1
|
Wan T, Wei A, Ding Z, Gonzalez SC, Wang J, Hou X, Luo X, He L, Song Z. Inflammation and macrophage infiltration exacerbate adult incision response by early life injury. BMC Anesthesiol 2025; 25:165. [PMID: 40211125 PMCID: PMC11983940 DOI: 10.1186/s12871-025-03029-7] [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: 02/10/2025] [Accepted: 03/26/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Neonatal hindpaw incision can evoke long-lasting changes in nociceptive processing following repeat injury in adulthood. Studies have focused on the effects and mechanisms in the spinal cord and brain, however changes in inflammation and macrophages in the periphery, especially at the site of early life injury, remain poorly defined. In this paper, we investigated the role of macrophages in the injured tissue in pain hypersensitivity caused by repeat hindpaw incisions and primed by neonatal injury. METHODS Hindpaw incision was performed in anesthetized adult rats. Among them, some had neonatal hindpaw incisions on postnatal day 3. To assess the role of inflammatory response in the priming of adult incision pain, the rats were treated with clodronate liposome, a macrophage depletion agent, and ketorolac tromethamine, the commonly used anti-inflammatory drug following surgery. Their mechanical pain sensitivity was measured via von Frey filaments. Inflammation induced by hindpaw incision was evaluated via Enzyme-linked Immunosorbent Assay, H&E, and immunofluorescence staining. The phenotypes of macrophages were examined by analyzing their surface markers by flow cytometry. RESULTS Mechanical pain hypersensitivity caused by the hindpaw incision in the adult rats was enhanced by previous neonatal injury, which also significantly increased microglial activation in the spinal dorsal horn, aggravation of inflammation, and infiltration of both M1 and M2 macrophages in damaged hindpaw tissue after the repeat incision in the adult rats on POD 5. Intraperitoneal injection of clodronate liposome alleviates nociceptive and inflammatory responses in neonatal injured rats. Intramuscular injection of ketorolac tromethamine decreased mechanical hyperalgesia and inflammatory responses primed by prior neonatal injury. CONCLUSIONS Neonatal tissue injury exacerbated mechanical hypersensitivity, infiltration, and activation of macrophages evoked by repeat hindpaw incision in adulthood.
Collapse
Affiliation(s)
- Tong Wan
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Anqi Wei
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
- Department of Anesthesiology, Guiqian International General Hospital, Guiyang, 550024, China
| | - Zhuofeng Ding
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China.
| | - Sarel Chavarria Gonzalez
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Jian Wang
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Xinran Hou
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Xiao Luo
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Liqiong He
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Zongbin Song
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China.
- Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China.
| |
Collapse
|
2
|
Moulin D, Sellam J, Berenbaum F, Guicheux J, Boutet MA. The role of the immune system in osteoarthritis: mechanisms, challenges and future directions. Nat Rev Rheumatol 2025; 21:221-236. [PMID: 40082724 DOI: 10.1038/s41584-025-01223-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 03/16/2025]
Abstract
Osteoarthritis (OA) is a chronic joint disease that has long been considered a simple wear-and-tear condition. Over the past decade, research has revealed that various inflammatory features of OA, such as low-grade peripheral inflammation and synovitis, contribute substantially to the pathophysiology of the disease. Technological advances in the past 5 years have revealed a large diversity of innate and adaptive immune cells in the joints, particularly in the synovium and infrapatellar fat pad. Notably, the presence of synovial lymphoid structures, circulating autoantibodies and alterations in memory T cell and B cell populations have been documented in OA. These data indicate a potential contribution of self-reactivity to the disease pathogenesis, blurring the often narrow and inaccurate line between chronic inflammatory and autoimmune diseases. The diverse immune changes associated with OA pathogenesis can vary across disease phenotypes, and a better characterization of their underlying molecular endotypes will be key to stratifying patients, designing novel therapeutic approaches and ultimately ameliorating treatment allocation. Furthermore, examining both articular and systemic alterations, including changes in the gut-joint axis and microbial dysbiosis, could open up novel avenues for OA management.
Collapse
Affiliation(s)
- David Moulin
- Université de Lorraine, CNRS, IMoPA, Nancy, France.
- CHRU-Nancy, IHU INFINY, Nancy, France.
| | - Jérémie Sellam
- Department of Rheumatology, Saint-Antoine Hospital, Centre de Recherche Saint-Antoine, Inserm, Sorbonne Université UMRS 938, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Francis Berenbaum
- Department of Rheumatology, Saint-Antoine Hospital, Centre de Recherche Saint-Antoine, Inserm, Sorbonne Université UMRS 938, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, INSERM, CHU Nantes, UMR1229 Regenerative Medicine and Skeleton, RMeS, Nantes, France
| | - Marie-Astrid Boutet
- Nantes Université, Oniris, INSERM, CHU Nantes, UMR1229 Regenerative Medicine and Skeleton, RMeS, Nantes, France.
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, UK.
| |
Collapse
|
3
|
Wu L, Cao X, Shen B. Development of a macrophage polarization-modulating therapeutic agent for osteoarthritis treatment. J Orthop Surg Res 2025; 20:279. [PMID: 40082923 PMCID: PMC11908040 DOI: 10.1186/s13018-025-05679-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Osteoarthritis (OA) is a common chronic degenerative joint disease. Recent studies have emphasized the crucial role of macrophages, particularly tissue-resident macrophages (Tissue-Resident Macrophages, TRMs), in the pathogenesis and progression of OA. Under physiological conditions, TRMs maintain joint homeostasis, but under various stimuli, they can polarize into pro-inflammatory M1 or anti-inflammatory M2 phenotypes. An imbalance in macrophage polarization, favoring the M1 phenotype, leads to sustained inflammation, cartilage degradation, and osteophyte formation, further exacerbating OA symptoms and structural damage. This article reviews the current understanding of macrophage polarization in OA, with a particular emphasis on the mechanisms by which TRMs influence the joint microenvironment. It explores the therapeutic potential of drug molecular platforms aimed at regulating macrophage polarization, shifting the balance from pro-inflammatory M1 to anti-inflammatory M2. The discussion includes various pharmacological agents such as corticosteroids, hyaluronic acid derivatives, monoclonal antibodies, and bioactive molecules like Squid Type II Collagen (SCII) in modulating macrophage function and slowing OA progression. Additionally, the article examines advancements in gene therapy methods targeting macrophages, utilizing nanotechnology-based delivery systems to enhance the specificity and efficiency of macrophage phenotype regulation. Targeting TRMs through sophisticated drug molecular platforms presents a promising strategy for developing novel diagnostic and therapeutic interventions for osteoarthritis.
Collapse
Affiliation(s)
- Limin Wu
- Department of Orthopaedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaotao Cao
- Early-Phase Clinical Research Unit, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Shen
- Department of Orthopaedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Malange KF, de Souza DM, Lemes JBP, Fagundes CC, Oliveira ALL, Pagliusi MO, Carvalho NS, Nishijima CM, da Silva CRR, Consonni SR, Sartori CR, Tambeli CH, Parada CA. The Implications of Brain-Derived Neurotrophic Factor in the Biological Activities of Platelet-Rich Plasma. Inflammation 2025; 48:426-446. [PMID: 38904872 DOI: 10.1007/s10753-024-02072-9] [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: 03/24/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 06/22/2024]
Abstract
Platelet-rich plasma (PRP) is a biological blood-derived therapeutic obtained from whole blood that contains higher levels of platelets. PRP has been primarily used to mitigate joint degeneration and chronic pain in osteoarthritis (OA). This clinical applicability is based mechanistically on the release of several proteins by platelets that can restore joint homeostasis. Platelets are the primary source of brain-derived neurotrophic factor (BDNF) outside the central nervous system. Interestingly, BDNF and PRP share key biological activities with clinical applicability for OA management, such as anti-inflammatory, anti-apoptotic, and antioxidant. However, the role of BDNF in PRP therapeutic activities is still unknown. Thus, this work aimed to investigate the implications of BDNF in therapeutic outcomes provided by PRP therapy in vitro and in-vivo, using the MIA-OA animal model in male Wistar rats. Initially, the PRP was characterized, obtaining a leukocyte-poor-platelet-rich plasma (LP-PRP). Our assays indicated that platelets activated by Calcium release BDNF, and suppression of M1 macrophage polarization induced by LP-PRP depends on BDNF full-length receptor, Tropomyosin Kinase-B (TrkB). OA animals were given LP-PRP intra-articular and showed functional recovery in gait, joint pain, inflammation, and tissue damage caused by MIA. Immunohistochemistry for activating transcriptional factor-3 (ATF-3) on L4/L5 dorsal root ganglia showed the LP-PRP decreased the nerve injury induced by MIA. All these LP-PRP therapeutic activities were reversed in the presence of TrkB receptor antagonist. Our results suggest that the therapeutic effects of LP-PRP in alleviating OA symptoms in rats depend on BDNF/TrkB activity.
Collapse
Affiliation(s)
- Kaue Franco Malange
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Douglas Menezes de Souza
- Department of Pharmacology, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, CEP 13083-862, Brazil
| | - Julia Borges Paes Lemes
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Cecilia Costa Fagundes
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Anna Lethicia Lima Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Marco Oreste Pagliusi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Nathalia Santos Carvalho
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Catarine Massucato Nishijima
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Cintia Rizoli Ruiz da Silva
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, CEP 13083-862, Brazil
| | - Silvio Roberto Consonni
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, CEP 13083-862, Brazil
| | - Cesar Renato Sartori
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Claudia Herrera Tambeli
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Carlos Amilcar Parada
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil.
| |
Collapse
|
5
|
Geraghty T, Ishihara S, Obeidat AM, Adamczyk NS, Hunter RS, Li J, Wang L, Lee H, Ko FC, Malfait AM, Miller RE. Acute systemic macrophage depletion in osteoarthritic mice alleviates pain-related behaviors and does not affect joint damage. Arthritis Res Ther 2024; 26:224. [PMID: 39707543 PMCID: PMC11660666 DOI: 10.1186/s13075-024-03457-9] [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: 08/17/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a painful degenerative joint disease and a leading source of years lived with disability globally due to inadequate treatment options. Neuroimmune interactions reportedly contribute to OA pain pathogenesis. Notably, in rodents, macrophages in the DRG are associated with onset of persistent OA pain. Our objective was to determine the effects of acute systemic macrophage depletion on pain-related behaviors and joint damage using surgical mouse models in both sexes. METHODS We depleted CSF1R + macrophages by treating male macrophage Fas-induced apoptosis (MaFIA) transgenic mice 8- or 16-weeks post destabilization of the medial meniscus (DMM) with AP20187 or vehicle control (10 mg/kg i.p., 1x/day for 5 days), or treating female MaFIA mice 12 weeks post partial meniscectomy (PMX) with AP20187 or vehicle control. We measured pain-related behaviors 1-3 days before and after depletion, and, 3-4 days after the last injection we examined joint histopathology and performed flow cytometry of the dorsal root ganglia (DRGs). In a separate cohort of male 8-week DMM mice or age-matched naïve vehicle controls, we conducted DRG bulk RNA-sequencing analyses after the 5-day vehicle or AP20187 treatment. RESULTS Eight- and 16-weeks post DMM in male mice, AP20187-induced macrophage depletion resulted in attenuated mechanical allodynia and knee hyperalgesia. Female mice showed alleviation of mechanical allodynia, knee hyperalgesia, and weight bearing deficits after macrophage depletion at 12 weeks post PMX. Macrophage depletion did not affect the degree of cartilage degeneration, osteophyte width, or synovitis in either sex. Flow cytometry of the DRG revealed that macrophages and neutrophils were reduced after AP20187 treatment. In addition, in the DRG, only MHCII + M1-like macrophages were significantly decreased, while CD163 + MHCII- M2-like macrophages were not affected in both sexes. DRG bulk RNA-seq revealed that Cxcl10 and Il1b were upregulated with DMM surgery compared to naïve mice, and downregulated in DMM after acute macrophage depletion. CONCLUSIONS Acute systemic macrophage depletion reduced the levels of pro-inflammatory macrophages in the DRG and alleviated pain-related behaviors in established surgically induced OA in mice of both sexes, without affecting joint damage. Overall, these studies provide insight into immune cell regulation in the DRG during OA.
Collapse
Affiliation(s)
- Terese Geraghty
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Natalie S Adamczyk
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Rahel S Hunter
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Jun Li
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Lai Wang
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Hoomin Lee
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Frank C Ko
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA.
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA.
| |
Collapse
|
6
|
Sasaki Y, Kijima K, Yoshioka K. Validity evaluation of a rat model of monoiodoacetate-induced osteoarthritis with clinically effective drugs. BMC Musculoskelet Disord 2024; 25:975. [PMID: 39609755 PMCID: PMC11605887 DOI: 10.1186/s12891-024-08083-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is the most common type of joint disease in elderly people and is characterized by pain and dysfunction. Although the monoiodoacetate (MIA)-induced model is widely used as a rodent KOA model, it is important to acknowledge the inherent limitations of this model, as the MIA model develops complex pathological phases on a daily basis. An accurate understanding of this model and the selection of an appropriate time point according to the target for drug candidates can lead to the development of clinically effective drugs. METHODS Changes in the pathological state of the MIA model were assessed via histopathological evaluation. Clodronate, a bisphosphonate, and diclofenac, a nonsteroidal anti-inflammatory drug (NSAID), were selected as models of clinically effective drugs due to their different mechanisms of action. The analgesic effects of both drugs on the MIA model were evaluated. The long-term effect of clodronate on subchondral bone osteoclasts was also evaluated. RESULTS Histopathological evaluation revealed that MIA-induced symptomatic behavior occurred in the early and late phases and was accompanied by synovial inflammation and osteoclast-related joint degeneration, respectively. Although clodronate inhibited symptomatic behavior and prevented cartilage degeneration from the early to late phases, diclofenac inhibited symptomatic behavior only in the early phase. Clodronate acted locally and inhibited the activation of subchondral osteoclasts. CONCLUSIONS Pathological changes, such as synovial changes in the early phase and knee joint degeneration in the late phase, in the MIA model are similar to those in human KOA. Our results indicate that the early phase in the MIA model is appropriate for evaluating the effects of anti-inflammatory agents such as NSAIDs and corticosteroids. The late phase in the MIA model is appropriate for evaluating the effects of drugs that act on cartilage and subchondral bone.
Collapse
Affiliation(s)
- Yamato Sasaki
- Central Research Laboratory, Research & Development Division, Seikagaku Corporation, Tateno 3-1253, Higashiyamato-shi, Tokyo, 207-0021, Japan.
| | - Kei Kijima
- Central Research Laboratory, Research & Development Division, Seikagaku Corporation, Tateno 3-1253, Higashiyamato-shi, Tokyo, 207-0021, Japan
| | - Keiji Yoshioka
- Central Research Laboratory, Research & Development Division, Seikagaku Corporation, Tateno 3-1253, Higashiyamato-shi, Tokyo, 207-0021, Japan
| |
Collapse
|
7
|
Gaigeard N, Cardon A, Le Goff B, Guicheux J, Boutet MA. Unveiling the macrophage dynamics in osteoarthritic joints: From inflammation to therapeutic strategies. Drug Discov Today 2024; 29:104187. [PMID: 39306233 DOI: 10.1016/j.drudis.2024.104187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 09/29/2024]
Abstract
Osteoarthritis (OA) is an incurable, painful, and debilitating joint disease affecting over 500 million people worldwide. The OA joint tissues are infiltrated by various immune cells, particularly macrophages, which are able to induce or perpetuate inflammation. Notably, synovitis and its macrophage component represent a target of interest for developing treatments. In this review, we describe the latest advances in understanding the heterogeneity of macrophage origins, phenotypes, and functions in the OA joint and the effect of current symptomatic therapies on these cells. We then highlight the therapeutic potential of anticytokines/chemokines, nano- and microdrug delivery, and future strategies to modulate macrophage functions in OA.
Collapse
Affiliation(s)
- Nicolas Gaigeard
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Anaïs Cardon
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Benoit Le Goff
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France
| | - Marie-Astrid Boutet
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR1229, F-44000 Nantes, France; Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, EC1M6BQ London, UK.
| |
Collapse
|
8
|
Zou Y, Liu C, Wang Z, Li G, Xiao J. Neural and immune roles in osteoarthritis pain: Mechanisms and intervention strategies. J Orthop Translat 2024; 48:123-132. [PMID: 39220678 PMCID: PMC11363721 DOI: 10.1016/j.jot.2024.07.010] [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: 04/14/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Pain is the leading symptom for most individuals with osteoarthritis (OA), a complex condition marked by joint discomfort. Recently, the dynamic interplay between the nervous and immune systems has become a focal point for understanding pain regulation. Despite this, there is still a substantial gap in our comprehensive understanding of the neuroimmune interactions and their effects on pain in OA. This review examines the bidirectional influences between immune cells and nerves in OA progression. It explores current approaches that target neuroimmune pathways, including promoting M2 macrophage polarization and specific neuronal receptor targeting, for effective pain reduction. Translational potential statement This review provides a comprehensive overview of the mechanisms underlying the interplay between the immune system and nervous system during the progression of OA, as well as their contributions to pain. Additionally, it compiles existing intervention strategies targeting neuroimmunity for the treatment of OA pain. This information offers valuable insights for researchers seeking to address the challenge of OA pain.
Collapse
Affiliation(s)
- Yi Zou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Changyu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Zhenggang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Guanghui Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| |
Collapse
|
9
|
Binvignat M, Sellam J, Berenbaum F, Felson DT. The role of obesity and adipose tissue dysfunction in osteoarthritis pain. Nat Rev Rheumatol 2024; 20:565-584. [PMID: 39112603 DOI: 10.1038/s41584-024-01143-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 08/29/2024]
Abstract
Obesity has a pivotal and multifaceted role in pain associated with osteoarthritis (OA), extending beyond the mechanistic influence of BMI. It exerts its effects both directly and indirectly through various modifiable risk factors associated with OA-related pain. Adipose tissue dysfunction is highly involved in OA-related pain through local and systemic inflammation, immune dysfunction, and the production of pro-inflammatory cytokines and adipokines. Adipose tissue dysfunction is intricately connected with metabolic syndrome, which independently exerts specific effects on OA-related pain, distinct from its association with BMI. The interplay among obesity, adipose tissue dysfunction and metabolic syndrome influences OA-related pain through diverse pain mechanisms, including nociceptive pain, peripheral sensitization and central sensitization. These complex interactions contribute to the heightened pain experience observed in individuals with OA and obesity. In addition, pain management strategies are less efficient in individuals with obesity. Importantly, therapeutic interventions targeting obesity and metabolic syndrome hold promise in managing OA-related pain. A deeper understanding of the intricate relationship between obesity, metabolic syndrome and OA-related pain is crucial and could have important implications for improving pain management and developing innovative therapeutic options in OA.
Collapse
Affiliation(s)
- Marie Binvignat
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Sorbonne University, INSERM UMRS_959, I3 Lab Immunology Immunopathology Immunotherapy, Paris, France
| | - Jérémie Sellam
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France.
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.
| | - Francis Berenbaum
- Department of Rheumatology, Sorbonne University, AP-HP Saint-Antoine hospital, Paris, France
- Sorbonne University, INSERM UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - David T Felson
- Boston University School of Medicine, Department of Medicine, Section of Rheumatology, Boston, MA, USA
| |
Collapse
|
10
|
Geraghty T, Ishihara S, Obeidat AM, Adamczyk NS, Hunter RS, Li J, Wang L, Lee H, Ko FC, Malfait AM, Miller RE. Acute systemic macrophage depletion in osteoarthritic mice alleviates pain-related behaviors and does not affect joint damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608301. [PMID: 39229102 PMCID: PMC11370380 DOI: 10.1101/2024.08.16.608301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background Osteoarthritis (OA) is a painful degenerative joint disease and a leading source of years lived with disability globally due to inadequate treatment options. Neuroimmune interactions reportedly contribute to OA pain pathogenesis. Notably, in rodents, macrophages in the DRG are associated with onset of persistent OA pain. Our objective was to determine the effects of acute systemic macrophage depletion on pain-related behaviors and joint damage using surgical mouse models in both sexes. Methods We depleted CSF1R+ macrophages by treating male macrophage Fas-induced apoptosis (MaFIA) transgenic mice 8- or 16-weeks post destabilization of the medial meniscus (DMM) with AP20187 or vehicle control (10 mg/kg i.p., 1x/day for 5 days), or treating female MaFIA mice 12 weeks post partial meniscectomy (PMX) with AP20187 or vehicle control. We measured pain-related behaviors 1-3 days before and after depletion, and, 3-4 days after the last injection we examined joint histopathology and performed flow cytometry of the dorsal root ganglia (DRGs). In a separate cohort of male 8-week DMM mice or age-matched naïve vehicle controls, we conducted DRG bulk RNA-sequencing analyses after the 5-day vehicle or AP20187 treatment. Results Eight- and 16-weeks post DMM in male mice, AP20187-induced macrophage depletion resulted in attenuated mechanical allodynia and knee hyperalgesia. Female mice showed alleviation of mechanical allodynia, knee hyperalgesia, and weight bearing deficits after macrophage depletion at 12 weeks post PMX. Macrophage depletion did not affect the degree of cartilage degeneration, osteophyte width, or synovitis in either sex. Flow cytometry of the DRG revealed that macrophages and neutrophils were reduced after AP20187 treatment. In addition, in the DRG, only MHCII+ M1-like macrophages were significantly decreased, while CD163+MHCII- M2-like macrophages were not affected in both sexes. DRG bulk RNA-seq revealed that Cxcl10 and Il1b were upregulated with DMM surgery compared to naïve mice, and downregulated in DMM after acute macrophage depletion. Conclusions Acute systemic macrophage depletion reduced the levels of pro-inflammatory macrophages in the DRG and alleviated pain-related behaviors in established surgically induced OA in mice of both sexes, without affecting joint damage. Overall, these studies provide insight into immune cell regulation in the DRG during OA.
Collapse
Affiliation(s)
- Terese Geraghty
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Shingo Ishihara
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Alia M Obeidat
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Natalie S Adamczyk
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Rahel S Hunter
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Jun Li
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Lai Wang
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Hoomin Lee
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Frank C Ko
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Anne-Marie Malfait
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Rachel E Miller
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| |
Collapse
|
11
|
Onodera T, Iwasaki K, Matsuoka M, Morioka Y, Matsubara S, Kondo E, Iwasaki N. The alterations in nerve growth factor concentration in plasma and synovial fluid before and after total knee arthroplasty. Sci Rep 2024; 14:8943. [PMID: 38637604 PMCID: PMC11026423 DOI: 10.1038/s41598-024-59685-1] [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: 02/09/2024] [Accepted: 04/13/2024] [Indexed: 04/20/2024] Open
Abstract
Total knee arthroplasty (TKA) is an effective procedure for pain relief; however, the emergence of postsurgical pain remains a concern. In this study, we investigated the production of nerve growth factor (NGF) and mediators that affect NGF production and their function in the synovial fluid and plasma after TKA. This study included 19 patients (20 knees) who had rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and knee osteoarthritis (OA) who underwent TKA, categorized into OA and non-OA groups. The levels of NGF, inflammatory cytokines, and lipid mediators were analyzed before and after surgery. The intraoperative synovial fluid NGF concentration was more than seven times higher in the non-OA group than in the OA group. The intra-articular NGF levels increased significantly by more than threefold postoperatively in the OA group but not in the non-OA group. Moreover, the levels of inflammatory cytokines and lipid mediators were increased in the synovial fluid of both groups. The intra-articular cytokines or NGF concentrations positively correlated with postoperative pain. Targeted NGF control has the potential to alleviate postsurgical pain in TKA, especially in patients with OA, emphasizing the importance of understanding NGF dynamics under different knee conditions.
Collapse
Affiliation(s)
- Tomohiro Onodera
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Sapporo, Japan.
| | - Koji Iwasaki
- Department of Functional Reconstruction for the Knee Joint, Hokkaido University, N15W7, Sapporo, Japan
| | - Masatake Matsuoka
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Sapporo, Japan
| | - Yasuhide Morioka
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd, Osaka, Japan
| | - Shinji Matsubara
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Sapporo, Japan
| | - Eiji Kondo
- Centre for Sports Medicine, Hokkaido University, N14W5, Sapporo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Sapporo, Japan
| |
Collapse
|
12
|
Blackler G, Lai-Zhao Y, Klapak J, Philpott HT, Pitchers KK, Maher AR, Fiset B, Walsh LA, Gillies ER, Appleton CT. Targeting STAT6-mediated synovial macrophage activation improves pain in experimental knee osteoarthritis. Arthritis Res Ther 2024; 26:73. [PMID: 38509602 PMCID: PMC10953260 DOI: 10.1186/s13075-024-03309-6] [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: 01/31/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Pain from osteoarthritis (OA) is one of the top causes of disability worldwide, but effective treatment is lacking. Nociceptive factors are released by activated synovial macrophages in OA, but depletion of synovial macrophages paradoxically worsens inflammation and tissue damage in previous studies. Rather than depleting macrophages, we hypothesized that inhibiting macrophage activation may improve pain without increasing tissue damage. We aimed to identify key mechanisms mediating synovial macrophage activation and test the role of STAT signaling in macrophages on pain outcomes in experimental knee OA. METHODS We induced experimental knee OA in rats via knee destabilization surgery, and performed RNA sequencing analysis on sorted synovial tissue macrophages to identify macrophage activation mechanisms. Liposomes laden with STAT1 or STAT6 inhibitors, vehicle (control), or clodronate (depletion control) were delivered selectively to synovial macrophages via serial intra-articular injections up to 12 weeks after OA induction. Treatment effects on knee and hindpaw mechanical pain sensitivity were measured during OA development, along with synovitis, cartilage damage, and synovial macrophage infiltration using histopathology and immunofluorescence. Lastly, crosstalk between drug-treated synovial tissue and articular chondrocytes was assessed in co-culture. RESULTS The majority of pathways identified by transcriptomic analyses in OA synovial macrophages involve STAT signaling. As expected, macrophage depletion reduced pain, but increased synovial tissue fibrosis and vascularization. In contrast, STAT6 inhibition in macrophages led to marked, sustained improvements in mechanical pain sensitivity and synovial inflammation without worsening synovial or cartilage pathology. During co-culture, STAT6 inhibitor-treated synovial tissue had minimal effects on healthy chondrocyte gene expression, whereas STAT1 inhibitor-treated synovium induced changes in numerous cartilage turnover-related genes. CONCLUSION These results suggest that STAT signaling is a major mediator of synovial macrophage activation in experimental knee OA. STAT6 may be a key mechanism mediating the release of nociceptive factors from macrophages and the development of mechanical pain sensitivity. Whereas therapeutic depletion of macrophages paradoxically increases inflammation and fibrosis, blocking STAT6-mediated synovial macrophage activation may be a novel strategy for OA-pain management without accelerating tissue damage.
Collapse
Affiliation(s)
- Garth Blackler
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 5B5, Canada
| | - Yue Lai-Zhao
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 5B5, Canada
- Bone and Joint Institute, Western University, London, ON, N6A 5B5, Canada
| | - Joseph Klapak
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 5B5, Canada
| | - Holly T Philpott
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 5B5, Canada
- Bone and Joint Institute, Western University, London, ON, N6A 5B5, Canada
| | - Kyle K Pitchers
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 5B5, Canada
| | - Andrew R Maher
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 5B5, Canada
| | - Benoit Fiset
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Elizabeth R Gillies
- Department of Chemistry, Western University, London, ON, N6A 5B5, Canada
- Department of Chemical and Biochemical Engineering, Western University, London, ON, N6A 5B5, Canada
| | - C Thomas Appleton
- Department of Physiology and Pharmacology, Western University, London, ON, N6A 5B5, Canada.
- Bone and Joint Institute, Western University, London, ON, N6A 5B5, Canada.
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, N6A 5C1, Canada.
| |
Collapse
|
13
|
Gordon C, Trainor J, Shah RJ, Studholme K, Gelman A, Doswell F, Sadar F, Giovannetti A, Gershenson J, Khan A, Nicholson J, Huang Z, Spurgat M, Tang SJ, Wang H, Ojima I, Carlson D, Komatsu DE, Kaczocha M. Fatty acid binding protein 5 inhibition attenuates pronociceptive cytokine/chemokine expression and suppresses osteoarthritis pain: A comparative human and rat study. Osteoarthritis Cartilage 2024; 32:266-280. [PMID: 38035977 PMCID: PMC11283882 DOI: 10.1016/j.joca.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 10/20/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is often accompanied by debilitating pain that is refractory to available analgesics due in part to the complexity of signaling molecules that drive OA pain and our inability to target these in parallel. Fatty acid binding protein 5 (FABP5) is a lipid chaperone that regulates inflammatory pain; however, its contribution to OA pain has not been characterized. DESIGN This combined clinical and pre-clinical study utilized synovial tissues obtained from subjects with end-stage OA and rats with monoiodoacetate-induced OA. Cytokine and chemokine release from human synovia incubated with a selective FABP5 inhibitor was profiled with cytokine arrays and ELISA. Immunohistochemical analyses were conducted for FABP5 in human and rat synovium. The efficacy of FABP5 inhibitors on pain was assessed in OA rats using incapacitance as an outcome. RNA-seq was then performed to characterize the transcriptomic landscape of synovial gene expression in OA rats treated with FABP5 inhibitor or vehicle. RESULTS FABP5 was expressed in human synovium and FABP5 inhibition reduced the secretion of pronociceptive cytokines (interleukin-6 [IL6], IL8) and chemokines (CCL2, CXCL1). In rats, FABP5 was upregulated in the OA synovium and its inhibition alleviated incapacitance. The transcriptome of the rat OA synovium exhibited >6000 differentially expressed genes, including the upregulation of numerous pronociceptive cytokines and chemokines. FABP5 inhibition blunted the upregulation of the majority of these pronociceptive mediators. CONCLUSIONS FABP5 is expressed in the OA synovium and its inhibition suppresses pronociceptive signaling and pain, indicating that FABP5 inhibitors may constitute a novel class of analgesics to treat OA.
Collapse
Affiliation(s)
- Chris Gordon
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - James Trainor
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Rohan J Shah
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Keith Studholme
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Alex Gelman
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Faniya Doswell
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Faisal Sadar
- Department of Orthopaedics and Rehabilitation, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Allessio Giovannetti
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Josh Gershenson
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Ayesha Khan
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - James Nicholson
- Department of Orthopaedics and Rehabilitation, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - ZeYu Huang
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Michael Spurgat
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Shao-Jun Tang
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA; Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Hehe Wang
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
| | - Iwao Ojima
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA; Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - David Carlson
- Genomics Core Facility and Institute for Advanced Computational Sciences, Stony Brook University, Stony Brook, NY, USA
| | - David E Komatsu
- Department of Orthopaedics and Rehabilitation, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Martin Kaczocha
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA; Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA; Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
14
|
Hu S, Li H, Jiang H, Liu X, Ke J, Long X. Macrophage Activation in Synovitis and Osteoarthritis of Temporomandibular Joint and Its Relationship with the Progression of Synovitis and Bone Remodeling. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:296-306. [PMID: 38245251 DOI: 10.1016/j.ajpath.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 01/22/2024]
Abstract
This study investigates the regulatory mechanisms of synovial macrophages and their polarization in the progression of temporomandibular joint osteoarthritis (TMJOA). Macrophage depletion models were established by intra-articular injection of clodronate liposomes and unloaded liposomes. TMJOA was induced by intra-articular injection of 50 μL Complete Freund's Adjuvant and the surgery of disc perforation. The contralateral joint was used as the control group. The expression of F4/80, CD86, and CD206 in the synovium was detected by immunofluorescence staining analysis. Hematoxylin and eosin staining and TMJOA synovial score were detected to show the synovial changes in rat joints after TMJOA induction and macrophage depletion. Changes in rat cartilage after TMJOA induction and macrophage depletion were shown by safranin fast green staining. The bone-related parameters of rats' joints were evaluated by micro-computed tomography analysis. The TMJOA model induced by Complete Freund's Adjuvant injection and disc perforation aggravated synovial hyperplasia and showed a significant up-regulation of expression of F4/80-, CD86-, and CD206-positive cells. F4/80, CD86, and CD206 staining levels were significantly decreased in macrophage depletion rats, whereas the synovitis score further increased and cartilage and subchondral bone destruction was slightly aggravated. Macrophages were crucially involved in the progression of TMJOA, and macrophage depletion in TMJOA synoviocytes promoted synovitis and cartilage destruction.
Collapse
Affiliation(s)
- Shiyu Hu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China; The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Province Key Laboratory of Oral Biomedicine. Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang, China
| | - Huimin Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Henghua Jiang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xin Liu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jin Ke
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Xing Long
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
15
|
De Roover A, Escribano-Núñez A, Monteagudo S, Lories R. Fundamentals of osteoarthritis: Inflammatory mediators in osteoarthritis. Osteoarthritis Cartilage 2023; 31:1303-1311. [PMID: 37353140 DOI: 10.1016/j.joca.2023.06.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/25/2023]
Abstract
OBJECTIVES As more has become known of the pathophysiology of osteoarthritis (OA), evidence that inflammation plays a critical role in its development and progression has accumulated. Here, we aim to review current knowledge of the complex inflammatory network in the OA joint. DESIGN This narrative review is presented in three main sections: local inflammation, systemic inflammation, and therapeutic implications. We focused on inflammatory mediators and their link to OA structural changes in the joint. RESULTS OA is characterized by chronic and low-grade inflammation mediated mostly by the innate immune system, which results in cartilage degradation, bone remodeling and synovial changes. Synovitis is regarded as an OA characteristic and associated with increased severity of symptoms and joint dysfunction. However, the articular cartilage and the subchondral bone also produce several pro-inflammatory mediators thus establishing a complex interplay between the different tissues of the joint. In addition, systemic low-grade inflammation induced by aging, obesity and metabolic syndrome can contribute to OA development and progression. The main inflammatory mediators associated with OA include cytokines, chemokines, growth factors, adipokines, and neuropeptides. CONCLUSIONS Future research is needed to deeper understand the molecular pathways mediating the inflammation in OA to provide new therapeutics that target these pathways, or to repurpose existing drugs.
Collapse
Affiliation(s)
- Astrid De Roover
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Ana Escribano-Núñez
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Silvia Monteagudo
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Rik Lories
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; Division of Rheumatology, University Hospitals Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
16
|
Tong S, Fang S, Ying K, Chen W. Titanium particles inhibit bone marrow mesenchymal stem cell osteogenic differentiation through the MAPK signaling pathway. FEBS Open Bio 2023; 13:1699-1708. [PMID: 37483149 PMCID: PMC10476562 DOI: 10.1002/2211-5463.13678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/23/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023] Open
Abstract
Metallic implants have great application in clinical orthopedics. Implants wear out in vivo due to long-term mechanical loading. The formation of wear debris is one of the long-term complications of prosthesis. In the case of artificial joint replacement in particular, aseptic loosening is the most common reason for secondary revision surgery. Previous studies suggested that wear debris caused aseptic loosening mainly by promoting osteolysis around the prosthesis. In this study, titanium particles, the most commonly used particles in clinical practice, were selected to simulate wear debris and explore the influence of titanium particles on osteogenic differentiation of mesenchymal stem cells. Our results show that titanium particles can significantly inhibit osteogenic differentiation in a dose-dependent manner. While engaged in preliminary exploration of the underlying mechanisms, we found that titanium particles significantly affect phosphorylation of ERK1/2, a key component of MAPK signaling. This suggests that the MAPK signaling pathway is involved in the inhibition of osteogenic differentiation by titanium particles.
Collapse
Affiliation(s)
- Shunyi Tong
- Department of Orthopaedic SurgeryLanxi People's HospitalChina
| | - Sanhua Fang
- Department of Orthopaedic SurgeryLanxi People's HospitalChina
| | - Kangjie Ying
- Department of Orthopaedic SurgeryLanxi People's HospitalChina
| | - Weiwei Chen
- Department of Orthopaedic SurgeryLanxi People's HospitalChina
| |
Collapse
|
17
|
Morioka N, Tsuruta M, Masuda N, Yamano K, Nakano M, Kochi T, Nakamura Y, Hisaoka-Nakashima K. Inhibition of Nuclear Receptor Related Orphan Receptor γ Ameliorates Mechanical Hypersensitivity Through the Suppression of Spinal Microglial Activation. Neuroscience 2023; 526:223-236. [PMID: 37419402 DOI: 10.1016/j.neuroscience.2023.07.002] [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: 01/29/2023] [Revised: 06/21/2023] [Accepted: 07/01/2023] [Indexed: 07/09/2023]
Abstract
Microglia are crucial in induction of central sensitization under a chronic pain state. Therefore, control of microglial activity is important to ameliorate nociceptive hypersensitivity. The nuclear receptor retinoic acid related orphan receptor γ (RORγ) contributes to the regulation of inflammation-related gene transcription in some immune cells, including T cells and macrophages. Their role and function in regulation of microglial activity and nociceptive transduction have yet to be elaborated. Treatment of cultured microglia with specific RORγ inverse agonists, SR2211 or GSK2981278, significantly suppressed lipopolysaccharide (LPS)-induced mRNA expression of pronociceptive molecules interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor (TNF). Intrathecal treatment of naïve male mice with LPS markedly induced mechanical hypersensitivity and upregulation of ionized calcium-biding adaptor molecule (Iba1) in the spinal dorsal horn, indicating microglial activation. In addition, intrathecal treatment with LPS significantly induced mRNA upregulation of IL-1β and IL-6 in the spinal dorsal horn. These responses were prevented by intrathecal pretreatment with SR2211. In addition, intrathecal administration of SR2211 significantly ameliorated established mechanical hypersensitivity and upregulation of Iba1 immunoreactivity in the spinal dorsal horn of male mice following peripheral sciatic nerve injury. The current findings demonstrate that blockade of RORγ in spinal microglia exerts anti-inflammatory effects, and that RORγ may be an appropriate target for the treatment of chronic pain.
Collapse
Affiliation(s)
- Norimitsu Morioka
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan.
| | - Maho Tsuruta
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Nao Masuda
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kiichi Yamano
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Manaya Nakano
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Takahiro Kochi
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| |
Collapse
|
18
|
Hayashi K, Lesnak JB, Plumb AN, Rasmussen LA, Sluka KA. P2X7-NLRP3-Caspase-1 signaling mediates activity-induced muscle pain in male but not female mice. Pain 2023; 164:1860-1873. [PMID: 36930885 PMCID: PMC10363217 DOI: 10.1097/j.pain.0000000000002887] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/23/2022] [Indexed: 03/19/2023]
Abstract
ABSTRACT We developed an animal model of activity-induced muscle pain that is dependent on local macrophage activation and release of interleukin-1β (IL-1β). Activation of purinergic type 2X (P2X) 7 receptors recruits the NOD-like receptor protein (NLRP) 3 and activates Caspase-1 to release IL-1β. We hypothesized that pharmacological blockade of P2X7, NLRP3, and Caspase-1 would prevent development of activity-induced muscle pain in vivo and release of IL-1β from macrophages in vitro. The decrease in muscle withdrawal thresholds in male, but not female, mice was prevented by the administration of P2X7, NLRP3, and Caspase-1 inhibitors before induction of the model, whereas blockade of IL-1β before induction prevented muscle hyperalgesia in both male and female mice. Blockade of P2X7, NLRP3, Capsase-1, or IL-1β 24 hours, but not 1 week, after induction of the model alleviated muscle hyperalgesia in male, but not female, mice. mRNA expression of P2X7, NLRP3, Caspase-1, and IL-1β from muscle was increased 24 hours after induction of the model in both male and female mice. Using multiplex, increases in IL-1β induced by combining adenosine triphosphate with pH 6.5 in lipopolysaccharide-primed male and female macrophages were significantly lower with the presence of inhibitors of P2X7 (A740003), NLRP3 (MCC950), and Caspase-1 (Z-WEHD-FMK) when compared with the vehicle. The current data suggest the P2X7/NLRP3/Caspase-1 pathway contributed to activity-induced muscle pain initiation and early maintenance phases in male but not female, and not in late maintenance phases in male mice.
Collapse
Affiliation(s)
- Kazuhiro Hayashi
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Joseph B. Lesnak
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Ashley N. Plumb
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Lynn A. Rasmussen
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Kathleen A. Sluka
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
19
|
Murakami T, Ishida T, Tanaka S, Nakayama J, Tsurugizawa T, Takahashi Y, Kato F, Kawamata M. Inflammation and subsequent nociceptor sensitization in the bone marrow are involved in an animal model of osteoarthritis pain. Life Sci 2023; 324:121736. [PMID: 37121542 DOI: 10.1016/j.lfs.2023.121736] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/02/2023]
Abstract
AIMS This study aimed to determine whether pathological changes in the bone marrow cause Osteoarthritis (OA) pain based on magnetic resonance imaging (MRI), immunohistochemistry, and electrophysiology. MAIN METHODS Adjuvant-induced arthritis (AIA) was achieved by injecting 150 μL of complete Freund's adjuvant into the right knee joints of male Sprague-Dawley rats. AIA rats were compared with saline-injected rats. KEY FINDINGS AIA significantly induced mechanical hyperalgesia and spontaneous pain in the right hind paw 1-14 days after induction. Intratibial injection of 50 μL of 1 % lidocaine significantly suppressed AIA-induced mechanical hyperalgesia (p = 0.0001) and spontaneous pain (p = 0.0006) 3 days after induction. In T2-weighted MRI, AIA induced high-signal intensity within the proximal tibial metaphysis, and the mean T2 values in this area significantly increased on days 3 (p = 0.0043) and 14 (p = 0.0012) after induction. AIA induced intraosseous edema and significantly increased the number of intraosseous granulocytes on days 3 (p < 0.0001) and 14 (p < 0.0001) after induction. The electrophysiological study on days 3-7 after induction showed significantly increased spontaneous firing rates (p = 0.0166) and evoked responses to cutaneous stimuli (brush, p < 0.0001; pinching, p = 0.0359) in the right hind paw plantar surface and intratibial stimuli (p = 0.0002) in wide-dynamic-range neurons of the spinal dorsal horn. SIGNIFICANCE Intraosseous changes caused by OA induce hypersensitivity in the sensory afferents innervating bone marrow may be involved in OA pain. Novel bone marrow-targeted therapies could be beneficial for treating OA pain.
Collapse
Affiliation(s)
- Toru Murakami
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takashi Ishida
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan.
| | - Satoshi Tanaka
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Tomokazu Tsurugizawa
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Yukari Takahashi
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| | - Fusao Kato
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| | - Mikito Kawamata
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| |
Collapse
|
20
|
Zhang S, Wang L, Kang Y, Wu J, Zhang Z. Nanomaterial-based Reactive Oxygen Species Scavengers for Osteoarthritis Therapy. Acta Biomater 2023; 162:1-19. [PMID: 36967052 DOI: 10.1016/j.actbio.2023.03.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/17/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Reactive oxygen species (ROS) play distinct but important roles in physiological and pathophysiological processes. Recent studies on osteoarthritis (OA) have suggested that ROS plays a crucial role in its development and progression, serving as key mediators in the degradation of the extracellular matrix, mitochondrial dysfunction, chondrocyte apoptosis, and OA progression. With the continuous development of nanomaterial technology, the ROS-scavenging ability and antioxidant effects of nanomaterials are being explored, with promising results already achieved in OA treatment. However, current research on nanomaterials as ROS scavengers for OA is relatively non-uniform and includes both inorganic and functionalized organic nanomaterials. Although the therapeutic efficacy of nanomaterials has been reported to be conclusive, there is still no uniformity in the timing and potential of their use in clinical practice. This paper reviews the nanomaterials currently used as ROS scavengers for OA treatment, along with their mechanisms of action, with the aim of providing a reference and direction for similar studies, and ultimately promoting the early clinical use of nanomaterials for OA treatment. STATEMENT OF SIGNIFICANCE: Reactive oxygen species (ROS) play an important role in the pathogenesis of osteoarthritis (OA). Nanomaterials serving as promising ROS scavengers have gained increasing attention in recent years. This review provides a comprehensive overview of ROS production and regulation, as well as their role in OA pathogenesis. Furthermore, this review highlights the applications of various types of nanomaterials as ROS scavengers in OA treatment and their mechanisms of action. Finally, the challenges and future prospects of nanomaterial-based ROS scavengers in OA therapy are discussed.
Collapse
|
21
|
Lattimer J, Fabiani M, Gaschen L, Aulakh K, Selting K. Clinical effectiveness and safety of intraarticular administration of a 117m Tin radiocolloid (Synovetin OA TM ) for treatment of early and intermediate grade osteoarthritis of the elbow in a dose finding study conducted in 44 dogs. Vet Radiol Ultrasound 2023; 64:351-359. [PMID: 36507585 DOI: 10.1111/vru.13198] [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: 04/26/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis of the elbow joint secondary to elbow dysplasia is common in dogs. Intraarticular radionuclide injection is thought to suppress both synovitis and inflammatory pain mediators in the joint which are not directly addressed by current treatments. This dose-finding investigation was a longitudinal, prospective, experimental parallel group, post-test study with repeated measures. Forty-four dogs, with low to intermediate-grade osteoarthritis, received a single injection into their most clinically affected elbow joint and were randomized into three treatment cohorts; 37 MBq, 64.75 MBq, or 92.5 MBq (normalized to the body surface area of a 22 kg dog) of 117m Sn radiocolloid. Dogs were assessed monthly by owners, using the canine Brief Pain Inventory (cBPI), and at 1, 3, 6, 9, and 12 months intervals by investigators. Positive responses to treatment were observed by both owners and clinicians in all dose groups with the medium dose group having the highest and most durable response rate based on cBPI scores. The results of this study support the use of 117m Sn radiocolloid as a primary treatment of osteoarthritis in low to intermediate-grade osteoarthritis of the canine elbow.
Collapse
Affiliation(s)
- Jimmy Lattimer
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, USA
| | | | - Lori Gaschen
- Veterinary Clinical Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Karanvir Aulakh
- Veterinary Clinical Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Kimberly Selting
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana-Champaign, Illinois, USA
| |
Collapse
|
22
|
GLX351322, a Novel NADPH Oxidase 4 Inhibitor, Attenuates TMJ Osteoarthritis by Inhibiting the ROS/MAPK/NF- κB Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:1952348. [PMID: 36756301 PMCID: PMC9902131 DOI: 10.1155/2023/1952348] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/27/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023]
Abstract
As a degenerative disease in joints, temporomandibular joint osteoarthritis (TMJOA) is characterized by progressive cartilage degradation, subchondral bone remodeling, and chronic synovitis, severely undermining functions and quality of life in patients. NADPH oxidase 4 (NOX4) contributes to reactive oxygen species (ROS) production and inflammatory pathway activation in osteoarthritis, which has attracted increasing attention in research in recent years. GLX351322 (GLX), a novel NOX4 inhibitor, exerts a protective effect on chondrocytes. However, whether it has a therapeutic effect on ROS production and inflammatory responses in synovial macrophages remains to be evaluated. In this study, we examined the effect of GLX on LPS-induced ROS production and inflammatory responses in vitro and on complete Freund's adjuvant (CFA)-induced TMJ inflammation in vivo. We found that GLX could depress LPS-induced intracellular ROS production and inflammatory response without cytotoxicity by inhibiting the ROS/MAPK/NF-κB signaling pathways. In line with in vitro observations, GLX markedly attenuated the synovial inflammatory reaction in the TMJ, thus protecting the condylar structure from severe damage. Taken together, our results suggest that GLX intervention or NOX4 inhibition is a promising curative strategy for TMJOA and other inflammatory diseases.
Collapse
|
23
|
Koya Y, Tanaka H, Yoshimi E, Takeshita N, Morita S, Morio H, Mori K, Fushiki H, Kamohara M. A novel anti-NGF PEGylated Fab' provides analgesia with lower risk of adverse effects. MAbs 2023; 15:2149055. [PMID: 36458900 PMCID: PMC9721442 DOI: 10.1080/19420862.2022.2149055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Nerve growth factor (NGF) has emerged as a key driver of pain perception in several chronic pain conditions, including osteoarthritis (OA), and plays an important role in the generation and survival of neurons. Although anti-NGF antibodies improve pain control and physical function in patients with clinical chronic pain conditions, anti-NGF IgGs are associated with safety concerns such as effects on fetal and postnatal development and the risk of rapidly progressive osteoarthritis. To overcome these drawbacks, we generated a novel anti-NGF PEGylated Fab' antibody. The anti-NGF PEGylated Fab' showed specific binding to and biological inhibitory activity against NGF, and analgesic effects in adjuvant-induced arthritis model mice in a similar manner to an anti-NGF IgG. In collagen-induced arthritis model mice, the anti-NGF PEGylated Fab' showed higher accumulation in inflamed foot pads than the anti-NGF IgG. In pregnant rats and non-human primates, the anti-NGF PEGylated Fab' was undetectable in fetuses, while the anti-NGF IgG was detected and caused abnormal postnatal development. The PEGylated Fab' and IgG also differed in their ability to form immune complexes in vitro. Additionally, while both PEGylated Fab' and IgG showed analgesic effects in sodium monoiodoacetate-induced arthritic model rats, their effects on edema were surprisingly quite different. While the anti-NGF IgG promoted edema over time, the anti-NGF PEGylated Fab' did not. The anti-NGF PEGylated Fab' (ASP6294) may thus be a potential therapeutic candidate with lower risk of adverse effects for various diseases in which NGF is involved such as OA and chronic back pain.
Collapse
Affiliation(s)
- Yukari Koya
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan,CONTACT Yukari Koya Astellas Pharma Inc, 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Hirotsugu Tanaka
- Incubation Lab, Astellas Innovation Management LLC, Cambridge, MA, USA
| | - Eiji Yoshimi
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | | | - Shuji Morita
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | - Hiroki Morio
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | - Kanako Mori
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | - Hiroshi Fushiki
- Drug Discovery Research, Astellas Pharma Inc, Tsukuba, Japan
| | | |
Collapse
|
24
|
Phytochemical Evaluation and Anti-Inflammatory Potential of Miconia albicans (Sw.) Triana Extracts. Molecules 2022; 27:molecules27185954. [PMID: 36144693 PMCID: PMC9500825 DOI: 10.3390/molecules27185954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
The plant Miconia albicans (Sw.) Triana has been popularly used in Brazil to treat chronic inflammatory disturbances, such as osteoarthritis. This disease affects 250 million people worldwide, and is associated with intense pain and loss of articular function. There is a lack of information about the phytochemistry and bioactivity of M. albicans. Therefore, this study determined the chemical composition of some extracts and evaluated their cytotoxicity, along with their antioxidant and anti-inflammatory, activities using in vitro models. Aqueous and ethanolic extracts were prepared. Afterwards, a liquid–liquid partition was developed using chloroform, ethyl acetate, and n-butanol. The extracts were characterized by LC–MS, and their biological activities were evaluated on epithelial cells (Vero), tumoral hepatic cells (Hep-G2), and THP-1 macrophages. LC–MS analyses identified several flavonoids in all fractions, such as quercetin, myricetin, and their glycosides. The crude extracts and n-butanol fractions did not present cytotoxicity to the cells. The non-toxic fractions presented significant antioxidant activity when evaluated in terms of DPPH scavenging activity, lipid peroxidation, and ROS inhibition. THP-1 macrophages treated with the n-butanol fraction (250 µg/mL) released fewer pro-inflammatory cytokines, even in the presence of LPS. In the future, it will be necessary to identify the phytochemicals that are responsible for anti-inflammatory effects for the discovery of new drugs. In vivo studies on M. albicans extracts are still required to confirm their possible mechanisms of action.
Collapse
|
25
|
Phenotype Diversity of Macrophages in Osteoarthritis: Implications for Development of Macrophage Modulating Therapies. Int J Mol Sci 2022; 23:ijms23158381. [PMID: 35955514 PMCID: PMC9369350 DOI: 10.3390/ijms23158381] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/23/2022] [Accepted: 07/23/2022] [Indexed: 02/04/2023] Open
Abstract
Chronic inflammation is implicated in numerous human pathologies. In particular, low-grade inflammation is currently recognized as an important mechanism of osteoarthritis (OA), at least in some patients. Among the signs of the inflammatory process are elevated macrophage numbers detected in the OA synovium compared to healthy controls. High macrophage counts also correlate with clinical symptoms of the disease. Macrophages are central players in the development of chronic inflammation, pain, cartilage destruction, and bone remodeling. However, macrophages are also involved in tissue repair and remodeling, including cartilage. Therefore, reduction of macrophage content in the joints correlates with deleterious effects in OA models. Macrophage population is heterogeneous and dynamic, with phenotype transitions being induced by a variety of stimuli. In order to effectively use the macrophage inflammatory circuit for treatment of OA, it is important to understand macrophage heterogeneity and interactions with surrounding cells and tissues in the joint. In this review, we discuss functional phenotypes of macrophages and specific targeting approaches relevant for OA treatment development.
Collapse
|
26
|
Liu Y, Lu T, Liu Z, Ning W, Li S, Chen Y, Ge X, Guo C, Zheng Y, Wei X, Wang H. Six macrophage-associated genes in synovium constitute a novel diagnostic signature for osteoarthritis. Front Immunol 2022; 13:936606. [PMID: 35967352 PMCID: PMC9368762 DOI: 10.3389/fimmu.2022.936606] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022] Open
Abstract
Background Synovial macrophages play important roles in the formation and progression of osteoarthritis (OA). This study aimed to explore the biological and clinical significance of macrophage-associated genes (MAGs) in OA. Methods The OA synovial gene expression profiles GSE89408 and GSE82107 were obtained from the GEO database. Single-sample gene set enrichment analysis (ssGSEA) and GSEA were employed to decipher differences in immune infiltration and macrophage-associated biological pathways, respectively. Protein–protein interaction (PPI) network analysis and machine learning were utilized to establish a macrophage-associated gene diagnostic signature (MAGDS). RT-qPCR was performed to test the expression of key MAGs in murine models. Results OA synovium presented high levels of immune infiltration and activation of macrophage-associated biological pathways. A total of 55 differentially expressed MAGs were identified. Using PPI analysis and machine learning, a MAGDS consisting of IL1B, C5AR1, FCGR2B, IL10, IL6, and TYROBP was established for OA diagnosis (AUC = 0.910) and molecular pathological evaluation. Patients with high MAGDS scores may possess higher levels of immune infiltration and expression of matrix metalloproteinases (MMPs), implying poor biological alterations. The diagnostic value of MAGDS was also validated in an external cohort (AUC = 0.886). The expression of key MAGs was validated in a murine model using RT-qPCR. Additionally, a competitive endogenous RNA network was constructed to reveal the potential posttranscriptional regulatory mechanisms. Conclusions We developed and validated a MAGDS model with the ability to accurately diagnose and characterize biological alterations in OA. The six key MAGs may also be latent targets for immunoregulatory therapy.
Collapse
Affiliation(s)
- Yiying Liu
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Taoyuan Lu
- Department of Cerebrovascular Disease, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenhua Ning
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siying Li
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanru Chen
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyong Ge
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Youyang Zheng
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangyang Wei
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Haiming Wang, ; Xiangyang Wei,
| | - Haiming Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Medical College of Zhengzhou University of Industrial technology, Zhengzhou, China
- *Correspondence: Haiming Wang, ; Xiangyang Wei,
| |
Collapse
|
27
|
Bai LK, Su YZ, Wang XX, Bai B, Zhang CQ, Zhang LY, Zhang GL. Synovial Macrophages: Past Life, Current Situation, and Application in Inflammatory Arthritis. Front Immunol 2022; 13:905356. [PMID: 35958604 PMCID: PMC9361854 DOI: 10.3389/fimmu.2022.905356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Inflammatory arthritis is an inflammatory disease that involves the joints and surrounding tissues. Synovial hyperplasia often presents when joints become inflamed due to immune cell infiltration. Synovial membrane is an important as well as a highly specific component of the joint, and its lesions can lead to degeneration of the joint surface, causing pain and joint disability or affecting the patients’ quality of life in severe cases. Synovial macrophages (SMs) are one of the cellular components of the synovial membrane, which not only retain the function of macrophages to engulf foreign bodies in the joint cavity, but also interact with synovial fibroblasts (SFs), T cells, B cells, and other inflammatory cells to promote the production of a variety of pro-inflammatory cytokines and chemokines, such as TNF-α, IL-1β, IL-8, and IL-6, which are involved in the pathogenic process of inflammatory arthritis. SMs from different tissue sources have differently differentiated potentials and functional expressions. This article provides a summary on studies pertaining to SMs in inflammatory arthritis, and explores their role in its treatment, in order to highlight novel treatment modalities for the disease.
Collapse
Affiliation(s)
- Lin-Kun Bai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Ya-Zhen Su
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Xue-Xue Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Bing Bai
- First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Cheng-Qiang Zhang
- Fifth Hospital of Shanxi Medical University, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
| | - Li-Yun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Gai-Lian Zhang
- Fifth Hospital of Shanxi Medical University, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
- *Correspondence: Gai-Lian Zhang,
| |
Collapse
|
28
|
Gao W, Shen L, Long DD, Pan TT, Wang D, Chai XQ, Hu SS. Angiotensin II type 2 receptor pharmacological agonist, C21, reduces the inflammation and pain hypersensitivity in mice with joint inflammatory pain. Int Immunopharmacol 2022; 110:108921. [PMID: 35724606 DOI: 10.1016/j.intimp.2022.108921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 11/15/2022]
Abstract
Primary and secondary hyperalgesia develop in response to chronic joint inflammation due to peripheral and central mechanisms. Synovial macrophage and spinal microglia are involved in pain sensitization in arthritis. The level of angiotensin II type 2 receptor (AT2R) is related to the severity of arthritis. This study aimed to determine the role of AT2R in primary and secondary hyperalgesia in joint inflammatory pain in mice. After intra-articular CFA injection, primary hyperalgesia in the ipsilateral knee joint was measured by pressure application meter and gait analysis, secondary hypersensitivity in ipsilateral hind-paw was measured by von-Frey and Hargreaves tests following a combination of global AT2R-deficient (Agtr2-/-) mice and AT2R pharmacological agonist C21. Synovial macrophage and spinal microglia were collected for flow cytometry. Morphological reconstruction of microglia was detected by immunostaining. AT2R expression was investigated by quantitative polymerase chain reaction and western blot. Neuronal hyperactivity was evaluated by c-Fos and CGRP immunostaining. We found that pain hypersensitivity and synovial inflammation in Agtr2-/- mice were significantly exacerbated compared with wild-type mice; conversely, systemically administrated C21 attenuated both of the symptoms. Additionally, spinal microglia were activated, and an abundant increase of spinal AT2R was expressed on activated microglia in response to peripheral joint inflammation. Intrathecally-administrated C21 reversed the secondary hypersensitivity, accompanied by alleviation of spinal microglial activation, spinal neuronal hyperactivity, and calcitonin gene-related peptide content. These findings revealed a beneficial role of AT2R activating stimulation against pain hypersensitivity in joint inflammatory pain via direct modulation of synovial macrophage and spinal microglial activity.
Collapse
Affiliation(s)
- Wei Gao
- Anhui Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Liang Shen
- Department of Anesthesiology, Anhui Provincial Hospital Affiliated to Medical University, Hefei 230036, China
| | - Dan-Dan Long
- Department of Anesthesiology, Anhui Provincial Hospital Affiliated to Medical University, Hefei 230036, China
| | - Ting-Ting Pan
- Department of Anesthesiology, Anhui Provincial Hospital Affiliated to Medical University, Hefei 230036, China
| | - Di Wang
- Department of Anesthesiology, Anhui Provincial Hospital Affiliated to Medical University, Hefei 230036, China
| | - Xiao-Qing Chai
- Anhui Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Shan-Shan Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
29
|
Gao Y, Mei C, Chen P, Chen X. The contribution of neuro-immune crosstalk to pain in the peripheral nervous system and the spinal cord. Int Immunopharmacol 2022; 107:108700. [DOI: 10.1016/j.intimp.2022.108700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/23/2022] [Accepted: 03/10/2022] [Indexed: 12/16/2022]
|
30
|
Ohashi Y, Uchida K, Fukushima K, Satoh M, Koyama T, Tsuchiya M, Saito H, Uchiyama K, Takahira N, Inoue G, Takaso M. Correlation between CD163 expression and resting pain in patients with hip osteoarthritis: Possible contribution of CD163+ monocytes/macrophages to pain pathogenesis. J Orthop Res 2022; 40:1365-1374. [PMID: 34370345 DOI: 10.1002/jor.25157] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/15/2021] [Accepted: 07/30/2021] [Indexed: 02/04/2023]
Abstract
Expression of CD163, a scavenger receptor specifically expressed by monocytes and macrophages, is elevated in the synovial tissue of patients with knee osteoarthritis (OA) compared with healthy controls. However, the association between CD163 expression in the synovium and pain in OA patients is unclear. We investigated the correlation between synovial CD163 expression and resting and active pain levels in patients with hip osteoarthritis (HOA). To investigate the possible contribution of CD163+ subsets to pain pathogenesis, we compared pain-related cytokine expression and M1/M2 macrophage marker expression in CD163+ and CD163- cells. We performed flow cytometric analysis to study the CD163+ cell population. We also examined pain-related cytokine expression and M1/M2 macrophage marker expression on CD163+ CD14high and CD163+ CD14low cells using cell sorting. Synovial CD163 expression significantly correlated with resting pain levels (p = 0.006; R = 0.321), but not active pain levels (p = 0.155; R = 0.169). Expression of the M1 macrophage marker CD80 was significantly higher in CD163+ than CD163- cells (p = 0.010), as was the expression of M2 macrophage markers CD206 and IL10 (CD206, p = 0.014; IL10, p = 0.005), and TNFA and IL1B (TNFA, p = 0.002; IL1B, p = 0.001). TNFA expression was significantly higher in CD163+ CD14low than CD163+ CD14high cells, while IL1B, IL10, and CD206 expression were comparable among these subsets. Our findings suggest that CD163 expression is associated with higher resting pain scores. As TNF-α plays a role in the pain process, CD163+ CD14low cells expressing TNFA may be a potent contributor to the pathogenesis of resting pain in HOA.
Collapse
Affiliation(s)
- Yoshihisa Ohashi
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| | - Kensuke Fukushima
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| | - Masashi Satoh
- Department of Immunology, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| | - Tomohisa Koyama
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| | - Maho Tsuchiya
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| | - Hiroki Saito
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| | - Katsufumi Uchiyama
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| | - Naonobu Takahira
- Department of Rehabilitation, Kitasato University School of Allied Health Sciences, Sagamihara City, Kanagawa, Japan
| | - Gen Inoue
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara City, Kanagawa, Japan
| |
Collapse
|
31
|
Valdrighi N, Vago JP, Blom AB, van de Loo FA, Blaney Davidson EN. Innate Immunity at the Core of Sex Differences in Osteoarthritic Pain? Front Pharmacol 2022; 13:881500. [PMID: 35662714 PMCID: PMC9160873 DOI: 10.3389/fphar.2022.881500] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022] Open
Abstract
Osteoarthritis (OA) is a progressive whole-joint disease; no disease-modifying drugs are currently available to stop or slow its process. Symptoms alleviation is the only treatment option. OA is the major cause of chronic pain in adults, with pain being the main symptom driving patients to seek medical help. OA pathophysiology is closely associated with the innate immune system, which is also closely linked to pain mediators leading to joint pain. Pain research has shown sex differences in the biology of pain, including sexually dimorphic responses from key cell types in the innate immune system. Not only is OA more prevalent in women than in men, but women patients also show worse OA outcomes, partially due to experiencing more pain symptoms despite having similar levels of structural damage. The cause of sex differences in OA and OA pain is poorly understood. This review provides an overview of the involvement of innate immunity in OA pain in joints and in the dorsal root ganglion. We summarize the emerging evidence of sex differences regarding innate immunity in OA pain. Our main goal with this review was to provide a scientific foundation for future research leading to alternative pain relief therapies targeting innate immunity that consider sex differences. This will ultimately lead to a more effective treatment of pain in both women and men.
Collapse
|
32
|
Vasconcelos DP, Jabangwe C, Lamghari M, Alves CJ. The Neuroimmune Interplay in Joint Pain: The Role of Macrophages. Front Immunol 2022; 13:812962. [PMID: 35355986 PMCID: PMC8959978 DOI: 10.3389/fimmu.2022.812962] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/15/2022] [Indexed: 12/29/2022] Open
Abstract
Chronic pain associated with joint disorders, such as rheumatoid arthritis (RA), osteoarthritis (OA) and implant aseptic loosening (AL), is a highly debilitating symptom that impacts mobility and quality of life in affected patients. The neuroimmune crosstalk has been demonstrated to play a critical role in the onset and establishment of chronic pain conditions. Immune cells release cytokines and immune mediators that can activate and sensitize nociceptors evoking pain, through interaction with receptors in the sensory nerve terminals. On the other hand, sensory and sympathetic nerve fibers release neurotransmitters that bind to their specific receptor expressed on surface of immune cells, initiating an immunomodulatory role. Macrophages have been shown to be key players in the neuroimmune crosstalk. Moreover, macrophages constitute the dominant immune cell population in RA, OA and AL. Importantly, the targeting of macrophages can result in anti-nociceptive effects in chronic pain conditions. Therefore, the aim of this review is to discuss the nature and impact of the interaction between the inflammatory response and nerve fibers in these joint disorders regarding the genesis and maintenance of pain. The role of macrophages is highlighted. The alteration in the joint innervation pattern and the inflammatory response are also described. Additionally, the immunomodulatory role of sensory and sympathetic neurotransmitters is revised.
Collapse
Affiliation(s)
- Daniela P Vasconcelos
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto- Associação, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Clive Jabangwe
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto- Associação, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto- Associação, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Instituto Ciências Biomédicas Abel Salazar, Universidade de Porto, Porto, Portugal
| | - Cecília J Alves
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto- Associação, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| |
Collapse
|
33
|
Wang D, Chai XQ, Hu SS, Pan F. Joint synovial macrophages as a potential target for intra-articular treatment of osteoarthritis-related pain. Osteoarthritis Cartilage 2022; 30:406-415. [PMID: 34861384 DOI: 10.1016/j.joca.2021.11.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/19/2021] [Accepted: 11/22/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis is the most common form of joint disease and is one of the leading causes of chronic pain. Given the multi-factorial nature, numerous efforts have been made to clarify the multiple factors impacting the pain symptoms and joint pathology, including synovial macrophages in particular. Accumulating evidence from studies involving human participants and experimental animal models suggests that accumulating macrophages in synovial tissue are implicated in peripherally mediated pain sensitization of affected joints in osteoarthritis. Crosstalk between synovial macrophages and the innervating primary nociceptive neurons is thought to contribute to this facilitated pain processing by the peripheral nervous system. Due to high plasticity and complexity of synovial macrophages in the joint, safe therapies targeting single cells or molecules are currently lacking. Using advanced technologies (such as single-cell RNA sequencing and mass cytometry), studies have shown that diverse subpopulations of synovial macrophages exist in the distinct synovial microenvironments of specific osteoarthritis subtypes. Considerable progress has been made in delineating the molecular mechanisms of various subsets of synovial macrophages in the development of osteoarthritis. To develop a novel intra-articular treatment paradigm targeting synovial macrophages, we have summarized in this review the recent advances in identifying the functional consequences of synovial macrophage sub-populations and understanding of the molecular mechanisms driving macrophage-mediated remodeling.
Collapse
Affiliation(s)
- D Wang
- Pain Clinic, Department of Anesthesiology, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei 230001, China.
| | - X-Q Chai
- Pain Clinic, Department of Anesthesiology, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei 230001, China.
| | - S-S Hu
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, PR China (Anhui Medical University), Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China.
| | - F Pan
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS 7000, Australia.
| |
Collapse
|
34
|
Paglia DN, Kanjilal D, Kadkoy Y, Moskonas S, Wetterstrand C, Lin A, Galloway J, Tompson J, Culbertson MD, O’Connor JP. Naproxen treatment inhibits articular cartilage loss in a rat model of osteoarthritis. J Orthop Res 2021; 39:2252-2259. [PMID: 33274763 PMCID: PMC8175455 DOI: 10.1002/jor.24937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/27/2020] [Accepted: 12/01/2020] [Indexed: 02/04/2023]
Abstract
The effects of naproxen, a nonsteroidal anti-inflammatory drug (NSAID), on articular cartilage degeneration in female Sprague-Dawley rats was examined. Osteoarthritis (OA) was induced by destabilization of the medial meniscus (DMM) in each knee. Rats were treated with acetaminophen (60 mg/kg), naproxen (8 mg/kg), or 1% carboxymethylcellulose (placebo) by oral gavage twice daily for 3 weeks, beginning 2 weeks after surgery. OA severity was assessed by histological Osteoarthritis Research Society International (OARSI) scoring and by measuring proximal tibia cartilage depth using contrast enhanced µCT (n = 6 per group) in specimens collected at 2, 5, and 7 weeks after surgery as well as on pristine knees. Medial cartilage OARSI scores from the DMM knees of naproxen-treated rats were statistically lower (i.e., better) than the medial cartilage OARSI scores from the DMM knees of placebo-treated rats at 5-weeks (8.7 ± 3.6 vs. 13.2 ± 2.4, p = 0.025) and 7-weeks (9.5 ± 1.2 vs. 12.5 ± 2.5, p = 0.024) after surgery. At 5 weeks after DMM surgery, medial articular cartilage depth in the proximal tibia specimens was significantly greater in the naproxen (1.78 ± 0.26 mm, p = 0.005) and acetaminophen (1.94 ± 0.12 mm, p < 0.001) treated rats as compared with placebo-treated rats (1.34 ± 0.24 mm). However, at 7 weeks (2 weeks after drug withdrawal), medial articular cartilage depth for acetaminophen-treated rats (1.36 ± 0.29 mm) was significantly reduced compared with specimens from the naproxen-treated rats (1.88 ± 0.14 mm; p = 0.004). The results indicate that naproxen treatment reduced articular cartilage degradation in the rat DMM model during and after naproxen treatment.
Collapse
Affiliation(s)
| | | | - Yazan Kadkoy
- Rutgers-New Jersey Medical School, Newark, NJ, USA
| | | | | | - Anthony Lin
- Rutgers-New Jersey Medical School, Newark, NJ, USA
| | | | | | | | - J. Patrick O’Connor
- Rutgers-New Jersey Medical School, Newark, NJ, USA
- School of Graduate Studies, Newark, NJ, USA
| |
Collapse
|
35
|
Li S, Wang H, Zhang Y, Qiao R, Xia P, Kong Z, Zhao H, Yin L. COL3A1 and MMP9 Serve as Potential Diagnostic Biomarkers of Osteoarthritis and Are Associated With Immune Cell Infiltration. Front Genet 2021; 12:721258. [PMID: 34512730 PMCID: PMC8430221 DOI: 10.3389/fgene.2021.721258] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/02/2021] [Indexed: 12/19/2022] Open
Abstract
Background Osteoarthritis (OA) is one of the most common age-related degenerative diseases. In recent years, some studies have shown that pathological changes in the synovial membrane occur earlier than those in the cartilage in OA. However, the molecular mechanism of synovitis in the pathological process of OA has not been elucidated. This study aimed to identify novel biomarkers associated with OA and to emphasize the role of immune cells in the pathogenesis of OA. Methods Microarray datasets were obtained from the Gene Expression Omnibus (GEO) and ArrayExpress databases and were then analyzed using R software. To determine differential immune cell subtype infiltration, the CIBERSORT deconvolution algorithm was used. Quantitative reverse transcription PCR (qRT-PCR) was used to determine the relative expressions of selected genes. Besides, Western blotting was used to assess the protein expression levels in osteoarthritic chondrocytes. Results After analyzing the database profiles, two potential biomarkers, collagen type 3 alpha 1 chain (COL3A1), and matrix metalloproteinase 9 (MMP9), associated with OA were discovered, which were confirmed by qRT-PCR and Western blotting. Specifically, the results revealed that, as the concentration of IL-1β increased, so did the gene and protein expression levels of COL3A1 and MMP9. Conclusion The findings provide valuable information and direction for future research into novel targets for OA immunotherapy and diagnosis and aids in the discovery of the underlying biological mechanisms of OA pathogenesis.
Collapse
Affiliation(s)
- Shushan Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haitao Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Renqiu Qiao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peige Xia
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiheng Kong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongbo Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Yin
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
PTX-3 Secreted by Intra-Articular-Injected SMUP-Cells Reduces Pain in an Osteoarthritis Rat Model. Cells 2021; 10:cells10092420. [PMID: 34572070 PMCID: PMC8466059 DOI: 10.3390/cells10092420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are accessible, abundantly available, and capable of regenerating; they have the potential to be developed as therapeutic agents for diseases. However, concerns remain in their further application. In this study, we developed a SMall cell+Ultra Potent+Scale UP cell (SMUP-Cell) platform to improve whole-cell processing, including manufacturing bioreactors and xeno-free solutions for commercialization. To confirm the superiority of SMUP-Cell improvements, we demonstrated that a molecule secreted by SMUP-Cells is capable of polarizing inflammatory macrophages (M1) into their anti-inflammatory phenotype (M2) at the site of injury in a pain-associated osteoarthritis (OA) model. Lipopolysaccharide-stimulated macrophages co-cultured with SMUP-Cells expressed low levels of M1-phenotype markers (CD11b, tumor necrosis factor-α, interleukin-1α, and interleukin-6), but high levels of M2 markers (CD163 and arginase-1). To identify the paracrine action underlying the anti-inflammatory effect of SMUP-Cells, we employed a cytokine array and detected increased levels of pentraxin-related protein-3 (PTX-3). Additionally, PTX-3 mRNA silencing was applied to confirm PTX-3 function. PTX-3 silencing in SMUP-Cells significantly decreased their therapeutic effects against monosodium iodoacetate (MIA)-induced OA. Thus, PTX-3 expression in injected SMUP-Cells, applied as a therapeutic strategy, reduced pain in an OA model.
Collapse
|
37
|
Soga M, Izumi T, Nanchi I, Horita N, Yamamoto M, Kawasaki S, Ogawa K, Fujita M, Morioka Y. Suppression of joint pain in transient receptor potential vanilloid 4 knockout rats with monoiodoacetate-induced osteoarthritis. Pain Rep 2021; 6:e951. [PMID: 34396019 PMCID: PMC8357256 DOI: 10.1097/pr9.0000000000000951] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/16/2021] [Accepted: 07/01/2021] [Indexed: 11/25/2022] Open
Abstract
Supplemental Digital Content is Available in the Text. Knee joint pain in osteoarthritis model rats is caused by the sensitization of transient receptor potential vanilloid 4 in the dorsal root ganglion neurons Introduction: Transient receptor potential vanilloid 4 (TRPV4) modulates osteoarthritic (OA) pain in animal models. However, the pathophysiological function of TRPV4 in regulating OA pain remains poorly understood. Methods: We developed TRPV4-knockout (TRPV4-KO) rats and assessed the effects of Trpv4 gene deficiency in a monoiodoacetate (MIA)-induced OA pain model (MIA rats) by examining pain-related behavior, pathological changes, and electrophysiological changes in dorsal root ganglion (DRG) neurons. The changes detected in TRPV4-KO rats were confirmed in wild-type rats using a TRPV4 antagonist. Results: Transient receptor potential vanilloid 4–KO rats showed the same pain threshold as wild-type rats for thermal or pressure stimuli under normal conditions. Trpv4 gene deletion did not suppress the development of osteoarthritis pathologically in MIA rats. However, the OA-related mechanical pain behaviors observed in MIA rats, including decreased grip strength, increased mechanical allodynia, and reduced weight-bearing on the ipsilateral side, were completely suppressed in TRPV4-KO rats. The DRG neurons in wild-type but not TRPV4-KO MIA rats were depolarized with increased action potentials. Transient receptor potential vanilloid 4 antagonist treatments recapitulated the effects of genetic Trpv4 deletion. Conclusion: Transient receptor potential vanilloid 4 was sensitized in the DRG neurons of MIA rats and played a critical role in the development of OA pain. These results suggest that the inhibition of TRPV4 might be a novel potent analgesic strategy for treating OA pain.
Collapse
Affiliation(s)
- Masahiko Soga
- Department of Pharmacological Efficacy Evaluation, Shionogi TechnoAdvance Research Co. Ltd., Toyonaka, Japan
| | - Takaya Izumi
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Isamu Nanchi
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Narumi Horita
- Department of Pharmacological Efficacy Evaluation, Shionogi TechnoAdvance Research Co. Ltd., Toyonaka, Japan
| | - Miyuki Yamamoto
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Shiori Kawasaki
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Koichi Ogawa
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Masahide Fujita
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Yasuhide Morioka
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| |
Collapse
|
38
|
Tazawa R, Kenmoku T, Uchida K, Arendt-Nielsen L, Nagura N, Nakawaki M, Matsumoto T, Inoue G, Takeuchi H, Jimbo T, Nakazawa T, Fukuda M, Takaso M. Increased nerve growth factor expression in the synovial tissues of patients with rotator cuff tears. Mol Pain 2021; 17:17448069211021252. [PMID: 34074169 PMCID: PMC8175831 DOI: 10.1177/17448069211021252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background Rotator cuff tears (RCTs) are often associated with severe shoulder pain. Non-steroidal anti-inflammatory drugs, not recommended for long-term use, do not effectively manage RCT-induced pain, resulting in reduced quality of life. To improve management, a better understanding of the fundamental properties of RCT pain is needed. Here, we aimed to compare the expression levels of nerve growth factor (NGF) and cyclooxygenase-2 (COX-2) mRNA in the synovial tissues of patients with RCT-induced pain and patients with non-painful recurrent shoulder dislocation (RSD). Methods The study included 32 patients with RCT who underwent arthroscopic rotator cuff repair and 28 patients with non-painful RSD who underwent arthroscopic Bankart repair. Synovial tissue samples were harvested from subacromial bursa and rotator interval of RCT patients and from the rotator interval of RSD patients. Samples were analyzed quantitatively expression levels for NGF and COX2 mRNA and NGF protein. Results NGF mRNA and protein levels were significantly higher in the rotator interval of RCT patients than in the rotator interval of RSD patients (p = 0.0017, p = 0.012, respectively), while COX2 mRNA levels did not differ significantly between the two patient groups. In RCT patients, COX2 mRNA was more highly expressed in the rotator interval than in the subacromial bursa (p = 0.038), whereas the mRNA and protein levels of NGF did not differ between the two tissues. The expression of NGF mRNA in the synovium of the rotator interval was significantly correlated with the numeric rating scale of pain (ρ = 0.38, p = 0.004). Conclusion NGF mRNA and protein levels were elevated in patients with painful RCT compared with those in patients with non-painful RSD, whereas COX-2 levels were comparable in the two patient groups. These findings provide insights into novel potential strategies for clinical management of RCT.
Collapse
Affiliation(s)
- Ryo Tazawa
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Tomonori Kenmoku
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Lars Arendt-Nielsen
- Department of Health Science and Technology, Center for Neuroplasticity and Pain, School of Medicine, Aalborg University, Aalborg, Denmark
| | - Naoshige Nagura
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Mitsufumi Nakawaki
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Gen Inoue
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hiroto Takeuchi
- Department of Rehabilitation, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takenori Jimbo
- Department of Rehabilitation, Kitasato University School of Medicine, Sagamihara, Japan
| | - Toshiyuki Nakazawa
- Department of Rehabilitation, Kitasato University School of Medicine, Sagamihara, Japan
| | - Michinari Fukuda
- Department of Rehabilitation, Kitasato University School of Medicine, Sagamihara, Japan
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
39
|
Kawasaki S, Soga M, Sakurai Y, Nanchi I, Yamamoto M, Imai S, Takahashi T, Tsuno N, Asaki T, Morioka Y, Fujita M. Selective blockade of transient receptor potential vanilloid 4 reduces cyclophosphamide-induced bladder pain in mice. Eur J Pharmacol 2021; 899:174040. [PMID: 33737012 DOI: 10.1016/j.ejphar.2021.174040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/30/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective cation channel activated by various physical stimuli such as cell swelling and shear stress. TRPV4 is expressed in bladder sensory nerves and epithelium, and its activation produces urinary dysfunction in rodents. However, there have been few reports regarding its involvement in bladder pain. Therefore, we investigated whether TRPV4 is involved in bladder pain in mouse cystitis model. Intraperitoneal injection of cyclophosphamide (CYP; 300 mg/kg) produced mechanical hypersensitivity in the lower abdomen associated with a severe inflammatory bladder in mice. The mechanical threshold was reversed significantly in Trpv4-knockout (KO) mice. Repeated injections of CYP (150 mg/kg) daily for 4 days provoked mild bladder inflammation and persistent mechanical hypersensitivity in mice. Trpv4-KO mice prevented a reduction of the mechanical threshold without an alteration in bladder inflammation. A selective TRPV4 antagonist also reversed the mechanical threshold in chronic cystitis mice. Although expression of Trpv4 was unchanged in the bladders of chronic cystitis mice, the level of phosphorylated TRPV4 was increased significantly. These results suggest involvement of TRPV4 in bladder pain of cystitis mice. A TRPV4 antagonist might be useful for patients with irritable bladder pain such as those with interstitial cystitis/painful bladder syndrome.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animals
- Behavior, Animal/drug effects
- Cells, Cultured
- Cyclophosphamide
- Cystitis, Interstitial/chemically induced
- Cystitis, Interstitial/metabolism
- Cystitis, Interstitial/physiopathology
- Cystitis, Interstitial/prevention & control
- Disease Models, Animal
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Nociceptive Pain/chemically induced
- Nociceptive Pain/metabolism
- Nociceptive Pain/physiopathology
- Nociceptive Pain/prevention & control
- Pain Threshold/drug effects
- Phosphorylation
- TRPV Cation Channels/antagonists & inhibitors
- TRPV Cation Channels/genetics
- TRPV Cation Channels/metabolism
- Urinary Bladder/drug effects
- Urinary Bladder/metabolism
- Urinary Bladder/physiopathology
- Mice
Collapse
Affiliation(s)
- Shiori Kawasaki
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Masahiko Soga
- Animal Production Technology for Animal Models, Shionogi Techno Advance Research Co. Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Yusuke Sakurai
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Isamu Nanchi
- Laboratory for Innovative Therapy Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Miyuki Yamamoto
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Sunao Imai
- Laboratory for Advanced Medicine Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Tatsuya Takahashi
- Laboratory for Advanced Medicine Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Naoki Tsuno
- API R&D Laboratory, CMC R&D Division, Shionogi & Co., Ltd., 1-3, Kuise terajima 2-chome, Amagasaki, Hyogo, 660-0813, Japan
| | - Toshiyuki Asaki
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Yasuhide Morioka
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Masahide Fujita
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan.
| |
Collapse
|
40
|
Pan TT, Pan F, Gao W, Hu SS, Wang D. Involvement of Macrophages and Spinal Microglia in Osteoarthritis Pain. Curr Rheumatol Rep 2021; 23:29. [PMID: 33893883 DOI: 10.1007/s11926-021-00997-w] [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] [Accepted: 02/23/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Chronic pain in osteoarthritis (OA) is characterized by pain sensitization, which involves both peripheral and central mechanisms. Studies suggest synovial macrophage and spinal microglia are implicated in pain sensitization in OA. We, therefore, reviewed the evidence of whether synovial macrophage and spinal microglia facilitated pain sensitization at diverse levels and how this event occurred in OA. RECENT FINDINGS Peripherally, joint inflammation is now believed to be a source of OA-related pain. Synovial macrophages accumulate in OA inflamed synovium and display a pro-inflammatory phenotype. Abundant macrophage-derived pro-inflammatory cytokines and other pain-causing substance facilitate hyperexcitation of primary sensory neuron in OA-related pain. Thus, activated synovial macrophage was considered a predictor for phenotyping of OA pain clinically. In response to affected joint-derived strong nociception, aberrant neuronal excitability is often associated with the hyperactivity of microglia in the spinal dorsal horn, thereby leading to central sensitization. Hyperactivity of synovial macrophage and spinal microglia underlies the mechanisms of pain sensitization at the peripheral and central level in OA. This concept provides not only a clinically relevant strategy for identifying the phenotype of OA-related pain but also has the potential to develop individualized interventions for OA, particularly in those patients with hyperactivity of macrophage and microglia.
Collapse
Affiliation(s)
- Ting-Ting Pan
- Department of Anesthesiology, Pain Clinic, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Feng Pan
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, TAS, 7000, Australia
| | - Wei Gao
- Department of Anesthesiology, Pain Clinic, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Shan-Shan Hu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
| | - Di Wang
- Department of Anesthesiology, Pain Clinic, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
41
|
Neuroimmune interactions and osteoarthritis pain: focus on macrophages. Pain Rep 2021; 6:e892. [PMID: 33981927 PMCID: PMC8108586 DOI: 10.1097/pr9.0000000000000892] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/01/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022] Open
Abstract
Bidirectional interactions between the immune system and the nervous system are increasingly appreciated as playing a pathogenic role in chronic pain. Unraveling the mechanisms by which inflammatory pain is mediated through communication between nerves and immune cells may lead to exciting new strategies for therapeutic intervention. In this narrative review, we focus on the role of macrophages in the pathogenesis of osteoarthritis (OA) pain. From regulating homeostasis to conducting phagocytosis, and from inducing inflammation to resolving it, macrophages are plastic cells that are highly adaptable to their environment. They rely on communicating with the environment through cytokines, growth factors, neuropeptides, and other signals to respond to inflammation or injury. The contribution of macrophages to OA joint damage has garnered much attention in recent years. Here, we discuss how macrophages may participate in the initiation and maintenance of pain in OA. We aim to summarize what is currently known about macrophages in OA pain and identify important gaps in the field to fuel future investigations.
Collapse
|
42
|
Yuan G, Yang S, Gautam M, Luo W, Yang S. Macrophage regulator of G-protein signaling 12 contributes to inflammatory pain hypersensitivity. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:448. [PMID: 33850845 PMCID: PMC8039686 DOI: 10.21037/atm-20-5729] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Pain is a predominant symptom in rheumatoid arthritis (RA) patients that results from joint inflammation and is augmented by central sensitization. Regulator of G-protein signaling 12 (RGS12) is the largest protein in the RGS protein family and plays a key role in the development of inflammation. This study investigated the regulation of RGS12 in inflammatory pain and explored the underlying mechanisms and potential RA pain targets. Methods Macrophage-specific RGS12-deficient (LysM-Cre+;RGS12fl/fl) mice were generated by mating RGS12fl/fl mice with LysM-Cre+ transgenic mice. Collagen antibody-induced arthritis (CAIA) models were induced in LysM-Cre+;RGS12fl/fl mice by the administration of a cocktail of five monoclonal antibodies and LPS. Mouse nociception was examined using the von Frey and heat plate tests. Primary macrophages and RAW264.7 cells were used to analyze the regulatory function and mechanism of RGS12 in vitro. The expression and function of RGS12 and COX2 (cyclooxygenase 2) were determined by real-time PCR, ELISA, and luciferase assays. Results Ablation of RGS12 in macrophages decreased pain-related phenotypes, such as paw swelling, the clinical score, and the inflammatory score, in the CAIA model. LysM-Cre+;RGS12fl/fl mice displayed increased resistance to thermal and mechanical stimulation from day 3 to day 9 during CAIA, indicating the inhibition of inflammatory pain. Overexpression of COX2 and PGE2 in macrophages enhanced RGS12 expression, and PGE2 regulated RGS12 expression through the G-protein-coupled receptors EP2 and EP4. Furthermore, RGS12 or the RGS12 PTB domain strengthened the transcriptional regulation of COX2 by NF-κB, whereas inhibiting NF-κB suppressed RGS12-mediated regulation of COX2 in macrophages. Conclusions Our results demonstrate that the deletion of RGS12 in macrophages attenuates inflammatory pain, which is likely due to impaired regulation of the COX2/PGE2 signaling pathway.
Collapse
Affiliation(s)
- Gongsheng Yuan
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuting Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mayank Gautam
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuying Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.,The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
43
|
da Silva Nascimento FG, de Souza Ferreira Bringel PH, Maia FWS, Lima CPC, Alves RC, Feitosa JPA, Mota MRL, Assreuy AMS, Castro RR. Galactomannan of Delonix regia seeds reduces nociception and morphological damage in the rat model of osteoarthritis induced by sodium monoiodoacetate. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:491-501. [PMID: 33057779 DOI: 10.1007/s00210-020-01996-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/08/2020] [Indexed: 01/06/2023]
Abstract
This study investigated the effects of the protein-free galactomannan obtained from Delonix regia seeds (GM-DR) in an experimental osteoarthritis (OA) model. GM-DR was obtained from water-homogenized endosperms by collection of the supernatant and precipitation with ethanol. The remaining proteins in the galactomannan were removed by alkaline hydrolysis. Weight average molar mass (Mw) of the galactomannan was estimated in 5.8 × 105 g mol-1, presenting mannose:galactose ratio of 2.39:1. Rats received sodium monoiodoacetate (OA groups, 1 mg/25 μL) or saline (sham group) in the right tibio-tarsal joint. GM-DR (30-300 μg) was administered by intra-articular route at days 14 and 21 after OA induction. Hypernociception was evaluated daily by the measurement of the mechanical threshold required to cause joint flexion and paw withdrawal reflex. The 56-day animal groups were euthanized for joint histopahological analysis using the OARSI score system. Lower doses of GM-DR (30 and 100 μg) promoted antinociception from day 15 until the endpoint at day 56. Joint damage was reduced by GM-DR administration (100 μg) in OA-subjected animals, compared to the vehicle-treated OA group (5.9 ± 1.8 vs 19.0 ± 1.8, respectively, p < 0.05). Conclusion: Both antinociception and damage reduction suggest that Delonix regia galactomannan is a promising approach for osteoarthritis therapy.
Collapse
Affiliation(s)
| | | | - Francisco Wildson Silva Maia
- Superior Institute of Biomedical Sciences, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, 60714-903, Brazil
| | - Carlos Pinheiro Chagas Lima
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Av. Mister Hull, s/n, Fortaleza, CE, 60451-970, Brazil
| | - Rômulo Couto Alves
- Catarinian Federal Institute of Education, Science and Technology, R. Vigário Frei João, 550, Luzerna, SC, 89609-000, Brazil
| | - Judith Pessoa Andrade Feitosa
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Av. Mister Hull, s/n, Fortaleza, CE, 60451-970, Brazil
| | - Mário Rogério Lima Mota
- Department of Dental Clinic, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceará, Street Alexandre Baraúna, 949, Fortaleza, CE, 60430-160, Brazil
| | - Ana Maria Sampaio Assreuy
- Superior Institute of Biomedical Sciences, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, 60714-903, Brazil
| | - Rondinelle Ribeiro Castro
- Superior Institute of Biomedical Sciences, State University of Ceará, Av. Dr. Silas Munguba, 1700, Fortaleza, CE, 60714-903, Brazil.
| |
Collapse
|
44
|
Liu JA, Yu J, Cheung CW. Immune Actions on the Peripheral Nervous System in Pain. Int J Mol Sci 2021; 22:ijms22031448. [PMID: 33535595 PMCID: PMC7867183 DOI: 10.3390/ijms22031448] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Pain can be induced by tissue injuries, diseases and infections. The interactions between the peripheral nervous system (PNS) and immune system are primary actions in pain sensitizations. In response to stimuli, nociceptors release various mediators from their terminals that potently activate and recruit immune cells, whereas infiltrated immune cells further promote sensitization of nociceptors and the transition from acute to chronic pain by producing cytokines, chemokines, lipid mediators and growth factors. Immune cells not only play roles in pain production but also contribute to PNS repair and pain resolution by secreting anti-inflammatory or analgesic effectors. Here, we discuss the distinct roles of four major types of immune cells (monocyte/macrophage, neutrophil, mast cell, and T cell) acting on the PNS during pain process. Integration of this current knowledge will enhance our understanding of cellular changes and molecular mechanisms underlying pain pathogenies, providing insights for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Jessica Aijia Liu
- Correspondence: (J.A.L.); (C.W.C.); Tel.: +852-2255-3303 (J.A.L. & C.W.C.); Fax: +852-2855-1654 (J.A.L. & C.W.C.)
| | | | - Chi Wai Cheung
- Correspondence: (J.A.L.); (C.W.C.); Tel.: +852-2255-3303 (J.A.L. & C.W.C.); Fax: +852-2855-1654 (J.A.L. & C.W.C.)
| |
Collapse
|
45
|
Ratneswaran A, Kapoor M. Osteoarthritis year in review: genetics, genomics, epigenetics. Osteoarthritis Cartilage 2021; 29:151-160. [PMID: 33227439 DOI: 10.1016/j.joca.2020.11.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/23/2020] [Accepted: 11/13/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In this review, we have highlighted advances in genetics, genomics and epigenetics in the field of osteoarthritis (OA) over the past year. METHODS A literature search was performed using PubMed and the criteria: "osteoarthritis" and one of the following terms "genetic(s), genomic(s), epigenetic(s), epigenomic(s), noncoding RNA, microRNA, long noncoding RNA, lncRNA, circular RNA, RNA sequencing, single cell sequencing, or DNA methylation between April 1, 2019 and April 30, 2020. RESULTS We identified 653 unique publications, many studies spanned multiple search terms. We summarized advances relating to evolutionary genetics, pain, ethnicity specific risk factors, functional studies of gene variants, and interactions between coding and non-coding RNAs in OA pathogenesis. CONCLUSIONS Studies have identified variants contributing to OA susceptibility, candidate biomarkers for diagnosis and prognosis, as well as promising therapeutic candidates. Validation in multiple cohorts, multi-omics strategies, and machine learning aided computational analyses have all contributed to the strength of published literature. Open access data-sets, greater sample sizes to capture broader populations and understanding disease mechanisms by investigating the interactions between multiple tissue types will further aid in progress towards understanding and curing OA.
Collapse
Affiliation(s)
- A Ratneswaran
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada; Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - M Kapoor
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada; Krembil Research Institute, University Health Network, Toronto, ON, Canada; Department of Surgery, Faculty of Medicine, University of Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
46
|
An Emerging Target in the Battle against Osteoarthritis: Macrophage Polarization. Int J Mol Sci 2020; 21:ijms21228513. [PMID: 33198196 PMCID: PMC7697192 DOI: 10.3390/ijms21228513] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is one of the most prevalent chronic joint diseases worldwide, which causes a series of problems, such as joint pain, muscle atrophy, and joint deformities. Benefiting from some advances in the clinical treatment of OA, the quality of life of OA patients has been improved. However, the clinical need for more effective treatments for OA is still very urgent. Increasing findings show that macrophages are a critical breakthrough in OA therapy. Stimulated by different factors, macrophages are differentiated into two phenotypes: the pro-inflammatory M1 type and anti-inflammatory M2 type. In this study, various therapeutic reagents for macrophage-dependent OA treatment are summarized, including physical stimuli, chemical compounds, and biological molecules. Subsequently, the mechanisms of action of various approaches to modulating macrophages are discussed, and the signaling pathways underlying these treatments are interpreted. The NF-κB signaling pathway plays a vital role in the occurrence and development of macrophage-mediated OA, as NF-κB signaling pathway agonists promote the occurrence of OA, whereas NF-κB inhibitors ameliorate OA. Besides, several signaling pathways are also involved in the process of OA, including the JNK, Akt, MAPK, STAT6, Wnt/β-catenin, and mTOR pathways. In summary, macrophage polarization is a critical node in regulating the inflammatory response of OA. Reagents targeting the polarization of macrophages can effectively inhibit inflammation in the joints, which finally relieves OA symptoms. Our work lays the foundation for the development of macrophage-targeted therapeutic molecules and helps to elucidate the role of macrophages in OA.
Collapse
|
47
|
Li B, Jing L, Jia L, Qian T, Jianyi C, Zhongsheng H, Xiaohong Z, Guowei C. Acupuncture reduces pain in rats with osteoarthritis by inhibiting MCP2/CCR2 signaling pathway. Exp Biol Med (Maywood) 2020; 245:1722-1731. [PMID: 32878462 DOI: 10.1177/1535370220952342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Acupuncture is an emerging alternative therapy that has been beneficial for the pain of osteoarthritis (OA). However, the underlying mechanism of protective effect remains unclear. MCP1/CCR2 axis can be stimulated in various periods of OA, and we hypothesize that acupuncture may treat OA by regulating the MCP1/CCR2 axis. This study aimed to explore the effect of acupuncture at points ST35 and ST36 on the effects of hyperalgesia and cartilage in OA rats including the expression of chemokines, nerve growth factor (NGF), and inflammatory-related proteins. OA was induced in male Sprague-Dawley rats by anterior cruciate ligament transection at the right knee. The first acupuncture intervention was performed on the seventh day after surgery and once a day for seven weeks. The knee-pain-related behaviors, histology, and related protein were examined in this study. We have found that electroacupuncture at ST35 and ST36 can significantly alleviate the hyperalgesia and cartilage degeneration as well as reducing nerve sprouting in OA knee joint. Moreover, acupuncture treatment may inhibit the MCP1/CCR2 axis as well as down-regulate inflaming factor and NGF in cartilage and synovial tissue. The data presented here indicate that acupuncture exerts a protective effect against hyperalgesia and cartilage degeneration, and the mechanism might involve in chemokines and NGF pathway.
Collapse
Affiliation(s)
- Bocun Li
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Jing
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Jia
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan 430061, China
| | - Tan Qian
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chen Jianyi
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan 430061, China
| | - Huang Zhongsheng
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan 430061, China
| | - Zhou Xiaohong
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan 430061, China
| | - Cai Guowei
- Department of Acupuncture, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
48
|
Therapeutic Manipulation of Macrophages Using Nanotechnological Approaches for the Treatment of Osteoarthritis. NANOMATERIALS 2020; 10:nano10081562. [PMID: 32784839 PMCID: PMC7466380 DOI: 10.3390/nano10081562] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is the most common joint pathology causing severe pain and disability. Macrophages play a central role in the pathogenesis of OA. In the joint microenvironment, macrophages with an M1-like pro-inflammatory phenotype induce chronic inflammation and joint destruction, and they have been correlated with the development and progression of the disease, while the M2-like anti-inflammatory macrophages support the recovery of the disease, promoting tissue repair and the resolution of inflammation. Nowadays, the treatment of OA in the clinic relies on systemic and/or intra-articular administration of anti-inflammatory and pain relief drugs, as well as surgical interventions for the severe cases (i.e., meniscectomy). The disadvantages of the pharmacological therapy are related to the chronic nature of the disease, requiring prolonged treatments, and to the particular location of the pathology in joint tissues, which are separated anatomical compartments with difficult access for the drugs. To overcome these challenges, nanotechnological approaches have been investigated to improve the delivery of drugs toward macrophages into the diseased joint. This strategy may offer advantages by reducing off-target toxicities and improving long-term therapeutic efficacy. In this review, we describe the nanomaterial-based approaches designed so far to directly or indirectly manipulate macrophages for the treatment of osteoarthritis.
Collapse
|
49
|
Mechanism of aspirin-induced inhibition on the secondary hyperalgesia in osteoarthritis model rats. Heliyon 2020; 6:e03963. [PMID: 32478188 PMCID: PMC7248669 DOI: 10.1016/j.heliyon.2020.e03963] [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: 12/04/2019] [Revised: 04/24/2020] [Accepted: 05/06/2020] [Indexed: 01/23/2023] Open
Abstract
Aims The daily activity of osteoarthritis (OA) patients is limited by chronic pain and central sensitization. Although non-steroidal anti-inflammatory drugs (NSAIDs) and acetaminophen are the first-line drugs for the treatment of OA-related pain, their efficacy on central sensitization remains unclear. In the present study, we evaluated the effect of acetylsalicylic acid (ASA, Aspirin) using an OA model induced by monosodium iodoacetate (MIA), which has a similar disease progression to human OA. Main methods Secondary hyperalgesia was assessed at the plantar surface of the hind paw by Von Frey test. We evaluated the expression of acid-sensing ion channel 3 (ASIC3) in dorsal root ganglia and that of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the spinal cord, which may cause secondary hyperalgesia in OA, by immunohistochemical analysis and real-time qPCR. Key findings The administration of ASA attenuated secondary hyperalgesia at 1–3 weeks after MIA, while celecoxib, a selective cyclooxygenase (COX)-2 inhibitor, failed to attenuate secondary hyperalgesia at week 2 after MIA injection, suggesting that ASA exerts its analgesic effect through a COX-2-independent pathway. Immunohistochemical analysis of the dorsal root ganglia indicated that ASA reduced the expression of ASIC3 during OA progression. Expression of TNF-α mRNA, but not IL-1β mRNA, in the spinal cord following MIA injection was suppressed by ASA administration. Significance These findings suggest that ASA may have the ability to attenuate secondary hyperalgesia through suppression of ASIC3 and/or TNF-α expression. ASA is therefore a clinically useful analgesic drug for treatment of secondary hyperalgesia in OA.
Collapse
|
50
|
An update on targets for treating osteoarthritis pain: NGF and TRPV1. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2020; 6:129-145. [PMID: 34178580 DOI: 10.1007/s40674-020-00146-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Purpose of review a)Osteoarthritis (OA) is the most common form of arthritis, and pain is the primary symptom of the disease, yet analgesic options for treating OA pain remain limited. In this review, we aimed to give an update on the current clinical and preclinical studies targeting two pathways that are being investigated for treating OA pain: the nerve growth factor (NGF) pathway and the transient receptor potential vanilloid-1 (TRPV1) pathway. Recent findings b)Antibodies against NGF, small molecule inhibitors of TrkA, TRPV1 agonists, and TRPV1 antagonists are all in different stages of clinical and pre-clinical testing for the treatment of OA pain. NGF antibodies have shown efficacy in the primary endpoints tested compared to placebo, however, rapidly progressive OA has been consistently observed in a subset of patients and the cause remains unclear. TRPV1 agonists have also demonstrated reduced pain with no serious adverse events - the most common adverse events include a burning or warming sensation upon administration. Summary c)Targeting the NGF and TRPV1 pathways appear effective for reducing OA pain, but further work is needed to better understand which patients may benefit most from these treatments. The anti-NGF antibody tanezumab and the TRPV1 agonist CNTX-4975 have both received fast-track designation from the FDA for the treatment of OA pain.
Collapse
|