1
|
Chen X, Qin Y, Wang L, Zhu Y, Zhang H, Liu W, Zeng M, Dai Q. Co-amorphous systems of sulfasalazine with matrine-type alkaloids: Enhanced solubility behaviors and synergistic therapeutic potential. Eur J Pharm Biopharm 2024; 203:114475. [PMID: 39216557 DOI: 10.1016/j.ejpb.2024.114475] [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: 06/03/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Sulfasalazine (SULF), a sulfonamide antibiotic, has been utilized in the treatment of rheumatoid arthritis (RA) and inflammatory bowel disease (IBD) since its discovery. However, its poor water solubility causes the high daily doses (1---3 g) for patients, which may lead to the intolerable toxic and side effects for their lifelong treatment for RA and IBD. In this work, two water-soluble natural anti-inflammatory alkaloids, matrine (MAR) and sophoridine (SPD), were employed to construct the co-amorphous systems of SULF for addressing its solubility issue. These newly obtained co-amorphous forms of SULF were comprehensively characterized by powder X-ray diffraction (PXRD), temperature-modulated differential scanning calorimetry (mDSC), Fourier-transform infrared spectroscopy (FTIR), and X-ray photoelectron spectroscopy (XPS). We also investigated their dissolution behavior, including powder dissolution, in vitro release, and intrinsic dissolution rate. Both co-amorphous systems exhibited superior dissolution performance compared to crystalline SULF. The underlying mechanism responsible for the enhanced dissolution behaviors in co-amorphous systems were also elucidated. These mechanisms include the inhibition of nucleation, complexation, increased hydrophilicity, and robust intermolecular interactions in aqueous solutions. Importantly, these co-amorphous systems demonstrated satisfactory physical stability under various storage conditions. Network pharmacological analysis was utilized to investigate the potential therapeutic targets of both co-amorphous systems against RA, revealing similar yet distinct multi-target synergistic therapeutic mechanisms in the treatment of this condition. Our study suggests these drug-drug co-amorphous systems hold promise for optimizing SULF dosage in the future and providing a potential drug combination strategy.
Collapse
Affiliation(s)
- Xin Chen
- Innovation Centre for Science and Technology, North Sichuan Medical College, Nanchong, Sichuan 637100, P. R. China.
| | - Yirui Qin
- Innovation Centre for Science and Technology, North Sichuan Medical College, Nanchong, Sichuan 637100, P. R. China; Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637100, P. R. China
| | - Lijun Wang
- Innovation Centre for Science and Technology, North Sichuan Medical College, Nanchong, Sichuan 637100, P. R. China; Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637100, P. R. China
| | - Yujing Zhu
- Laboratory of Pharmaceutical Solid-State Chemistry, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, P. R. China
| | - Hailu Zhang
- Laboratory of Pharmaceutical Solid-State Chemistry, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, P. R. China; Interdisciplinary Institute of NMR and Molecular Sciences (NMR-X), School of Chemistry and Chemical Engineering, State Key Laboratory of Refractories and Metallurgy, Wuhan University of Science and Technology, Wuhan 430081, P. R. China
| | - Wenhu Liu
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan 637100, P. R. China.
| | - Mei Zeng
- Innovation Centre for Science and Technology, North Sichuan Medical College, Nanchong, Sichuan 637100, P. R. China; Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637100, P. R. China
| | - Qian Dai
- Institute of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637100, P. R. China.
| |
Collapse
|
2
|
Lin CY, Law YY, Yu CC, Wu YY, Hou SM, Chen WL, Yang SY, Tsai CH, Lo YS, Fong YC, Tang CH. NAMPT enhances LOX expression and promotes metastasis in human chondrosarcoma cells by inhibiting miR-26b-5p synthesis. J Cell Physiol 2024; 239:e31345. [PMID: 38940190 DOI: 10.1002/jcp.31345] [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: 12/13/2023] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024]
Abstract
Chondrosarcoma is a malignant bone tumor that emerges from abnormalities in cartilaginous tissue and is related with lung metastases. Nicotinamide phosphoribosyltransferase (NAMPT) is an adipocytokine reported to enhance tumor metastasis. Our results from clinical samples and the Gene Expression Omnibus data set reveal that NAMPT levels are markedly higher in chondrosarcoma patients than in normal individuals. NAMPT stimulation significantly increased lysyl oxidase (LOX) production in chondrosarcoma cells. Additionally, NAMPT increased LOX-dependent cell migration and invasion in chondrosarcoma by suppressing miR-26b-5p generation through the c-Src and Akt signaling pathways. Overexpression of NAMPT promoted chondrosarcoma metastasis to the lung in vivo. Furthermore, knockdown of LOX counteracted NAMPT-facilitated metastasis. Thus, the NAMPT/LOX axis presents a novel target for treating the metastasis of chondrosarcoma.
Collapse
Affiliation(s)
- Chih-Yang Lin
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yat-Yin Law
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Cheng-Chieh Yu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Yu-Ying Wu
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Orthopedics, Penghu Hospital, Ministry of Health and Welfare, Penghu, Taiwan
| | - Sheng-Mou Hou
- The Director's Office, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Department of Research, Taiwan Blood Services Foundation, Taipei, Taiwan
| | - Wei-Li Chen
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Shang-Yu Yang
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yuan-Shun Lo
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yunlin, Taiwan
- Graduate Institute of Precision Engineering, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu, Taiwan
| |
Collapse
|
3
|
Balendran T, Lim K, Hamilton JA, Achuthan AA. Targeting transcription factors for therapeutic benefit in rheumatoid arthritis. Front Immunol 2023; 14:1196931. [PMID: 37457726 PMCID: PMC10339812 DOI: 10.3389/fimmu.2023.1196931] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is a destructive inflammatory autoimmune disease that causes pain and disability. Many of the currently available drugs for treating RA patients are aimed at halting the progression of the disease and alleviating inflammation. Further, some of these treatment options have drawbacks, including disease recurrence and adverse effects due to long-term use. These inefficiencies have created a need for a different approach to treating RA. Recently, the focus has shifted to direct targeting of transcription factors (TFs), as they play a vital role in the pathogenesis of RA, activating key cytokines, chemokines, adhesion molecules, and enzymes. In light of this, synthetic drugs and natural compounds are being explored to target key TFs or their signaling pathways in RA. This review discusses the role of four key TFs in inflammation, namely NF-κB, STATs, AP-1 and IRFs, and their potential for being targeted to treat RA.
Collapse
Affiliation(s)
- Thivya Balendran
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Keith Lim
- Department of Medicine, Western Health, The University of Melbourne, St Albans, VIC, Australia
| | - John A. Hamilton
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Adrian A. Achuthan
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
4
|
Mulberroside A alleviates osteoarthritis via restoring impaired autophagy and suppressing MAPK/NF-κB/PI3K-AKT-mTOR signaling pathways. iScience 2023; 26:105936. [PMID: 36698724 PMCID: PMC9868682 DOI: 10.1016/j.isci.2023.105936] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/11/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023] Open
Abstract
Osteoarthritis (OA) is a trauma-/age-related degenerative disease characterized by chronic inflammation as one of its pathogenic mechanisms. Mulberroside A (MA), a natural bioactive withanolide, demonstrates anti-inflammatory properties in various diseases; however, little is known about the effect of MA on OA. We aim to examine the role of MA on OA and to identify the potential mechanisms through which it protects articular cartilage. In vitro, MA improved inflammatory response, anabolism, and catabolism in IL-1β-induced OA chondrocytes. The chondroprotective effects of MA were attributed to suppressing the MAPK, NF-κB, and PI3K-AKT-mTOR signaling pathways, as well as promoting the autophagy process. In vivo, intra-articular injection of MA reduced the cartilage destruction and reversed the change of anabolic and catabolic-related proteins in destabilized medial meniscus (DMM)-induced OA models. Thus, the study indicates that MA exhibits a chondroprotective effect and might be a promising agent for OA treatment.
Collapse
|
5
|
Aroke EN, Hobson JM, Ptacek T, Jackson P, Goodin BR. Genome-wide DNA methylation study identifies significant epigenomic changes associated with internalized stigma in adults with non-specific chronic low back pain. FRONTIERS IN PAIN RESEARCH 2022; 3:1021963. [PMID: 36518098 PMCID: PMC9742283 DOI: 10.3389/fpain.2022.1021963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/26/2022] [Indexed: 11/29/2022] Open
Abstract
Non-specific chronic low back pain (cLBP) represents a common musculoskeletal condition with no identifiable cause. It cannot be diagnosed with conventional neuroimaging techniques such as computerized tomography (CT). The diagnostic uncertainty that characterizes non-specific cLBP can lead to stigmatizing responses from others that can become internalized Among individuals with non-specific cLBP, internalized stigma is associated with greater pain intensity and disability. Yet, no study has examined the biological mechanism linking high internalized stigma to worse outcomes in individuals with non-specific cLBP. We aimed to identify differentially methylated loci (DML), enrichment pathways, and associated network interactions among individuals with non-specific cLBP experiencing low vs. high internalized stigma. We examined DNA methylation in whole blood samples from 48 adults, ages 19-85, using reduced representation bisulfite sequencing (RRBS). After controlling for age, sex, race, and multiple testing, differentially methylated loci (DML) differed in adults with low vs. high internalized stigma by at least 10% and q < 0.01 in 3,665 CpG sites: 2,280 hypomethylated and 1,385 hypermethylated. Gene ontology (GO) analyses of the annotated genes from these sites revealed significant enrichment of 274 biological processes, 29 cellular components, and 24 molecular functions (adjusted p < 0.05). The top enriched molecular functions regulate protein binding and DNA binding of transcription factor activity. Pathway analyses indicated that many functional genomic pathways, including Hippo Signaling, Melanogenesis, and Pathways in Cancer, were enriched with differentially methylated genes. Also, there was a significant interaction between relevance pathways such as P53, mTOR, PI3K-Akt, and Wnt signaling pathways. These pathways have previously been associated with neuroinflammation, neurodegeneration, and stress-related conditions. Thus, findings point to possible stress-induced DNAm changes as the link between high levels of internalized stigma and worse outcomes in adults with non-specific cLBP.
Collapse
Affiliation(s)
- Edwin N. Aroke
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joanna M. Hobson
- Biobehavioral Pain Lab, Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Travis Ptacek
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Pamela Jackson
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Burel R. Goodin
- Biobehavioral Pain Lab, Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, United States
- Center for Addiction and Pain Prevention and Intervention (CAPPI), University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
6
|
Lu Z, Zhang A, Wang J, Han K, Gao H. Estrogen alleviates post-traumatic osteoarthritis progression and decreases p-EGFR levels in female mouse cartilage. BMC Musculoskelet Disord 2022; 23:685. [PMID: 35854298 PMCID: PMC9295391 DOI: 10.1186/s12891-022-05608-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 11/16/2022] Open
Abstract
Objective To investigate the effect of estrogen on the progression of post-traumatic osteoarthritis (PTOA) in mice and its possible mechanism. Methods Twelve-week-old ICR mice were divided into Group A (female control group), group B (ovariectomized(OVX) group), group C (OVX group supplemented with estrogen), and group D (male group) by destabilization of the medial meniscus (DMM)or sham operation. Safranin O staining was performed at 8 weeks and 12 weeks after operation, and the degree of articular cartilage lesion was evaluated using Mankin score. Twelve weeks after the operation, tissue sections were stained to analyze the matrix metalloproteinase 13(MMP13), phosphorylated epidermal growth factor receptor (p-EGFR) expression and apoptosis of chondrocytes. Results Decreased estrogen can significantly increase the weight of mice in female mice. The degree of cartilage damage in the knee joint on the DMM side of female mice was significantly severer than that on the Sham side. The DMM side also showed higher MMP13 expression and increased apoptotic chondrocytes. The degree of cartilage damage in the knee joint on the DMM side of female mice was significantly reduced after estrogen supplementation, and cartilage damage in the knee joint on the DMM side of female mice was less serious than that of male mice. As estrogen levels decreased, the severity of cartilage erosion in the knee joint on the DMM side was aggravated, and p-EGFR expression in the cartilage surface was also higher in female mice contrast to that in male mice. However, minimal changes in p-EGFR expression in the cartilage surface of bilateral knee joints of male mice were observe. Conclusion Estrogen has a regulatory effect on PTOA and its inhibits the expression of p-EGFR in cartilage on the knee joint surface and has a protective effect on articular cartilage in female mice.
Collapse
Affiliation(s)
- Zhihua Lu
- Yangzhou Polytechnic College, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Aihua Zhang
- Department of Rehabilitation Medicine, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, People's Republic of China
| | - Jingcheng Wang
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, People's Republic of China
| | - Kuijing Han
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, People's Republic of China.
| | - Han Gao
- Department of Doppler Ultrasonic, Clinical Medical College of Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, Jiangsu, 225001, People's Republic of China.
| |
Collapse
|
7
|
Wu XC, Zhu ZH, Zhang JP, Shao FM, Peng JQ, Chen Y, Wang XZ, Li WY, Cao YL, Feng W, Xu JG, Ding DF. Identification of thrombin as a key regulator of chondrocyte catabolic activity through RNA-Seq and experimental verification. Gene X 2022; 823:146327. [PMID: 35219816 DOI: 10.1016/j.gene.2022.146327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/29/2021] [Accepted: 02/11/2022] [Indexed: 11/13/2022] Open
Abstract
The present study was designed to explore the relationship between thrombin and catabolic activity in chondrocytes. Primary rat chondrocytes were cultured for 24 h with rat serum (RS), rat plasma (RP), or rat plasma supplemented with thrombin (RPT). RNA-sequencing was then performed. Cell proliferation was analyzed by EdU uptake, CCK-8 assays and protein-protein interaction (PPI) network of proliferation-related genes. Heatmaps were used to visualize differences in gene expression. Gene Ontology (GO) enrichment analyses of up- and down-regulated differentially expressed genes were conducted. Molecular probes were used to label the endoplasmic reticulum in chondrocytes from three treatment groups. Immunofluorescence and Safranin O staining were used to assess type II collagen (Col2a1) expression and proteoglycan synthesis, whereas Lox expression was assessed by immunocytochemistry. The expression of enzymes involved in the synthesis and maturation of extracellular matrix (ECM) components and chemokines were measured by qPCR while matrix metalloproteinases (MMPs) levels were evaluated by Western blotting. Relevant nodules were selected through further PPI network analyses. A total of 727 and 1162 genes were up- and down-regulated based on the Venn diagrams comparison among groups. Thrombin was thus able to promote chondrocyte proliferation and a shift towards fibrotic morphology, while upregulating MMPs and chemokines linked to ECM degradation. In addition, thrombin decreased the enzyme expression involved in the synthesis and maturation of ECM.
Collapse
Affiliation(s)
- Xi-Chen Wu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhi-Heng Zhu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun-Peng Zhang
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fu-Ming Shao
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing-Qiu Peng
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Chen
- Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xue-Zong Wang
- Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wen-Yao Li
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue-Long Cao
- Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei Feng
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian-Guang Xu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Dao-Fang Ding
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
8
|
Chen Z, Zhou L, Ge Y, Chen J, Du W, Xiao L, Tong P, Huang J, Shan L, Efferth T. Fuzi decoction ameliorates pain and cartilage degeneration of osteoarthritic rats through PI3K-Akt signaling pathway and its clinical retrospective evidence. PHYTOMEDICINE 2022; 100:154071. [PMID: 35378415 DOI: 10.1016/j.phymed.2022.154071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/17/2022] [Accepted: 03/19/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is a difficult disease but the clinic lacks effective therapy. As a classic formula of traditional Chinese medicine (TCM), Fuzi decoction (FZD) has been clinically applied for treating OA-related syndromes, but its anti-OA efficacy and mechanism remain unclear. PURPOSE To experimentally and clinically determine the anti-OA efficacy of FZD and clarify the underlying mechanism. METHODS UPLC/MS/MS was applied to identify the main components of FZD. A monoiodoacetate (MIA)-induced OA rat model was employed to evaluate the in vivo efficacy of FZD against OA, by using pain behavior assessment, histopathological observation, and immunohistochemical analysis. Primary rat chondrocytes were isolated to determine the in vitro effects of FZD by using cell viability assay, wound healing assay, and real-time PCR (qPCR) analysis on anabolic/catabolic mRNA expressions. RNA sequencing (RNA-seq) and network pharmacology analysis were conducted and the overlapping data were used to predict the mechanism of FZD, followed by verification with qPCR and Western blot assays. Finally, a retrospective analysis was performed to confirm FZD's efficacy and safety in OA patients. RESULTS The UPLC/MS/MS result showed that FZD contained atractylenolide I, benzoylhypaconitine, benzoylmesaconitine, benzoylaconitine, hypaconitine, mesaconitine, aconitine, lobetyolin, paeoniflorin, and pachymic acid. The in vivo data showed that FZD restored the cartilage degeneration in MIA-induced OA rats by ameliorating pain behavior parameters, recovering histopathological alterations, benefitting cartilage anabolism (up-regulating Col2 expression), and suppressing catabolism (down-regulating MMP13 and Col10 expressions). The in vitro data showed that FZD increased cell viability and wound healing capacity of chondrocytes, and restored the altered expressions of anabolic and catabolic genes of chondrocytes. The overlapping results of RNA-seq and network pharmacology analysis suggested that PI3K/Akt signaling mediated the anti-OA mechanism of FZD, which was verified by qPCR and Western blot experiments. Clinically, the anti-OA efficacy and safety of FZD were confirmed by the retrospective analysis on OA patients. CONCLUSION The scientific innovation of this study was the determination of anti-OA efficacy of FZD by experimental and clinical evidence and the discovery of its mechanism by integrated RNA-seq, network pharmacology, and molecular experiments, which suggests FZD as a promising TCM agency for OA treatment.
Collapse
Affiliation(s)
- Zuxiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Yanzhi Ge
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Junjie Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Wenxi Du
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Luwei Xiao
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Peijian Tong
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Jiefeng Huang
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Johannes Gutenberg University, Mainz, 55128, Germany
| |
Collapse
|
9
|
Khezri MR, Varzandeh R, Ghasemnejad-Berenji M. The probable role and therapeutic potential of the PI3K/AKT signaling pathway in SARS-CoV-2 induced coagulopathy. Cell Mol Biol Lett 2022; 27:6. [PMID: 35016612 PMCID: PMC8751460 DOI: 10.1186/s11658-022-00308-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/05/2022] [Indexed: 02/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is associated with a high mortality rate. The majority of deaths in this disease are caused by ARDS (acute respiratory distress syndrome) followed by cytokine storm and coagulation complications. Although alterations in the level of the number of coagulation factors have been detected in samples from COVID-19 patients, the direct molecular mechanism which has been involved in this pathologic process has not been explored yet. The PI3K/AKT signaling pathway is an intracellular pathway which plays a central role in cell survival. Also, in recent years the association between this pathway and coagulopathies has been well clarified. Therefore, based on the evidence on over-activity of the PI3K/AKT signaling pathway in SARS-CoV-2 infection, in the current review, the probable role of this cellular pathway as a therapeutic target for the prevention of coagulation complications in patients with COVID-19 is discussed.
Collapse
Affiliation(s)
- Mohammad Rafi Khezri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Sero Road, 5715799313, Urmia, Iran.
| | - Reza Varzandeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Sero Road, 5715799313, Urmia, Iran
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Sero Road, 5715799313, Urmia, Iran. .,Research Center for Experimental and Applied Pharmaceutical Sciences, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
10
|
Cartilage degeneration is associated with activation of the PI3K/AKT signaling pathway in a growing rat experimental model of developmental trochlear dysplasia. J Adv Res 2022; 35:109-116. [PMID: 35003796 PMCID: PMC8721235 DOI: 10.1016/j.jare.2021.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 01/25/2023] Open
Abstract
Established a new experimental rat model of the developmental trochlear dysplasia; Using the macroscopic morphological and micro-CT to assess trochlear dysplasia; Using Histological staining to investigate the cartilage degradation of the model; Investigated the relationship of the PI3K/AKT signaling pathway with trochlear dysplasia cartilage degeneration; Using immunohistochemistry and qPCR to investigate the PI3K/AKT and the marker of the cartilage degeneration.
Introduction Trochlear dysplasia is a commonly encountered lower extremity deformity in humans. However, the molecular mechanism of cartilage degeneration in trochlear dysplasia is unclear thus far. Objectives The PI3K/AKT signaling pathway is known to be important for regulating the pathophysiology of cartilage degeneration. The aim of this study was to investigate the relationship of the PI3K/AKT signaling pathway with trochlear dysplasia cartilage degeneration. Methods In total, 120 female Sprague-Dawley rats (4 weeks of age) were randomly separated into control and experimental groups. Distal femurs were isolated from the experimental group at 4, 8, and 12 weeks after surgery; they were isolated from the control group at the same time points. Micro-computed tomography and histological examination were performed to investigate trochlear anatomy and changes in trochlear cartilage. Subsequently, expression patterns of PI3K/AKT, TGFβ1, and ADAMTS-4 in cartilage were investigated by immunohistochemistry and quantitative polymerase chain reaction. Results In the experimental group, the trochlear dysplasia model was successfully established at 8 weeks after surgery. Moreover, cartilage degeneration was observed beginning at 8 weeks after surgery, with higher protein and mRNA expression levels of PI3K/AKT, TGFβ1, and ADAMTS-4, relative to the control group. Conclusion Patellar instability might lead to trochlear dysplasia in growing rats. Moreover, trochlear dysplasia may cause patellofemoral osteoarthritis; cartilage degeneration in trochlear dysplasia might be associated with activation of the PI3K/AKT signaling pathway. These results provide insights regarding the high incidence of osteoarthritis in patients with trochlear dysplasia. However, more research is needed to clarify the underlying mechanisms.
Collapse
|
11
|
Ferrao Blanco MN, Domenech Garcia H, Legeai-Mallet L, van Osch GJVM. Tyrosine kinases regulate chondrocyte hypertrophy: promising drug targets for Osteoarthritis. Osteoarthritis Cartilage 2021; 29:1389-1398. [PMID: 34284112 DOI: 10.1016/j.joca.2021.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is a major health problem worldwide that affects the joints and causes severe disability. It is characterized by pain and low-grade inflammation. However, the exact pathogenesis remains unknown and the therapeutic options are limited. In OA articular chondrocytes undergo a phenotypic transition becoming hypertrophic, which leads to cartilage damage, aggravating the disease. Therefore, a therapeutic agent inhibiting hypertrophy would be a promising disease-modifying drug. The therapeutic use of tyrosine kinase inhibitors has been mainly focused on oncology, but the Food and Drug Administration (FDA) approval of the Janus kinase inhibitor Tofacitinib in Rheumatoid Arthritis has broadened the applicability of these compounds to other diseases. Interestingly, tyrosine kinases have been associated with chondrocyte hypertrophy. In this review, we discuss the experimental evidence that implicates specific tyrosine kinases in signaling pathways promoting chondrocyte hypertrophy, highlighting their potential as therapeutic targets for OA.
Collapse
Affiliation(s)
- M N Ferrao Blanco
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - H Domenech Garcia
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - L Legeai-Mallet
- Université de Paris, INSERM U1163, Institut Imagine, Paris, France.
| | - G J V M van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, the Netherlands.
| |
Collapse
|
12
|
Lee KT, Chen BC, Liu SC, Lin YY, Tsai CH, Ko CY, Tang CH, Tung KC. Nesfatin-1 facilitates IL-1β production in osteoarthritis synovial fibroblasts by suppressing miR-204-5p synthesis through the AP-1 and NF-κB pathways. Aging (Albany NY) 2021; 13:22490-22501. [PMID: 34560673 PMCID: PMC8507299 DOI: 10.18632/aging.203559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022]
Abstract
The progression of osteoarthritis (OA) is mediated by adipokines, one of which is nesfatin-1, which is responsible for the production of inflammatory cytokines. However, how this molecule may affect the synthesis of the proinflammatory cytokine interleukin 1 beta (IL-1β) in OA is unclear. Our analyses of records from the Gene Expression Omnibus (GEO) dataset and clinical specimens of synovial tissue revealed higher levels of nesfatin-1 and IL-1β in OA samples compared with normal healthy tissue. We found that nesfatin-1 facilitates IL-1β synthesis in human OA synovial fibroblasts (OASFs) and suppresses the generation of micro-RNA (miR)-204-5p, as the miR-204-5p levels in OA patients were lower than those in healthy controls. Nesfatin-1-induced stimulation of IL-1β in human OASFs occurred via the suppression of miR-204-5p synthesis by the PI3K, Akt, AP-1 and NF-κB pathways. We suggest that nesfatin-1 is worth targeting in OA treatment.
Collapse
Affiliation(s)
- Kun-Tsan Lee
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan.,Department of Orthopedics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Bo-Cheng Chen
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Yen-You Lin
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Yuan Ko
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Kwong-Chung Tung
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| |
Collapse
|
13
|
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: 8] [Impact Index Per Article: 2.0] [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
|
14
|
Yi D, Yu H, Lu K, Ruan C, Ding C, Tong L, Zhao X, Chen D. AMPK Signaling in Energy Control, Cartilage Biology, and Osteoarthritis. Front Cell Dev Biol 2021; 9:696602. [PMID: 34239878 PMCID: PMC8258395 DOI: 10.3389/fcell.2021.696602] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022] Open
Abstract
The adenosine monophosphate (AMP)-activated protein kinase (AMPK) was initially identified as an enzyme acting as an "energy sensor" in maintaining energy homeostasis via serine/threonine phosphorylation when low cellular adenosine triphosphate (ATP) level was sensed. AMPK participates in catabolic and anabolic processes at the molecular and cellular levels and is involved in appetite-regulating circuit in the hypothalamus. AMPK signaling also modulates energy metabolism in organs such as adipose tissue, brain, muscle, and heart, which are highly dependent on energy consumption via adjusting the AMP/ADP:ATP ratio. In clinics, biguanides and thiazolidinediones are prescribed to patients with metabolic disorders through activating AMPK signaling and inhibiting complex I in the mitochondria, leading to a reduction in mitochondrial respiration and elevated ATP production. The role of AMPK in mediating skeletal development and related diseases remains obscure. In this review, in addition to discuss the emerging advances of AMPK studies in energy control, we will also illustrate current discoveries of AMPK in chondrocyte homeostasis, osteoarthritis (OA) development, and the signaling interaction of AMPK with other pathways, such as mTOR (mechanistic target of rapamycin), Wnt, and NF-κB (nuclear factor κB) under OA condition.
Collapse
Affiliation(s)
- Dan Yi
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huan Yu
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ke Lu
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Changshun Ruan
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Liping Tong
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaoli Zhao
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Di Chen
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
15
|
Li L, Zhou D, Liu Q, Li D, Wang Q, Shi X, Wen C, Huang L. Network analysis indicating the pharmacological mechanism of Yunpi-Qufeng-Chushi-prescription in prophylactic treatment of rheumatoid arthritis. BMC Complement Med Ther 2021; 21:142. [PMID: 33992108 PMCID: PMC8122573 DOI: 10.1186/s12906-021-03311-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/28/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA), is an autoimmune inflammatory disease with increasing global morbidity and high disability. Early treatment is an effective intervention to slow down joint deformation. However, as for early RA and pre-RA patients, it sometimes takes a long time to make a definite diagnosis and few guidelines have made suggestion for these suspected or early phrase individuals. Yunpi-Qufeng-Chushi-Prescription (YQCP) is an optimization of the traditional formula, Cangzhu Fangfeng Tang which is effective for arthromyodynia management. METHODS In this study, LC-MS identify the main component of YQCP. Ingredients of the 11 herbs were collected from Traditional Chinese Medicine Integrated Database (TCMID). Targets of these ingredients were collected from two source, TCMID and PharmMapper. Microarray of 20 early untreated RA patients and corresponding health control were download from NCBI Gene Expression Omnibus (GEO) database to defined the differential expressed genes. Gene ontology analysis and KEGG enrichment analysis were carried out for the YQCP. Protein-protein interactions (PPIs) networks were constructed to identify the hub targets. At last, molecular docking (MD) were conducted to further verified the the possibility of YQCP for RA therapy. RESULT The study indicated that by acting on hub targets such as C3, EGFR, SRC and MMP9, YQCP may influence the mature of B cells and inhibit B cell-related IgG production, regulate oxidative stress and modulate activity of several enzymes including peroxidase and metallopeptidase to delay the occurrence and progress of RA and benefit the pre-RA or early RA patients. CONCLUSION YQCP is a potential effective therapy for prophylactic treatment of RA.
Collapse
Affiliation(s)
- Lin Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310000, Zhejiang, China
| | - Donghai Zhou
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Qiuping Liu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310000, Zhejiang, China
| | - Dianming Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310000, Zhejiang, China
| | - Qiao Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310000, Zhejiang, China
| | - Xiaowei Shi
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310000, Zhejiang, China
| | - Chengping Wen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310000, Zhejiang, China.
| | - Lin Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
16
|
Neefjes M, van Caam APM, van der Kraan PM. Transcription Factors in Cartilage Homeostasis and Osteoarthritis. BIOLOGY 2020; 9:biology9090290. [PMID: 32937960 PMCID: PMC7563835 DOI: 10.3390/biology9090290] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, and it is characterized by articular cartilage loss. In part, OA is caused by aberrant anabolic and catabolic activities of the chondrocyte, the only cell type present in cartilage. These chondrocyte activities depend on the intra- and extracellular signals that the cell receives and integrates into gene expression. The key proteins for this integration are transcription factors. A large number of transcription factors exist, and a better understanding of the transcription factors activated by the various signaling pathways active during OA can help us to better understand the complex etiology of OA. In addition, establishing such a profile can help to stratify patients in different subtypes, which can be a very useful approach towards personalized therapy. In this review, we discuss crucial transcription factors for extracellular matrix metabolism, chondrocyte hypertrophy, chondrocyte senescence, and autophagy in chondrocytes. In addition, we discuss how insight into these factors can be used for treatment purposes.
Collapse
|
17
|
Pathogenesis of Osteoarthritis: Risk Factors, Regulatory Pathways in Chondrocytes, and Experimental Models. BIOLOGY 2020; 9:biology9080194. [PMID: 32751156 PMCID: PMC7464998 DOI: 10.3390/biology9080194] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/28/2022]
Abstract
As the most common chronic degenerative joint disease, osteoarthritis (OA) is the leading cause of pain and physical disability, affecting millions of people worldwide. Mainly characterized by articular cartilage degradation, osteophyte formation, subchondral bone remodeling, and synovial inflammation, OA is a heterogeneous disease that impacts all component tissues of the articular joint organ. Pathological changes, and thus symptoms, vary from person to person, underscoring the critical need of personalized therapies. However, there has only been limited progress towards the prevention and treatment of OA, and there are no approved effective disease-modifying osteoarthritis drugs (DMOADs). Conventional treatments, including non-steroidal anti-inflammatory drugs (NSAIDs) and physical therapy, are still the major remedies to manage the symptoms until the need for total joint replacement. In this review, we provide an update of the known OA risk factors and relevant mechanisms of action. In addition, given that the lack of biologically relevant models to recapitulate human OA pathogenesis represents one of the major roadblocks in developing DMOADs, we discuss current in vivo and in vitro experimental OA models, with special emphasis on recent development and application potential of human cell-derived microphysiological tissue chip platforms.
Collapse
|
18
|
Activation of Protein Kinase Cδ Contributes to the Induction of Src/EGF Receptor/ERK Signaling in Ammonia-treated Astrocytes. J Mol Neurosci 2020; 70:1110-1119. [PMID: 32125625 DOI: 10.1007/s12031-020-01517-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 02/19/2020] [Indexed: 02/06/2023]
Abstract
Previously, we showed that Src-mediated EGF receptor transactivation/ERK activation mediates ammonia-induced astrocyte swelling, which represents a major component of brain edema in hyperammonemic disorders. Here, we tested the role of PKC in the induction of this signaling pathway and its involvement in ammonia-mediated cell swelling. We found that incubating astrocytes with bisindolylmaleimide (BIM, an inhibitor of classical and novel PKC isoforms) or rottlerin, a PKCδ-specific inhibitor, attenuated the ammonia-induced phosphorylation of EGFR, while GF109203X had no effect on this pathway. We further found that BIM or rottlerin pretreatment inhibited the ammonia-induced phosphorylation of Src and that ammonia significantly increased the level of PKCδ pulled down by a Src antibody. AG1478, a specific EGFR kinase activity inhibitor, effectively inhibited phosphorylation at Tyr1068 but had no discernable effect on phosphorylation at Tyr845. Moreover, BIM or rottlerin abrogated ammonia-induced ERK phosphorylation. BIM-, rottlerin-, or GF109203X-treated astrocytes showed a significant reduction in cell swelling compared to that observed after treatment with ammonia alone. Finally, it was found that AG1478 attenuated ammonia-induced PKCα translocation to the particulate fraction. Taken together, our results indicate that PKCδ mediates ammonia-induced astrocyte swelling by activating Src and downstream EGF receptor/ERK signaling, which may contribute to the pathogenesis of neuropsychiatric disorders associated with hyperammonemia.
Collapse
|
19
|
Wang K, Chu M, Wang F, Zhao Y, Chen H, Dai X. Putative functional variants of PI3K/AKT/mTOR pathway are associated with knee osteoarthritis susceptibility. J Clin Lab Anal 2020; 34:e23240. [PMID: 32052902 PMCID: PMC7307371 DOI: 10.1002/jcla.23240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 01/07/2020] [Accepted: 01/22/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative musculoskeletal disease which causes joint deformity and pain and finally leads to limb dysfunction. Knee osteoarthritis (KOA) has the highest incidence among all kinds of OA. Strong evidence leads to the understanding that P13K/AKT/mTOR signaling is very important in cartilage degeneration. METHODS This research sought to understand the association between genetic variation of PI3K/AKT/mTOR genes and KOA susceptibility among Chinese population. All the genetic variants of PI3K/AKT/mTOR pathway were graded and selected using RegulomeDB database, and then, an association study including 278 osteoarthritis patients and 289 controls was conducted. RESULTS Finally, eight SNPs' genotypes' distributions and susceptibility to KOA were presented. AKT1 rs2498789 was associated with KOA susceptibility in dominate genetic model (AA + GA vs GG) after adjusted for BMI, age, and gender: OR = 1.46, 95% CI: 1.03-2.05, P = .03. PIK3CA rs7646409 was also associated with KOA susceptibility (TC vs TT) after adjusted for BMI, age, and gender: OR = 0.58, 95% CI: 0.36-0.93, P = .02. PIK3CA rs7646409 (TC vs TT) with KOA risk was more significant in age < 60 group (P for heterogeneity was .03). Risk score showed significant association with KOA susceptibility after cumulative analysis (OR = 2.45, 95% CI: 1.35-4.45, P = .003). CONCLUSIONS This study shows that genetic variation of PI3K/AKT/mTOR is associated with KOA susceptibility in Chinese Han population, indicating that PI3K/AKT/mTOR is very important in KOA pathogenesis.
Collapse
Affiliation(s)
- Kejie Wang
- Department of Orthopaedics, Changzhou First People's Hospital, Changzhou, Jiangsu, China.,Department of Orthopaedics, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Minjie Chu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Feng Wang
- Department of Orthopaedics, Changzhou First People's Hospital, Changzhou, Jiangsu, China.,Department of Orthopaedics, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yiwen Zhao
- Department of Orthopaedics, Changzhou First People's Hospital, Changzhou, Jiangsu, China.,Department of Orthopaedics, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Haifeng Chen
- Department of Orthopaedics, Changzhou First People's Hospital, Changzhou, Jiangsu, China.,Department of Orthopaedics, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaoyu Dai
- Department of Orthopaedics, Changzhou First People's Hospital, Changzhou, Jiangsu, China.,Department of Orthopaedics, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
20
|
Liu SC, Tsai CH, Wu TY, Tsai CH, Tsai FJ, Chung JG, Huang CY, Yang JS, Hsu YM, Yin MC, Wu YC, Tang CH. Soya-cerebroside reduces IL-1β-induced MMP-1 production in chondrocytes and inhibits cartilage degradation: implications for the treatment of osteoarthritis. FOOD AGR IMMUNOL 2019. [DOI: 10.1080/09540105.2019.1611745] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Shan-Chi Liu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Chun-Hao Tsai
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Tung-Ying Wu
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chang-Hai Tsai
- China Medical University Children's Hospital, China Medical University, Taichung, Taiwan
- Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- China Medical University Children's Hospital, China Medical University, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Chih-Yang Huang
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Mei-Chin Yin
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan
| | - Yang-Chang Wu
- Graduate Institute of Natural Products and Research Center for Natural Products & Drug Development, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
21
|
Heuberger DM, Schuepbach RA. Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb J 2019; 17:4. [PMID: 30976204 PMCID: PMC6440139 DOI: 10.1186/s12959-019-0194-8] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
Inflammatory diseases have become increasingly prevalent with industrialization. To address this, numerous anti-inflammatory agents and molecular targets have been considered in clinical trials. Among molecular targets, protease-activated receptors (PARs) are abundantly recognized for their roles in the development of chronic inflammatory diseases. In particular, several inflammatory effects are directly mediated by the sensing of proteolytic activity by PARs. PARs belong to the seven transmembrane domain G protein-coupled receptor family, but are unique in their lack of physiologically soluble ligands. In contrast with classical receptors, PARs are activated by N-terminal proteolytic cleavage. Upon removal of specific N-terminal peptides, the resulting N-termini serve as tethered activation ligands that interact with the extracellular loop 2 domain and initiate receptor signaling. In the classical pathway, activated receptors mediate signaling by recruiting G proteins. However, activation of PARs alternatively lead to the transactivation of and signaling through receptors such as co-localized PARs, ion channels, and toll-like receptors. In this review we consider PARs and their modulators as potential therapeutic agents, and summarize the current understanding of PAR functions from clinical and in vitro studies of PAR-related inflammation.
Collapse
Affiliation(s)
- Dorothea M Heuberger
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Surgical Research Division, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reto A Schuepbach
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Hodgson D, Rowan AD, Falciani F, Proctor CJ. Systems biology reveals how altered TGFβ signalling with age reduces protection against pro-inflammatory stimuli. PLoS Comput Biol 2019; 15:e1006685. [PMID: 30677026 PMCID: PMC6363221 DOI: 10.1371/journal.pcbi.1006685] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 02/05/2019] [Accepted: 11/26/2018] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative condition caused by dysregulation of multiple molecular signalling pathways. Such dysregulation results in damage to cartilage, a smooth and protective tissue that enables low friction articulation of synovial joints. Matrix metalloproteinases (MMPs), especially MMP-13, are key enzymes in the cleavage of type II collagen which is a vital component for cartilage integrity. Transforming growth factor beta (TGFβ) can protect against pro-inflammatory cytokine-mediated MMP expression. With age there is a change in the ratio of two TGFβ type I receptors (Alk1/Alk5), a shift that results in TGFβ losing its protective role in cartilage homeostasis. Instead, TGFβ promotes cartilage degradation which correlates with the spontaneous development of OA in murine models. However, the mechanism by which TGFβ protects against pro-inflammatory responses and how this changes with age has not been extensively studied. As TGFβ signalling is complex, we used systems biology to combine experimental and computational outputs to examine how the system changes with age. Experiments showed that the repressive effect of TGFβ on chondrocytes treated with a pro-inflammatory stimulus required Alk5. Computational modelling revealed two independent mechanisms were needed to explain the crosstalk between TGFβ and pro-inflammatory signalling pathways. A novel meta-analysis of microarray data from OA patient tissue was used to create a Cytoscape network representative of human OA and revealed the importance of inflammation. Combining the modelled genes with the microarray network provided a global overview into the crosstalk between the different signalling pathways involved in OA development. Our results provide further insights into the mechanisms that cause TGFβ signalling to change from a protective to a detrimental pathway in cartilage with ageing. Moreover, such a systems biology approach may enable restoration of the protective role of TGFβ as a potential therapy to prevent age-related loss of cartilage and the development of OA.
Collapse
Affiliation(s)
- David Hodgson
- Institute of Cellular Medicine, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
| | - Andrew D. Rowan
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Francesco Falciani
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Carole J. Proctor
- Institute of Cellular Medicine, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
- MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Wu TJ, Lin CY, Tsai CH, Huang YL, Tang CH. Glucose suppresses IL-1β-induced MMP-1 expression through the FAK, MEK, ERK, and AP-1 signaling pathways. ENVIRONMENTAL TOXICOLOGY 2018; 33:1061-1068. [PMID: 30098273 DOI: 10.1002/tox.22618] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/12/2018] [Accepted: 06/17/2018] [Indexed: 06/08/2023]
Abstract
Osteoarthritis (OA) commonly affects the synovial joint and is characterized by degradation of articular cartilage. Increased matrix metalloproteinase (MMP) activity plays a major role in this degradation. Dextrose (D-glucose) prolotherapy has shown promising activity in the treatment of different musculoskeletal disorders, including OA. However, little is known about the role of glucose on MMP inhibition in OA therapy. We found that stimulating chondrocytes with the proinflammatory cytokine interleukin-1β (IL-1β) increased the expression of MMP-1, MMP-3, and MMP-13. Glucose reduced this increase in MMP-1 expression, but had no effect upon MMP-3 or MMP-13 expression. Analyses using a focal adhesion kinase (FAK) inhibitor, MEK inhibitors (U0126 and PD98059), an ERK inhibitor, AP-1 inhibitors (curcumin and tanshinone), or siRNAs demonstrated that the FAK, MEK, ERK, and AP-1 pathways mediate IL-1β-induced increases in MMP-1 expression. Glucose antagonized IL-1β-promoted phosphorylation of FAK, MEK, ERK, and c-Jun. Thus, glucose decreased IL-1β-induced MMP-1 expression through the FAK, MEK, ERK, and AP-1 signaling cascades. These findings may provide a better understanding of the mechanisms of prolotherapy on inhibiting MMP expression.
Collapse
Affiliation(s)
- Tsung-Ju Wu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Physical Medicine and Rehabilitation, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Yang Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chun-Hao Tsai
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
24
|
Wang W, Qiao Y, Li Z. New Insights into Modes of GPCR Activation. Trends Pharmacol Sci 2018; 39:367-386. [DOI: 10.1016/j.tips.2018.01.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
|
25
|
Lin C, Shao Y, Zeng C, Zhao C, Fang H, Wang L, Pan J, Liu L, Qi W, Feng X, Qiu H, Zhang H, Chen Y, Wang H, Cai D, Xian CJ. Blocking PI3K/AKT signaling inhibits bone sclerosis in subchondral bone and attenuates post-traumatic osteoarthritis. J Cell Physiol 2018; 233:6135-6147. [PMID: 29323710 DOI: 10.1002/jcp.26460] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/05/2018] [Indexed: 01/05/2023]
Abstract
PI3K/AKT signaling is essential in regulating pathophysiology of osteoarthritis (OA). However, its potential modulatory role in early OA progression has not been investigated yet. Here, a mouse destabilization OA model in the tibia was used to investigate roles of PI3K/AKT signaling in the early subchondral bone changes and OA pathological process. We revealed a significant increase in PI3K/AKT signaling activation which was associated with aberrant bone formation in tibial subchondral bone following destabilizing the medial meniscus (DMM), which was effectively prevented by treatment with PI3K/AKT signaling inhibitor LY294002. PI3K/AKT signaling inhibition attenuated articular cartilage degeneration. Serum and bone biochemical analyses revealed increased levels of MMP-13, which was found expressed mainly by osteoblastic cells in subchondral bone. However, this MMP-13 induction was attenuated by LY294002 treatment. Furthermore, PI3K/AKT signaling was found to enhance preosteoblast proliferation, differentiation, and expression of MMP-13 by activating NF-κB pathway. In conclusion, inhibition of PI3K/AKT/NF-κB axis was able to prevent aberrant bone formation and attenuate cartilage degeneration in OA mice.
Collapse
Affiliation(s)
- Chuangxin Lin
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Yan Shao
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Chun Zeng
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Chang Zhao
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Hang Fang
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Liping Wang
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Jianying Pan
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Liangliang Liu
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Weizhong Qi
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Xiaofeng Feng
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Hong Qiu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Haiyang Zhang
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Yuhui Chen
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Hong Wang
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Daozhang Cai
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Academy of Orthopaedics of Guangdong Province, Guangzhou, Guangdong, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, Guangdong, China
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
26
|
Visfatin Promotes IL-6 and TNF-α Production in Human Synovial Fibroblasts by Repressing miR-199a-5p through ERK, p38 and JNK Signaling Pathways. Int J Mol Sci 2018; 19:ijms19010190. [PMID: 29316707 PMCID: PMC5796139 DOI: 10.3390/ijms19010190] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 12/29/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022] Open
Abstract
Osteoarthritis (OA), an inflammatory form of arthritis, is characterized by synovial inflammation and cartilage destruction largely influenced by two key proinflammatory cytokines-interleukin-6 (IL-6) and tumor necrosis factor α (TNF-α). Notably, levels of visfatin (a proinflammatory adipokine) are elevated in patients with OA, although the relationship of visfatin to IL-6 and TNF-α expression in OA pathogenesis has been unclear. In this study, visfatin enhanced the expression of IL-6 and TNF-α in human OA synovial fibroblasts (OASFs) in a concentration-dependent manner and stimulation of OASFs with visfatin promoted phosphorylation of extracellular-signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK), while ERK, p38, and JNK inhibitors or siRNAs all abolished visfatin-induced increases in IL-6 and TNF-α production. Moreover, transfection with miR-199a-5p mimics reversed visfatin-induced increases in IL-6 and TNF-α production. Furthermore, we also found that visfatin-promoted IL-6 and TNF-α production is mediated via the inhibition of miR-199a-5p expression through the ERK, p38, and JNK signaling pathways. Visfatin may therefore be an appropriate target for drug intervention in OA treatment.
Collapse
|
27
|
FRA1 promotes squamous cell carcinoma growth and metastasis through distinct AKT and c-Jun dependent mechanisms. Oncotarget 2018; 7:34371-83. [PMID: 27144339 PMCID: PMC5085162 DOI: 10.18632/oncotarget.9110] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/11/2016] [Indexed: 12/14/2022] Open
Abstract
FRA1 (Fos-like antigen 1) is highly expressed in many epithelial cancers including squamous cell carcinoma of the skin (cSCC) and head and neck (HNSCC). However, the functional importance and the mechanisms mediating FRA1 function in these cancers are not fully understood. Here, we demonstrate that FRA1 gene silencing in HNSCC and cSCC cells resulted in two consequences – impaired cell proliferation and migration. FRA1 regulation of cell growth was distinct from that of c-Jun, a prominent Jun group AP-1 factor. While c-Jun was required for the expression of the G1/S phase cell cycle promoter CDK4, FRA1 was essential for AKT activation and AKT-dependent expression of CyclinB1, a molecule required for G2-M progression. Exogenous expression of a constitutively active form of AKT rescued cancer cell growth defect caused by FRA1-loss. Additionally, FRA1 knockdown markedly slowed cell adhesion and migration, and conversely expression of an active FRA1 mutant (FRA1DD) expedited these processes in a JNK/c-Jun-dependent manner. Through protein and ChIP-PCR analyses, we identified KIND1, a cytoskeletal regulator of the cell adhesion molecule β1-integrin, as a novel FRA1 transcriptional target. Restoring KIND1 expression rescued migratory defects induced by FRA1 loss. In agreement with these in vitro data, HNSCC cells with FRA1 loss displayed markedly reduced rates of subcutaneous tumor growth and pulmonary metastasis. Together, these results indicate that FRA1 promotes cancer growth through AKT, and enhances cancer cell migration through JNK/c-Jun, pinpointing FRA1 as a key integrator of JNK and AKT signaling pathways and a potential therapeutic target for cSCC and HNSCC.
Collapse
|
28
|
Huang BR, Chen TS, Bau DT, Chuang IC, Tsai CF, Chang PC, Lu DY. EGFR is a pivotal regulator of thrombin-mediated inflammation in primary human nucleus pulposus culture. Sci Rep 2017; 7:8578. [PMID: 28819180 PMCID: PMC5561020 DOI: 10.1038/s41598-017-09122-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/19/2017] [Indexed: 01/23/2023] Open
Abstract
We found that the coagulation and cytokine pathways were important mechanisms involve in the degeneration of intervertebral discs (IVD) using a microarray approach to analyze gene expression in different grades of specimens. Furthermore, using a cytokine/chemokine array, a significant increase in CXCL8 expression was observed in human nucleus pulposus (NP) cells after thrombin treatment. The enhancement of CXCL8 expression by thrombin was activated by the PAR1 receptor. Importantly, analysis of degenerated human NP tissue samples showed that EGFR expression positively correlated with the grade of tissue degeneration. In NP cells, thrombin caused an increase in phosphorylation of the EGFR at the Tyr1068, and treatment with the pharmacological EGFR inhibitor, AG1473 effectively blocked thrombin-enhanced CXCL8 production. Surprisingly, inhibition of STAT3 for 24 h decreased expression of EGFR. Treatment with thrombin also increased Akt and GSK3α/β activation; this activation was also blocked by EGFR inhibitor. Although c-Src, ERK, and FAK were activated by thrombin, only c-Src and ERK were involved in the STAT3/CXCL8 induction. Our findings indicate that stimulation of an inflammatory response in NP cells by thrombin is part of a specific pathophysiology that modulates the EGFR activation through activation of Src/ERK/STAT3 signaling.
Collapse
Affiliation(s)
- Bor-Ren Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tzu-Sheng Chen
- Department of Pathology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Da-Tian Bau
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - I-Chen Chuang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics, Asia University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan. .,Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan.
| |
Collapse
|
29
|
Chen J, Zhang J, Tachie-Menson T, Shukla N, Garrod DR, Robinson C. Allergen-dependent oxidant formation requires purinoceptor activation of ADAM 10 and prothrombin. J Allergy Clin Immunol 2017; 139:2023-2026.e9. [PMID: 28111309 PMCID: PMC5457034 DOI: 10.1016/j.jaci.2016.12.954] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/14/2016] [Accepted: 12/14/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Jie Chen
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Jihui Zhang
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Theresa Tachie-Menson
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Neha Shukla
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - David R Garrod
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Clive Robinson
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom.
| |
Collapse
|
30
|
Bianchi A, Guibert M, Cailotto F, Gasser A, Presle N, Mainard D, Netter P, Kempf H, Jouzeau JY, Reboul P. Fibroblast Growth Factor 23 drives MMP13 expression in human osteoarthritic chondrocytes in a Klotho-independent manner. Osteoarthritis Cartilage 2016; 24:1961-1969. [PMID: 27307356 DOI: 10.1016/j.joca.2016.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Fibroblast Growth Factor 23 (FGF23) may represent an attractive candidate that could participate to the osteoarthritic (OA)-induced phenotype switch of chondrocytes. To address this hypothesis, we investigated the expression of FGF23, its receptors (FGFRs) and co-receptor (Klotho) in human cartilage and studied the effects of rhFGF23 on OA chondrocytes. METHOD Gene expression or protein levels were analysed by RT-PCR and immunohistochemistry. Collagenase 3 (MMP13) activity was measured by a fluorescent assay. MAPK signalling pathways were investigated by phosphoprotein array, immunoblotting and the use of selective inhibitors. RNA silencing was performed to confirm the respective contribution of FGFR1 and Klotho. RESULTS We showed that the expression of FGF23, FGFR1 and Klotho was up-regulated at both mRNA and protein levels in OA chondrocytes when compared to healthy ones. These overexpressions were markedly elevated in the damaged regions of OA cartilage. When stimulated with rhFGF23, OA chondrocytes displayed an extended expression of FGF23 and of markers of hypertrophy such as MMP13, COL10A1, and VEGF. We demonstrated that FGF23 auto-stimulation was both FGFR1-and Klotho-dependent, whereas the expression of markers of hypertrophy was mainly dependent on FGFR1 alone. Finally, we showed that FGF23-induced MMP13 expression was strongly regulated by the MEK/ERK cascade and to a lesser extent, by the PI-3K/AKT pathway. CONCLUSION These results demonstrate that FGF23 sustains differentiation of OA chondrocytes in a Klotho-independent manner.
Collapse
Affiliation(s)
- A Bianchi
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - M Guibert
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - F Cailotto
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - A Gasser
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - N Presle
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - D Mainard
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France; Département de Chirurgie Orthopédique et Traumatologique, Centre Hospitalier Universitaire, Nancy, France.
| | - P Netter
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France; Département de Pharmacologie Clinique et Toxicologie, Centre Hospitalier Universitaire, Nancy, France.
| | - H Kempf
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - J-Y Jouzeau
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France; Département de Pharmacologie Clinique et Toxicologie, Centre Hospitalier Universitaire, Nancy, France.
| | - P Reboul
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
31
|
Jiao K, Zeng G, Niu LN, Yang HX, Ren GT, Xu XY, Li FF, Tay FR, Wang MQ. Activation of α2A-adrenergic signal transduction in chondrocytes promotes degenerative remodelling of temporomandibular joint. Sci Rep 2016; 6:30085. [PMID: 27452863 PMCID: PMC4958971 DOI: 10.1038/srep30085] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/28/2016] [Indexed: 12/29/2022] Open
Abstract
This study tested whether activation of adrenoreceptors in chondrocytes has roles in degenerative remodelling of temporomandibular joint (TMJ) and to determine associated mechanisms. Unilateral anterior crossbite (UAC) was established to induce TMJ degeneration in rats. Saline vehicle, α2- and β-adrenoreceptor antagonists or agonists were injected locally into the TMJ area of UAC rats. Cartilage degeneration, subchondral bone microarchitecture and the expression of adrenoreceptors, aggrecans, matrix metalloproteinases (MMPs) and RANKL by chondrocytes were evaluated. Chondrocytes were stimulated by norepinephrine to investigate signal transduction of adrenoreceptors. Increased α2A-adrenoreceptor expression was observed in condylar cartilage of UAC rats, together with cartilage degeneration and subchondral bone loss. Norepinephrine depresses aggrecans expression but stimulates MMP-3, MMP-13 and RANKL production by chondrocytes through ERK1/2 and PKA pathway; these effects were abolished by an α2A-adrenoreceptor antagonist. Furthermore, inhibition of α2A-adrenoreceptor attenuated degenerative remodelling in the condylar cartilage and subchondral bone, as revealed by increased cartilage thickness, proteoglycans and aggrecan expression, and decreased MMP-3, MMP-13 and RANKL expressions in cartilage, increased BMD, BV/TV, and decreased Tb.Sp in subchondral bone. Conversely, activation of α2A-adrenoreceptor intensified aforementioned degenerative changes in UAC rats. It is concluded that activation of α2A-adrenergic signal in chondrocytes promotes TMJ degenerative remodelling by chondrocyte-mediated pro-catabolic activities.
Collapse
Affiliation(s)
- Kai Jiao
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, 145 Changle Western Road, Xi'an, 710032, China
| | - Guang Zeng
- Department of Dentistry, Tangdu Hospital, Forth Military Medical University, Shannxi, Xi'an, 710038, China
| | - Li-Na Niu
- State Key Laboratory of Military Stomatology, Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Changle Western Road No.145, Xi'an, 710032, China
| | - Hong-Xu Yang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, 145 Changle Western Road, Xi'an, 710032, China
| | - Gao-Tong Ren
- Undergraduate Department of Oral Science, Fourth Military Medical University, Changle Western Road No.145, Xi'an, 710032, China
| | - Xin-Yue Xu
- Undergraduate Department of Oral Science, Fourth Military Medical University, Changle Western Road No.145, Xi'an, 710032, China
| | - Fei-Fei Li
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, Fourth Military Medical University, 145 Changle Western Road, Xi'an, 710032, China
| | - Franklin R Tay
- The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Mei-Qing Wang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, 145 Changle Western Road, Xi'an, 710032, China
| |
Collapse
|
32
|
Yang CC, Hsiao LD, Yang CM, Lin CC. Thrombin Enhanced Matrix Metalloproteinase-9 Expression and Migration of SK-N-SH Cells via PAR-1, c-Src, PYK2, EGFR, Erk1/2 and AP-1. Mol Neurobiol 2016; 54:3476-3491. [PMID: 27181591 DOI: 10.1007/s12035-016-9916-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 05/03/2016] [Indexed: 01/30/2023]
Abstract
Neuroinflammation is a hallmark of neurodegenerative disorders in the central nerve system (CNS). Thrombin has been known as one of the factors in pathological processes including migration, blood-brain barrier breakdown, brain edema formation, neuroinflammation, and neuronal death. Thrombin has been shown to be a regulator of matrix metalloproteinase (MMPs) expression leading to cell migration. Among MMPs, the elevated expression of MMP-9 has been observed in patients with brain diseases, which may contribute to the pathology of neuroinflammatory and neurodegenerative diseases. However, the mechanisms underlying thrombin-induced MMP-9 expression in SK-N-SH cells were not completely understood. Here, we used gelatin zymography, Western blot, real-time PCR, promoter activity assay, and cell migration assay to demonstrate that thrombin induced the expression of pro-form MMP-9 protein and messenger RNA (mRNA), and promoter activity in SK-N-SH cells, which were attenuated by pretreatment with the pharmacological inhibitor of protease-activated receptor-1 (PAR-1, SCH79797), Gi-coupled receptor (GPA2), c-Src (PP1), Pyk2 (PF431396), EGFR (AG1478), PI3K (LY294002), Akt (SH-5), MEK1/2 (U0126), or AP-1 (TanshinoneIIA) and transfection with small interfering RNA (siRNA) of PAR-1, Gi, c-Src, Pyk2, EGFR, Akt, p44, p42, or c-Jun. Moreover, thrombin-stimulated c-Src, Pyk2, EGFR, Akt, p42/p44 MAPK, or c-Jun phosphorylation was attenuated by their respective inhibitor of PP1, PF431396, AG1478, SH-5, U0126, or TanshinoneIIA. Finally, pretreatment with these inhibitors also blocked thrombin-induced SK-N-SH cell migration. Our results concluded that thrombin binding to PAR-1 receptor activated Gi-protein/c-Src/Pyk2/EGFR/PI3K/Akt/p42/p44 MAPK cascade, which in turn elicited AP-1 activation and ultimately evoked MMP-9 expression and cell migration in SK-N-SH cells.
Collapse
Affiliation(s)
- Chien-Chung Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.,Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Lin-Kou, Kwei-San, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Ageing Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan. .,Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan. .,Research Center for Industry of Human Ecology and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan. .,Department of Pharmacology, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-San, Tao-Yuan, Taiwan.
| | - Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan. .,Department of Anesthetics, Chang Gung Memorial Hospital at Lin-Kou, 5 Fu-Hsin Street, Kwei-San, Tao-Yuan, Taiwan.
| |
Collapse
|
33
|
Protein pathways working in human follicular fluid: the future for tailored IVF? Expert Rev Mol Med 2016; 18:e9. [DOI: 10.1017/erm.2016.4] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human follicular fluid (HFF) contains molecules and proteins that may affect follicle growth, oocyte maturation and competence acquiring. Despite the numerous studies, an integrated broad overview on biomolecular and patho/physiological processes that are proved or supposed to take place in HFF during folliculogenesis and oocyte development is still missing. In this review we report, for the first time, all the proteins unambiguously detected in HFF and, applying DAVID (Database for Annotation, Visualization and Integrated Discovery) and MetaCore bioinformatic resources, we shed new lights on their functional correlation, delineating protein patterns and pathways with reasonable potentialities for oocyte quality estimation in in vitro fertilisation (IVF) programs. Performing a rigorous PubMed search, we redacted a list of 617 unique proteins unambiguously-annotated as HFF components. Their functional processing suggested the occurrence in HFF of a tight and highly dynamic functional-network, which is balanced by specific effectors, primarily involved in extracellular matrix degradation and remodelling, inflammation and coagulation. Metalloproteinases, thrombin and vitamin-D-receptor/retinoid-X-receptor-alpha resulted as the main key factors in the nets and their differential activity may be indicative of ovarian health and oocyte quality. Despite future accurate clinical investigations are absolutely needed, the present analysis may provide a starting point for more accurate oocyte quality estimation and for defining personalised therapies in reproductive medicine.
Collapse
|
34
|
Chen JH, Tsai CH, Lin HY, Huang CF, Leung YM, Lai SW, Tsai CF, Chang PC, Lu DY, Lin C. Interlukin-18 Is a Pivot Regulatory Factor on Matrix Metalloproteinase-13 Expression and Brain Astrocytic Migration. Mol Neurobiol 2015; 53:6218-6227. [PMID: 26558633 DOI: 10.1007/s12035-015-9529-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/05/2015] [Indexed: 01/28/2023]
Abstract
The expression of matrix metalloproteinase-13 (MMP-13) has been shown to be elevated in some pathophysiological conditions and is involved in the degradation of extracellular matrix in astrocytes. In current study, the function of MMP-13 was further investigated. The conditioned medium (CM) collected from activated microglia increased interleukin (IL)-18 production and enhanced MMP-13 expression in astrocytes. Furthermore, treatment with recombinant IL-18 increased MMP-13 protein and mRNA levels in astrocytes. Recombinant IL-18 stimulation also increased the enzymatic activity of MMP-13 and the migratory activity of astrocytes, while administration of MMP-13 or pan-MMP inhibitors antagonized IL-18-induced migratory activity of astrocytes. In addition, administration of recombinant IL-18 to astrocytes led to the phosphorylation of JNK, Akt, or PKCδ, and treatment of astrocytes with JNK, PI3 kinase/Akt, or PKCδ inhibitors significantly decreased the IL-18-induced migratory activity. Taken together, the results suggest that IL-18-induced MMP-13 expression in astrocytes is regulated by JNK, PI3 kinase/Akt, and PKCδ signaling pathways. These findings also indicate that IL-18 is an important regulator leading to MMP-13 expression and cell migration in astrocytes.
Collapse
Affiliation(s)
- Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chon-Haw Tsai
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Hsiao-Yun Lin
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Chien-Fang Huang
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Yuk-Man Leung
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Sheng-Wei Lai
- Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics, Asia University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan. .,Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan.
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan.
| |
Collapse
|
35
|
Cattaneo F, Guerra G, Parisi M, De Marinis M, Tafuri D, Cinelli M, Ammendola R. Cell-surface receptors transactivation mediated by g protein-coupled receptors. Int J Mol Sci 2014; 15:19700-28. [PMID: 25356505 PMCID: PMC4264134 DOI: 10.3390/ijms151119700] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/30/2014] [Accepted: 10/13/2014] [Indexed: 12/17/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are seven transmembrane-spanning proteins belonging to a large family of cell-surface receptors involved in many intracellular signaling cascades. Despite GPCRs lack intrinsic tyrosine kinase activity, tyrosine phosphorylation of a tyrosine kinase receptor (RTK) occurs in response to binding of specific agonists of several such receptors, triggering intracellular mitogenic cascades. This suggests that the notion that GPCRs are associated with the regulation of post-mitotic cell functions is no longer believable. Crosstalk between GPCR and RTK may occur by different molecular mechanism such as the activation of metalloproteases, which can induce the metalloprotease-dependent release of RTK ligands, or in a ligand-independent manner involving membrane associated non-receptor tyrosine kinases, such as c-Src. Reactive oxygen species (ROS) are also implicated as signaling intermediates in RTKs transactivation. Intracellular concentration of ROS increases transiently in cells stimulated with GPCR agonists and their deliberated and regulated generation is mainly catalyzed by enzymes that belong to nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family. Oxidation and/or reduction of cysteine sulfhydryl groups of phosphatases tightly controls the activity of RTKs and ROS-mediated inhibition of cellular phosphatases results in an equilibrium shift from the non-phosphorylated to the phosphorylated state of RTKs. Many GPCR agonists activate phospholipase C, which catalyze the hydrolysis of phosphatidylinositol 4,5-bis-phosphate to produce inositol 1,4,5-triphosphate and diacylglicerol. The consequent mobilization of Ca2+ from endoplasmic reticulum leads to the activation of protein kinase C (PKC) isoforms. PKCα mediates feedback inhibition of RTK transactivation during GPCR stimulation. Recent data have expanded the coverage of transactivation to include Serine/Threonine kinase receptors and Toll-like receptors. Herein, we discuss the main mechanisms of GPCR-mediated cell-surface receptors transactivation and the pathways involved in intracellular responses induced by GPCR agonists. These studies may suggest the design of novel strategies for therapeutic interventions.
Collapse
Affiliation(s)
- Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy.
| | - Melania Parisi
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Marta De Marinis
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Domenico Tafuri
- Department of Sport Science and Wellness, University of Naples Parthenope, Naples 80133, Italy.
| | - Mariapia Cinelli
- Department of Public Health, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| |
Collapse
|
36
|
The characteristics of thrombin in osteoarthritic pathogenesis and treatment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:407518. [PMID: 25313362 PMCID: PMC4182002 DOI: 10.1155/2014/407518] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/01/2014] [Indexed: 01/08/2023]
Abstract
Osteoarthritis (OA) is a mechanical abnormality associated with degradation of joints. It is characterized by chronic, progressive degeneration of articular cartilage, abnormalities of bone, and synovial change. The most common symptom of OA is local inflammation resulting from exogenous stress or endogenous abnormal cytokines. Additionally, OA is associated with local and/or systemic activation of coagulation and anticoagulation pathways. Thrombin plays an important role in the stimulation of fibrin deposition and the proinflammatory processes in OA. Thrombin mediates hemostatic and inflammatory responses and guides the immune response to tissue damage. Thrombin activates intracellular signaling pathways by interacting with transmembrane domain G protein coupled receptors (GPCRs), known as protease-activated receptors (PARs). In pathogenic mechanisms, PARs have been implicated in the development of acute and chronic inflammatory responses in OA. Therefore, discovery of thrombin signaling pathways would help us to understand the mechanism of OA pathogenesis and lead us to develop therapeutic drugs in the future.
Collapse
|