 10/05/2024] Open
Abstract
Aim: We synthesized MgO NPs via sol-gel reaction and investigated them as carriers to deliver Mg2+ to the affected joint for osteoarthritis (OA).Materials & methods: The physicochemical properties of samples were characterized by transmission electron microscope (TEM), dynamic light scattering (DLS) and x-ray diffraction (XRD). The release of Mg2+ was monitored by ICP-MS. The potential cytotoxicity was evaluated using MTT assay. The efficacy and biosafety were evaluated in a rabbit OA model.Results: MgO NPs can prolong the Mg2+ release time from 0.5 h to 12 h. No significant cytotoxicity was observed when concentrations below 250 μg/ml. Intra-articular samples could effectively alleviate the degeneration and destruction of the cartilage.Conclusion: this study demonstrates the potential of MgO NPs as a safe and effective treatment of OA. Simultaneously, the size of the particles may play a significant role in influencing the therapeutic outcome.
Collapse
Affiliation(s)
- Sen Mei
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Fangchao Jiang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Na Liu
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Zhizi Feng
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Yu Zheng
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Wei Yang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Weizhong Zhang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Yingna Cui
- Department of Chemistry, Dalian University, Dalian, Liaoning, 116000, China
| | - Weiming Wang
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Nan Zhang
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| |
Collapse
|
4
|
Wen ZH, Tang CC, Lin YY, Yao ZK, Hsieh SP, Gar-Hwa-Lai, Chen WF, Jean YH. Effects of Etanercept on Experimental Osteoarthritis in Rats: Role of Histone Deacetylases. Cartilage 2024:19476035241264012. [PMID: 39057748 PMCID: PMC11569629 DOI: 10.1177/19476035241264012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/03/2024] [Accepted: 06/09/2024] [Indexed: 07/28/2024] Open
Abstract
OBJECTIVE Mounting evidence suggests that histone deacetylases (HDAC) inhibitors reduce cartilage destruction in animal models of osteoarthritis (OA). Tumor necrosis factor (TNF)-α-blocking treatment for OA may provide effective joint protection by slowing joint damage. To investigate the effects of intraperitoneal administration of etanercept (a TNF-α inhibitor) on OA development in rats and changes in the nociceptive behavior of rats and expression of HDACs, RUNX2, and MMP13 in cartilage. METHODS Induction of OA in Wistar rats was accomplished through anterior cruciate ligament transection (ACLT). One or five milligrams (mg) of etanercept was administered intraperitoneally for 5 consecutive weeks after ACLT to the ACLT + etanercept (1 and 5 mg/kg) groups. Nociceptive behavior and changes in knee joint width were analyzed. Cartilage was evaluated histologically and immunohistochemically. RESULTS ACLT + etanercept significantly improved mechanical allodynia and weight-bearing distribution compared to ACLT alone. In OA rats treated with etanercept, cartilage degeneration and synovitis were significantly less pronounced than those in ACLT rats. OA-affected cartilage also showed reduced expression of HDAC 6, 7, RUNX-2, and MMP-13 in response to etanercept but increased expression of HDAC4. CONCLUSION Our study demonstrated that etanercept therapy (1) attenuated the development of OA and synovitis in rats, (2) reduced nociception, and (3) regulated chondrocyte metabolism, possibly by inhibiting cell HDAC6 and HDAC7, RUNX2, and MMP13 and increasing HDAC4 expression. Based on new evidence, etanercept may have therapeutic potential in OA.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chi-Chieh Tang
- Department of Early Childhood Education, National Pintung University, Pingtung, Taiwan
| | - Yen-You Lin
- Department of Sports Medicine, China Medical University, Taichung, Taiwan
| | - Zhi-Kang Yao
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Orthopedics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Shih-Peng Hsieh
- Section of Pathology, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Gar-Hwa-Lai
- Section of Orthopedic Surgery, Pingtung Veterans General Hospital, Pingtung, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Neurosurgery, College of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung, Taiwan
| | - Yen-Hsuan Jean
- Section of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| |
Collapse
|
5
|
Hung YC, Chen LJ, Wang JH, Ho TJ, Tseng GF, Chen HP. The Therapeutic Potential of Intra-Articular Injection of Synthetic Deer Antler Peptides in a Rat Model of Knee Osteoarthritis. Int J Mol Sci 2024; 25:6041. [PMID: 38892229 PMCID: PMC11172866 DOI: 10.3390/ijms25116041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Synthetic deer antler peptides (TSKYR, TSK, and YR) stimulate the proliferation of human chondrocytes and osteoblasts and increase the chondrocyte content of collagen and glycosamino-glycan in vitro. This study investigated the peptide mixture's pain relief and chondroprotective effect in a rat model of collagenase-induced osteoarthritis. Thirty-six adult male Sprague-Dawley rats were divided into three groups: control (saline), positive control (hyaluronic acid), and ex-perimental (peptides). Intra-articular collagenase injections were administered on days 1 and 4 to induce osteoarthritis in the left knees of the rats. Two injections of saline, hyaluronic acid, or the peptides were injected into the same knees of each corresponding group at the beginning of week one and two, respectively. Joint swelling, arthritic pain, and histopathological changes were evaluated. Injection of the peptides significantly reduced arthritic pain compared to the control group, as evidenced by the closer-to-normal weight-bearing and paw withdrawal threshold test results. Histological analyses showed reduced cartilage matrix loss and improved total cartilage degeneration score in the experimental versus the control group. Our findings suggest that intra-articular injection of synthetic deer antler peptides is a promising treatment for osteoarthritis.
Collapse
Affiliation(s)
- Yu-Chou Hung
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan
- School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
- Department of Physical Medicine and Rehabilitation, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan
| | - Li-Jin Chen
- Department of Anatomy, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| | - Jen-Hung Wang
- Department of Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan;
| | - Tsung-Jung Ho
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 970374, Taiwan
| | - Guo-Fang Tseng
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan
- Department of Anatomy, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| | - Hao-Ping Chen
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| |
Collapse
|
6
|
Wen ZH, Wu ZS, Huang SY, Chou TL, Cheng HJ, Lo YH, Jean YH, Sung CS. Local Magnesium Sulfate Administration Ameliorates Nociception, Peripheral Inflammation, and Spinal Sensitization in a Rat Model of Incisional Pain. Neuroscience 2024; 547:98-107. [PMID: 38657727 DOI: 10.1016/j.neuroscience.2024.03.033] [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: 01/20/2024] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024]
Abstract
OBJECTIVE Postoperative pain remains one of the most common complaints after surgery, and appropriate treatments are limited. METHODS We therefore investigated the effect of the anti-nociceptive properties of magnesium sulfate (MgSO4), an N-methyl-D-aspartate (NMDA) receptor antagonist, on incision-induced postoperative pain and peripheral and central nervous system inflammation. RESULTS We found that local MgSO4 administration dose-dependently increases paw withdrawal latency, indicating reduced peripheral postoperative pain. Furthermore, MgSO4 inhibited the expression of interleukin-1β (IL-1β) and inducible nitric oxide synthase (iNOS) and phosphorylation of the NMDA receptor NR1 subunit in injured paw tissue and significantly attenuated microglial and astrocytic activation in the ipsilateral lumbar spinal cord dorsal horn. CONCLUSION Locally administered MgSO4 has potential for development as an adjunctive therapy for preventing central nociceptive sensitization.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804201, Taiwan; Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Zong-Sheng Wu
- Division of Pain Management, Department of Anesthesiology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Shi-Ying Huang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Tung-Lin Chou
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Hao-Jung Cheng
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Yi-Hao Lo
- Department of Family Medicine, Zouying Armed Forces General Hospital, Kaohsiung 813204, Taiwan; Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
| | - Yen-Hsuan Jean
- Department of Orthopedic Surgery, Pingtung Christian Hospital, No. 60 Dalian Road, Pingtung 900026, Taiwan
| | - Chun-Sung Sung
- Division of Pain Management, Department of Anesthesiology, Taipei Veterans General Hospital, Taipei 112201, Taiwan; School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan.
| |
Collapse
|
7
|
Neto EDS, Pedro PPDA, Cartágenes MDSDS, Neto JOB, Garcia JBS. The effect of low dose intra-articular S(+) ketamine on osteoarthritis in rats: an experimental study. BRAZILIAN JOURNAL OF ANESTHESIOLOGY (ELSEVIER) 2024; 74:844502. [PMID: 38604407 PMCID: PMC11031719 DOI: 10.1016/j.bjane.2024.844502] [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: 03/21/2023] [Revised: 02/16/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND This study aimed to investigate the analgesic impact of S(+)-ketamine on pain behavior and synovial inflammation in an osteoarthritis (OA) model. METHODS Animals were grouped as follows: OA-Saline (n = 24) and OA-Ketamine (n = 24), OA induced via intra-articular sodium monoiodoacetate (MIA); a Non-OA group (n = 24) served as the control. On the 7th day post OA induction, animals received either saline or S(+)-ketamine (0.5 mg.kg-1). Behavioral and histopathological assessments were conducted up to day 28. RESULTS S(+)-ketamine reduced allodynia from day 7 to 28 and hyperalgesia from day 10 to 28. It notably alleviated weight distribution deficits from day 10 until the end of the study. Significant walking improvement was observed on day 14 in S(+)-ketamine-treated rats. Starting on day 14, OA groups showed grip force decline, which was countered by S(+)-ketamine on day 21. However, S(+)-ketamine did not diminish synovial inflammation. CONCLUSION Low Intra-articular (IA) doses of S(+)-ketamine reduced MIA-induced OA pain but did not reverse synovial histopathological changes. IRB APPROVAL NUMBER 23115 012030/2009-05.
Collapse
Affiliation(s)
| | | | - Maria do Socorro de Sousa Cartágenes
- Universidade Federal do Maranhão, Departamento de Ciências Fisiológicas; Universidade Federal do Maranhão, Laboratório Experimental para Estudo da Dor, São Luiz, MA, Brazil; Faculdade de Medicina da Universidade Ceuma, São Luiz, MA, Brazil
| | - José Osvaldo Barbosa Neto
- Universidade Federal do Maranhão, Laboratório Experimental para Estudo da Dor, São Luiz, MA, Brazil; Faculdade de Medicina da Universidade Ceuma, São Luiz, MA, Brazil.
| | - João Batista Santos Garcia
- Universidade Federal do Maranhão, Laboratório Experimental para Estudo da Dor, São Luiz, MA, Brazil; Universidade Federal do Maranhão, Departamento de Anestesiologia, Dor e Paliativos, São Luiz, MA, Brazil
| |
Collapse
|
8
|
Zheng L, Zhao S, Li Y, Xu J, Yan W, Guo B, Xu J, Jiang L, Zhang Y, Wei H, Jiang Q. Engineered MgO nanoparticles for cartilage-bone synergistic therapy. SCIENCE ADVANCES 2024; 10:eadk6084. [PMID: 38457498 PMCID: PMC10923500 DOI: 10.1126/sciadv.adk6084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
The emerging therapeutic strategies for osteoarthritis (OA) are shifting toward comprehensive approaches that target periarticular tissues, involving both cartilage and subchondral bone. This shift drives the development of single-component therapeutics capable of acting on multiple tissues and cells. Magnesium, an element essential for maintaining skeletal health, shows promise in treating OA. However, the precise effects of magnesium on cartilage and subchondral bone are not yet clear. Here, we investigated the therapeutic effect of Mg2+ on OA, unveiling its protective effects on both cartilage and bone at the cellular and animal levels. The beneficial effect on the cartilage-bone interaction is primarily mediated by the PI3K/AKT pathway. In addition, we developed poly(lactic-co-glycolic acid) (PLGA) microspheres loaded with nano-magnesium oxide modified with stearic acid (SA), MgO&SA@PLGA, for intra-articular injection. These microspheres demonstrated remarkable efficacy in alleviating OA in rat models, highlighting their translational potential in clinical applications.
Collapse
Affiliation(s)
- Liming Zheng
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Sheng Zhao
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Yixuan Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Wenjin Yan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Baosheng Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jianbin Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Lifeng Jiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Yifeng Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Hui Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| |
Collapse
|
9
|
Pinto ACMD, de Melo Nunes R, de Freitas Carvalho WV, Girão VCC, Rocha FAC. Systemic and local antiinflammatory effect of magnesium chloride in experimental arthritis. Adv Rheumatol 2024; 64:6. [PMID: 38178208 DOI: 10.1186/s42358-023-00346-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024] Open
Abstract
OBJECTIVE Despite some knowledge gaps in scientific evidence, MgCl2 is largely used for pain relief in musculoskeletal diseases. Mg salts were shown to provide analgesia postoperatively in orthopedic surgery and low Mg levels were linked to arthritis development and severity. We determined the anti-inflammatory activity of MgCl2 in an acute arthritis model. METHODS Mice received 0.1 mg/25µL Zymosan (Zy) or saline into the knees. Joint pain was evaluated using von Frey test; cell influx, and interleukin (IL)-1 level were assessed in joint lavage at 6 h. Synovia were excised for histopathology and analysis of immunoexpression of nuclear factor kappa B (NFκB) and tumor necrosis factor (TNF)-α. Groups (n = 6/group) received either 90 mg/kg MgCl2/100 µL or saline per os (systemic) or 500 µg/25 µL MgCl2 or saline intra-articularly (i.a.) 30 min prior to Zy. RESULTS MgCl2 given either systemically or locally significantly reduced cell influx (p = 0.0012 and p = 0.0269, respectively), pain (p = 0.0005 and p = 0.0038, respectively), and intra-articular IL-1 level (p = 0.0391), as compared to saline. Systemic MgCl2 significantly decreased NFκB (p < 0.05) immmunoexpression, as compared to saline. CONCLUSION MgCl2 given systemically or locally displayed anti-inflammatory activity in a severe acute arthritis model reducing cell influx, pain, and cytokine release. MgCl2 operates at least partially via inhibiting NFκB activation. This is the first in vivo demonstration that MgCl2 decreases cytokine release in arthritis, prompting reduction of inflammation and pain relief.
Collapse
Affiliation(s)
| | - Rodolfo de Melo Nunes
- Departamento de Medicina Interna da Faculdade de Medicina da Universidade Federal do Ceará, Fortaleza - Ceará, Brazil
| | | | | | - Francisco Airton Castro Rocha
- Departamento de Medicina Interna da Faculdade de Medicina da Universidade Federal do Ceará, Fortaleza - Ceará, Brazil.
- Instituto de Biomedicina - Laboratório de Investigação em Osteoartropatias, Rua Coronel Nunes de Melo, 1315 -1º. Andar Rodolfo Teofilo, Fortaleza, CE, CEP: 60430-270, Brazil.
| |
Collapse
|
10
|
Zhang Y, Chen T, Luo P, Li S, Zhu J, Xue S, Cao P, Zhu Z, Li J, Wang X, Wluka AE, Cicuttini F, Ruan G, Ding C. Associations of Dietary Macroelements with Knee Joint Structures, Symptoms, Quality of Life, and Comorbid Conditions in People with Symptomatic Knee Osteoarthritis. Nutrients 2022; 14:nu14173576. [PMID: 36079832 PMCID: PMC9460692 DOI: 10.3390/nu14173576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/20/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Osteoarthritis (OA), the most common joint disease in the elderly, has no cure. Macroelements are vital in human health and their relationships with OA are not clear. Clarifying the relationships between macroelements and OA may assist knee OA management. Methods: This study was a post-hoc analysis using data from a two-year randomized controlled trial among 392 participants with knee OA. Dietary macroelements, including calcium, magnesium, potassium, and phosphorus were computed-based on a semi-quantitative food frequency questionnaire at baseline. Knee joint structures (including cartilage volume, cartilage defect, bone marrow lesions, and effusion-synovitis volume), OA symptoms, quality of life, and OA comorbid conditions (including lower limb muscle strength and depressive symptoms) were assessed at baseline and month 24. Western Ontario and McMaster Universities (WOMAC) Index and depressive symptoms were assessed at baseline and months 3, 6, 12, and 24. Quality of life and lower limb muscle strength were assessed at baseline and months 6, 12, and 24. All analyses were conducted using mixed-effects models. Results: Higher dietary magnesium and potassium were associated with fewer OA symptoms, higher quality of life, greater lower limb muscle strength, and fewer depressive symptoms, but not with knee joint structures. Higher dietary calcium and phosphorus was not associated with any of the OA-related outcomes, except that dietary phosphorus was associated with greater lower limb muscle strength. Conclusions: In the longitudinal analyses, higher dietary magnesium and potassium intake are associated with fewer OA symptoms, higher quality of life, and milder comorbid conditions in patients with knee OA, suggesting dietary magnesium and potassium may have beneficial effects on OA and could be used for knee OA management.
Collapse
Affiliation(s)
- Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Tianyu Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
| | - Ping Luo
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Department of Spinal Surgery, Changsha Hospital of Hunan Normal University, Changsha 410000, China
| | - Shengfa Li
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Jianwei Zhu
- Department of Orthopedics, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
| | - Song Xue
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei 230000, China
| | - Peihua Cao
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Jia Li
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Xiaoshuai Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Anita E. Wluka
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3006, Australia
| | - Flavia Cicuttini
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3006, Australia
| | - Guangfeng Ruan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
- Clinical Research Centre, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
- Correspondence: (G.R.); (C.D.)
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3006, Australia
- Clinical Research Centre, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510000, China
- Correspondence: (G.R.); (C.D.)
| |
Collapse
|
11
|
Ita ME, Singh S, Troche HR, Welch RL, Winkelstein BA. Intra-articular MMP-1 in the spinal facet joint induces sustained pain and neuronal dysregulation in the DRG and spinal cord, and alters ligament kinematics under tensile loading. Front Bioeng Biotechnol 2022; 10:926675. [PMID: 35992346 PMCID: PMC9382200 DOI: 10.3389/fbioe.2022.926675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/27/2022] [Indexed: 12/03/2022] Open
Abstract
Chronic joint pain is a major healthcare challenge with a staggering socioeconomic burden. Pain from synovial joints is mediated by the innervated collagenous capsular ligament that surrounds the joint and encodes nociceptive signals. The interstitial collagenase MMP-1 is elevated in painful joint pathologies and has many roles in collagen regulation and signal transduction. Yet, the role of MMP-1 in mediating nociception in painful joints remains poorly understood. The goal of this study was to determine whether exogenous intra-articular MMP-1 induces pain in the spinal facet joint and to investigate effects of MMP-1 on mediating the capsular ligament’s collagen network, biomechanical response, and neuronal regulation. Intra-articular MMP-1 was administered into the cervical C6/C7 facet joints of rats. Mechanical hyperalgesia quantified behavioral sensitivity before, and for 28 days after, injection. On day 28, joint tissue structure was assessed using histology. Multiscale ligament kinematics were defined under tensile loading along with microstructural changes in the collagen network. The amount of degraded collagen in ligaments was quantified and substance P expression assayed in neural tissue since it is a regulatory of nociceptive signaling. Intra-articular MMP-1 induces behavioral sensitivity that is sustained for 28 days (p < 0.01), absent any significant effects on the structure of joint tissues. Yet, there are changes in the ligament’s biomechanical and microstructural behavior under load. Ligaments from joints injected with MMP-1 exhibit greater displacement at yield (p = 0.04) and a step-like increase in the number of anomalous reorganization events of the collagen fibers during loading (p ≤ 0.02). Collagen hybridizing peptide, a metric of damaged collagen, is positively correlated with the spread of collagen fibers in the unloaded state after MMP-1 (p = 0.01) and that correlation is maintained throughout the sub-failure regime (p ≤ 0.03). MMP-1 injection increases substance P expression in dorsal root ganglia (p < 0.01) and spinal cord (p < 0.01) neurons. These findings suggest that MMP-1 is a likely mediator of neuronal signaling in joint pain and that MMP-1 presence in the joint space may predispose the capsular ligament to altered responses to loading. MMP-1-mediated pathways may be relevant targets for treating degenerative joint pain in cases with subtle or no evidence of structural degeneration.
Collapse
Affiliation(s)
- Meagan E. Ita
- Spine Pain Research Laboratory, Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Sagar Singh
- Spine Pain Research Laboratory, Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Harrison R. Troche
- Spine Pain Research Laboratory, Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Rachel L. Welch
- Spine Pain Research Laboratory, Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Beth A. Winkelstein
- Spine Pain Research Laboratory, Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Beth A. Winkelstein,
| |
Collapse
|
12
|
Bai R, Miao MZ, Li H, Wang Y, Hou R, He K, Wu X, Jin H, Zeng C, Cui Y, Lei G. Increased Wnt/β-catenin signaling contributes to autophagy inhibition resulting from a dietary magnesium deficiency in injury-induced osteoarthritis. Arthritis Res Ther 2022; 24:165. [PMID: 35804467 PMCID: PMC9264717 DOI: 10.1186/s13075-022-02848-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
Background Dietary magnesium deficiency, which is common in modern diet, has been associated with osteoarthritis (OA) susceptibility. Despite this clinical association, no study has addressed if dietary magnesium deficiency accelerates OA development, especially at molecular level. This study aimed to explore aggravating effects of dietary magnesium deficiency on cartilage damage in an injury-induced murine OA model and to determine the underlying mechanism. Methods Twelve-week-old C57BL/6J mice subject to injury-induced OA modeling were randomized into different diet groups in which the mice were fed a diet with daily recommended magnesium content (500 mg/kg) or diets with low magnesium content (100 or 300 mg/kg). Articular cartilage damage was evaluated using the OARSI score. To determine molecular mechanisms in vitro, mouse chondrocytes were treated with media of low magnesium conditions at 0.1 and 0.4 mM, compared with normal magnesium condition at 0.7 mM as control. Anabolic and catabolic factors, autophagy markers, β-catenin, Wnt ligands, and a magnesium channel transient receptor potential cation channel subfamily member 7 (TRPM7) were analyzed by quantitative real-time PCR and immunoblotting. Autolysosomes were detected by DALGreen staining via fluorescence microscopy and autophagosomes were evaluated by transmission electron microscopy. Autophagy markers, β-catenin, and TRPM7 were assessed in vivo in the mouse cartilage, comparing between dietary magnesium deficiency and normal diet, by immunohistochemistry. Results Dietary magnesium deficiency aggravated injury-induced cartilage damage, indicated by significant higher OARSI scores. Autophagy markers LC3-II and Beclin-1 were decreased both in low magnesium diet-fed mice and low magnesium-treated chondrocytes. The number of autolysosomes and autophagosomes was also reduced under low magnesium conditions. Moreover, magnesium deficiency induced decreased anabolic and increased catabolic effect of chondrocytes which could be restored by autophagy activator rapamycin. In addition, reduced autophagy under low magnesium conditions is mediated by activated Wnt/β-catenin signaling. The expression of TRPM7 also decreased in low magnesium diet-fed mice, indicating that downstream changes could be regulated through this channel. Conclusions Dietary magnesium deficiency contributes to OA development, which is mediated by reduced autophagy through Wnt/β-catenin signaling activation. These findings indicated potential benefits of adequate dietary magnesium for OA patients or those individuals at high risk of OA. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02848-0.
Collapse
Affiliation(s)
- Ruijun Bai
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Michael Z Miao
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Curriculum in Oral and Craniofacial Biomedicine, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hui Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Yiqing Wang
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ruixue Hou
- Department of Population Health Science and Policy, Icahn School of Medical at Mount Sinai, New York, NY, USA
| | - Ke He
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Xuan Wu
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Hongyu Jin
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.,Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China.,Hunan Engineering Research Center of Osteoarthritis, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Cui
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. .,Xiangya International Medical Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China. .,Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China. .,Hunan Engineering Research Center of Osteoarthritis, Changsha, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
13
|
Proton pump inhibitor therapy and risk of knee replacement surgery: a general population-based cohort study. Osteoarthritis Cartilage 2022; 30:559-569. [PMID: 35031493 PMCID: PMC8940684 DOI: 10.1016/j.joca.2021.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/29/2021] [Accepted: 12/27/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Proton pump inhibitors (PPIs) are among the most commonly used medications for patients with osteoarthritis (OA). Various types of PPIs have different impacts on lowering serum magnesium level that may affect knee OA progression. We aimed to compare the risk of clinically relevant endpoint of knee replacement (KR) among initiators of five different PPIs with that among histamine-2 receptor antagonist (H2RA) initiators. DESIGN Among patients with knee OA (≥50 years) in The Health Improvement Network database in the UK we conducted five sequential propensity-score matched cohort studies to compare the risk of KR over 5-year among patients who initiated omeprazole (n = 2,672), pantoprazole (n = 664), lansoprazole (n = 3,747), rabeprazole (n = 751), or esomeprazole (n = 827) with those who initiated H2RA. RESULTS The prevalence of PPI prescriptions among participants with knee OA increased from 12.7% in 2000-44.0% in 2017. Two-hundred-and-seventy-four KRs (30.8/1,000 person-years) occurred in omeprazole initiators and 230 KRs (25.4/1,000 person-years) in H2RA initiators. Compared with H2RA initiators, the risk of KR was 21% higher in omeprazole initiators (hazard ratio [HR] = 1.21,95% confidence interval [CI]:1.01-1.44). Similar results were observed when pantoprazole use was compared with H2RA use (HR = 1.38,95%CI:1.00-1.90). No such an increased risk of KR was observed among lansoprazole (HR = 1.06,95%CI:0.92-1.23), rabeprazole (HR = 0.97,95%CI:0.73-1.30), or esomeprazole (HR = 0.83,95%CI:0.60-1.15) initiators compared with that among H2RA initiators. CONCLUSIONS In this population-based cohort study, initiation of omeprazole or pantoprazole use was associated with a higher risk of KR than initiation of H2RA use. This study raises concern regarding an unexpected risk of omeprazole and pantoprazole on accelerating OA progression.
Collapse
|
14
|
Li G, Cheng T, Yu X. The Impact of Trace Elements on Osteoarthritis. Front Med (Lausanne) 2022; 8:771297. [PMID: 35004740 PMCID: PMC8732765 DOI: 10.3389/fmed.2021.771297] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/30/2021] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is a progressive degenerative disease characterized by cartilage degradation, synovial inflammation, subchondral sclerosis and osteophyte formation. It has a multifactorial etiology with potential contributions from heredity, endocrine function, abnormal mechanical load and nutrition. Of particular considerations are trace element status. Several trace elements, such as boron and magnesium are essential for normal development of the bone and joint in human. While cadmium correlates with the severity of OA. The present review focuses on the roles of trace elements (boron, cadmium, copper, iron, magnesium, manganese, selenium, zinc) in OA and explores the mechanisms by which they act.
Collapse
Affiliation(s)
- Guoyong Li
- Department of Orthopaedics, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Cheng
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xuefeng Yu
- Department of Orthopaedics, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Research status of biodegradable metals designed for oral and maxillofacial applications: A review. Bioact Mater 2021; 6:4186-4208. [PMID: 33997502 PMCID: PMC8099919 DOI: 10.1016/j.bioactmat.2021.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/10/2021] [Accepted: 01/10/2021] [Indexed: 01/08/2023] Open
Abstract
The oral and maxillofacial regions have complex anatomical structures and different tissue types, which have vital health and aesthetic functions. Biodegradable metals (BMs) is a promising bioactive materials to treat oral and maxillofacial diseases. This review summarizes the research status and future research directions of BMs for oral and maxillofacial applications. Mg-based BMs and Zn-based BMs for bone fracture fixation systems, and guided bone regeneration (GBR) membranes, are discussed in detail. Zn-based BMs with a moderate degradation rate and superior mechanical properties for GBR membranes show great potential for clinical translation. Fe-based BMs have a relatively low degradation rate and insoluble degradation products, which greatly limit their application and clinical translation. Furthermore, we proposed potential future research directions for BMs in the oral and maxillofacial regions, including 3D printed BM bone scaffolds, surface modification for BMs GBR membranes, and BMs containing hydrogels for cartilage regeneration, soft tissue regeneration, and nerve regeneration. Taken together, the progress made in the development of BMs in oral and maxillofacial regions has laid a foundation for further clinical translation.
Collapse
|
16
|
Kuang X, Chiou J, Lo K, Wen C. Magnesium in joint health and osteoarthritis. Nutr Res 2021; 90:24-35. [PMID: 34023805 DOI: 10.1016/j.nutres.2021.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 01/31/2021] [Accepted: 03/16/2021] [Indexed: 12/28/2022]
Abstract
Osteoarthritis (OA) is a prevalent debilitating age-related skeletal disease. The hallmark of OA is the degradation of articular cartilage that cushions the joint during movement. It is characterized by chronic pain and disability. Magnesium, a critical trace element in the human body, plays a pivotal role in metabolism homeostasis and the energy balance. Humans obtain magnesium mainly from the diet. However, inadequate magnesium intake is not uncommon. Moreover, the magnesium status deteriorates with ageing. There has been a growing body of clinical studies pointing to an intimate relationship between dietary magnesium and OA although the conclusion remains controversial. As reported, the magnesium ion concentration is essential to determine cell fate. Firstly, the low-concentration magnesium ions induced human fibroblasts senescence. Magnesium supplementation was also able to mitigate chondrocyte apoptosis, and to facilitate chondrocyte proliferation and differentiation. In this literature review, we will outline the existing evidence in animals and humans. We will also discuss the controversies on plasma or intracellular level of magnesium as the indicator of magnesium status. In addition, we put forward the interplay between dietary magnesium intake and intestinal microbiome to modulate the inflammatory milieu in the conjecture of OA pathogenesis. This leads to an emerging hypothesis that the synergistic effect of magnesium and probiotics may open a new avenue for the prevention and treatment of OA.
Collapse
Affiliation(s)
- Xiaoqing Kuang
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Jiachi Chiou
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Kenneth Lo
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong.
| |
Collapse
|
17
|
Lin YY, Chen NF, Yang SN, Jean YH, Kuo HM, Chen PC, Feng CW, Liu YW, Lai YC, Wen ZH. Effects of Streptococcus thermophilus on anterior cruciate ligament transection-induced early osteoarthritis in rats. Exp Ther Med 2021; 21:222. [PMID: 33603831 PMCID: PMC7851616 DOI: 10.3892/etm.2021.9653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disorder and is classically defined as a progressively degenerative disease of articular cartilage. It manifests as joint pain and disability and currently has no comprehensive treatments. The primary purpose of the present study was to test the effects of probiotics, Streptococcus thermophilus (TCI633), on anterior cruciate ligament transection (ACLT)-induced experimental osteoarthritis (OA) in rats. In the current study, the experimental groups were given TCI633 (5x109, 5x1010 and 5x1011 CFU/kg/day) and glucosamine sulfate (250 mg/kg) between week 8 and 20 following ACLT. The results showed that oral administration of TCI633 and glucosamine had significant therapeutic effects on pain behaviors and knee swelling. Dose-dependent effects of TCI633 were also observed in ACLT-treated rats. Histopathological analysis demonstrated that ACLT+TCI633 (5x109, 5x1010 and 5x1011 CFU/kg/day) improved the synovial inflammation and cartilage damage of ACLT rats. Histology evaluation using the Osteoarthritis Research Society International system and synovial inflammatory score analysis showed the dose-dependent inhibition of TCI633 on synovial inflammation and cartilage damage. Immunohistochemical staining and TUNEL apoptosis staining showed that TCI633 could effectively increase the expression of type II collagen and reduce the amount of chondrocyte apoptosis in cartilage. Therefore, the present study demonstrated that oral intake of TCI633 could significantly suppressing pain behavior, reduce joint swelling and synovial tissue inflammation and increase type II collagen expression in cartilage. There was also a reduction in chondrocyte apoptosis and decreased progression of OA in ACLT-treated rats.
Collapse
Affiliation(s)
- Yen-You Lin
- Department of Sports Medicine, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan, R.O.C.,Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - San-Nan Yang
- Department of Pediatrics, E-DA Hospital, School of Medicine, College of Medicine I-Shou University, Kaohsiung 82445, Taiwan, R.O.C
| | - Yen-Hsuan Jean
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Pingtung 90059, Taiwan, R.O.C
| | - Hsiao-Mei Kuo
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - Pei-Chin Chen
- Department of Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan, R.O.C
| | - Chien-Wei Feng
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan, R.O.C
| | - Yu-Wei Liu
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - Yu-Cheng Lai
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan, R.O.C.,Department of Orthopedics, Asia University Hospital, Taichung 41354, Taiwan, R.O.C
| | - Zhi-Hong Wen
- Department of Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan, R.O.C.,Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan, R.O.C.,Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| |
Collapse
|
18
|
Abstract
Brain-derived neurotrophic factor (BDNF) and the high-affinity receptor tropomyosin receptor kinase B (TrkB) have important roles in neuronal survival and in spinal sensitization mechanisms associated with chronic pain. Recent clinical evidence also supports a peripheral role of BDNF in osteoarthritis (OA), with synovial expression of TrkB associated with higher OA pain. The aim of this study was to use clinical samples and animal models to explore the potential contribution of knee joint BDNF/TrkB signalling to chronic OA pain. Brain-derived neurotrophic factor and TrkB mRNA and protein were present in knee synovia from OA patients (16 women, 14 men, median age 67 years [interquartile range: 61-73]). There was a significant positive correlation between mRNA expression of NTRK2 (TrkB) and the proinflammatory chemokine fractalkine in the OA synovia. Using the surgical medial meniscal transection (MNX) model and the chemical monosodium iodoacetate (MIA) model of OA pain in male rats, the effects of peripheral BDNF injection, vs sequestering endogenous BDNF with TrkB-Fc chimera, on established pain behaviour were determined. Intra-articular injection of BDNF augmented established OA pain behaviour in MIA rats, but had no effect in controls. Intra-articular injection of the TrkB-Fc chimera acutely reversed pain behaviour to a similar extent in both models of OA pain (weight-bearing asymmetry MIA: -11 ± 4%, MNX: -12 ± 4%), compared to vehicle treatment. Our data suggesting a contribution of peripheral knee joint BDNF/TrkB signalling in the maintenance of chronic OA joint pain support further investigation of the therapeutic potential of this target.
Collapse
|
19
|
Magnesium sulfate prophylaxis attenuates the postpartum effects of preeclampsia by promoting M2 macrophage polarization. Hypertens Res 2020; 44:13-22. [PMID: 32719464 DOI: 10.1038/s41440-020-0511-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/13/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022]
Abstract
Preeclampsia is a complex disorder that is characterized by new onset hypertension and proteinuria at or after 20 weeks of gestation. Preeclampsia is a leading cause of maternal and fetal morbidity and mortality. MgSO4 is commonly used to treat severe preeclampsia, but its mechanism of action is poorly understood, and investigations into the effects of MgSO4 during the postpartum period are lacking. In this study, timed-pregnant Sprague-Dawley rats received low-dose lipopolysaccharide (LPS) on gestational day 14 to induce preeclampsia. Maternal and fetal outcomes and the macrophage profile 1 week after delivery were explored. On postpartum day (PD) 7, the maternal systolic blood pressure and urinary protein level were significantly increased, the number of M1 macrophages was increased and the number of M2 macrophages was decreased in the maternal kidney and brain; the median duration of gestation, the number of live fetuses, and the fetal weight/placenta weight ratio were significantly decreased; and the percentage of growth-restricted pups and fetal mortality were significantly increased in preeclampsia rats compared to normal pregnant control rats. Prophylactic MgSO4 decreased blood pressure at PD7, improved pregnancy outcomes, and promoted the polarization of M2 macrophages in the kidney and of M2 microglia in the brain of preeclampsia rats. These findings confirm that the pathophysiology of preeclampsia involves the dysregulation of the inflammatory response and the activation of M1 macrophages in several target organs during pregnancy. MgSO4 prophylaxis attenuates the postpartum effects of preeclampsia by promoting M2 macrophage polarization in the maternal kidney and brain.
Collapse
|
20
|
Yang Y, Li P, Zhu S, Bi R. Comparison of early-stage changes of osteoarthritis in cartilage and subchondral bone between two different rat models. PeerJ 2020; 8:e8934. [PMID: 32341895 PMCID: PMC7179570 DOI: 10.7717/peerj.8934] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/17/2020] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease and the major cause of joint pain and disability in the elderly. It is mainly characterized by articular cartilage degradation and subchondral bone remodeling. There are two main types of OA: natural occurring OA and secondary OA, mainly associated with aging and trauma, respectively. In this study, we established two OA models in rat knee joints to simulate the two types of OA, using the type II collagenase injection (CI) and anterior cruciate ligament transection (ACLT), respectively. After intervention for 2-6 weeks, cartilage and subchondral bone changes were detected in histological staining, immunochemistry, and micro-CT. Results showed that both models with typical pathology changes of OA were successfully induced, while the development and severity of OA process in the models were different. In ACLT rats, the cartilage damage was milder, lasted for a shorter time, and subchondral bone reconstruction occurred earlier, compared with the changes in CI rats. The cartilage damage was secondary to subchondral bone change in ACLT rats, while subchondral bone change was secondary to cartilage degeneration in CI rats. In conclusion, the interaction between cartilage and subchondral bone is different between the natural-occurring and secondary OA models. These two models not only suggest potential different mechanisms of the two types of OA, but also provide new directions for OA treatment and prevention.
Collapse
Affiliation(s)
- Yutao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Peiran Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Songsong Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ruiye Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Li H, Ding X, Terkeltaub R, Lin H, Zhang Y, Zhou B, He K, Li K, Liu Z, Wei J, Yang Y, Xie H, Zeng C, Lei G. Exploration of metformin as novel therapy for osteoarthritis: preventing cartilage degeneration and reducing pain behavior. Arthritis Res Ther 2020; 22:34. [PMID: 32087740 PMCID: PMC7036179 DOI: 10.1186/s13075-020-2129-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/12/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Metformin could activate adenosine monophosphate-activated protein kinase (AMPK) which was postulated as a potential therapeutic target for osteoarthritis. This study aimed to examine the effects of metformin on cartilage and pain in osteoarthritis mouse model. METHODS Eighty 10-week-old male C57BL/6 mice were randomized to 6 groups: non-operation, sham-operation, destabilization of the medial meniscus (DMM)-operation with intragastric saline/metformin, and DMM-operation with intraarticular saline/metformin. Articular cartilage degeneration was examined by scanning electron microscopy (SEM) and graded using the scoring system recommended by Osteoarthritis Research Society International (OARSI). Mechanical withdrawal threshold and hind paw weight distribution were measured to assess the pain-related behavior. Cell Counting Kit-8 assay, quantificational real-time polymerase chain reaction, and western blot analysis were conducted to examine the anabolic and anti-catabolic effect of metformin and the role of AMPK in mediating its effects on interleukin-1β stimulated primary mice chondrocytes. RESULTS Compared with mice receiving intragastric and intraarticular saline, mice in both intragastric and intraarticular metformin displayed attenuated articular cartilage degeneration, indicated by less cartilage damage under SEM and significantly lower OARSI scores. A higher paw withdrawal threshold and a decreased weight-bearing asymmetry were observed in the intragastric and intraarticular metformin mice compared with their corresponding saline groups in DMM model of osteoarthritis. In vitro experiments showed that metformin not only decreased the level of matrix metalloproteinase 13, but also elevated type II collagen production through activating AMPK pathway. CONCLUSIONS Metformin attenuates osteoarthritis structural worsening and modulates pain, suggesting its potential for osteoarthritis prevention or treatment.
Collapse
Affiliation(s)
- Hui Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Xiang Ding
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Robert Terkeltaub
- Department of Medicine, University of California at San Diego, San Diego, USA.,VA San Diego Medical Center, San Diego, USA
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Yuqing Zhang
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA.,The Mongan Institute, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Bin Zhou
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Ke He
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Kun Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Zhichen Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Jie Wei
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA.,The Mongan Institute, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Yuanheng Yang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.,Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China.,Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center of Osteoarthritis, Changsha, China.,National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China. .,Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA. .,The Mongan Institute, Massachusetts General Hospital, Harvard Medical School, Boston, USA.
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China. .,Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China. .,Hunan Engineering Research Center of Osteoarthritis, Changsha, China. .,National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
22
|
Ekiz Yılmaz T, Taşdemir M, Kaya M, Arıcan N, Ahıshalı B. The effects of magnesium sulfate on cyclophosphamide-induced ovarian damage: Folliculogenesis. Acta Histochem 2020; 122:151470. [PMID: 31812447 DOI: 10.1016/j.acthis.2019.151470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 01/18/2023]
Abstract
Cyclophosphamide (CYP) is one of the alkylating chemotherapeutic agents and its adverse effects on folliculogenesis in the ovary are well-known due to the previous scientific research on this topic. Magnesium has various effects in organisms, including catalytic functions on the activation and inhibition of many enzymes, and regulatory functions on cell proliferation, cell cycle, and differentiation. In this study, the effects of magnesium sulfate (MgSO4) on CYP induced ovarian damage were investigated. Immature Wistar-Albino female rats of 28-days were treated with pregnant mare serum gonadotrophin (PMSG) to develop the first generation of preovulatory follicles. Rats of the experimental groups were then treated with either CYP (100 mg/kg, i.p) and MgSO4 (270 mg/kg loading dose; 27 mg/kg maintenance doseX12, i.p) solely or in combination. Following in-vivo 5-bromo-2-deoxyuridine (BrdU) labeling, animals were sacrificed and ovaries were embedded in paraffin and Epon. In the ovaries, added to the evaluation of general morphology and follicle count; BrdU and TUNEL-labeling, cleaved caspase-3 and p27 (cyclin-dependent kinase inhibitor) staining was also performed immunohistochemically and an ultrastructural evaluation was performed by transmission electron microscopy (TEM). The number of primordial follicles were decreased and multilaminar primary and atretic follicles were increased in CYP group. After MgSO4 treatment, while primordial follicle pool were elevated, the number of atretic follicles were decreased. Additionally, decreased BrdU-labeling, increased cleaved caspase 3 immunoreactivity and increased TUNEL labeling were observed in CYP group. In CYP treated animals, observations showed that while MgSO4 administration caused no alterations in BrdU proliferation index and caspase-3 immunoreactivity, it significantly reduced the TUNEL labeling. It was also observed that, while p27 immunoreactivity significantly increased in the nuclei of granulosa and theca cells in the CYP group; MgSO4 treatment significantly reduced these immunoreactivities. The ultrastructural observations showed frequent apoptotic profiles in granulosa and theca cells in both early and advanced stages of follicles in the CYP group and the MgSO4 treatment before the CYP application led to ultrastructural alleviation of the apoptotic process. In conclusion, our data suggest that MgSO4 may provide an option of pharmacologic treatment for fertility preservation owing to the beneficial effects of on chemotherapy-induced accelerated follicular apoptotic process, and the protection of the primordial follicle pool.
Collapse
Affiliation(s)
- Tuğba Ekiz Yılmaz
- Istanbul University, Istanbul Faculty of Medicine, Department of Histology and Embryology, Istanbul 34093, Turkey.
| | - Müge Taşdemir
- Istanbul University, Istanbul Faculty of Medicine, Department of Histology and Embryology, Istanbul 34093, Turkey.
| | - Mehmet Kaya
- Istanbul University, Istanbul Faculty of Medicine, Department of Physiology, Istanbul 34093, Turkey.
| | - Nadir Arıcan
- Istanbul University, Istanbul Faculty of Medicine, Department of Forensic Medicine, Istanbul 34093, Turkey.
| | - Bülent Ahıshalı
- Istanbul University, Istanbul Faculty of Medicine, Department of Histology and Embryology, Istanbul 34093, Turkey.
| |
Collapse
|
23
|
Magnesium and vitamin C supplementation attenuates steroid-associated osteonecrosis in a rat model. Biomaterials 2020; 238:119828. [PMID: 32045781 PMCID: PMC7185815 DOI: 10.1016/j.biomaterials.2020.119828] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/07/2020] [Accepted: 01/25/2020] [Indexed: 01/15/2023]
Abstract
Magnesium (Mg)-based biometal attracts clinical applications due to its biodegradability and beneficial biological effects on tissue regeneration, especially in orthopaedics, yet the underlying anabolic mechanisms in relevant clinical disorders are lacking. The present study investigated the effect of magnesium (Mg) and vitamin C (VC) supplementation for preventing steroid-associated osteonecrosis (SAON) in a rat experimental model. In SAON rats, 50 mg/kg Mg, or 100 mg/kg VC, or combination, or water control was orally supplemented daily for 2 or 6 weeks respectively. Osteonecrosis was evaluated by histology. Serum Mg, VC, and bone turnover markers were measured. Microfil-perfused samples prepared for angiography and trabecular architecture were evaluated by micro-CT. Primary bone marrow cells were isolated from each group to evaluate their potentials in osteoblastogenesis and osteoclastogenesis. The mechanisms were tested in vitro. Histological evaluation showed SAON lesions in steroid treated groups. Mg and VC supplementation synergistically reduced the apoptosis of osteocytes and osteoclast number, and increased osteoblast surface. VC supplementation significantly increased the bone formation marker PINP, and the combination significantly decreased the bone resorption marker CTX. TNFα expression and oxidative injury were decreased in bone marrow in Mg/VC/combination group. Mg significantly increased the blood perfusion in proximal tibia and decreased the leakage particles in distal tibia 2 weeks after SAON induction. VC significantly elevated the osteoblast differentiation potential of marrow cells and improved the trabecular architecture. The combination supplementation significantly inhibited osteoclast differentiation potential of marrow cells. In vitro study showed promoting osteoblast differentiation effect of VC, and anti-inflammation and promoting angiogenesis effect of Mg with underlying mechanisms. Mg and VC supplementation could synergistically alleviate SAON in rats, indicating great translational potentials of metallic minerals for preventing SAON.
Collapse
|
24
|
Understanding the Molecular Mechanisms Underlying the Pathogenesis of Arthritis Pain Using Animal Models. Int J Mol Sci 2020; 21:ijms21020533. [PMID: 31947680 PMCID: PMC7013391 DOI: 10.3390/ijms21020533] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/27/2019] [Accepted: 01/09/2020] [Indexed: 12/22/2022] Open
Abstract
Arthritis, including osteoarthritis (OA) and rheumatoid arthritis (RA), is the leading cause of years lived with disability (YLD) worldwide. Although pain is the cardinal symptom of arthritis, which is directly related to function and quality of life, the elucidation of the mechanism underlying the pathogenesis of pain in arthritis has lagged behind other areas, such as inflammation control and regulation of autoimmunity. The lack of therapeutics for optimal pain management is partially responsible for the current epidemic of opioid and narcotic abuse. Recent advances in animal experimentation and molecular biology have led to significant progress in our understanding of arthritis pain. Despite the inherent problems in the extrapolation of data gained from animal pain studies to arthritis in human patients, the critical assessment of molecular mediators and translational studies would help to define the relevance of novel therapeutic targets for the treatment of arthritis pain. This review discusses biological and molecular mechanisms underlying the pathogenesis of arthritis pain determined in animal models of OA and RA, along with the methodologies used.
Collapse
|
25
|
Yao H, Xu JK, Zheng NY, Wang JL, Mok SW, Lee YW, Shi L, Wang JY, Yue J, Yung SH, Hu PJ, Ruan YC, Zhang YF, Ho KW, Qin L. Intra-articular injection of magnesium chloride attenuates osteoarthritis progression in rats. Osteoarthritis Cartilage 2019; 27:1811-1821. [PMID: 31536815 DOI: 10.1016/j.joca.2019.08.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 08/22/2019] [Accepted: 08/30/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To explore the effects of Mg2+ on the expression of osteoarthritic markers in human cartilage and synovium tissue explants. To investigate the therapeutic effect of intra-articular injection of Mg2+ in an established rat OA (Osteoarthritis) model of anterior cruciate ligament transection with partial medial meniscectomy (ACLT + PMM). DESIGN Human cartilage and synovium explants were collected from total knee replacement surgeries and incubated with MgCl2 (20 mmol/L) in vitro. A rat OA model was established by ACLT + PMM surgery in 450-500 g male Sprague Dawley (SD) rats. To select the optimal dose, intra-articular injections of MgCl2 (0.05, 0.5, 5 mol/L) were performed at 4 weeks after the surgery every 3 days for 2 weeks. The effect of optimized MgCl2 was further determined by histology, immunohistochemistry, and quantitative real-time polymerase chain reaction. RESULTS The expressions of osteoarthritic markers in human cartilage and synovium explants were inhibited by Mg2+in vitro. Immunohistochemical analysis further suggested the inhibitory effects of Mg2+ on the expression of MMP-13 and IL-6 in the human tissue explants. Cartilage degeneration and synovitis in ACLT + PMM rats were significantly improved by intra-articular injections of Mg2+ (0.5 mol/L). Immunohistochemical analysis also showed the regulatory effects of Mg2+ on osteoarthritic markers in both cartilage and synovium in rats, consistent with in vitro results. CONCLUSION Intra-articular injections of Mg2+ at 0.5 mol/L attenuate the progression of OA in the ACLT + PMM rat model. Such effect was at least in part explained by the promotion of cartilage matrix synthesis and the suppression of synovial inflammation.
Collapse
Affiliation(s)
- H Yao
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - J K Xu
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - N Y Zheng
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - J L Wang
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - S W Mok
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - Y W Lee
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - L Shi
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - J Y Wang
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - J Yue
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - S H Yung
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - P J Hu
- Department of Biomedical Engineering, Polytechnic University of Hong Kong, Hong Kong SAR, PR China.
| | - Y C Ruan
- Department of Biomedical Engineering, Polytechnic University of Hong Kong, Hong Kong SAR, PR China.
| | - Y F Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, PR China.
| | - K W Ho
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| | - L Qin
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| |
Collapse
|
26
|
Wen ZH, Lin YY, Chang YC, Tang CC, Hsieh SP, Lee HP, Sung CS, Chen WF, Lee CH, Hsuan Jean Y. The COX-2 inhibitor etoricoxib reduces experimental osteoarthritis and nociception in rats: The roles of TGF-β1 and NGF expressions in chondrocytes. Eur J Pain 2019; 24:209-222. [PMID: 31495059 DOI: 10.1002/ejp.1478] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is the most common joint disease, especially affecting the knee joint. Etoricoxib, a highly selective cyclooxygenase (COX)-2 inhibitor which can reduce postoperative pain after orthopaedic surgery. The aim of this study was to investigate the effects of oral etoricoxib on the development of OA and to examine concomitant changes in the nociceptive behaviour of rats. METHOD OA was induced in wistar rats by anterior cruciate ligament transection (ACLT) of the right knee. The ACLT + etoricoxib groups received 6.7 or 33.3 mg/kg of oral etoricoxib three times a week for 12 consecutive weeks, starting at week 8 after ACLT. Nociceptive behaviours and changes in knee joint width during OA development were analyzed. Histopathological studies were then performed on the cartilage. Immunohistochemical analysis was performed to examine the effect of etoricoxib on the expression of transforming growth factor-beta (TGF-β) and nerve growth factor (NGF) in articular cartilage chondrocytes. RESULTS OA rats receiving etoricoxib showed a significantly lower degree of cartilage degeneration than the rats receiving placebo. Nociceptive behaviour studies showed significant improvement in the ACLT + etoricoxib groups compared to that in the ACLT group. Moreover, etoricoxib attenuated NGF expression, but increased TGF-β expression, in OA-affected cartilage. CONCLUSIONS Oral etoricoxib in a rat OA model (a) attenuates the development of OA, (b) concomitantly reduces nociception, and (c) modulates chondrocyte metabolism, possibly by inhibiting NGF expression and increasing TGF-β expression. SIGNIFICANCE Oral administration of etoricoxib can attenuate the development of OA, with an associated attenuation of nociceptive behaviour in an experimental rat OA model. Moreover, etoricoxib attenuated NGF expression, but enhanced TGF-β expression in OA-affected chondrocytes. These findings may pave the way for further investigations of etoricoxib as a potential therapeutic target for the treatment of the inflammatory component in OA.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yen-You Lin
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Yi-Chen Chang
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chi-Chieh Tang
- Department of Early Childhood Education, National Pintung University, Pingtung, Taiwan
| | - Shih-Peng Hsieh
- Section of Pathology, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Hsin-Pai Lee
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veteran General Hospital, Taipei, Taiwan
| | - Wu-Fu Chen
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chian-Her Lee
- Department of Orthopedic, School of Medicine, Taipei Medical University, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yen Hsuan Jean
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| |
Collapse
|
27
|
Wei J, Neogi T, Terkeltaub R, Fenves AZ, Zeng C, Misra D, Choi HK, Lei G, Zhang Y. Thiazide diuretics and risk of knee replacement surgery among patients with knee osteoarthritis: a general population-based cohort study. Osteoarthritis Cartilage 2019; 27:1454-1461. [PMID: 31181261 PMCID: PMC11482426 DOI: 10.1016/j.joca.2019.05.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/30/2019] [Accepted: 05/29/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Thiazide diuretic use is associated with higher bone mineral density (BMD) and possibly lower serum magnesium levels than loop diuretic use, and both high BMD and low serum magnesium have been linked to high prevalent knee osteoarthritis. This study aimed to compare the risk of a clinically relevant endpoint, knee replacement (KR) surgery, among initiators of thiazide and loop diuretics. DESIGN Among patients aged ≥50 years with a diagnosis of knee osteoarthritis in The Health Improvement Network (THIN) in United Kingdom, we conducted a propensity score-matched cohort study to examine the relation of thiazide diuretic initiation vs loop diuretic initiation to the risk of KR over 5 years. RESULTS Among thiazide and loop diuretic initiators (n = 3,488 for each group; mean age: 73 years; female ratio: 59%), 359 (28.6/1,000 person-years) and 283 (24.1/1,000 person-years) KRs occurred during the follow-up period, respectively. The hazard ratio (HR) of KR for thiazide diuretic initiation vs loop diuretic initiation was 1.26 (95% confidence interval [CI]: 1.08-1.47). The adherence-adjusted HR of KR for continuous use of thiazide diuretics was 1.44 (95% CI: 1.21-1.72). CONCLUSIONS In this population-based cohort of patients with knee osteoarthritis, thiazide diuretic use was associated with a higher risk of KR than loop diuretic use. This association may potentially be due to thiazide diuretics' effect on BMD and serum magnesium.
Collapse
Affiliation(s)
- J Wei
- Health Management Center, Xiangya Hospital, Central South University, Changsha, Hunan, China; Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - T Neogi
- Section of Rheumatology, Boston University School of Medicine, Boston, MA, USA.
| | - R Terkeltaub
- Department of Medicine, University of California at San Diego, San Diego, CA, USA; VA San Diego Medical Center, San Diego, CA, USA.
| | - A Z Fenves
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - C Zeng
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - D Misra
- Section of Rheumatology, Boston University School of Medicine, Boston, MA, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - H K Choi
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - G Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Y Zhang
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Liu Y, Qin X, Lu X, Jiang J. Effects of inhibiting the PI3K/Akt/mTOR signaling pathway on the pain of sciatic endometriosis in a rat model. Can J Physiol Pharmacol 2019; 97:963-970. [PMID: 31461309 DOI: 10.1139/cjpp-2019-0156] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This study investigated the relationship between the pain of sciatic endometriosis and the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway. Adult female Sprague–Dawley rats successfully received sciatic endometriosis induction. Mechanical paw withdrawal threshold and paw withdrawal latency were recorded to assess the mechanical hypersensitivity and thermal hyperalgesia. Quantitative real-time PCR, Western blotting, and enzyme-linked immunosorbent assays were used to detect PI3K, Akt, and mTOR expressions and their phosphorylation as well as the expressions of substance P, calcitonin gene-related peptide (CGRP), and nerve growth factor (NGF). Mechanical paw withdrawal threshold and paw withdrawal latency significantly decreased after sciatic endometriosis induction in rats; this decrease was ameliorated by inhibiting the PI3K/Akt/mTOR signaling pathway using LY294002. Compared with controls, rats with sciatic endometriosis showed increased PI3K, Akt, and mTOR expressions and elevated p-PI3K, p-Akt, and p-mTOR protein expressions. Higher NGF, substance P, and CGRP expressions were also found in the superficial dorsal horn of the spinal cord in rats with sciatic endometriosis than in control rats 21 days after surgery. Following the injection of LY294002 into rats with sciatic endometriosis, there was a significant decrease in the expressions of NGF, substance P, and CGRP. In conclusion, the inhibition of the PI3K/Akt/mTOR signaling pathway may alleviate endometriosis-associated sciatic nerve pain in a rat model of sciatic endometriosis.
Collapse
Affiliation(s)
- Yan Liu
- Department of Obstetrics and Gynecology, Liaocheng People’s Hospital, Liaocheng 252000, Shandong, China
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan 250000, Shandong, China
| | - Xuying Qin
- Department of Obstetrics and Gynecology, Liaocheng People’s Hospital, Liaocheng 252000, Shandong, China
| | - Xiaofen Lu
- Department of Obstetrics and Gynecology, Liaocheng People’s Hospital, Liaocheng 252000, Shandong, China
| | - Jie Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan 250000, Shandong, China
| |
Collapse
|
29
|
Yue J, Jin S, Gu S, Sun R, Liang Q. High concentration magnesium inhibits extracellular matrix calcification and protects articular cartilage via Erk/autophagy pathway. J Cell Physiol 2019; 234:23190-23201. [PMID: 31161622 DOI: 10.1002/jcp.28885] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/14/2019] [Accepted: 05/01/2019] [Indexed: 01/05/2023]
Abstract
The significant cytopathological changes of osteoarthritis are chondrocyte hypertrophy, proteoglycan loss, extracellular matrix (ECM) calcification, and terminally, the replacement of cartilage by bone. Meanwhile, magnesium ion (Mg2+ ), as the second most abundant divalent cation in the human body, has been proved to inhibit the ECM calcification of hBMSCs (human bone marrow stromal cells), hVSMCs (Human vascular smooth muscle cells), and TDSCs (tendon-derived stem cells) in vitro studies. The ATDC5 cell line, which holds chondrocyte characteristics, was used in this study as an in vitro subject. We found that Mg2+ can efficiently suppress the ECM calcification and downregulate both hypertrophy and matrix metalloproteinase-related genes. Meanwhile, Mg2+ inhibits the formation of autophagy by inhibiting Erk phosphorylation signaling and lowers the expression of LC3, and eventually effectively reduces the formation of ECM calcification in vitro. In this study, we also used destabilization of the medial meniscus (DMM)-induced osteoarthritis (OA) animal model to further confirm the protective effect of Mg2+ on articular cartilage. Compared with the control group (saline-injected), continuous intra-articular magnesium chloride (MgCl2 ) injection can significantly alleviate the severity of cartilage calcification in OA animal model. Immunofluorescence staining also revealed that saline-injected DMM group had a higher positive rate of LC3 expression in cartilage chondrocytes, compared with MgCl2 -injected DMM group. In general, Mg2+ can significantly downregulate the hypertrophic gene Runx2, MMP13, and Col10α1, upregulate the chondrogenic genes Sox9 and Col1α1, inhibit the Erk phosphorylation signaling, reduce the expression of autophagy protein LC3, and effectively inhibit the ECM calcification of ATDC5. In vivo study also proved that intra-articular injection of Mg2+ protected knee cartilage by inhibiting the autophagy formation.
Collapse
Affiliation(s)
- Jiaji Yue
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shanzi Jin
- Department of Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shizhong Gu
- Department of Sports Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Rui Sun
- Department of Sports Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Qingwei Liang
- Department of Sports Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
30
|
Chen R, Zhou X, Yin S, Lu Z, Nie J, Zhou W, Liu X. [Study on the protective mechanism of autophagy on cartilage by magnesium sulfate]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:1340-1345. [PMID: 30600669 DOI: 10.7507/1002-1892.201804015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective To investigate the mechanism of magnesium sulfate in protecting rabbit cartilage by initiating autophagy. Methods Twenty-four adult female New Zealand rabbits were used to prepare post-traumatic osteoarthritis (PTOA) models by anterior cruciate ligament transection. Then, the PTOA models were randomly divided into PTOA group, distilled water group, and magnesium sulfate group, with 8 rabbits in each group. Immediately after operation, the distilled water group and the magnesium sulfate group were injected with 0.5 mL distilled water and 20 mmol/L magnesium sulfate solution in the joint cavity 3 times a week for 4 weeks, respectively. The PTOA group was not treated. The general condition of the animals was observed after operation. After 4 weeks, the expressions of tumor necrosis factor α (TNF-α) and collagen typeⅡ in the joint fluid and the expression of collagen type Ⅱ in venous blood were detected by ELISA assay. The protein expressions of transient receptor potential channel vanilloid 5 (TRPV5) and microtubule associated protein 1 light chain 3 (LC3; LC3-Ⅱ/LC3-Ⅰ) in femoral cartilage were detected by Western blot. The mRNA expressions of interleukin 1β (IL-1β), TNF-α, matrix metalloproteinases 3 (MMP-3) in synovial tissue and collagen type Ⅱ, Aggrecan (AGN), SOX9 in cartilage tissue were detected by real-time fluorescence quantitative PCR. Cartilage tissue sections were stained with HE staining, Masson staining, and Alcian blue staining and scored according to the modified histological osteoarthritis (OA) score. Results All animals survived until the experiment was completed. Compared with the other two groups, the expression of TNF-α in joint effusion and collagen type Ⅱ in joint effusion and venous blood were decreased in magnesium sulfate group; the protein expression of TRPV5 decreased, and the ratio of LC3-Ⅱ/LC3-Ⅰ increased significantly; the mRNA expressions of IL-1β, TNF-α, and MMP-3 in synovial tissue were decreased, and the mRNA expressions of collagen type Ⅱ, AGN, and SOX9 in cartilage tissue were increased; OA scores also decreased significantly. All differences were statistically significant ( P<0.05). There was no significant difference in the above indicators between the PTOA group and the distilled water group ( P>0.05). Conclusion Intra-articular injection of magnesium sulfate can reduce intra-articular inflammation, reduce the loss of collagen type Ⅱ and AGN, and is beneficial to cartilage regeneration in rabbits. The mechanism may be related to the initiation of chondroautophagy by inhibiting the calcium channel TRPV5.
Collapse
Affiliation(s)
- Rong Chen
- Department of Traumatic Orthopedics, the People's Hospital of Shiyan, Affiliated to Hubei University of Medicine, Shiyan Hubei, 442000, P.R.China
| | - Xue Zhou
- The Fourth Hospital of Clinical Medicine, Hubei University of Medicine, Shiyan Hubei, 442000, P.R.China
| | - Shaomei Yin
- The Fourth Hospital of Clinical Medicine, Hubei University of Medicine, Shiyan Hubei, 442000, P.R.China
| | - Zeyu Lu
- The Fourth Hospital of Clinical Medicine, Hubei University of Medicine, Shiyan Hubei, 442000, P.R.China
| | - Jinpeng Nie
- The Fourth Hospital of Clinical Medicine, Hubei University of Medicine, Shiyan Hubei, 442000, P.R.China
| | - Wencheng Zhou
- The Fourth Hospital of Clinical Medicine, Hubei University of Medicine, Shiyan Hubei, 442000, P.R.China
| | - Xinghui Liu
- Department of Anatomy, Hubei University of Medicine, Shiyan Hubei, 442000,
| |
Collapse
|
31
|
Chondroprotective Effects and Mechanisms of Dextromethorphan: Repurposing Antitussive Medication for Osteoarthritis Treatment. Int J Mol Sci 2018. [PMID: 29534535 PMCID: PMC5877686 DOI: 10.3390/ijms19030825] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disorder and primarily affects older people. The ideal anti-OA drug should have a modest anti-inflammatory effect and only limited or no toxicity for long-term use. Because the antitussive medication dextromethorphan (DXM) is protective in atherosclerosis and neurological diseases, two common disorders in aged people, we examined whether DXM can be protective in pro-inflammatory cytokine-stimulated chondrocytes and in a collagen-induced arthritis (CIA) animal model in this study. Chondrocytes were prepared from cartilage specimens taken from pigs or OA patients. Western blotting, quantitative PCR, and immunohistochemistry were adopted to measure the expression of collagen II (Col II) and matrix metalloproteinases (MMP). DXM significantly restored tumor necrosis factor-alpha (TNF-α)-mediated reduction of collagen II and decreased TNF-α-induced MMP-13 production. To inhibit the synthesis of MMP-13, DXM blocked TNF-α downstream signaling, including I kappa B kinase (IKK)α/β-IκBα-nuclear factor-kappaB (NF-κB) and c-Jun N-terminal kinase (JNK)-activator protein-1 (AP-1) activation. Besides this, DXM protected the CIA mice from severe inflammation and cartilage destruction. DXM seemed to protect cartilage from inflammation-mediated matrix degradation, which is an irreversible status in the disease progression of osteoarthritis. The results suggested that testing DXM as an osteoarthritis therapeutic should be a focus in further research.
Collapse
|
32
|
Duan L, Zhang W, Zhang F, Cai H. Myrtol improves post-traumatic knee osteoarthritis by regulation of reactive oxygen species, transforming growth factor β1 and apoptosis in a mouse model. Exp Ther Med 2017; 15:393-399. [PMID: 29250157 DOI: 10.3892/etm.2017.5367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 02/10/2017] [Indexed: 12/21/2022] Open
Abstract
The present study tested whether myrtol improves post-traumatic knee osteoarthritis (PTKO) by regulating the reactive oxygen species (ROS), transforming growth factor β1 (TGF-β1) and apoptosis in a mouse model. PTKO model mice were administered with 150, 300 or 450 mg/kg myrtol for 8 weeks. ELISA analysis was used to measure tumor necrosis factor-α, interleukin-6, malondialdehyde, superoxide dismutase, reactive oxygen species and TGF-β1 levels. Caspase-3 and Bax protein expressions were analyzed using western blot analysis. In the current study, treatment with myrtol improved the tissue damage and osteoarthritis score, while it also reversed the subchondral bone thickness, subchondral bone density, trabecular bone volume/relative trabecular bone volume ratio and trabecular bone spacing in PTKO mice. The activity of tumor necrosis factor α, interleukin-6, TGF-β1, malondialdehyde, superoxide dismutase and ROS were effectively inhibited, and the protein expression of caspase-3 and Bax were clearly suppressed by treatment with myrtol in a mouse model of PTKO. In conclusion, the results demonstrated that myrtol treatment improved PTKO through the suppression of inflammation, oxidative stress, ROS, TGF-β1 and Bax/caspase-3 in mice, and myrtol may be a potential agent for clinical therapy.
Collapse
Affiliation(s)
- Liqun Duan
- Department of Orthopedics, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Wenzhi Zhang
- Department of Orthopedics, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Feng Zhang
- Department of Orthopedics, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| | - Haiping Cai
- Department of Orthopedics, Anhui Provincial Hospital, Hefei, Anhui 230001, P.R. China
| |
Collapse
|
33
|
Rubio-Martínez LM, Rioja E, Castro Martins M, Wipawee S, Clegg P, Peffers MJ. Local anaesthetics or their combination with morphine and/or magnesium sulphate are toxic for equine chondrocytes and synoviocytes in vitro. BMC Vet Res 2017; 13:318. [PMID: 29115971 PMCID: PMC5678813 DOI: 10.1186/s12917-017-1244-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 10/30/2017] [Indexed: 12/11/2022] Open
Abstract
Background Chondrotoxic effects of local anaesthetics are well reported in humans and some animal species but knowledge on their toxic effects on synoviocytes or equine chondrocytes or the effects on cellular production of inflammatory cytokines is limited. The purpose of this study was to evaluate the in vitro effects of local anaesthetics, morphine, magnesium sulphate (MgSO4) or their combinations on cell viability and pro-inflammatory cytokine gene expression of equine synoviocytes and chondrocytes. Equine synoviocytes and cartilage explants harvested from normal joints in a co-culture system were exposed to mepivacaine (4.4 mg/ml), bupivacaine (2.2 mg/ml), morphine (2.85 mg/ml) and MgSO4 (37 mg/ml) alone or each local anaesthetic plus morphine or MgSO4 or both together. Chondrocyte and synoviocyte cell viability was assessed by CellTiter-Glo Luminescent Cell Viability Assay. Synoviocyte gene expression of IL-1β, IL-6 or TNF-α was measured and compared using the ∆∆CT method. Results Morphine alone, MgSO4 alone or their combination did not alter cell viability or the expression of IL-1β, IL-6 or TNF-α. However, local anaesthetics alone or in combination with morphine and/or MgSO4 reduced cell viability and increased the gene expression of IL-1β, IL-6 or TNF-α. Single short exposure to local anaesthetics is toxic to both chondrocytes and synoviocytes and their combination with morphine and/or MgSO4 enhanced the cytotoxic effects. Conclusions This in vitro study gives further evidence of the absence of cytotoxic effects of morphine alone, MgSO4 alone or their combination on normal articular tissues. However, local anaesthetics alone or in combination with morphine and/or MgSO4 have cytotoxic effects on equine articular tissues. Electronic supplementary material The online version of this article (10.1186/s12917-017-1244-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- L M Rubio-Martínez
- Institute of Veterinary Science, University of Liverpool, Leahurst Campus, Chester High Road, CH647TE, Neston, UK.
| | - E Rioja
- Institute of Veterinary Science, University of Liverpool, Leahurst Campus, Chester High Road, CH647TE, Neston, UK
| | - M Castro Martins
- Institute of Veterinary Science, University of Liverpool, Leahurst Campus, Chester High Road, CH647TE, Neston, UK
| | - S Wipawee
- Faculty of Veterinary Science, Rajamangala University of Technology Srivijaya (Thailand) and Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, UK
| | - P Clegg
- Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, UK
| | - M J Peffers
- Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, UK
| |
Collapse
|
34
|
Miller RE, Malfait AM. Osteoarthritis pain: What are we learning from animal models? Best Pract Res Clin Rheumatol 2017; 31:676-687. [PMID: 30509413 PMCID: PMC6284232 DOI: 10.1016/j.berh.2018.03.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 02/27/2018] [Accepted: 03/08/2018] [Indexed: 12/15/2022]
Abstract
All experimental models of osteoarthritis (OA)-like joint damage are accompanied by behaviors indicative of pain. In experimental knee OA, evoked pain responses to exogenously applied stimuli suggest that animals become sensitized to mechanical stimuli. Neurobiological techniques including electrophysiology and in vivo calcium imaging confirm that joint damage is associated with mechanical stimuli through peripheral sensitization. Several mediators present in the OA joint can cause peripheral sensitization, most notably the neurotrophin nerve growth factor (NGF). Furthermore, experimental OA is associated with neuroinflammation in the peripheral nervous system and central nervous system (CNS), including macrophage infiltration of the dorsal root ganglia and microglial activation in the spinal cord. Increasingly, researchers are employing models that are slowly progressive, and this approach has revealed that distinct pain mechanisms operate in a time-dependent manner, which may have important translational significance. While the study of pain in experimental OA is rapidly evolving, with the application of increasingly sophisticated techniques to assess pain and unravel the neurobiology of its genesis, important gaps and limitations in our current approaches exist, which our research community needs to address.
Collapse
Affiliation(s)
- Rachel E Miller
- Department of Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, United States
| | - Anne-Marie Malfait
- Department of Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W Harrison Street, Suite 510, Chicago, IL, 60612, United States.
| |
Collapse
|
35
|
Wang P, Wang ZY. Metal ions influx is a double edged sword for the pathogenesis of Alzheimer's disease. Ageing Res Rev 2017; 35:265-290. [PMID: 27829171 DOI: 10.1016/j.arr.2016.10.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 09/08/2016] [Accepted: 10/17/2016] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is a common form of dementia in aged people, which is defined by two pathological characteristics: β-amyloid protein (Aβ) deposition and tau hyperphosphorylation. Although the mechanisms of AD development are still being debated, a series of evidence supports the idea that metals, such as copper, iron, zinc, magnesium and aluminium, are involved in the pathogenesis of the disease. In particular, the processes of Aβ deposition in senile plaques (SP) and the inclusion of phosphorylated tau in neurofibrillary tangles (NFTs) are markedly influenced by alterations in the homeostasis of the aforementioned metal ions. Moreover, the mechanisms of oxidative stress, synaptic plasticity, neurotoxicity, autophagy and apoptosis mediate the effects of metal ions-induced the aggregation state of Aβ and phosphorylated tau on AD development. More importantly, imbalance of these mechanisms finally caused cognitive decline in different experiment models. Collectively, reconstructing the signaling network that regulates AD progression by metal ions may provide novel insights for developing chelators specific for metal ions to combat AD.
Collapse
Affiliation(s)
- Pu Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, PR China.
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, PR China.
| |
Collapse
|
36
|
Ho MJ, Lee DR, Jung HJ, Song WH, Park JS, Kang MJ. Formulation and Analgesic Effect of Sodium Hyaluronate and Magnesium Sulfate Combination in Rats Following Intra-articular Injection. B KOREAN CHEM SOC 2017. [DOI: 10.1002/bkcs.11128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Myoung Jin Ho
- College of Pharmacy; Dankook University; Chungnam 31116 Korea
| | - Dae Ro Lee
- College of Pharmacy; Dankook University; Chungnam 31116 Korea
| | - Hyuck Jun Jung
- College of Pharmacy; Dankook University; Chungnam 31116 Korea
| | - Woo Heon Song
- R&D Center; GLPharmTech Corporation; Seongnam Gyeonggido 13202 South Korea
| | - Jun Sang Park
- R&D Center; GLPharmTech Corporation; Seongnam Gyeonggido 13202 South Korea
| | - Myung Joo Kang
- College of Pharmacy; Dankook University; Chungnam 31116 Korea
| |
Collapse
|
37
|
Bujalska-Zadrożny M, Tatarkiewicz J, Kulik K, Filip M, Naruszewicz M. Magnesium enhances opioid-induced analgesia – What we have learnt in the past decades? Eur J Pharm Sci 2017; 99:113-127. [PMID: 27884758 DOI: 10.1016/j.ejps.2016.11.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/15/2016] [Accepted: 11/19/2016] [Indexed: 02/07/2023]
|
38
|
Kızılcık N, Özler T, Menda F, Uluçay Ç, Köner Ö, Altıntaş F. The effects of intra-articular levobupivacain versus levobupivacain plus magnesium sulfate on postoperative analgesia in patients undergoing arthroscopic meniscectomy: A prospective randomized controlled study. ACTA ORTHOPAEDICA ET TRAUMATOLOGICA TURCICA 2017; 51:104-109. [PMID: 28314555 PMCID: PMC6197358 DOI: 10.1016/j.aott.2017.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/14/2016] [Accepted: 09/15/2016] [Indexed: 01/31/2023]
Abstract
Objective The aim of this study was to compared the effectiveness of intraarticular levobupivacain with levobupivacain and magnesium sulfate. Methods In this prospective randomized double blinded study, 96 patients (67 male, 29 female; age range: 18–65 years) with ASA (American Society of Anesthesiologist) score I and II, who had undergone arthroscopic meniscectomy operation, were divided to 3 groups that had postoperative analgesia with intra-articular saline injection (control group), levobupivacain injection (L group) or levobupivacain and magnesium sulfate injection (LM group). Patients were compared with postoperative VAS (Visual Analog Score) score during rest and activity, opioid analgesic need, non-opioid analgesic need and other medication needs. Results Postoperative VAS scores during rest and activation at early postoperative period were significantly lower at LM group when compared with L group and lower than control group at all time periods. Opioid analgesic need, non-opioid analgesic need and other medication needs for non-pain symptoms were lower at LM group when compared with L and control groups at all time periods. Conclusion Intraarticular magnesium sulfate plus Levobupivacain injection is a safe and effective method for post operative pain management after arthroscopic meniscectomy. Keywords: Intra-articular injection, Magnesium sulfate, Levobupivacain, Postoperative analgesia, Chondrocyte apoptosis, Pain management, Arthroscopic menisectomy Level of Evidence Level I, Therapeutic study
Collapse
Affiliation(s)
| | - Turhan Özler
- Yeditepe University Faculty of Medicine, Turkey.
| | - Ferdi Menda
- Yeditepe University Faculty of Medicine, Turkey
| | | | - Özge Köner
- Yeditepe University Faculty of Medicine, Turkey
| | | |
Collapse
|
39
|
Zeng C, Li YS, Wei J, Xie DX, Xie X, Li LJ, Gao SG, Luo W, Xiong YL, Xiao WF, Lei GH. Analgesic effect and safety of single-dose intra-articular magnesium after arthroscopic surgery: a systematic review and meta-analysis. Sci Rep 2016; 6:38024. [PMID: 27901095 PMCID: PMC5128786 DOI: 10.1038/srep38024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 11/03/2016] [Indexed: 12/17/2022] Open
Abstract
To examine the analgesic effect and safety of single-dose intra-articular (IA) magnesium (Mg) after arthroscopic surgery. Pubmed, Embase and Cochrane library were searched through in January 2016. Eight RCTs and eight experimental studies were included. The IA Mg exhibited a significantly lower pain score when compared with placebo (MD, -0.41, 95% CI, -0.78 to -0.05, p = 0.03). There was no significant difference between Mg and bupivacaine in terms of pain relief and the time to first analgesic request. Furthermore, statistically significant differences both in pain score (MD, -0.62, 95% CI, -0.81 to -0.42, p < 0.00001) and time to first analgesic request (MD, 6.25, 95% CI, 5.22 to 7.29, p < 0.00001) were observed between Mg plus bupivacaine and bupivacaine alone. There was no statistically significant difference among the various groups with respect to adverse reactions. Most of the included in vitro studies reported the chondrocyte protective effect of Mg supplementation. There were also two in vivo studies showing the cartilage protective effect of IA Mg. The single-dose IA Mg following arthroscopic surgery was effective in pain relief without increasing adverse reactions, and it could also enhance the analgesic effect of bupivacaine. In addition, Mg seemed to possess the cartilage or chondrocyte protective effect based on experimental studies.
Collapse
Affiliation(s)
- Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Yu-sheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Jie Wei
- Health Management Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- Department of Epidemiology and Health Statistics, School of Public Health, Central South University, Changsha, Hunan Province, 410008, China
| | - Dong-xing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Xi Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Liang-jun Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Shu-guang Gao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Wei Luo
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Yi-lin Xiong
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Wen-feng Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Guang-hua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| |
Collapse
|
40
|
Raines AL, Shih MS, Chua L, Su CW, Tseng SCG, O'Connell J. Efficacy of Particulate Amniotic Membrane and Umbilical Cord Tissues in Attenuating Cartilage Destruction in an Osteoarthritis Model. Tissue Eng Part A 2016; 23:12-19. [PMID: 27707109 DOI: 10.1089/ten.tea.2016.0088] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Osteoarthritis (OA) is a progressive degenerative joint disease, and to date, no disease-modifying OA drug exists. Amniotic membrane and umbilical cord products have been used clinically in several diseases due to their anti-inflammatory and antiscarring properties. In the present study, we sought to evaluate whether a particulate amniotic membrane and umbilical cord (AM/UC) matrix could aid in attenuating disease progression. Lewis rats underwent medial meniscus transection (MMT) to induce OA. Two weeks after surgery, animals received intra-articular injections (50 μL) of either 50 or 100 μg/μL particulate AM/UC or saline control and were subsequently euthanized 1 or 4 weeks later. Cartilage degeneration was assessed using both histological scoring methods and equilibrium partitioning of an ionic contrast agent-microcomputed tomography (EPIC-μCT). EPIC-μCT analysis demonstrated that overall cartilage destruction was attenuated, with a significant increase in both cartilage thickness and volume as well as a significant decrease in total lesion area in animals injected with either dose of particulate AM/UC at 1 week, but only a high dose at 4 weeks postinjection. Osteoarthritis Research Society International (OARSI) histology scores of tibial sections corroborated EPIC-μCT results. Overall joint destruction was attenuated in animals injected with either dose of AM/UC tissue compared with saline-injected control animals at 1 week postinjection. Only high-dose AM/UC-injected animals continued to show less overall joint destruction by 4 weeks postinjection. Intra-articular injection of particulate AM/UC tissue attenuates cartilage degradation in a rat MMT model of OA, suggesting that it may be able to slow joint destruction in patients with OA.
Collapse
|
41
|
Gaurilcikaite E, Renton T, Grant AD. The paradox of painless periodontal disease. Oral Dis 2016; 23:451-463. [PMID: 27397640 DOI: 10.1111/odi.12537] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/04/2016] [Accepted: 06/23/2016] [Indexed: 12/23/2022]
Abstract
Periodontal diseases, primarily gingivitis and periodontitis, are characterised by progressive inflammation and tissue destruction. However, they are unusual in that they are not also accompanied by the pain commonly seen in other inflammatory conditions. This suggests that interactions between periodontal bacteria and host cells create a unique environment in which the pro-algesic effects of inflammatory mediators and factors released during tissue damage are directly or indirectly inhibited. In this review, we summarise the evidence that periodontal disease is characterised by an accumulation of classically pro-algesic factors from bacteria and host cells. We then discuss several mechanisms by which inflammatory sensitisation of nociceptive fibres could be prevented through inactivation or inhibition of these factors. Further studies are necessary to fully understand the molecular processes underlying the endogenous localised hypoalgesia in human periodontal disease. This knowledge might provide a rational basis to develop future therapeutic interventions, such as host modulation therapies, against a wide variety of other human pain conditions.
Collapse
Affiliation(s)
- E Gaurilcikaite
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - T Renton
- Department of Oral Surgery, Dental Institute, King's College London, London, UK
| | - A D Grant
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
42
|
Wen ZH, Tang CC, Chang YC, Huang SY, Lin YY, Hsieh SP, Lee HP, Lin SC, Chen WF, Jean YH. Calcitonin attenuates cartilage degeneration and nociception in an experimental rat model of osteoarthritis: role of TGF-β in chondrocytes. Sci Rep 2016; 6:28862. [PMID: 27345362 PMCID: PMC4921823 DOI: 10.1038/srep28862] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
We investigated the role of the calcitonin (Miacalcin) in the progression of osteoarthritis (OA) and in nociceptive behavior in an experimental rat model of OA and osteoporosis. OA was induced by anterior cruciate ligament transection (ACLT) of the right knee and by bilateral ovariectomy (OVX) in Wistar rats. Nociceptive behaviors (secondary mechanical allodynia and weight-bearing distribution of the hind paws) were analyzed prior to surgery and every week, beginning at 12 weeks after surgery, up to 20 weeks. At 20 weeks, histopathological studies were performed on the cartilage of the knee joints. Immunohistochemical analysis was performed to examine the effect of calcitonin on transforming growth factor (TGF)-β1 expression in articular cartilage chondrocytes. Rats subjected to ACLT + OVX surgery showed obvious OA changes in the joints. Animals subjected to ACLT + OVX and treated with calcitonin showed significantly less cartilage degeneration and improved nociceptive tests compared with animals subjected to ACLT + OVX surgeries alone. Moreover, calcitonin increased TGF-β1 expression in chondrocytes in ACLT + OVX-affected cartilage. Subcutaneous injection of calcitonin (1) attenuated the development of OA, (2) concomitantly reduced nociception, and (3) modulated chondrocyte metabolism, possibly by increasing cellular TGF-β1 expression.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology &Resources, and Center for Translational Biopharmaceuticals, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chi-Chieh Tang
- Department of Early Childhood Education, National Pintung University, Taiwan
| | - Yi-Chen Chang
- Department of Marine Biotechnology &Resources, and Center for Translational Biopharmaceuticals, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Shi-Ying Huang
- Department of Marine Biotechnology &Resources, and Center for Translational Biopharmaceuticals, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yen-You Lin
- Department of Marine Biotechnology &Resources, and Center for Translational Biopharmaceuticals, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Shih-Peng Hsieh
- Section of Pathology, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Hsin-Pai Lee
- Section of Pathology, Pingtung Christian Hospital, Pingtung, Taiwan.,Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Sung-Chun Lin
- Section of Pathology, Pingtung Christian Hospital, Pingtung, Taiwan.,Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Wu-Fu Chen
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan.,Department of Neurosurgery, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Taiwan
| | - Yen-Hsuan Jean
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| |
Collapse
|
43
|
Lu W, Wang L, Wo C, Yao J. Ketamine attenuates osteoarthritis of the knee via modulation of inflammatory responses in a rabbit model. Mol Med Rep 2016; 13:5013-20. [PMID: 27109206 PMCID: PMC4878578 DOI: 10.3892/mmr.2016.5164] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 03/31/2016] [Indexed: 12/02/2022] Open
Abstract
The aim of the present study was to investigate the efficacy of ketamine in attenuating osteoarthritis (OA) and modulating the expression of inflammatory mediators. A rabbit OA model was established by knee immobilization using plaster bandages. After six weeks, rabbits were randomly allocated into four groups (n=6/group): Normal saline, Ket60, Ket100, and Ket200 and twice a week for four weeks the rabbits received an intra-articular injection of saline, or 60, 100 or 200 µmol/l ketamine, respectively. One week after the final injection, samples of synovial membrane, synovial fluid and articular cartilage were isolated. The pathological changes were assessed by general observation, hematoxylin and eosin staining and Alcian blue/periodic-acid Schiff staining. Cartilage pathology was assessed using Mankin's scoring system. Tumor necrosis factor (TNF)-α and interleukin (IL)-10 levels in the synovial fluid were measured by enzyme-linked immunosorbent assays. The nuclear factor (NF)-κB p65 subunit expression level in cartilage samples was determined by immunohistochemistry. OA was characterized by morphological changes in the articular surface, cartilage lesions, infiltration of inflammatory cells and a significantly increased Mankin's score. Elevated TNF-α and reduced IL-10 levels in the synovial fluid, along with increased p65 expression levels in the cartilage were observed in OA rabbits. Intra-articular injection of ketamine ameliorated the pathological characteristics of OA, reduced the Mankin's score, decreased TNF-α and NF-κB p65 expression levels, and increased the level of IL-10 expression in a dose-dependent manner. Thus is was demonstrated that Ketamine suppresses the inflammatory response in OA by modulating inflammatory mediator expression levels in a rabbit model of OA.
Collapse
Affiliation(s)
- Wei Lu
- Department of Anesthesiology, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Lin Wang
- Department of Anesthesiology, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Chunxin Wo
- Department of Anesthesiology, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Jing Yao
- Department of Anesthesiology, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
44
|
Vasconcelos DM, Santos SG, Lamghari M, Barbosa MA. The two faces of metal ions: From implants rejection to tissue repair/regeneration. Biomaterials 2016; 84:262-275. [DOI: 10.1016/j.biomaterials.2016.01.046] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/20/2022]
|
45
|
Li Y, Yue J, Yang C. Unraveling the role of Mg(++) in osteoarthritis. Life Sci 2016; 147:24-9. [PMID: 26800786 DOI: 10.1016/j.lfs.2016.01.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/03/2016] [Accepted: 01/18/2016] [Indexed: 12/29/2022]
Abstract
Mg(++) is widely involved in human physiological processes that may play key roles in the generation and progression of diseases. Osteoarthritis (OA) is a complex joint disorder characterized by articular cartilage degradation, abnormal mineralization and inflammation. Magnesium deficiency is considered to be a major risk factor for OA development and progression. Magnesium deficiency is active in several pathways that have been implicated in OA, including increased inflammatory mediators, cartilage damage, defective chondrocyte biosynthesis, aberrant calcification and a weakened effect of analgesics. Abundant in vitro and in vivo evidence in animal models now suggests that the nutritional supplementation or local infiltration of Mg(++) represent effective therapies for OA. The goal of this review is to summarize the current understanding of the role of Mg(++) in OA with particular emphasis on the related molecular mechanisms involved in OA progression.
Collapse
Affiliation(s)
- Yaqiang Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; School of medicine, Tongji University, Shanghai, China
| | - Jiaji Yue
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; School of medicine, Tongji University, Shanghai, China
| | - Chunxi Yang
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; School of medicine, Tongji University, Shanghai, China.
| |
Collapse
|
46
|
Lewis R, Barrett-Jolley R. Changes in Membrane Receptors and Ion Channels as Potential Biomarkers for Osteoarthritis. Front Physiol 2015; 6:357. [PMID: 26648874 PMCID: PMC4664663 DOI: 10.3389/fphys.2015.00357] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/11/2015] [Indexed: 01/01/2023] Open
Abstract
Osteoarthritis (OA), a degenerative joint condition, is currently difficult to detect early enough for any of the current treatment options to be completely successful. Early diagnosis of this disease could increase the numbers of patients who are able to slow its progression. There are now several diseases where membrane protein biomarkers are used for early diagnosis. The numbers of proteins in the membrane is vast and so it is a rich source of potential biomarkers for OA but we need more knowledge of these before they can be considered practical biomarkers. How are they best measured and are they selective to OA or even certain types of OA? The first step in this process is to identify membrane proteins that change in OA. Here, we summarize several ion channels and receptors that change in OA models and/or OA patients, and may thus be considered candidates as novel membrane biomarkers of OA.
Collapse
Affiliation(s)
- Rebecca Lewis
- Faculty of Health and Medical Sciences, School of Veterinary Medicine and Science, University of Surrey Guildford, UK
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease, University of Liverpool Liverpool, UK
| |
Collapse
|
47
|
Chenopodium ambrosioides L. Reduces Synovial Inflammation and Pain in Experimental Osteoarthritis. PLoS One 2015; 10:e0141886. [PMID: 26524084 PMCID: PMC4629912 DOI: 10.1371/journal.pone.0141886] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/14/2015] [Indexed: 11/24/2022] Open
Abstract
The chronicity of osteoarthritis (OA), characterized by pain and inflammation in the joints, is linked to a glutamate receptor, N-methyl-D-aspartate (NMDA). The use of plant species such as Chenopodium ambrosioides L. (Amaranthaceae) as NMDA antagonists offers a promising perspective. This work aims to analyze the antinociceptive and anti-inflammatory responses of the crude hydroalcoholic extract (HCE) of C. ambrosioides leaves in an experimental OA model. Wistar rats were separated into six groups (n = 24): clean (C), negative control (CTL-), positive control (CTL+), HCE0.5, HCE5 and HCE50. The first group received no intervention. The other groups received an intra-articular injection of sodium monoiodoacetate (MIA) (8 mg/kg) on day 0. After six hours, they were orally treated with saline, Maxicam plus (meloxicam + chondroitin sulfate) and HCE at doses of 0.5 mg/kg, 5 mg/kg and 50 mg/kg, respectively. After three, seven and ten days, clinical evaluations were performed (knee diameter, mechanical allodynia, mechanical hyperalgesia and motor activity). On the tenth day, after euthanasia, synovial fluid and draining lymph node were collected for cellular quantification, and cartilage was collected for histopathological analysis. Finally, molecular docking was performed to evaluate the compatibility of ascaridole, a monoterpene found in HCE, with the NMDA receptor. After the third day, HCE reduced knee edema. HCE5 showed less cellular infiltrate in the cartilage and synovium and lower intensities of allodynia from the third day and of hyperalgesia from the seventh day up to the last treatment day. The HCE5 and HCE50 groups improved in forced walking. In relation to molecular docking, ascaridole showed NMDA receptor binding affinity. C. ambrosioides HCE was effective in the treatment of OA because it reduced synovial inflammation and behavioral changes due to pain. This effect may be related to the antagonistic effect of ascaridole on the NMDA receptor.
Collapse
|
48
|
Wen ZH, Chang YC, Jean YH. Excitatory amino acid glutamate: role in peripheral nociceptive transduction and inflammation in experimental and clinical osteoarthritis. Osteoarthritis Cartilage 2015; 23:2009-16. [PMID: 26521747 DOI: 10.1016/j.joca.2015.03.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/15/2015] [Accepted: 03/18/2015] [Indexed: 02/02/2023]
Abstract
Although a large proportion of patients with osteoarthritis (OA) show inflammation in their affected joints, the pathological role of inflammation in the development and progression of OA has yet to be clarified. Glutamate is considered an excitatory amino acid (EAA) neurotransmitter in the mammalian central nervous system (CNS). There are cellular membrane glutamate receptors and transporters for signal input modulation and termination as well as vesicular glutamate transporters (VGLUTs) for signal output through exocytotic release. Glutamate been shown to mediate intercellular communications in bone cells in a manner similar to synaptic transmission within the CNS. Glutamate-mediated events may also contribute to the pathogenesis and ongoing processes of peripheral nociceptive transduction and inflammation of experimental arthritis models as well as human arthritic conditions. This review will discuss the differential roles of glutamate signaling and blockade in peripheral neuronal and non-neuronal joint tissues, including bone remodeling systems and their potentials to impact OA-related inflammation and progression. This will serve to identify several potential targets to direct novel therapies for OA. Future studies will further elucidate the role of glutamate in the development and progression of OA, as well as its association with the clinical features of the disease.
Collapse
Affiliation(s)
- Z-H Wen
- Marine Biomedical Laboratory & Center for Translational Biopharmaceuticals, Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Taiwan
| | - Y-C Chang
- Marine Biomedical Laboratory & Center for Translational Biopharmaceuticals, Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Taiwan
| | - Y-H Jean
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan.
| |
Collapse
|
49
|
Miller RE, Tran PB, Sondoqah A, Raghu P, Ishihara S, Miller RJ, Malfait AM. The Role of Peripheral Nociceptive Neurons in the Pathophysiology of Osteoarthritis Pain. Curr Osteoporos Rep 2015; 13:318-26. [PMID: 26233284 PMCID: PMC4596062 DOI: 10.1007/s11914-015-0280-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Knee osteoarthritis is characterized by progressive damage and remodeling of all tissues in the knee joint. Pain is the main symptom associated with knee osteoarthritis. Recent clinical and pre-clinical studies have provided novel insights into the mechanisms that drive the pain associated with joint destruction. In this narrative review, we describe current knowledge regarding the changes in the peripheral and central nervous systems that occur during the progression of osteoarthritis and discuss how therapeutic interventions may provide pain relief.
Collapse
Affiliation(s)
- Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
- Department of Biochemistry, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Phuong B. Tran
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Alia Sondoqah
- Department of Biochemistry, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Padmanabhan Raghu
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
| | - Richard J. Miller
- Department of Pharmacology, Northwestern University, Lurie 8-125, 303 E. Superior St, Chicago, IL 60611
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
- Department of Biochemistry, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL 60612
- Corresponding author Anne-Marie Malfait, MD, PhD, Associate Professor of Medicine, , T: 312-563-2925, F: 312-563-2267
| |
Collapse
|
50
|
Uppstu P, Paakki C, Rosling A. In vitro hydrolysis and magnesium release of poly(d,l-lactide-co-glycolide)-based composites containing bioresorbable glasses and magnesium hydroxide. J Appl Polym Sci 2015. [DOI: 10.1002/app.42646] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Peter Uppstu
- Laboratory of Polymer Technology; Centre of Excellence in Functional Materials at Biological Interfaces; Åbo Akademi University; Biskopsgatan 8 FI-20500 Åbo Finland
| | - Charlotta Paakki
- Laboratory of Polymer Technology; Centre of Excellence in Functional Materials at Biological Interfaces; Åbo Akademi University; Biskopsgatan 8 FI-20500 Åbo Finland
| | - Ari Rosling
- Laboratory of Polymer Technology; Centre of Excellence in Functional Materials at Biological Interfaces; Åbo Akademi University; Biskopsgatan 8 FI-20500 Åbo Finland
| |
Collapse
|