1
|
Wei RF, Chen XY, Zhou JH, Zhang Y, Xie PY, Teng YL, Zong SH, Zeng GF. Single-cell RNA sequencing reveals B cell dynamics and osteoclast activation in Talaromycosis-related bone destruction. Cell Signal 2025; 131:111708. [PMID: 40032159 DOI: 10.1016/j.cellsig.2025.111708] [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: 11/26/2024] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
OBJECTIVE To explore the changes in bone destruction caused by Talaromycosis and its potential mechanisms. METHODS We assessed bone destruction and its severity using Micro CT, hematoxylin and eosin staining, tartrate-resistant acid phosphatase staining, F-actin staining, and Western blotting. We evaluated the biomechanical properties and pain perception in mice through biomechanical testing and the von Frey test. Single-cell RNA sequencing was used to analyze B cell composition in mouse tibial bone marrow, with findings verified by flow cytometry and qRT-PCR. Inflammatory cytokine levels, reactive oxygen species (ROS), and mitochondrial membrane potential were measured via enzyme-linked immunosorbent assay, immunofluorescence, and flow cytometry. Key proteins in the MAPK signaling pathway were also evaluated using Western blot. RESULTS Talaromyces marneffei (TM) infection led to increased osteoclast activity and significant bone destruction, accompanied by a reduction in weight gain, increased pain, and diminished bone biomechanical properties in mice. Post-infection, an increase in the number of B cells, particularly Naïve-B, ProB, and mature B cells, was observed, potentially linked to oxidative phosphorylation processes. TM infection elevated inflammatory cytokines production, ROS production and decreased mitochondrial membrane potential in vivo and in vitro. Furthermore, TM infection enhanced osteoclast differentiation through the activation of MAPK signaling pathways, including p38, ERK, and JNK. CONCLUSION TM infection induces B cell maturation and promotes bone destruction in the tibia of mice. This effect may be associated with mitochondrial apoptosis and ROS production during oxidative phosphorylation, potentially through MAPK pathway activation.
Collapse
Affiliation(s)
- Ru-Fei Wei
- School of Public Health of Guangxi Medical University, No. 22, Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region 530021, China; School of Basic Medical Sciences of Youjiang Medical University for Nationalities, No. 98, Chengxiang Road, Youjiang District, Baise, Guangxi Zhuang Autonomous Region 531400, China.
| | - Xin-Ying Chen
- School of Public Health of Guangxi Medical University, No. 22, Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region 530021, China
| | - Jun-Hong Zhou
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region 530021, China
| | - Yi Zhang
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region 530021, China
| | - Pei-Ya Xie
- Pharmaceutical College of Guangxi Medical University, No. 22, Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region 530021, China
| | - Yi-Lin Teng
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region 530021, China
| | - Shao-Hui Zong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region 530021, China; Wuming Hospital of Guangxi Medical University, No. 26, Yongning Road, Wuming District, Nanning City, Guangxi Zhuang Autonomous Region 530021, China.
| | - Gao-Feng Zeng
- School of Public Health of Guangxi Medical University, No. 22, Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region 530021, China.
| |
Collapse
|
2
|
Meng X, Qin L, Wang X. Biased agonism of G protein-coupled receptors as a novel strategy for osteoarthritis therapy. Bone Res 2025; 13:52. [PMID: 40355428 PMCID: PMC12069619 DOI: 10.1038/s41413-025-00435-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 05/14/2025] Open
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disorder marked by chronic pain, inflammation, and cartilage loss, with current treatments limited to symptom relief. G protein-coupled receptors (GPCRs) play a pivotal role in OA progression by regulating inflammation, chondrocyte survival, and matrix homeostasis. However, their multifaceted signaling, via G proteins or β-arrestins, poses challenges for precise therapeutic targeting. Biased agonism, where ligands selectively activate specific GPCR pathways, emerges as a promising approach to optimize efficacy and reduce side effects. This review examines biased signaling in OA-associated GPCRs, including cannabinoid receptors (CB1, CB2), chemokine receptors (CCR2, CXCR4), protease-activated receptors (PAR-2), adenosine receptors (A1R, A2AR, A2BR, A3R), melanocortin receptors (MC1R, MC3R), bradykinin receptors (B2R), prostaglandin E2 receptors (EP-2, EP-4), and calcium-sensing receptors (CaSR). We analyze ligands in clinical trials and explore natural products from Traditional Chinese Medicine as potential biased agonists. These compounds, with diverse structures and bioactivities, offer novel therapeutic avenues. By harnessing biased agonism, this review underscores the potential for developing targeted, safer OA therapies that address its complex pathology, bridging molecular insights with clinical translation.
Collapse
Affiliation(s)
- Xiangbo Meng
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Ling Qin
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Xinluan Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- University of Chinese Academy of Sciences, Beijing, PR China.
- Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
3
|
Shah H, Fairlie DP, Lim J. Protease-activated receptor 2: A promising therapeutic target for women's cancers. J Pharmacol Exp Ther 2025; 392:100016. [PMID: 39892996 DOI: 10.1124/jpet.124.002176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/26/2024] [Accepted: 04/08/2024] [Indexed: 01/22/2025] Open
Abstract
Cancers affecting women, such as breast, uterine, ovarian, endometrial, and cervical cancers, have become increasingly prevalent. The growing incidence and death rates associated with these cancers warrant the development of innovative and alternative approaches to current treatments. This article investigates the association of women's cancers with a molecular target known as protease-activated receptor 2 (PAR2), a G protein-coupled receptor that is expressed on the surface of cancer cells. Expression levels of the PAR2 gene were curated from publicly available databases, and PAR2 was found to be significantly overexpressed in tissues from patients with breast, uterine, ovarian, endometrial, or cervical cancer compared with normal tissues. PAR2 overexpression has been previously linked to tumor progression and, in some cases, tumor growth. Activation of PAR2 by either endogenous proteases or synthetic agonists triggers certain downstream intracellular signaling pathways that have been associated with tumor progression, cell migration and invasion, angiogenesis, and apoptosis of cancer cells. Although recent advances have led to identification of several PAR2 antagonists, none has yet been developed for human use. Additionally, PAR2 inhibition has been shown to increase the efficacy of chemotherapeutic drugs, allowing them to be potentially used at less toxic doses in combination therapies for cancer. The present work briefly summarizes the current status of PAR2 as a potential therapeutic target for treating women's cancers. SIGNIFICANCE STATEMENT: This article highlights potential roles for protease-activated receptor 2 (PAR2) in cancers affecting women. Overexpression of the PAR2 gene in women's cancers is associated with various oncogenic processes, such as tumor progression, cell migration, and invasion, ultimately contributing to poorer patient prognoses. Given the increasing incidence of women's cancers, there is an urgent need to develop novel therapeutic drugs, and PAR2 represents a promising target for developing new treatments.
Collapse
Affiliation(s)
- Himani Shah
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
| | - Junxian Lim
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
4
|
Patnaik R, Varghese R, Jannati S, Naidoo N, Banerjee Y. Targeting PAR2-mediated inflammation in osteoarthritis: a comprehensive in vitro evaluation of oleocanthal's potential as a functional food intervention for chondrocyte protection and anti-inflammatory effects. BMC Musculoskelet Disord 2024; 25:769. [PMID: 39354427 PMCID: PMC11446003 DOI: 10.1186/s12891-024-07888-y] [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: 03/26/2024] [Accepted: 09/20/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by chronic inflammation and progressive cartilage degradation, ultimately leading to joint dysfunction and disability. Oleocanthal (OC), a bioactive phenolic compound derived from extra virgin olive oil, has garnered significant attention due to its potent anti-inflammatory properties, which are comparable to those of non-steroidal anti-inflammatory drugs (NSAIDs). This study pioneers the investigation into the effects of OC on the Protease-Activated Receptor-2 (PAR-2) mediated inflammatory pathway in OA, aiming to validate its efficacy as a functional food-based therapeutic intervention. METHODS To simulate cartilage tissue in vitro, human bone marrow-derived mesenchymal stem cells (BMSCs) were differentiated into chondrocytes. An inflammatory OA-like environment was induced in these chondrocytes using lipopolysaccharide (LPS) to mimic the pathological conditions of OA. The therapeutic effects of OC were evaluated by treating these inflamed chondrocytes with various concentrations of OC. The study focused on assessing key inflammatory markers, catabolic enzymes, and mitochondrial function to elucidate the protective mechanisms of OC. Mitochondrial function, specifically mitochondrial membrane potential (ΔΨm), was assessed using Rhodamine 123 staining, a fluorescent dye that selectively accumulates in active mitochondria. The integrity of ΔΨm serves as an indicator of mitochondrial and bioenergetic function. Additionally, Western blotting was employed to analyze protein expression levels, while real-time polymerase chain reaction (RT-PCR) was used to quantify gene expression of inflammatory cytokines and catabolic enzymes. Flow cytometry was utilized to measure cell viability and apoptosis, providing a comprehensive evaluation of OC's therapeutic effects on chondrocytes. RESULTS The results demonstrated that OC significantly downregulated PAR-2 expression in a dose-dependent manner, leading to a substantial reduction in pro-inflammatory cytokines, including TNF-α, IL-1β, and MCP-1. Furthermore, OC attenuated the expression of catabolic markers such as SOX4 and ADAMTS5, which are critically involved in cartilage matrix degradation. Importantly, OC was found to preserve mitochondrial membrane potential (ΔΨm) in chondrocytes subjected to inflammatory stress, as evidenced by Rhodamine 123 staining, indicating a protective effect on cellular bioenergetics. Additionally, OC modulated the Receptor Activator of Nuclear Factor Kappa-Β Ligand (RANKL)/Receptor Activator of Nuclear Factor Kappa-Β (RANK) pathway, suggesting a broader therapeutic action against the multifactorial pathogenesis of OA. CONCLUSIONS This study is the first to elucidate the modulatory effects of OC on the PAR-2 mediated inflammatory pathway in OA, revealing its potential as a multifaceted therapeutic agent that not only mitigates inflammation but also protects cartilage integrity. The preservation of mitochondrial function and modulation of the RANKL/RANK pathway further underscores OC's comprehensive therapeutic potential in counteracting the complex pathogenesis of OA. These findings position OC as a promising candidate for integration into nutritional interventions aimed at managing OA. However, further research is warranted to fully explore OC's therapeutic potential across different stages of OA and its long-term effects in musculoskeletal disorders.
Collapse
|
5
|
Nag JK, Appasamy P, Malka H, Sedley S, Bar-Shavit R. New Target(s) for RNF43 Regulation: Implications for Therapeutic Strategies. Int J Mol Sci 2024; 25:8083. [PMID: 39125653 PMCID: PMC11311281 DOI: 10.3390/ijms25158083] [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: 06/13/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Cancer cells depend on specific oncogenic pathways or present a genetic alteration that leads to a particular disturbance. Still, personalized and targeted biological therapy remains challenging, with current efforts generally yielding disappointing results. Carefully assessing onco-target molecular pathways can, however, potently assist with such efforts for the selection of patient populations that would best respond to a given drug treatment. RNF43, an E3 ubiquitin ligase that negatively regulates Wnt/frizzled (FZD) receptors by their ubiquitination, internalization, and degradation, controls a key pathway in cancer. Recently, additional target proteins of RNF43 were described, including p85 of the PI3K/AKT/mTOR signaling pathway and protease-activated receptor 2 (PAR2), a G-protein-coupled receptor that potently induces β-catenin stabilization, independent of Wnts. RNF43 mutations with impaired E3 ligase activity were found in several types of cancers (e.g., gastrointestinal system tumors and endometrial and ovarian cancer), pointing to a high dependency on FZD receptors and possibly PAR2 and the PI3K/AKT/mTOR signaling pathway. The development of drugs toward these targets is essential for improved treatment of cancer patients.
Collapse
Affiliation(s)
| | | | | | | | - Rachel Bar-Shavit
- Sharett Institute of Oncology, Hadassah Medical Center, Hebrew University, Jerusalem 91120, Israel; (J.K.N.); (P.A.); (H.M.); (S.S.)
| |
Collapse
|
6
|
Saleh AS, Abdel-Gabbar M, Gabr H, Shams A, Tamur S, Mahdi EA, Ahmed OM. Ameliorative effects of undifferentiated and differentiated BM-MSCs in MIA-induced osteoarthritic Wistar rats: roles of NF-κB and MMPs signaling pathways. Am J Transl Res 2024; 16:2793-2813. [PMID: 39114694 PMCID: PMC11301505 DOI: 10.62347/fghv2647] [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: 01/02/2024] [Accepted: 05/15/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVES Osteoarthritis (OA) is a degenerative joint condition that is persistent. OA affects millions of people throughout the world. Both people and society are heavily economically burdened by osteoarthritis. There is currently no medication that can structurally alter the OA processes or stop the disease from progressing. Stem cells have the potential to revolutionize medicine due to their capacity to differentiate into chondrocytes, capacity to heal tissues and organs including osteoarthritic joints, and immunomodulatory capabilities. Therefore, the goal of the current investigation was to determine how bone marrow-derived mesenchymal stem cells (BM-MSCs) and chondrogenic differentiated mesenchymal stem cells (CD-MSCs) affected the treatment of OA in rats with monosodium iodoacetate (MIA)-induced osteoarthritis. METHODS Male Wistar rats were injected three times with MIA (1 mg)/100 µL isotonic saline to induce osteoarthritis in the ankle joint of the right hind leg. Following the MIA injection, the osteoarthritic rats were given weekly treatments of 1 × 106 BM-MSCs and CD-MSCs into the tail vein for three weeks. RESULTS The obtained results showed that in osteoarthritic rats, BM-MSCs and CD-MSCs dramatically decreased ankle diameter measurements, decreased oxidized glutathione (GSSG) level, and boosted glutathione peroxidase (GPx) and glutathione reductase (GR) activities. Additionally, in rats with MIA-induced OA, BM-MSCs and CD-MSCs dramatically boosted interleukin-10 (IL-10) serum levels while considerably decreasing serum anticitrullinated protein antibodies (ACPA), tumour necrosis factor-α (TNF-α), and interleukin-17 (IL-17) levels as well as ankle transforming growth factor-β1 (TGF-β1) expression. Analysis of histology, immunohistochemistry, and western blots in osteoarthritic joints showed that cartilage breakdown and joint inflammation gradually decreased over time. CONCLUSIONS It is possible to conclude from these results that BM-MSCs and CD-MSCs have anti-arthritic potential in MIA-induced OA, which may be mediated via inhibitory effects on oxidative stress, MMPs and inflammation through suppressing the NF-κB pathway. In osteoarthritis, using CD-MSCs as a treatment is more beneficial therapeutically than using BM-MSCs.
Collapse
Affiliation(s)
- Ablaa S Saleh
- Department of Biochemistry, Faculty of Science, Beni-Suef UniversityBeni-Suef 62521, Egypt
| | - Mohammed Abdel-Gabbar
- Department of Biochemistry, Faculty of Science, Beni-Suef UniversityBeni-Suef 62521, Egypt
| | - Hala Gabr
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo UniversityCairo 11435, Egypt
| | - Anwar Shams
- Department of Pharmacology, College of Medicine, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif UniversityTaif 26432, Saudi Arabia
- High Altitude Research Center, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
| | - Shadi Tamur
- Department of Pediatric, College of Medicine, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
| | - Emad A Mahdi
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef UniversityBeni-Suef 62521, Egypt
| | - Osama M Ahmed
- Division of Physiology, Department of Zoology, Faculty of Science, Beni-Suef UniversityBeni-Suef 62521, Egypt
| |
Collapse
|
7
|
Reches G, Piran R. Par2-mediated responses in inflammation and regeneration: choosing between repair and damage. Inflamm Regen 2024; 44:26. [PMID: 38816842 PMCID: PMC11138036 DOI: 10.1186/s41232-024-00338-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/19/2024] [Indexed: 06/01/2024] Open
Abstract
The protease activated receptor 2 (Par2) plays a pivotal role in various damage models, influencing injury, proliferation, inflammation, and regeneration. Despite extensive studies, its binary roles- EITHER aggravating injury or promoting recovery-make a conclusive translational decision on its modulation strategy elusive. Analyzing two liver regeneration models, autoimmune hepatitis and direct hepatic damage, we discovered Par2's outcome depends on the injury's nature. In immune-mediated injury, Par2 exacerbates damage, while in direct tissue injury, it promotes regeneration. Subsequently, we evaluated the clinical significance of this finding by investigating Par2's expression in the context of autoimmune diabetes. We found that the absence of Par2 in all lymphocytes provided full protection against the autoimmune destruction of insulin-producing β-cells in mice, whereas the introduction of a β-cell-specific Par2 null mutation accelerated the onset of autoimmune diabetes. This pattern led us to hypothesize whether these observations are universal. A comprehensive review of recent Par2 publications across tissues and systems confirms the claim drafted above: Par2's initial activation in the immune system aggravates inflammation, hindering recovery, whereas its primary activation in the damaged tissue fosters regeneration. As a membrane-anchored receptor, Par2 emerges as an attractive drug target. Our findings highlight a crucial translational modulation strategy in regenerative medicine based on injury type.
Collapse
Affiliation(s)
- Gal Reches
- The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| | - Ron Piran
- The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel.
| |
Collapse
|
8
|
Costa TFR, Catta-Preta CMC, Goundry A, Carvalho DB, Rodrigues NS, Vivarini AC, de Abreu MF, Reis FCG, Lima APCA. The ecotin-like peptidase inhibitor of Trypanosoma cruzi prevents TMPRSS2-PAR2-TLR4 crosstalk downmodulating infection and inflammation. FASEB J 2024; 38:e23566. [PMID: 38526868 DOI: 10.1096/fj.202302091rr] [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: 10/15/2023] [Revised: 02/24/2024] [Accepted: 03/06/2024] [Indexed: 03/27/2024]
Abstract
Trypanosoma cruzi is the causative agent of Chagas disease, a chronic pathology that affects the heart and/or digestive system. This parasite invades and multiplies in virtually all nucleated cells, using a variety of host cell receptors for infection. T. cruzi has a gene that encodes an ecotin-like inhibitor of serine peptidases, ISP2. We generated ISP2-null mutants (Δisp2) in T. cruzi Dm28c using CRISPR/Cas9. Epimastigotes of Δisp2 grew normally in vitro but were more susceptible to lysis by human serum compared to parental and ISP2 add-back lines. Tissue culture trypomastigotes of Δisp2 were more infective to human muscle cells in vitro, which was reverted by the serine peptidase inhibitors aprotinin and camostat, suggesting that host cell epitheliasin/TMPRSS2 is the target of ISP2. Pretreatment of host cells with an antagonist to the protease-activated receptor 2 (PAR2) or an inhibitor of Toll-like receptor 4 (TLR4) selectively counteracted the increased cell invasion by Δisp2, but did not affect invasion by parental and add-back lines. The same was observed following targeted gene silencing of PAR2, TLR4 or TMPRSS2 in host cells by siRNA. Furthermore, Δisp2 caused increased tissue edema in a BALB/c mouse footpad infection model after 3 h differently to that observed following infection with parental and add-back lines. We propose that ISP2 contributes to protect T. cruzi from the anti-microbial effects of human serum and to prevent triggering of PAR2 and TLR4 in host cells, resulting in the modulation of host cell invasion and contributing to decrease inflammation during acute infection.
Collapse
Affiliation(s)
- Tatiana F R Costa
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina M C Catta-Preta
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amy Goundry
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle B Carvalho
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nathalia S Rodrigues
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aislan C Vivarini
- Departamento de Biologia Celular e Molecular, Insituto de Biologia, Universidade Federal Fluminense, Niteroi, Brazil
| | - Mayra Fonseca de Abreu
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavia C G Reis
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula C A Lima
- Laboratório de Bioquímica e Biologia Molecular de Proteases, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Furuya K, Nakajima M, Tsunedomi R, Nakagami Y, Xu M, Matsui H, Tokumitsu Y, Shindo Y, Watanabe Y, Tomochika S, Maeda N, Iida M, Suzuki N, Takeda S, Hazama S, Ioka T, Hoshii Y, Ueno T, Nagano H. High serum proteinase-3 levels predict poor progression-free survival and lower efficacy of bevacizumab in metastatic colorectal cancer. BMC Cancer 2024; 24:165. [PMID: 38308214 PMCID: PMC10835931 DOI: 10.1186/s12885-024-11924-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/26/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND To improve the prognosis of patients with metastatic colorectal cancer (mCRC), investigating predictive biomarkers of their prognosis and chemotherapeutic responsiveness is necessary. This study aimed to analyze the clinical significance of serum proteinase-3 (PRTN3) as a predictor for prognosis and chemosensitivity, especially to bevacizumab therapy, in mCRC. METHODS This single-center retrospective observational study enrolled 79 patients with mCRC in our hospital and 353 patients with colorectal cancer in the TCGA database. Preoperative serum PRTN3 levels were measured using an enzyme-linked immunosorbent assay. The clinicopathological characteristics and prognosis according to serum PRTN3 levels were then evaluated. PRTN3 expression in tumor and stromal cells was evaluated immunohistochemically. The impact of PRTN3 levels on angiogenesis and bevacizumab sensitivity was evaluated using the tube formation assay. RESULTS Serum PRTN3 levels were an independent poor prognostic factor for progression-free survival (PFS) (hazard ratio, 2.082; 95% confidence interval, 1.118-3.647; P=0.010) in patients with mCRC. Similarly, prognostic analysis with TCGA data sets showed poorer overall survival in patients with PRTN3 expression than that in patients without PRTN3 expression, especially in patients with stage IV. Immunohistochemical analysis of resected specimens revealed that stromal neutrophils expressed PRTN3, and their expression level was significantly correlated with serum PRTN3 levels. Interestingly, the effectiveness of first-line chemotherapy was significantly poorer in the high serum PRTN3 level group. High serum PRTN3 was significantly associated with poor PFS (hazard ratio, 3.027; 95% confidence interval, 1.175-7.793; P=0.0161) in patients treated with bevacizumab, an anti-angiogenic inhibitor. The tube formation assay revealed that PRTN3 administration notably augmented angiogenesis while simultaneously attenuating the anti-angiogenic influence exerted by bevacizumab therapy. CONCLUSIONS Serum PRTN3 levels could be a novel predictive biomarker of PFS of first-line chemotherapy, especially for bevacizumab therapy, in patients with mCRC.
Collapse
Affiliation(s)
- Kei Furuya
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Masao Nakajima
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yuki Nakagami
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ming Xu
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hiroto Matsui
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yukio Tokumitsu
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yoshitaro Shindo
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yusaku Watanabe
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shinobu Tomochika
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Noriko Maeda
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Michihisa Iida
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shigeru Takeda
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shoichi Hazama
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Tatsuya Ioka
- Oncology Center, Yamaguchi University Hospital, Ube, Yamaguchi, 755-8505, Japan
| | - Yoshinobu Hoshii
- Department of Diagnostic Pathology, Yamaguchi University Hospital, Ube, Yamaguchi, 755-8505, Japan
| | - Tomio Ueno
- Department of Digestive Surgery, Kawasaki Medical School, Kurashiki, Okayama, 701-0192, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|
10
|
Han LL, Lu QQ, Zheng WW, Li YL, Song YY, Zhang XZ, Long SR, Liu RD, Wang ZQ, Cui J. A novel trypsin of Trichinella spiralis mediates larval invasion of gut epithelium via binding to PAR2 and activating ERK1/2 pathway. PLoS Negl Trop Dis 2024; 18:e0011874. [PMID: 38166153 PMCID: PMC10786404 DOI: 10.1371/journal.pntd.0011874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/12/2024] [Accepted: 12/19/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Proteases secreted by Trichinella spiralis intestinal infective larvae (IIL) play an important role in larval invasion and pathogenesis. However, the mechanism through which proteases mediate larval invasion of intestinal epithelial cells (IECs) remains unclear. A novel T. spiralis trypsin (TsTryp) was identified in IIL excretory/secretory (ES) proteins. It was an early and highly expressed protease at IIL stage, and had the potential as an early diagnostic antigen. The aim of this study was to investigate the biological characteristics of this novel TsTryp, its role in larval invasion of gut epithelium, and the mechanisms involved. METHODOLOGY/PRINCIPAL FINDING TsTryp with C-terminal domain was cloned and expressed in Escherichia coli BL21 (DE3), and the rTsTryp had the enzymatic activity of natural trypsin, but it could not directly degrade gut tight junctions (TJs) proteins. qPCR and western blotting showed that TsTryp was highly expressed at the invasive IIL stage. Immunofluorescence assay (IFA), ELISA and Far Western blotting revealed that rTsTryp specifically bound to IECs, and confocal microscopy showed that the binding of rTsTryp with IECs was mainly localized in the cytomembrane. Co-immunoprecipitation (Co-IP) confirmed that rTsTryp bound to protease activated receptors 2 (PAR2) in Caco-2 cells. rTsTryp binding to PAR2 resulted in decreased expression levels of ZO-1 and occludin and increased paracellular permeability in Caco-2 monolayers by activating the extracellular regulated protein kinases 1/2 (ERK1/2) pathway. rTsTryp decreased TJs expression and increased epithelial permeability, which could be abrogated by the PAR2 antagonist AZ3451 and ERK1/2 inhibitor PD98059. rTsTryp facilitated larval invasion of IECs, and anti-rTsTryp antibodies inhibited invasion. Both inhibitors impeded larval invasion and alleviated intestinal inflammation in vitro and in vivo. CONCLUSIONS TsTryp binding to PAR2 activated the ERK1/2 pathway, decreased the expression of gut TJs proteins, disrupted epithelial integrity and barrier function, and consequently mediated larval invasion of the gut mucosa. Therefore, rTsTryp could be regarded as a potential vaccine target for blocking T. spiralis invasion and infection.
Collapse
Affiliation(s)
- Lu Lu Han
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Qi Qi Lu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Wen Wen Zheng
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Yang Li Li
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Yan Yan Song
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Xin Zhuo Zhang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Peterson AA, Liu DR. Small-molecule discovery through DNA-encoded libraries. Nat Rev Drug Discov 2023; 22:699-722. [PMID: 37328653 PMCID: PMC10924799 DOI: 10.1038/s41573-023-00713-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 06/18/2023]
Abstract
The development of bioactive small molecules as probes or drug candidates requires discovery platforms that enable access to chemical diversity and can quickly reveal new ligands for a target of interest. Within the past 15 years, DNA-encoded library (DEL) technology has matured into a widely used platform for small-molecule discovery, yielding a wide variety of bioactive ligands for many therapeutically relevant targets. DELs offer many advantages compared with traditional screening methods, including efficiency of screening, easily multiplexed targets and library selections, minimized resources needed to evaluate an entire DEL and large library sizes. This Review provides accounts of recently described small molecules discovered from DELs, including their initial identification, optimization and validation of biological properties including suitability for clinical applications.
Collapse
Affiliation(s)
- Alexander A Peterson
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
12
|
Sui X, Xie T, Xu Y, Zhang A, Zhang Y, Gu F, Li L, Xu Z, Chen J. Protease-Activated Receptor-2 and Phospholipid Metabolism Analysis in Hyperuricemia-Induced Renal Injury. Mediators Inflamm 2023; 2023:5007488. [PMID: 37484603 PMCID: PMC10359134 DOI: 10.1155/2023/5007488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 04/30/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023] Open
Abstract
Interstitial inflammation is an important mechanism of pathological damage in renal injury caused by hyperuricemia. Protease-activated receptor-2 (PAR2) is a class of targets that act upstream of the PI3K/AKT/NF-κB pathway and is involved in various inflammatory diseases. We induced a hyperuricemia model in rats by adenine and ethambutol gavage in an in vivo experiment. We demonstrated that PAR2 and PI3K/AKT/NF-κB pathway expression were significantly upregulated in renal tissues, with massive inflammatory cell infiltration in the renal interstitium and renal tissue injury. Treating hyperuricemic rats with AZ3451, a selective metabotropic antagonist of PAR2, we demonstrated that PAR2 antagonism inhibited the PI3K/AKT/NF-κB pathway and attenuated tubular dilation and tubulointerstitial inflammatory cell infiltration. The phospholipid metabolism profiles provided a perfect separation between the normal and hyperuricemic rats. In addition, we also found that AZ3451 can affect phospholipid metabolism. Our work suggests that PAR2 may mediate hyperuricemia-mediated renal injury by activating the PI3K/AKT/NF-κB pathway. The PAR2 antagonist AZ3451 may be a promising therapeutic strategy for hyperuricemia-induced inflammatory responses.
Collapse
Affiliation(s)
- Xiaolu Sui
- Department of Nephrology, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Tingfei Xie
- Department of Nephrology, The People's Hospital of Baoan Shenzhen, The Second School of Clinical Medicine, Southern Medical University, Shenzhen 518000, Guangdong, China
| | - Yunpeng Xu
- Department of Nephrology, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Aisha Zhang
- Department of Nephrology, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Yanzi Zhang
- Department of Nephrology, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Fengjuan Gu
- Department of Nephrology, Shenzhen Baoan People's Hospital (Group) The Second People's Hospital, Shenzhen 518000, Guangdong, China
| | - Lixiang Li
- Department of Nephrology, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518000, Guangdong, China
| | - Zibin Xu
- Department of Nephrology, The People's Hospital of Baoan Shenzhen, The Second School of Clinical Medicine, Southern Medical University, Shenzhen 518000, Guangdong, China
| | - Jihong Chen
- Department of Nephrology, The Second Affiliated Hospital of Shenzhen University, Shenzhen 518000, Guangdong, China
- Department of Nephrology, The People's Hospital of Baoan Shenzhen, The Second School of Clinical Medicine, Southern Medical University, Shenzhen 518000, Guangdong, China
| |
Collapse
|
13
|
Huang S, Cao G, Dai D, Xu Q, Ruiz S, Shindo S, Nakamura S, Kawai T, Lin J, Han X. Porphyromonas gingivalis outer membrane vesicles exacerbate retinal microvascular endothelial cell dysfunction in diabetic retinopathy. Front Microbiol 2023; 14:1167160. [PMID: 37250057 PMCID: PMC10213754 DOI: 10.3389/fmicb.2023.1167160] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/14/2023] [Indexed: 05/31/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the leading causes of blindness. Periodontitis is one of the highest oral incidences and has been closely related to various systemic conditions through Porphyromonas gingivalis (P. gingivalis). P. gingivalis OMVs, derived from P. gingivalis, can cause endothelial dysfunction and potentially affect microvascular diseases. Current epidemiological studies provide limited evidence suggesting that periodontitis is associated with DR. However, there is a lack of basic research elucidating how periodontitis affects the severity of DR. This study aimed to explore the potential of P. gingivalis OMVs to contribute to the pathogenesis of DR and explore how it affect the retinal microvascular endothelium. The results demonstrated that P. gingivalis OMVs accelerated the blood-retinal barrier damage in DR mice. In vitro studies showed that the expression of inflammatory factors in human retinal microvascular endothelial cells (HRMECs) was increased after P. gingivalis OMVs stimulation, and the increased reactive oxygen species production, mitochondrial dysfunction, apoptosis, and altered endothelial permeability were observed in HRMECs under P. gingivalis OMVs stimulation. In addition, we found that protease-activated receptor-2 (PAR-2) regulated OMVs-induced TNF-α, MMP-9 mRNA expression, cell death, and endothelial permeability. Overall, we suggested that P. gingivalis OMVs induced mitochondria-related cell death of HRMECs and accelerated endothelial dysfunction, thus aggravating DR, in which PAR-2 plays a potential role. This study is the first research report to delineate the potential molecular mechanism of P. gingivalis OMVs on DR pathogenesis, which uniquely focused on elucidating the possible impact of periodontal pathogen derivatives on DR progression.
Collapse
Affiliation(s)
- Shengyuan Huang
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Guoqin Cao
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Dong Dai
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qiuping Xu
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Sunniva Ruiz
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Satoru Shindo
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Shin Nakamura
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Toshihisa Kawai
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Jiang Lin
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaozhe Han
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| |
Collapse
|
14
|
Shpakov AO. Allosteric Regulation of G-Protein-Coupled Receptors: From Diversity of Molecular Mechanisms to Multiple Allosteric Sites and Their Ligands. Int J Mol Sci 2023; 24:6187. [PMID: 37047169 PMCID: PMC10094638 DOI: 10.3390/ijms24076187] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Allosteric regulation is critical for the functioning of G protein-coupled receptors (GPCRs) and their signaling pathways. Endogenous allosteric regulators of GPCRs are simple ions, various biomolecules, and protein components of GPCR signaling (G proteins and β-arrestins). The stability and functional activity of GPCR complexes is also due to multicenter allosteric interactions between protomers. The complexity of allosteric effects caused by numerous regulators differing in structure, availability, and mechanisms of action predetermines the multiplicity and different topology of allosteric sites in GPCRs. These sites can be localized in extracellular loops; inside the transmembrane tunnel and in its upper and lower vestibules; in cytoplasmic loops; and on the outer, membrane-contacting surface of the transmembrane domain. They are involved in the regulation of basal and orthosteric agonist-stimulated receptor activity, biased agonism, GPCR-complex formation, and endocytosis. They are targets for a large number of synthetic allosteric regulators and modulators, including those constructed using molecular docking. The review is devoted to the principles and mechanisms of GPCRs allosteric regulation, the multiplicity of allosteric sites and their topology, and the endogenous and synthetic allosteric regulators, including autoantibodies and pepducins. The allosteric regulation of chemokine receptors, proteinase-activated receptors, thyroid-stimulating and luteinizing hormone receptors, and beta-adrenergic receptors are described in more detail.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
15
|
Zhang D, Ma L, Tan X, Deng W, Wen S, Li Y, Qin B, Cao S, Yu T. Intradermal miR-16-5p targets Akt3 and reduces RTX-induced postherpetic neuralgia-mimic pain in mice. Eur J Pharmacol 2023; 946:175665. [PMID: 36940911 DOI: 10.1016/j.ejphar.2023.175665] [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: 08/06/2022] [Revised: 02/27/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
The molecular mechanisms of refractory pain in postherpetic neuralgia (PHN) patients are not fully understood. PHN may be related to skin abnormality after herpes zoster induced skin lesions. We previously reported 317 differentially expressed microRNAs (miRNAs) in PHN skin compared with the contralateral normal mirror skin. In this study, 19 differential miRNAs were selected and the expression was validated in other 12 PHN patients. The expression levels of miR-16-5p, miR-20a-5p, miR-505-5p, miR-3664-3p, miR-4714-3p and let-7a-5p are lower in PHN skin, which is the same as those in microarray experiment. To evaluate the effects of cutaneous miRNA on PHN, the expression of candidate miRNAs is further observed in resiniferatoxin (RTX) induced PHN-mimic mice model. In the plantar skin of RTX mice, miR-16-5p and let-7a-5p are downregulated, with the same expression trend of PHN patients. In addition, intraplantar injection of agomir-16-5p reduced mechanical hyperalgesia, and improved thermal hypoalgesia in RTX mice. Furthermore, agomir-16-5p down-regulated the expression levels of Akt3, which is the target gene of agomir-16-5p. These results suggest that intraplantar miR-16-5p may alleviate RTX induced PHN-mimic pain by inhibiting the expression of Akt3 in the skin.
Collapse
Affiliation(s)
- Dexin Zhang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Lulin Ma
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xinran Tan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Wenwen Deng
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Song Wen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ying Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Bangyong Qin
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Song Cao
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, Guizhou, China.
| | - Tian Yu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
16
|
Involvement of Mast-Cell-Tryptase- and Protease-Activated Receptor 2-Mediated Signaling and Urothelial Barrier Dysfunction with Reduced Uroplakin II Expression in Bladder Hyperactivity Induced by Chronic Bladder Ischemia in the Rat. Int J Mol Sci 2023; 24:ijms24043982. [PMID: 36835398 PMCID: PMC9966957 DOI: 10.3390/ijms24043982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
We aimed to investigate the relationship between mast cell (MC) infiltration into the bladder with urothelial barrier dysfunction and bladder hyperactivity in a chronic bladder ischemia (CBI) rat model. We compared CBI rats (CBI group; n = 10) with normal rats (control group; n = 10). We measured the expression of mast cell tryptase (MCT) and protease-activated receptor 2 (PAR2), which are correlated with C fiber activation via MCT, and Uroplakins (UP Ia, Ib, II and III), which are critical to urothelial barrier function, via Western blotting. The effects of FSLLRY-NH2, a PAR2 antagonist, administered intravenously, on the bladder function of CBI rats were evaluated with a cystometrogram. In the CBI group, the MC number in the bladder was significantly greater (p = 0.03), and the expression of MCT (p = 0.02) and PAR2 (p = 0.02) was significantly increased compared to that of the control group. The 10 μg/kg FSLLRY-NH2 injection significantly increased the micturition interval of CBI rats (p = 0.03). The percentage of UP-II-positive cells on the urothelium with immunohistochemical staining was significantly lower in the CBI group than in the control group (p < 0.01). Chronic ischemia induces urothelial barrier dysfunction via impairing UP II, consequently inducing MC infiltration into the bladder wall and increased PAR2 expression. PAR2 activation by MCT may contribute to bladder hyperactivity.
Collapse
|
17
|
Wang H, Yan Y, Pathak JL, Hong W, Zeng J, Qian D, Hao B, Li H, Gu J, Jaspers RT, Wu G, Shao M, Peng G, Lan H. Quercetin prevents osteoarthritis progression possibly via regulation of local and systemic inflammatory cascades. J Cell Mol Med 2023; 27:515-528. [PMID: 36722313 PMCID: PMC9930437 DOI: 10.1111/jcmm.17672] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 02/02/2023] Open
Abstract
Due to the lack of effective treatments, osteoarthritis (OA) remains a challenge for clinicians. Quercetin, a bioflavonoid, has shown potent anti-inflammatory effects. However, its effect on preventing OA progression and the underlying mechanisms are still unclear. In this study, Sprague-Dawley male rats were divided into five groups: control group, OA group (monosodium iodoacetate intra-articular injection), and three quercetin-treated groups. Quercetin-treated groups were treated with intragastric quercetin once a day for 28 days. Gross observation and histopathological analysis showed cartilage degradation and matrix loss in the OA group. High-dose quercetin-group joints showed failure in OA progression. High-dose quercetin inhibited the OA-induced expression of MMP-3, MMP-13, ADAMTS4, and ADAMTS5 and promoted the OA-reduced expression of aggrecan and collagen II. Levels of most inflammatory cytokines and growth factors tested in synovial fluid and serum were upregulated in the OA group and these increases were reversed by high-dose quercetin. Similarly, subchondral trabecular bone was degraded in the OA group and this effect was reversed in the high-dose quercetin group. Our findings indicate that quercetin has a protective effect against OA development and progression possibly via maintaining the inflammatory cascade homeostasis. Therefore, quercetin could be a potential therapeutic agent to prevent OA progression in risk groups.
Collapse
Affiliation(s)
- Haiyan Wang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina,Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement SciencesVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Yongyong Yan
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina,Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement SciencesVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Janak L. Pathak
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Wei Hong
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina,GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Jing Zeng
- Liwan Central Hospital of GuangzhouGuangzhouChina
| | - Dongyang Qian
- Department of Orthopaedics, The First Affiliated HospitalGuangzhou Medical University/Guangdong Key Laboratory of Orthopaedic Technology and Implant MaterialsGuangzhouChina
| | - Binwei Hao
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina,Department of Pulmonary and Critical Care Medicine, Shanxi Bethune HospitalShanxi Academy of Medical SciencesTaiyuanChina
| | - Haiqing Li
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jinlan Gu
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Richard T. Jaspers
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina,Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement SciencesVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Centre for Dentistry Amsterdam (ACTA)Vrije Universiteit Amsterdam, Amsterdam Movement ScienceAmsterdamThe Netherlands
| | - Ming Shao
- Department of Orthopaedic SurgeryThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Gongyong Peng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Haifeng Lan
- Department of Orthopaedic SurgeryThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
18
|
Loucks A, Maerz T, Hankenson K, Moeser A, Colbath A. The multifaceted role of mast cells in joint inflammation and arthritis. Osteoarthritis Cartilage 2023; 31:567-575. [PMID: 36682447 DOI: 10.1016/j.joca.2023.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To review current knowledge surrounding the role of mast cells in joint inflammation and arthritis. METHOD Narrative review. RESULTS Mast cells (MCs) are commonly observed in the synovium of the joint, particularly surrounding blood vessels and nerve endings. Some studies have reported increased MC number and degranulation in patients with osteoarthritis (OA). In two studies, MCs were the only immune cell type found in higher concentrations in synovium of OA patients compared to rheumatoid arthritis patients. Activation of MCs in OA includes signaling pathways such as immunoglobulin E/Fc epsilon Receptor 1 (IgE/FcεR1), immunoglobulin G/Fc gamma receptor (IgG/FcγR), complement, and toll-like cell surface receptor-mediated signaling, resulting in context-dependent release of either pro-inflammatory and/or anti-inflammatory mediators within the joint. Activation of MCs results in the release of pro-inflammatory mediators that ultimately contribute to inflammation of the synovium, bone remodeling, and cartilage damage. However, some studies have proposed that MCs can also exhibit anti-inflammatory effects by secreting mediators that inactivate pro-inflammatory cytokines such as interleukin 6 (IL-6). CONCLUSIONS MCs may play a role in mediating synovial inflammation and OA progression. However, the mechanisms governing MC activation, the downstream pro- and/or anti-inflammatory effects, and their impact on osteoarthritis pathogenesis remains to be elucidated and requires extensive further study. Furthermore, it is important to establish the pathways of MC activation in OA to determine whether MCs exhibit varying phenotypes as a function of disease stage. Ultimately, such research is needed before understanding whether MCs could be targeted in OA treatments.
Collapse
Affiliation(s)
- A Loucks
- Department of Clinical Sciences, Cornell University, Ithaca, NY, USA.
| | - T Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - K Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - A Moeser
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA.
| | - A Colbath
- Department of Clinical Sciences, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
19
|
GB83, an Agonist of PAR2 with a Unique Mechanism of Action Distinct from Trypsin and PAR2-AP. Int J Mol Sci 2022; 23:ijms231810631. [PMID: 36142527 PMCID: PMC9506296 DOI: 10.3390/ijms231810631] [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: 08/22/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Protease-activated receptor 2 (PAR2) is a G-protein-coupled receptor (GPCR) activated by proteolytic cleavage of its N-terminal domain. Once activated, PAR2 is rapidly desensitized and internalized by phosphorylation and β-arrestin recruitment. Due to its irreversible activation mechanism, some agonists that rapidly desensitized PAR2 have been misconceived as antagonists, and this has impeded a better understanding of the pathophysiological role of PAR2. In the present study, we found that GB83, initially identified as a PAR2 antagonist, is a bona fide agonist of PAR2 that induces unique cellular signaling, distinct from trypsin and PAR2-activating peptide (AP). Activation of PAR2 by GB83 markedly elicited an increase in intracellular calcium levels and phosphorylation of MAPKs, but in a delayed and sustained manner compared to the rapid and transient signals induced by trypsin and PAR2-AP. Interestingly, unlike PAR2-AP, GB83 and trypsin induced sustained receptor endocytosis and PAR2 colocalization with β-arrestin. Moreover, the recovery of the localization and function of PAR2 was significantly delayed after stimulation by GB83, which may be the reason why GB83 is recognized as an antagonist of PAR2. Our results revealed that GB83 is a bona fide agonist of PAR2 that uniquely modulates PAR2-mediated cellular signaling and is a useful pharmacological tool for studying the pathophysiological role of PAR2.
Collapse
|
20
|
Hamdalla HM, Ahmed RR, Galaly SR, Ahmed OM, Naguib IA, Alghamdi BS, Abdul-Hamid M. Assessment of the Efficacy of Bone Marrow-Derived Mesenchymal Stem Cells against a Monoiodoacetate-Induced Osteoarthritis Model in Wistar Rats. Stem Cells Int 2022; 2022:1900403. [PMID: 36017131 PMCID: PMC9398859 DOI: 10.1155/2022/1900403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 11/18/2022] Open
Abstract
Osteoarthritis (OA) of the knee is a debilitating condition that can severely limit an individual's mobility and quality of life. This study was designed to evaluate the efficacy of bone marrow-derived mesenchymal stem cell (BM-MSC) treatment in cartilage repair using a rat model of monoiodoacetate- (MIA-) induced knee OA. OA was induced in the knee joint of rats by an intracapsular injection of MIA (2 mg/50 μL) on day zero. The rats were divided into three groups (n = 6): a normal control group, an osteoarthritic control group, and an osteoarthritic group receiving a single intra-articular injection of BM-MSCs (5 × 106 cells/rat). The knee diameter was recorded once per week. By the end of the performed experiment, X-ray imaging and enzyme-linked immunosorbent assay analysis of serum inflammatory cytokines interleukin-1beta (IL-β), IL-6, and tumor necrosis factor-α (TNF-α) and anti-inflammatory cytokines interleukin-10 and transforming growth factor-beta (TGF-β) were carried out. In addition, RT-PCR was used to measure nuclear factor-kappa B (NF-κB), inducible nitric oxide synthase (iNOS), and type II collagen mRNA levels and Western blot analysis was used to determine caspase-3 protein levels in all treated groups. Finally, hematoxylin/and eosin stains were used for histopathological investigation. Administration of BM-MSCs significantly downregulated knee joint swelling and MIA-induced (IL-1β, IL-6, and TNF-α) and upregulated IL-10 and TGF-β as well. Moreover, BM-MSC-treated osteoarthritic rats exhibited decreased expression of NF-κB, iNOS, and apoptotic mediator (caspase-3) and increased expression of type II collagen when compared to rats treated with MIA alone. The hematoxylin/eosin-stained sections revealed that BM-MSC administration ameliorated the knee joint alterations in MIA-injected rats. BM-MSCs could be an effective treatment for inflamed knee joints in the MIA-treated rat model of osteoarthritis, and the effect may be mediated via its anti-inflammatory and antioxidant potential.
Collapse
Affiliation(s)
- Hadeer Mohamed Hamdalla
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni Suef, Egypt
| | - Rasha Rashad Ahmed
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni Suef, Egypt
| | - Sanaa Rida Galaly
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni Suef, Egypt
| | - Osama Mohamed Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni Suef, Egypt
| | - Ibrahim A. Naguib
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Manal Abdul-Hamid
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni Suef, Egypt
| |
Collapse
|
21
|
Loucks A, Maerz T, Hankenson K, Moeser A, Colbath A. WITHDRAWN: The Multifaceted Role of Mast Cells in Joint Inflammation and Arthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2022. [DOI: 10.1016/j.ocarto.2022.100309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
22
|
Rivas CM, Yee MC, Addison KJ, Lovett M, Pal K, Ledford JG, Dussor G, Price TJ, Vagner J, DeFea KA, Boitano S. Proteinase-activated receptor-2 antagonist C391 inhibits Alternaria-induced airway epithelial signalling and asthma indicators in acute exposure mouse models. Br J Pharmacol 2022; 179:2208-2222. [PMID: 34841515 DOI: 10.1111/bph.15745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/19/2021] [Accepted: 11/04/2021] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Despite the availability of a variety of treatment options, many asthma patients have poorly controlled disease with frequent exacerbations. Proteinase-activated receptor-2 (PAR2) has been identified in preclinical animal models as important to asthma initiation and progression following allergen exposure. Proteinase activation of PAR2 raises intracellular Ca2+ , inducing MAPK and β-arrestin signalling in the airway, leading to inflammatory and protective effects. We have developed C391, a potent PAR2 antagonist effective in blocking peptidomimetic- and trypsin-induced PAR2 signalling in vitro as well as reducing inflammatory PAR2-associated pain in vivo. We hypothesized that PAR2 antagonism by C391 would attenuate allergen-induced acutely expressed asthma indicators in murine models. EXPERIMENTAL APPROACH We evaluated the ability of C391 to alter Alternaria alternata-induced PAR2 signalling pathways in vitro using a human airway epithelial cell line that naturally expresses PAR2 (16HBE14o-) and a transfected embryonic cell line (HEK 293). We next evaluated the ability for C391 to reduce A. alternata-induced acutely expressed asthma indicators in vivo in two murine strains. KEY RESULTS C391 blocked A. alternata-induced, PAR2-dependent Ca2+ and MAPK signalling in 16HBE14o- cells, as well as β-arrestin recruitment in HEK 293 cells. C391 effectively attenuated A. alternata-induced inflammation, mucus production, mucus cell hyperplasia and airway hyperresponsiveness in acute allergen-challenged murine models. CONCLUSIONS AND IMPLICATIONS To our best knowledge, this is the first demonstration of pharmacological intervention of PAR2 to reduce allergen-induced asthma indicators in vivo. These data support further development of PAR2 antagonists as potential first-in-class allergic asthma drugs.
Collapse
Affiliation(s)
- Candy M Rivas
- Physiological Sciences Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA.,Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Michael C Yee
- Biomedical Sciences, University of California Riverside, Riverside, California, USA
| | - Kenneth J Addison
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, Arizona, USA.,Department of Cellular and Molecular Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Marissa Lovett
- Physiological Sciences Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA
| | - Kasturi Pal
- Biomedical Sciences, University of California Riverside, Riverside, California, USA
| | - Julie G Ledford
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, Arizona, USA.,Department of Cellular and Molecular Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas, USA
| | - Josef Vagner
- Bio5 Collaborative Research Institute, University of Arizona, Tucson, Arizona, USA
| | - Kathryn A DeFea
- Biomedical Sciences, University of California Riverside, Riverside, California, USA.,Corporate Headquarters, PARMedics, Inc., Temecula, California, USA
| | - Scott Boitano
- Physiological Sciences Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA.,Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, Arizona, USA.,Department of Cellular and Molecular Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA.,Bio5 Collaborative Research Institute, University of Arizona, Tucson, Arizona, USA.,Department of Physiology, University of Arizona Health Sciences, Tucson, Arizona, USA
| |
Collapse
|
23
|
Carpintero-Fernández P, Varela-Eirín M, García-Yuste A, López-Díaz I, Caeiro JR, Mayán MD. Osteoarthritis: Mechanistic Insights, Senescence, and Novel Therapeutic Opportunities. Bioelectricity 2022; 4:39-47. [PMID: 39355566 PMCID: PMC11441363 DOI: 10.1089/bioe.2021.0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disease. In the last years, the research community has focused on understanding the molecular mechanisms that led to the pathogenesis of the disease, trying to identify different molecular and clinical phenotypes along with the discovery of new therapeutic opportunities. Different types of cell-to-cell communication mechanisms have been proposed to contribute to OA progression, including mechanisms mediated by connexin43 (Cx43) channels or by small extracellular vesicles. Furthermore, changes in the chondrocyte phenotype such as cellular senescence have been proposed as new contributors of the OA progression, changing the paradigm of the disease. The use of different drugs able to restore chondrocyte phenotype, to reduce cellular senescence and senescence-associated secretory phenotype components, and to modulate ion channel activity or Cx43 appears to be promising therapeutic strategies for the different types of OA. In this review, we aim to summarize the current knowledge in OA phenotypes related with aging and tissue damage and the new therapeutic opportunities currently available.
Collapse
Affiliation(s)
- Paula Carpintero-Fernández
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Marta Varela-Eirín
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Alejandro García-Yuste
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Iñaki López-Díaz
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - José Ramón Caeiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| |
Collapse
|
24
|
Li Z, Dai A, Yang M, Chen S, Deng Z, Li L. p38MAPK Signaling Pathway in Osteoarthritis: Pathological and Therapeutic Aspects. J Inflamm Res 2022; 15:723-734. [PMID: 35140502 PMCID: PMC8820459 DOI: 10.2147/jir.s348491] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/16/2022] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is an aging-related joint disease, pathologically featured with degenerated articular cartilage and deformation of subchondral bone. OA has become the fourth major cause of disability in the world, imposing a huge economic burden. At present, the pathogenesis and pathophysiology of OA are still unclear. Complex regulating networks containing different biochemical signaling pathways are involved in OA pathogenesis and progression. The p38MAPK signaling pathway is a member of the MAPK signaling pathway family, which participates in the induction of cellular senescence, the differentiation of chondrocytes, the synthesis of matrix metalloproteinase (MMPs) and the production of pro-inflammatory factors. In recent years, studies on the regulating role of p38MAPK signaling pathway and the application of its inhibitors have attracted growing attention, with an increasing number of in vivo and in vitro studies. One interesting finding is that the inhibition of p38MAPK could suppress chondrocyte inflammation and ameliorate OA, indicating its therapeutic role in OA treatment. Based on this, we reviewed the mechanisms of p38MAPK signaling pathway in the pathogenesis of OA, hoping to provide new ideas for future research and OA treatment.
Collapse
Affiliation(s)
- Zongchao Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, People’s Republic of China
| | - Aonan Dai
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, People’s Republic of China
| | - Ming Yang
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, People’s Republic of China
| | - Siyu Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
- School of Clinical Medicine, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
- School of Clinical Medicine, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
- Correspondence: Zhenhan Deng, Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, 3002 Sungang West Road, Shenzhen City, 518035, People’s Republic of China, Tel +86 13928440786, Fax +86 755-83366388, Email ; Liangjun Li, Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan South Road, Changsha City, 410018, People’s Republic of China, Tel +86 13875822004, Fax +86 731-85668156, Email
| | - Liangjun Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, People’s Republic of China
- Correspondence: Zhenhan Deng, Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, 3002 Sungang West Road, Shenzhen City, 518035, People’s Republic of China, Tel +86 13928440786, Fax +86 755-83366388, Email ; Liangjun Li, Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan South Road, Changsha City, 410018, People’s Republic of China, Tel +86 13875822004, Fax +86 731-85668156, Email
| |
Collapse
|
25
|
Wang F, Liu M, Wang N, Luo J. G Protein-Coupled Receptors in Osteoarthritis. Front Endocrinol (Lausanne) 2022; 12:808835. [PMID: 35154008 PMCID: PMC8831737 DOI: 10.3389/fendo.2021.808835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic joint disease characterized, for which there are no available therapies being able to modify the progression of OA and prevent long-term disability. Critical roles of G-protein coupled receptors (GPCRs) have been established in OA cartilage degeneration, subchondral bone sclerosis and chronic pain. In this review, we describe the pathophysiological processes targeted by GPCRs in OA, along with related preclinical model and/or clinical trial data. We review examples of GPCRs which may offer attractive therapeutic strategies for OA, including receptors for cannabinoids, hormones, prostaglandins, fatty acids, adenosines, chemokines, and discuss the main challenges for developing these therapies.
Collapse
Affiliation(s)
- Fanhua Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ning Wang
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, United Kingdom
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
26
|
Krumb M, Kammer LM, Badir SO, Cabrera-Afonso MJ, Wu VE, Huang M, Csakai A, Marcaurelle LA, Molander GA. Photochemical C-H arylation of heteroarenes for DNA-encoded library synthesis. Chem Sci 2022; 13:1023-1029. [PMID: 35211268 PMCID: PMC8790789 DOI: 10.1039/d1sc05683b] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/06/2021] [Indexed: 12/22/2022] Open
Abstract
DNA-encoded library (DEL) technology has emerged as a time- and cost-efficient technique for the identification of therapeutic candidates in the pharmaceutical industry. Although several reaction classes have been successfully validated in DEL environments, there remains a paucity of DNA-compatible reactions that harness building blocks (BBs) from readily available substructures bearing multifunctional handles for further library diversification under mild, dilute, and aqueous conditions. In this study, the direct C-H carbofunctionalization of medicinally-relevant heteroarenes can be accomplished via the photoreduction of DNA-conjugated (hetero)aryl halides to deliver reactive aryl radical intermediates in a regulated fashion within minutes of blue light illumination. A broad array of electron-rich and electron-poor heteroarene scaffolds undergo transformation in the presence of sensitive functional groups.
Collapse
Affiliation(s)
- Matthias Krumb
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania 231 South 34th Street Philadelphia PA 19104-6323 USA
| | - Lisa Marie Kammer
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania 231 South 34th Street Philadelphia PA 19104-6323 USA
| | - Shorouk O Badir
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania 231 South 34th Street Philadelphia PA 19104-6323 USA
| | - María Jesús Cabrera-Afonso
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania 231 South 34th Street Philadelphia PA 19104-6323 USA
| | - Victoria E Wu
- Encoded Library Technologies/NCE Molecular Discovery, R&D Medicinal Science and Technology, GlaxoSmithKline 200 Cambridge Park Drive Cambridge MA 02140 USA
| | - Minxue Huang
- Encoded Library Technologies/NCE Molecular Discovery, R&D Medicinal Science and Technology, GlaxoSmithKline 200 Cambridge Park Drive Cambridge MA 02140 USA
| | - Adam Csakai
- Encoded Library Technologies/NCE Molecular Discovery, R&D Medicinal Science and Technology, GlaxoSmithKline 200 Cambridge Park Drive Cambridge MA 02140 USA
| | - Lisa A Marcaurelle
- Encoded Library Technologies/NCE Molecular Discovery, R&D Medicinal Science and Technology, GlaxoSmithKline 200 Cambridge Park Drive Cambridge MA 02140 USA
| | - Gary A Molander
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania 231 South 34th Street Philadelphia PA 19104-6323 USA
| |
Collapse
|
27
|
Wen ZQ, Liu D, Zhang Y, Cai ZJ, Xiao WF, Li YS. G Protein-Coupled Receptors in Osteoarthritis: A Novel Perspective on Pathogenesis and Treatment. Front Cell Dev Biol 2021; 9:758220. [PMID: 34746150 PMCID: PMC8564363 DOI: 10.3389/fcell.2021.758220] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/27/2021] [Indexed: 11/30/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are transmembrane receptor proteins that trigger numerous intracellular signaling pathways in response to the extracellular stimuli. The GPCRs superfamily contains enormous structural and functional diversity and mediates extensive biological processes. Until now, critical roles have been established in many diseases, including osteoarthritis (OA). Existing studies have shown that GPCRs play an important role in some OA-related pathogenesis, such as cartilage matrix degradation, synovitis, subchondral bone remodeling, and osteophyte formation. However, current pharmacological treatments are mostly symptomatic and there is a paucity of disease-modifying OA drugs so far. Targeting GPCRs is capable of inhibiting cartilage matrix degradation and synovitis and up-regulating cartilage matrix synthesis, providing a new therapeutic strategy for OA. In this review, we have comprehensively summarized the structures, biofunctions, and the novel roles of GPCRs in the pathogenesis and treatment of OA, which is expected to lay the foundation for the development of novel therapeutics against OA. Even though targeting GPCRs may ameliorate OA progression, many GPCRs-related therapeutic strategies are still in the pre-clinical stage and require further investigation.
Collapse
Affiliation(s)
- Ze-qin Wen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zi-jun Cai
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-feng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Sun L, Gai J, Shi S, Zhao J, Bai X, Liu B, Li X. Protease-Activated Receptor 2 (PAR-2) Antagonist AZ3451 Mitigates Oxidized Low-Density Lipoprotein (Ox-LDL)-Induced Damage and Endothelial Inflammation. Chem Res Toxicol 2021; 34:2202-2208. [PMID: 34590836 DOI: 10.1021/acs.chemrestox.1c00154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Oxidized low-density lipoprotein (ox-LDL)-induced endothelial dysfunction plays an important role in the initiation and development of cardiovascular diseases, especially atherosclerosis (AS). Protease-activated receptor 2 (PAR-2) is a receptor for inflammatory proteases. However, the biological function of PAR-2 in endothelial cells and the pathophysiological process of AS are still unknown. In the current study, we found that treatment with ox-LDL increased the gene and protein expressions of PAR-2 in EA.hy926 endothelial cells. Interestingly, we found that antagonism of PAR-2 with its specific antagonist AZ3451 could ameliorate ox-LDL-induced lactate dehydrogenase (LDH) release. Treatment with AZ3451 considerably improved the mitochondrial function by restoring the mitochondrial membrane potential and increasing the levels of intracellular adenosine triphosphate (ATP). Also, we found that AZ3451 attenuated ox-LDL-induced expression and production of pro-inflammatory cytokines such as interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interleukin-8 (IL-8). Treatment with AZ3451 also mitigated the expression of matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9). Notably, our results demonstrated that the presence of AZ3451 alleviated ox-LDL-induced expression of the endothelial cell adhesion molecules vascular cell adhesion molecule-1 (VCAM-1) and intercellular cell adhesion molecule-1 (ICAM-1). Mechanistically, we found that AZ3451 attenuated ox-LDL-induced activation of nuclear factor-κB (NF-κB) by reducing the levels of intracellular NF-κB p65 and the luciferase activity of NF-κB promoter. Based on these findings, we conclude that PAR-2 might become a novel therapeutic target for the treatment of AS.
Collapse
Affiliation(s)
- Lixiu Sun
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Jiaxin Gai
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Shuai Shi
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Jia Zhao
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Xiaopeng Bai
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Bingchen Liu
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Xueqi Li
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| |
Collapse
|
29
|
Song J, He Z, Yang M, Yu T, Wang X, Liu B, Li J. HepaticIschemia/Reperfusion Injuryinvolves functional tryptase/PAR-2 signaling in liver sinusoidal endothelial cell population. Int Immunopharmacol 2021; 100:108052. [PMID: 34454294 DOI: 10.1016/j.intimp.2021.108052] [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: 04/04/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
Mast cells (MCs) are tissue-resident effector cells that could be the earliest responder to release a unique, stimulus-specific set of mediators in hepatic ischemia-reperfusion (IR) injury However, how MCs function in the hepatic IR has remained a formidable challenge due to the substantial redundancy and functional diverse of these mediators. Tryptase is the main protease for degranulation of MCs and its receptor-protease-activated receptor 2 (PAR-2) is widely expressed in endothelial cells. It is unclear whether and how tryptase/PAR-2 axis participates in hepatic IR. We employed an experimental warm 70% liver IR model in mice and found that tryptase was accumulated in the circulation during hepatic IR and positively correlated with liver injury. Tryptase inhibition by protamine can significantly down-regulate the expression of adhesion molecules and reduce neutrophil infiltration within the liver. The level of inflammatory factors and chemokines were also consistent with the pathological change of the liver. In addition, the treatment with exogeneous tryptase in MC-deficient mice can induce the damage observed in wild type mice in the context of liver IR. In vitro, neutrophil infiltration and inflammatory factor secretion were regulated by Tryptase/PAR-2, involving the adhesion molecule expression to regulate neutrophil adhesion dependent on NF-κB pathway. Conclusion: tryptase/PAR-2 participates in liver injury through the activation of LSECs in the early phase of liver IR.
Collapse
Affiliation(s)
- Jian Song
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China; Department of General Surgery, Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Zhigang He
- Department of Plastic and Constructive Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Muqing Yang
- Department of General Surgery, Shanghai Tenth People's Hospital School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Tianyu Yu
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China
| | - Xiaodong Wang
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China
| | - Bin Liu
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China
| | - Jiyu Li
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China.
| |
Collapse
|
30
|
Lucena F, McDougall JJ. Protease Activated Receptors and Arthritis. Int J Mol Sci 2021; 22:9352. [PMID: 34502257 PMCID: PMC8430764 DOI: 10.3390/ijms22179352] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
The catabolic and destructive activity of serine proteases in arthritic joints is well known; however, these enzymes can also signal pain and inflammation in joints. For example, thrombin, trypsin, tryptase, and neutrophil elastase cleave the extracellular N-terminus of a family of G protein-coupled receptors and the remaining tethered ligand sequence then binds to the same receptor to initiate a series of molecular signalling processes. These protease activated receptors (PARs) pervade multiple tissues and cells throughout joints where they have the potential to regulate joint homeostasis. Overall, joint PARs contribute to pain, inflammation, and structural integrity by altering vascular reactivity, nociceptor sensitivity, and tissue remodelling. This review highlights the therapeutic potential of targeting PARs to alleviate the pain and destructive nature of elevated proteases in various arthritic conditions.
Collapse
Affiliation(s)
| | - Jason J. McDougall
- Departments of Pharmacology and Anesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada;
| |
Collapse
|
31
|
The development of proteinase-activated receptor-2 modulators and the challenges involved. Biochem Soc Trans 2021; 48:2525-2537. [PMID: 33242065 PMCID: PMC7752072 DOI: 10.1042/bst20200191] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/13/2020] [Accepted: 11/02/2020] [Indexed: 11/30/2022]
Abstract
Protease-activated receptor-2 (PAR2) has been extensively studied since its discovery in the mid-1990. Despite the advances in understanding PAR2 pharmacology, it has taken almost 25 years for the first inhibitor to reach clinical trials, and so far, no PAR2 antagonist has been approved for human use. Research has employed classical approaches to develop a wide array of PAR2 agonists and antagonists, consisting of peptides, peptoids and antibodies to name a few, with a surge in patent applications over this period. Recent breakthroughs in PAR2 structure determination has provided a unique insight into proposed PAR2 ligand binding sites. Publication of the first crystal structures of PAR2 resolved in complex with two novel non-peptide small molecule antagonists (AZ8838 and AZ3451) revealed two distinct binding pockets, originally presumed to be allosteric sites, with a PAR2 antibody (Fab3949) used to block tethered ligand engagement with the peptide-binding domain of the receptor. Further studies have proposed orthosteric site occupancy for AZ8838 as a competitive antagonist. One company has taken the first PAR2 antibody (MEDI0618) into phase I clinical trial (NCT04198558). While this first-in-human trial is at the early stages of the assessment of safety, other research into the structural characterisation of PAR2 is still ongoing in an attempt to identify new ways to target receptor activity. This review will focus on the development of novel PAR2 modulators developed to date, with an emphasis placed upon the advances made in the pharmacological targeting of PAR2 activity as a strategy to limit chronic inflammatory disease.
Collapse
|
32
|
Chandrabalan A, Ramachandran R. Molecular mechanisms regulating Proteinase‐Activated Receptors (PARs). FEBS J 2021; 288:2697-2726. [DOI: 10.1111/febs.15829] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Arundhasa Chandrabalan
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| |
Collapse
|
33
|
Patel S, Badir SO, Molander GA. Developments in Photoredox-Mediated Alkylation for DNA-Encoded Libraries. TRENDS IN CHEMISTRY 2021; 3:161-175. [PMID: 33987530 PMCID: PMC8112611 DOI: 10.1016/j.trechm.2020.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recently, DNA-encoded library (DEL) technology has emerged as an innovative screening modality for the rapid discovery of therapeutic candidates in pharmaceutical settings. This platform enables a cost-effective, time-efficient, and large-scale assembly and interrogation of billions of small organic ligands against a biological target in a single experiment. An outstanding challenge in DEL synthesis is the necessity for water-compatible transformations under ambient conditions. To access uncharted chemical space, the adoption of photoredox catalysis in DELs, including Ni-catalyzed manifolds and radical/polar crossover reactions, has enabled the construction of novel structural scaffolds through regulated odd-electron intermediates. Herein, a critical discussion of the validation of photoredox-mediated alkylation in DEL environments is presented. Current synthetic gaps are highlighted and opportunities for further development are speculated upon.
Collapse
Affiliation(s)
- Shivani Patel
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | - Shorouk O. Badir
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | - Gary A. Molander
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| |
Collapse
|
34
|
Wang Y, Gao W. Effects of TNF-α on autophagy of rheumatoid arthritis fibroblast-like synoviocytes and regulation of the NF-κB signaling pathway. Immunobiology 2021; 226:152059. [PMID: 33561598 DOI: 10.1016/j.imbio.2021.152059] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/20/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022]
Abstract
Rheumatoid arthritis (RA) is a common chronic autoimmune disease, which seriously harms human health. The hyperplastic growth of fibroblast-like synoviocytes (FLSs) plays a key role in the pathogenesis of RA. However, the pathogenesis of RA remains unclear. In this experiment, we confirmed that Tumor necrosis factor alpha (TNF-α) could activate the autophagy of RA-FLSs. 3-Methyladenine (3-MA) and Chloroquine (CQ), two types of autophagy blocker, combined with TNF-α were used to treat FLSs. The results showed that this treatment caused a reduction in the level of autophagy-related protein, significant increases in the expression of apoptosis-related protein and the apoptosis rate, and significant inhibition of the proliferation-promoting ability of TNF-α. Ammonium pyrrolidinedithiocarbamate (PDTC), a specific nuclear factor kappa-B (NF-κB) activity blocker, significantly inhibited autophagy induced by TNF-α. Collectively, these findings showed, for the first time, that TNF-α can up-regulate autophagy activity and activate the NF-κB signal pathway. Inhibition of autophagy can improve the imbalance of proliferation/apoptosis of FLSs aggravated by TNF-α to some extent, thus delaying the progression of RA. The NF-κB signal pathway may be involved in the regulation of FLSs autophagy by TNF-α.
Collapse
Affiliation(s)
- Yu Wang
- Department of Rheumatology, the First Affiliated Hospital of Jinzhou Medical University, Liaoning, Jinzhou 121000, China
| | - Wei Gao
- Department of Rheumatology, the First Affiliated Hospital of Jinzhou Medical University, Liaoning, Jinzhou 121000, China.
| |
Collapse
|
35
|
Her JY, Lee Y, Kim SJ, Heo G, Choo J, Kim Y, Howe C, Rhee SH, Yu HS, Chung HY, Pothoulakis C, Im E. Blockage of protease-activated receptor 2 exacerbates inflammation in high-fat environment partly through autophagy inhibition. Am J Physiol Gastrointest Liver Physiol 2021; 320:G30-G42. [PMID: 33146548 DOI: 10.1152/ajpgi.00203.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Protease-activated receptor 2 (PAR2) regulates inflammatory responses and lipid metabolism. However, its precise role in colitis remains unclear. In this study, we aimed to investigate the function of PAR2 in high-fat diet-fed mice with colitis and its potential role in autophagy. PAR2+/+ and PAR2-/- mice were fed a high-fat diet (HFD) for 7 days before colitis induction with dextran sodium sulfate. Deletion of PAR2 and an HFD significantly exacerbated colitis, as shown by increased mortality, body weight loss, diarrhea or bloody stools, colon length shortening, and mucosal damage. Proinflammatory cytokine levels were elevated in HFD-fed PAR2-/- mice and in cells treated with the PAR2 antagonist GB83, palmitic acid (PA), and a cytokine cocktail (CC). Damaging effects of PAR2 blockage were associated with autophagy regulation by reducing the levels of YAP1, SIRT1, PGC-1α, Atg5, and LC3A/B-I/II. In addition, mitochondrial dysfunction was demonstrated only in cells treated with GB83, PA, and CC. Reduced cell viability and greater induction of apoptosis, as shown by increased levels of cleaved caspase-9, cleaved caspase-3, and cleaved poly(ADP-ribose) polymerase (PARP), were observed in cells treated with GB83, PA, and CC but not in those treated with only PA and CC. Collectively, protective effects of PAR2 were elucidated during inflammation accompanied by a high-fat environment by promoting autophagy and inhibiting apoptosis, suggesting PAR2 as a therapeutic target for inflammatory bowel disease co-occurring with metabolic syndrome.NEW & NOTEWORTHY Deletion of PAR2 with high-fat diet feeding exacerbates colitis in a murine colitis model. Proinflammatory effects of PAR2 blockage in a high-fat environment were associated with an altered balance between autophagy and apoptosis. Increased colonic levels of PAR2 represent as a therapeutic strategy for IBD co-occurring with metabolic syndrome.
Collapse
Affiliation(s)
- Ji Yun Her
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yunna Lee
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Su Jin Kim
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Gwangbeom Heo
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Jieun Choo
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yuju Kim
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Cody Howe
- Department of Biological Sciences, Oakland University, Rochester, Michigan
| | - Sang Hoon Rhee
- Department of Biological Sciences, Oakland University, Rochester, Michigan
| | - Hak Sun Yu
- Department of Parasitology and Tropical Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Hae Young Chung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Charalabos Pothoulakis
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Eunok Im
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
36
|
Zheng S, Ren J, Gong S, Qiao F, He J. CTRP9 protects against MIA-induced inflammation and knee cartilage damage by deactivating the MAPK/NF-κB pathway in rats with osteoarthritis. Open Life Sci 2020; 15:971-980. [PMID: 33817283 PMCID: PMC7874593 DOI: 10.1515/biol-2020-0105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 08/23/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
C1q/TNF-related protein 9 (CTRP9), the closest paralog of adiponectin, has been reported to protect against inflammation-related diseases. However, its role in regulating osteoarthritis (OA) has not been fully elucidated. First, a rat model of OA was generated. Furthermore, rats with OA were injected with different doses of recombinant CTRP9 protein (rCTRP9), and the knee cartilage damage was evaluated. Finally, the phosphorylation of p38 and the secretion of matrix metalloproteinases (MMPs) were detected by Western blotting and enzyme-linked immunosorbent assay. Results revealed that CTRP9 was highly expressed in adipose tissue, followed by skeletal muscle and cartilage tissue, and less expressed in liver, kidney and lung. Moreover, the expression of CTRP9 significantly decreased in the monosodium iodoacetate (MIA) group in the knee cartilage and knee synovial fluid, and the contents of interleukin-1β (IL-1β) and IL-6 significantly increased in knee synovial fluid. In addition, rCTRP9 alleviated MIA-induced inflammation, oxidative stress and knee cartilage damage in a dose-dependent way. In addition, rCTRP9 could attenuate the expression of p38MAPK and p-p38 and suppress the expression of nuclear factor-kappa B (NF-κB), p65 and MMPs. Collectively, the results of the present study suggested that CTRP9 alleviates the inflammation of MIA-induced OA through deactivating p38MAPK and NF-κB signaling pathways in rats.
Collapse
Affiliation(s)
- Shicheng Zheng
- Department of Integrated TCM & Western Medicine Orthopaedics, Xi’an Honghui Hospital, Xi’an Jiaotong University, 555 of Youyi East Road, Xi’an 710056, China
| | - Jing Ren
- Department of Integrated TCM & Western Medicine Orthopaedics, Xi’an Honghui Hospital, Xi’an Jiaotong University, 555 of Youyi East Road, Xi’an 710056, China
| | - Sihai Gong
- Department of Integrated TCM & Western Medicine Orthopaedics, Xi’an Honghui Hospital, Xi’an Jiaotong University, 555 of Youyi East Road, Xi’an 710056, China
| | - Feng Qiao
- Department of Integrated TCM & Western Medicine Orthopaedics, Xi’an Honghui Hospital, Xi’an Jiaotong University, 555 of Youyi East Road, Xi’an 710056, China
| | - Jinlong He
- Department of Integrated TCM & Western Medicine Orthopaedics, Xi’an Honghui Hospital, Xi’an Jiaotong University, 555 of Youyi East Road, Xi’an 710056, China
| |
Collapse
|
37
|
Tang Y, Mo Y, Xin D, Zeng L, Yue Z, Xu C. β-ecdysterone alleviates osteoarthritis by activating autophagy in chondrocytes through regulating PI3K/AKT/mTOR signal pathway. Am J Transl Res 2020; 12:7174-7186. [PMID: 33312358 PMCID: PMC7724317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 10/09/2020] [Indexed: 06/12/2023]
Abstract
PURPOSE To investigate the therapeutic effects of β-ecdysterone on osteoarthritis (OA) and the underlying mechanism. METHODS OA model was established on rats by injecting MIA. ELSA was used to determine the concentration of IL-1β, IL-6, NO and TNF-α in the chondrocytes and cartilage tissues. Immunofluorescence assay was used to determine the expression of collagen II in the chondrocytes. The survival rate of chondrocytes was evaluated by MTT assay. The apoptosis of chondrocytes was checked by AO/PI staining and flow cytometry assay. The expression level of Atg7, PI3K and caspase-3 was evaluated by qRT-PCR. Western Blot was used determine the expression of PI3K, p-AKT1, AKT1, p-mTOR, mTOR, p70S6K, p-p70S6K, LC3I, LC3II and caspase-3. HE staining was used to check the pathological state of cartilage tissues. RESULTS Chondrocytes were tolerable to rapamycin, 3-methyladenine and β-ecdysterone at the concentration of 10 mM, 100 nM and 40 μM, respectively. The apoptosis of chondrocytes was inhibited by rapamycin and β-ecdysterone, and induced by 3-methyladenine. PI3K, p-AKT1, p-mTOR, p-p70S6K and caspase-3 were down-regulated by rapamycin and β-ecdysterone, and up-regulated by 3-methyladenine in both the chondrocytes and the cartilage tissues. The expression of Atg7 and LC3II/LC3I were regulated in a opposite way. The inflammation state was improved by rapamycin and β-ecdysterone both the chondrocytes and the cartilage tissues. HE staining results showed that the pathological state of cartilage tissues was alleviated by β-ecdysterone. CONCLUSION β-ecdysterone might alleviate osteoarthritis by activating autophagy in chondrocytes through regulating PI3K/AKT/mTOR signal pathway.
Collapse
Affiliation(s)
- Yanghua Tang
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| | - Yafeng Mo
- Department of Orthopedics, Third Clinical College of Zhejiang Chinese Medical UniversityNo. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, China
| | - Dawei Xin
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| | - Linru Zeng
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| | - Zhenshuang Yue
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| | - Canda Xu
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| |
Collapse
|
38
|
Xiao P, Zhu X, Sun J, Zhang Y, Qiu W, Li J, Wu X. MicroRNA-613 alleviates IL-1β-induced injury in chondrogenic CHON-001 cells by targeting fibronectin 1. Am J Transl Res 2020; 12:5308-5319. [PMID: 33042421 PMCID: PMC7540165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 07/26/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is an aging-related chronic degenerative joint disease. A number of miRNAs have been found to be involved in the development of OA, but the role of miR-613 in OA remains unclear. Thus, this study aimed to investigate the role of miR-613 during the progression of OA. METHODS CHON-001 cells were transfected with miR-613 agonist for 48 h, and then exposed to 10 ng/mL IL-1β for 24 h. Cell viability, cell proliferation and cell apoptosis in CHON-001 cells were assessed by CCK-8, immunofluorescence, and flow cytometry assays, respectively. In addition, the dual luciferase reporter system assay was used to determine the interaction of miR-613 and fibronectin 1 in CHON-001 cells. RESULTS The level of miR-613 was significantly decreased in IL-1β-treated CHON-001 cells. Overexpression of miR-613 markedly inhibited IL-1β-induced apoptosis in CHON-001 cells. In addition, upregulation of miR-613 obviously alleviated IL-1β-induced inflammatory response and cartilage matrix degradation in CHON-001 cells. Meanwhile, fibronectin 1 was identified as a direct binding target of miR-613 in CHON-001 cells. Overexpression of miR-613 alleviated IL-1β-induced injury in CHON-001 cells via downregulating the expression of fibronectin 1. Furthermore, overexpression of miR-613 alleviated cartilage degradation, and reduced OARSI scores and subchondral bone thickness in a mouse model of OA. CONCLUSION Our data indicated that overexpression of miR-613 could inhibit IL-1β-induced injury in CHON-001 cells via decreasing the level fibronectin 1 in vitro, and alleviate the symptoms of OA in vivo. Therefore, miR-613 might be a potential therapeutic option for the treatment of OA.
Collapse
Affiliation(s)
- Peng Xiao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Xu Zhu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Jinpeng Sun
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Yuhang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Weijian Qiu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Jianqiang Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Xuejian Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| |
Collapse
|
39
|
Yang Y, You X, Cohen JD, Zhou H, He W, Li Z, Xiong Y, Yu T. Sex Differences in Osteoarthritis Pathogenesis: A Comprehensive Study Based on Bioinformatics. Med Sci Monit 2020; 26:e923331. [PMID: 32255771 PMCID: PMC7163332 DOI: 10.12659/msm.923331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Osteoarthritis (OA) is a common disorder in the elderly. OA influences the daily life of patients and has become a worldwide health problem. It is still unclear whether the pathogenesis mechanism is different between males and females. This study investigated the differentially expressed genes (DEGs) and explored the different signaling pathways of OA between males and females. Material/Methods Data sets of GSE55457, GSE55584, and GSE12021 were retrieved from Gene Expression Omnibus to conduct DEGs analysis. Enrichment analysis of Kyoto Encyclopedia of Genes and Genomes pathway and Gene Ontology term was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID) bioinformatics tool. The protein interaction network was constructed in Cytoscape 3.7.2. qRT-PCR was then performed to validate the expression of hub genes in OA patients and healthy people. Results In total, 4 co-upregulated and 10 co-downregulated genes were identified. We found that enriched pathways were different between males and females. BCL2L1, EEF1A1, EEF2, HNRNPD, and PABPN1 were considered as hub genes in OA pathogenesis in males, while EEF2, EEF1A1, RPL37A, FN1 were considered as hub genes in OA pathogenesis in females. Consistent with the bioinformatics analysis, the qRT-PCR analysis also showed that the gene expression of BCL2L1, HNRNPD, and PABPN1 was significantly lower in male OA patients. In contrast, EEF2, EEF1A1, and RPL37A were significantly lower in female OA patients. Conclusions The DEGs identified may be involved in different OA disease progression mechanisms between males and females, and they are considered as treatment targets or prognosis markers for males and females. The pathogenesis mechanism is sex-dependent.
Collapse
Affiliation(s)
- Yunfeng Yang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Xiaomeng You
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jordan Daniel Cohen
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Haichao Zhou
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Wenbao He
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Zihua Li
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Tao Yu
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|