1
|
Garau Paganella L, Badolato A, Labouesse C, Fischer G, Sänger CS, Kourouklis A, Giampietro C, Werner S, Mazza E, Tibbitt MW. Variations in fluid chemical potential induce fibroblast mechano-response in 3D hydrogels. BIOMATERIALS ADVANCES 2024; 163:213933. [PMID: 38972277 DOI: 10.1016/j.bioadv.2024.213933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Mechanical deformation of skin creates variations in fluid chemical potential, leading to local changes in hydrostatic and osmotic pressure, whose effects on mechanobiology remain poorly understood. To study these effects, we investigate the specific influences of hydrostatic and osmotic pressure on primary human dermal fibroblasts in three-dimensional hydrogel culture models. Cyclic hydrostatic pressure and hyperosmotic stress enhanced the percentage of cells expressing the proliferation marker Ki67 in both collagen and PEG-based hydrogels. Osmotic pressure also activated the p38 MAPK stress response pathway and increased the expression of the osmoresponsive genes PRSS35 and NFAT5. When cells were cultured in two-dimension (2D), no change in proliferation was observed with either hydrostatic or osmotic pressure. Furthermore, basal, and osmotic pressure-induced expression of osmoresponsive genes differed in 2D culture versus 3D hydrogels, highlighting the role of dimensionality in skin cell mechanotransduction and stressing the importance of 3D tissue-like models that better replicate in vivo conditions. Overall, these results indicate that fluid chemical potential changes affect dermal fibroblast mechanobiology, which has implications for skin function and for tissue regeneration strategies.
Collapse
Affiliation(s)
- Lorenza Garau Paganella
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland; Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Asia Badolato
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Céline Labouesse
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Gabriel Fischer
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Catharina S Sänger
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Andreas Kourouklis
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Costanza Giampietro
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland; EMPA, Swiss Federal Laboratories for Material Science and Technologies, Dubendorf, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Edoardo Mazza
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland; EMPA, Swiss Federal Laboratories for Material Science and Technologies, Dubendorf, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Rinotas V, Iliaki K, Pavlidi L, Meletakos T, Mosialos G, Armaka M. Cyld restrains the hyperactivation of synovial fibroblasts in inflammatory arthritis by regulating the TAK1/IKK2 signaling axis. Cell Death Dis 2024; 15:584. [PMID: 39122678 PMCID: PMC11316070 DOI: 10.1038/s41419-024-06966-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
TNF is a potent cytokine known for its involvement in physiology and pathology. In Rheumatoid Arthritis (RA), persistent TNF signals cause aberrant activation of synovial fibroblasts (SFs), the resident cells crucially involved in the inflammatory and destructive responses of the affected synovial membrane. However, the molecular switches that control the pathogenic activation of SFs remain poorly defined. Cyld is a major component of deubiquitination (DUB) machinery regulating the signaling responses towards survival/inflammation and programmed necrosis that induced by cytokines, growth factors and microbial products. Herein, we follow functional genetic approaches to understand how Cyld affects arthritogenic TNF signaling in SFs. We demonstrate that in spontaneous and induced RA models, SF-Cyld DUB deficiency deteriorates arthritic phenotypes due to increased levels of chemokines, adhesion receptors and bone-degrading enzymes generated by mutant SFs. Mechanistically, Cyld serves to restrict the TNF-induced hyperactivation of SFs by limiting Tak1-mediated signaling, and, therefore, leading to supervised NF-κB and JNK activity. However, Cyld is not critically involved in the regulation of TNF-induced death of SFs. Our results identify SF-Cyld as a regulator of TNF-mediated arthritis and inform the signaling landscape underpinning the SF responses.
Collapse
Affiliation(s)
- Vagelis Rinotas
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center (BSRC) "Alexander Fleming", Vari, Greece
| | - Kalliopi Iliaki
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center (BSRC) "Alexander Fleming", Vari, Greece
| | - Lydia Pavlidi
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center (BSRC) "Alexander Fleming", Vari, Greece
| | - Theodore Meletakos
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center (BSRC) "Alexander Fleming", Vari, Greece
| | - George Mosialos
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Marietta Armaka
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center (BSRC) "Alexander Fleming", Vari, Greece.
| |
Collapse
|
3
|
Jiang G, Cai Y, Cheng D, Wang H, Deng G, Xiang D. CYLD alleviates NLRP3 inflammasome-mediated pyroptosis in osteoporosis by deubiquitinating WNK1. J Orthop Surg Res 2024; 19:212. [PMID: 38561786 PMCID: PMC10983667 DOI: 10.1186/s13018-024-04675-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Osteoporosis (OP) is the result of bone mass reduction and bone structure disorder. Bone marrow mesenchymal stem cells (BMSCs) are the main source of osteogenic precursor cells involved in adult bone remodeling. The involvement of the deubiquitinating enzyme CYLD in OP has recently been discovered. However, the detailed role and mechanism of CYLD remain unknown. METHODS The OP mouse model was established by performing ovariectomy (OVX) on mice. Hematoxylin and eosin staining, Masson and Immunohistochemical staining were used to assess pathologic changes. Real-time quantitative PCR, Western blot, and immunofluorescence were employed to assess the expression levels of CYLD, WNK1, NLRP3 and osteogenesis-related molecules. The binding relationship between CYLD and WNK1 was validated through a co-immunoprecipitation assay. The osteogenic capacity of BMSCs was determined using Alkaline phosphatase (ALP) and alizarin red staining (ARS). Protein ubiquitination was evaluated by a ubiquitination assay. RESULTS The levels of both CYLD and WNK1 were decreased in bone tissues and BMSCs of OVX mice. Overexpression of CYLD or WNK1 induced osteogenic differentiation in BMSCs. Additionally, NLRP3 inflammation was activated in OVX mice, but its activation was attenuated upon overexpression of CYLD or WNK1. CYLD was observed to reduce the ubiquitination of WNK1, thereby enhancing its protein stability and leading to the inactivation of NLRP3 inflammation. However, the protective effects of CYLD on osteogenic differentiation and NLRP3 inflammation inactivation were diminished upon silencing of WNK1. CONCLUSION CYLD mitigates NLRP3 inflammasome-triggered pyroptosis in osteoporosis through its deubiquitination of WNK1.
Collapse
Affiliation(s)
- Guiyong Jiang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 15th Floor, Surgery Building, Southern Hospital, No.1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China
| | - Yu Cai
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Duo Cheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 15th Floor, Surgery Building, Southern Hospital, No.1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China
| | - Hao Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 15th Floor, Surgery Building, Southern Hospital, No.1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China
| | - Geyang Deng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 15th Floor, Surgery Building, Southern Hospital, No.1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China
| | - Dayong Xiang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, 15th Floor, Surgery Building, Southern Hospital, No.1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
4
|
Ren C, Chen J, Che Q, Jia Q, Lu H, Qi X, Zhang X, Shu Q. IL-37 alleviates TNF-α-induced pyroptosis of rheumatoid arthritis fibroblast-like synoviocytes by inhibiting the NF-κB/GSDMD signaling pathway. Immunobiology 2023; 228:152382. [PMID: 37075579 DOI: 10.1016/j.imbio.2023.152382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/09/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023]
Abstract
OBJECTIVE Pyroptosis is crucial to rheumatoid arthritis (RA) by inducing and aggravating inflammation. TNF-α is abundant in fibroblast-like synoviocytes of RA (RA-FLSs) and plays a key role in pyroptosis by inducing nuclear factor (NF)-κB activation. Additionally, interleukin (IL)-37 is involved in autoimmune diseases as an anti-inflammatory cytokine and innate and acquired immune response inhibitor. However, the effect of IL-37 on pyroptosis in RA-FLSs remains unclear. Therefore, this study investigated the effects and mechanism of IL-37 on RA-FLS pyroptosis induced by TNF-α. METHODS In this study, the serum cytokines in patients with RA and healthy controls were detected using ELISA. The RA-FLSs were then cultured with TNF-α, with or without various IL-37 concentrations, to test the cytokine levels in the cell supernatant. 5-Ethynyl-2'-Deoxyuridine (EdU) assay assessed the effects of IL-37 on RA FLS proliferation. RA-FLS apoptosis was assessed using flow cytometry and mitochondrial membrane potential (MMP) measurement. In addition, transmission electron microscopy (TEM) was used to examine cell pyroptosis. We selected the optimal concentration for the following experiments and detected the signal pathway of IL-37 on pyroptosis of RA FLSs by quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and Western blotting. Finally, we validated the therapeutic effects of IL-37 on CIA rat model in vivo. RESULTS IL-37 inhibited inflammation in vitro and in vivo and reduced pyroptosis-related protein expression in RA FLSs. Furthermore, we determined that nuclear factor κB (NF-κB) signaling is required for GSDMD-mediated pyroptosis in RA FLSs. CONCLUSION IL-37 alleviates TNF-α-induced pyroptosis of RA FLSs by inhibiting NF-κB/GSDMD signaling. Additionally, our data revealed a novel mechanism for IL-37 in RA FLSs, suggesting a new potential therapy for IL-37 to treat RA.
Collapse
|
5
|
Song T, Zhou M, Li W, Lv M, Zheng L, Zhao M. The anti-inflammatory effect of vasoactive peptides from soybean protein hydrolysates by mediating serum extracellular vesicles-derived miRNA-19b/CYLD/TRAF6 axis in the vascular microenvironment of SHRs. Food Res Int 2022; 160:111742. [DOI: 10.1016/j.foodres.2022.111742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/28/2022]
|
6
|
MiR-129-5p Inactivates NF-κB Pathway to Block Rheumatoid Arthritis Development via Targeting BRD4. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:8330659. [PMID: 35494514 PMCID: PMC9042608 DOI: 10.1155/2022/8330659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 12/29/2022]
Abstract
Methods The abundance of miR-129-5p was detected in the samples including normal tissues and RA tissues and cell lines including human fibroblast-like synoviocytes (hFLSs) and human rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs). The CCK-8 assay, flow cytometry, Transwell, and ELISA were used to observe the effects of miR-129-5p on the phenotype of RA-FLSs. Moreover, the potential targets of miR-129-5p were identified with TargetScan and dual-luciferase reporter gene assay. Besides, the abundances of the proteins were analyzed with western blot. Results Decreased miR-129-5p was observed in RA tissues and cells. Increased miR-129-5p obviously blocked the proliferation, inflammatory stress, and migration and remarkably promoted cellular apoptosis. Moreover, BRD4 was confirmed as targets of miR-129-5p, and BRD4 upregulation could partly rescue the inhibition of miR-129-5p on aggressive behaviors of RA-FLSs. Besides, the finding of this study also proved that upregulated miR-129-5p could impede the NF-κB pathway via targeting BRD4. Conclusion This study suggests that miR-129-5p suppresses the activation of NF-κB pathway to block the progression of RA via targeting BRD4.
Collapse
|
7
|
Yang Y, Liu Y, Yu H, Xie Q, Wang B, Jiang S, Su W, Mao Y, Li B, Peng C, Jian Y, Wang W. Sesquiterpenes from Kadsura coccinea attenuate rheumatoid arthritis-related inflammation by inhibiting the NF-κB and JAK2/STAT3 signal pathways. PHYTOCHEMISTRY 2022; 194:113018. [PMID: 34837762 DOI: 10.1016/j.phytochem.2021.113018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 06/13/2023]
Abstract
The roots of Kadsura coccinea is commonly used in Tujia ethnomedicine, named "heilaohu", having the effect of treating rheumatic arthritis (RA). Chemical investigation on the ethanol extract of heilaohu led to the isolation of one undescribed cuparane sesquiterpenoid, heilaohusesquiterpenoid A, one undescribed carotane sesquiterpenoids, heilaohusesquiterpenoid B, and eighteen sesquiterpene derivatives. Their structures were subsequently determined based on their 1D and 2D-NMR, HR-ESI-MS, and ECD spectroscopic data. Gaultheriadiolide was the most cytotoxic compound against the proliferation of rheumatoid arthritis-fibroblastoid synovial (RA-FLS) cells with an IC50 value of 9.37 μM. In the same line, nine compounds exhibited significant inhibition effects against TNF-α and IL-6 release in the LPS-induced RAW264.7 cells with IC50 values ranging between 1.03 and 10.99 μM. The potential molecular mechanisms of the active compounds against RA were established through pharmacological network analysis based on the initial screening results. Experimental validation showed that gaultheriadiolide suppressed inflammation by inhibiting the NF-kB and JAK2/STAT3 pathways. This study enriches the structural diversity of sesquiterpenes in K. coccinea and lays a foundation for further anti-RA and anti-inflammatory studies.
Collapse
Affiliation(s)
- Yupei Yang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Yongbei Liu
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Huanghe Yu
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Qingling Xie
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Bin Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Sai Jiang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Wei Su
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Yu Mao
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Bin Li
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Caiyun Peng
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Yuqing Jian
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
8
|
Parihar N, Bhatt LK. Deubiquitylating enzymes: potential target in autoimmune diseases. Inflammopharmacology 2021; 29:1683-1699. [PMID: 34792672 DOI: 10.1007/s10787-021-00890-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/28/2021] [Indexed: 12/28/2022]
Abstract
The ubiquitin-proteasome pathway is responsible for the turnover of different cellular proteins, such as transport proteins, presentation of antigens to the immune system, control of the cell cycle, and activities that promote cancer. The enzymes which remove ubiquitin, deubiquitylating enzymes (DUBs), play a critical role in central and peripheral immune tolerance to prevent the development of autoimmune diseases and thus present a potential therapeutic target for the treatment of autoimmune diseases. DUBs function by removing ubiquitin(s) from target protein and block ubiquitin chain elongation. The addition and removal of ubiquitin molecules have a significant impact on immune responses. DUBs and E3 ligases both specifically cleave target protein and modulate protein activity and expression. The balance between ubiquitylation and deubiquitylation modulates protein levels and also protein interactions. Dysregulation of the ubiquitin-proteasome pathway results in the development of various autoimmune diseases such as inflammatory bowel diseases (IBD), psoriasis, multiple sclerosis (MS), systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). This review summarizes the current understanding of ubiquitination in autoimmune diseases and focuses on various DUBs responsible for the progression of autoimmune diseases.
Collapse
Affiliation(s)
- Niraj Parihar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
9
|
Han Y, Wang J, Li S, Li Y, Zhang Y, Zhang R, Zhang Y, Fan H, Shi H, Pan J, Song G, Ge L, Wang L. Isopsoralen ameliorates rheumatoid arthritis by targeting MIF. Arthritis Res Ther 2021; 23:243. [PMID: 34535196 PMCID: PMC8447788 DOI: 10.1186/s13075-021-02619-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 08/27/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Isopsoralen (IPRN), one of the active ingredients of Psoralea corylifolia Linn, has anti-inflammatory properties. We attempted to investigate the inhibitory effects of IPRN on rheumatoid arthritis (RA) and characterize its potential mechanism. METHODS RA fibroblast-like synoviocytes (FLSs) and mice with collagen-induced arthritis (CIA) were used as in vitro and in vivo models to analyze the antiarthritic effect of IPRN. Histological analysis of the inflamed joints from mice with CIA was performed using microcomputed tomography (micro-CT) and hematoxylin-eosin (HE) staining. RNA sequencing (RNA-Seq), network pharmacology analysis, molecular docking, drug affinity responsive target stability (DARTS) assay, and cellular thermal shift assay (CETSA) were performed to evaluate the targets of IPRN. RESULTS IPRN ameliorated the inflammatory phenotype of RA FLSs by inhibiting their cytokine production, migration, invasion, and proangiogenic ability. IPRN also significantly reduced the severity of CIA in mice by decreasing paw thickness, arthritis score, bone damage, and serum inflammatory cytokine levels. A mechanistic study demonstrated that macrophage migration inhibitory factor (MIF), a key protein in the inflammatory process, was the specific target by which IPRN exerted its anti-inflammatory effects in RA FLSs. CONCLUSION Our study demonstrates the antiarthritic effect of IPRN, which suggests the therapeutic potential of IPRN in RA.
Collapse
Affiliation(s)
- Yi Han
- China Academy of Chinese Medical Sciences, Guang'anmen Hospital, Beijing, China
| | - Jinguang Wang
- Department of Orthopedics, Dezhou People's Hospital, Dezhou, Shandong, China
| | - Shufeng Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yi Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yongli Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ruojia Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China
| | - Huancai Fan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haojun Shi
- The Second Clinical Medical College, Henan University of Chinese Medicine, Jinan, China
| | - Jihong Pan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China
| | - Guanhua Song
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Luna Ge
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China.
| | - Lin Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China.
| |
Collapse
|
10
|
Ma K, Zhang C, Li W. Fascin1 mediated release of pro-inflammatory cytokines and invasion/migration in rheumatoid arthritis via the STAT3 pathway. Cell Cycle 2021; 20:2210-2220. [PMID: 34499588 DOI: 10.1080/15384101.2021.1974790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, multi-factorial disease characterized by Synovial hyperplasia, chronic inflammation, and autoimmune reaction. Fascin1 overexpression has been implicated in cancer, immune, and inflammatory diseases. However, the relationship between Fascin1 and rheumatoid arthritis (RA) has not yet been determined. We investigated whether Fascin1 could modulate pro-inflammatory cytokine secretion and the proliferation, apoptosis, and invasion/migration of fibroblast-like synoviocytes (RA-FLSs). Fascin 1 was suppressed with a short interfering (si)RNA approach. Functional analysis contained MTT assay, flow cytometry,Transwell™ assays, wound healing, Quantitative polymerase chain reaction and western blotting were used to detect cell proliferation,apoptosis ratio, invasion/ migration, the mRNA and protein expression of the realted markers, respectively. Overexpression of fascin1 was observed in RA-FLSs group compared with control group. Fascin1 expression positively correlated with changes in the expression of RA disease activity markers (RF, CRP, and DAB28, respectively). We also observed a significant positive correlation between Fascin1 and STAT3 mRNA levels in RA- FLSs.Fascin1 silencing attenuated the expression of pro-inflammatory cytokines; reduced FLS proliferation in vitro; and increased apoptosis ratio and bax, cleaved PARP, and caspase-3 expression. si- Fascin1 transfection delayed RA-FLS invasion/migration and reversed the epithelial- mesenchymal transition. These data suggest that Fascin1 exerts positive effects on the proliferation, cell cycle, and invasion/migration of RA-FLSs by activating signal transducer and activator of transcription 3 signaling.After all, Fascin1 contributed to RA development.
Collapse
Affiliation(s)
- Kun Ma
- Luoyang Orthopaedic Hospital of Henan Province & Orthopaedic Hospital of Henan Province, Luoyang, Henan, P.R. China
| | - Chuan Zhang
- Luoyang Orthopaedic Hospital of Henan Province & Orthopaedic Hospital of Henan Province, Luoyang, Henan, P.R. China
| | - Wuyin Li
- Luoyang Orthopaedic Hospital of Henan Province & Orthopaedic Hospital of Henan Province, Luoyang, Henan, P.R. China
| |
Collapse
|
11
|
Ren Z, Lv M, Yu Q, Bao J, Lou K, Li X. MicroRNA-370-3p shuttled by breast cancer cell-derived extracellular vesicles induces fibroblast activation through the CYLD/Nf-κB axis to promote breast cancer progression. FASEB J 2021; 35:e21383. [PMID: 33629796 DOI: 10.1096/fj.202001430rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer is a malignancy arising in the mammary epithelial tissues. Recent studies have indicated the abundance of microRNAs (miRNAs) in extracellular vesicles (EVs), and their interactions have been illustrated to exert crucial roles in the cell-to-cell communication. The present study focused on investigating whether EV-delivered miR-370-3p affects breast cancer. Initially, the miR-370-3p expression pattern was examined in the cancer-associated fibroblasts (CAFs), normal fibroblasts (NFs), and cancerous cells-derived EVs. The relation of miR-370-3p to CYLD was assessed using luciferase activity assay. Afterwards, based on ectopic expression and depletion experiments in the MCF-7 breast cancer cells, we evaluated stemness, migration, invasion, and sphere formation ability, and EMT, accompanied with measurement on the expression patterns of pro-inflammatory factors and nuclear factor-kappa B (NF-κB) signaling-related genes. Finally, tumorigenesis and proliferation were analyzed in vivo using a nude mouse xenograft model. The in vitro experiments revealed that breast cancer cell-derived EVs promoted NF activation, while activated fibroblasts contributed to enhanced stemness, migration, invasion, as well as EMT of cancerous cells. In addition, EVs could transfer miR-370-3p from breast cancer cells to NFs, and EV-encapsulated miR-370-3p was also found to facilitate fibroblast activation. Mechanistically, EV-encapsulated miR-370-3p downregulated the expression of CYLD through binding to its 3'UTR and activated the NF-κB signaling pathway, thereby promoting the cellular functions in vitro and in vivo in breast cancer. Taken together, EVs secreted by breast cancer cells could carry miR-370-3p to aggravate breast cancer through downregulating CYLD expression and activating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhaojun Ren
- Department of Pathology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research &, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Mengmeng Lv
- Department of Gynecologic Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research &, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Qiao Yu
- Department of General Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research &, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Jun Bao
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research &, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Kexin Lou
- Department of Pathology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research &, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Xiujuan Li
- Department of General Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research &, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
12
|
Chen Q, Lei YQ, Liu JF, Wang ZC, Cao H. Beneficial effects of chlorogenic acid treatment on neuroinflammation after deep hypothermic circulatory arrest may be mediated through CYLD/NF-κB signaling. Brain Res 2021; 1767:147572. [PMID: 34216581 DOI: 10.1016/j.brainres.2021.147572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/30/2021] [Accepted: 06/25/2021] [Indexed: 10/21/2022]
Abstract
Deep hypothermic circulatory arrest (DHCA) during heart surgery may induce neuroinflammation leading to neurocognitive dysfunction. Chlorogenic acid (CA) is a common phytochemical, which can attenuate neuroinflammation. Nevertheless, the underlying mechanism involved in the anti-inflammatory effect of CA after DHCA is unknown. The present study therefore characterized the anti-inflammatory functions of CA after DHCA using in vivo and in vitro DHCA models. The activation of microglia, inflammatory cytokine levels, and the NF-κB pathway were measured. The results showed that CA treatment ameliorated neurocognitive function and reduced the inflammatory cytokine levels in the brain and circulation. Furthermore, the microglial and NF-κB activations were suppressed after DHCA. CA exerted the same anti-inflammatory effect in hypothermia OGD microglial cells as the in vivo study. Additional studies indicated that the regulation of ubiquitin ligase activity of TRAF6 and RIP1 by CYLD was related to the mechanism involving inhibition of CA in the NF-κB pathway. Together, the results showed that CA may attenuate neuroinflammation after DHCA by modulating the signaling of CYLD/NF-κB.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China.
| | - Yu-Qing Lei
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Jian-Feng Liu
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Zeng-Chun Wang
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Hua Cao
- Department of Cardiac Surgery, Fujian Branch of Shanghai Children's Medical Cente, Fuzhou, China; Fujian Children's Hospital, Fuzhou, China; Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China
| |
Collapse
|
13
|
Alameda JP, García-García VA, López S, Hernando A, Page A, Navarro M, Moreno-Maldonado R, Paramio JM, Ramírez Á, García-Fernández RA, Casanova ML. CYLD Inhibits the Development of Skin Squamous Cell Tumors in Immunocompetent Mice. Int J Mol Sci 2021; 22:6736. [PMID: 34201751 PMCID: PMC8268443 DOI: 10.3390/ijms22136736] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 11/17/2022] Open
Abstract
Cylindromatosis (CYLD) is a deubiquitinase (DUB) enzyme that was initially characterized as a tumor suppressor of adnexal skin tumors in patients with CYLD syndrome. Later, it was also shown that the expression of functionally inactive mutated forms of CYLD promoted tumor development and progression of non-melanoma skin cancer (NMSC). However, the ability of wild-type CYLD to inhibit skin tumorigenesis in vivo in immunocompetent mice has not been proved. Herein, we generated transgenic mice that express the wild type form of CYLD under the control of the keratin 5 (K5) promoter (K5-CYLDwt mice) and analyzed the skin properties of these transgenic mice by WB and immunohistochemistry, studied the survival and proliferating characteristics of primary keratinocytes, and performed chemical skin carcinogenesis experiments. As a result, we found a reduced activation of the nuclear factor kappa B (NF-κB) pathway in the skin of K5-CYLDwt mice in response to tumor necrosis factor-α (TNF-α); accordingly, when subjected to insults, K5-CYLDwt keratinocytes are prone to apoptosis and are protected from excessive hyperproliferation. Skin carcinogenesis assays showed inhibition of tumor development in K5-CYLDwt mice. As a mechanism of this tumor suppressor activity, we found that a moderate increase in CYLD expression levels reduced NF-κB activation, which favored the differentiation of tumor epidermal cells and inhibited its proliferation; moreover, it decreased tumor angiogenesis and inflammation. Altogether, our results suggest that increased levels of CYLD may be useful for anti-skin cancer therapy.
Collapse
Affiliation(s)
- Josefa P. Alameda
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (J.P.A.); (V.A.G.-G.); (A.H.); (A.P.); (M.N.); (R.M.-M.); (J.M.P.); (Á.R.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Verónica A. García-García
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (J.P.A.); (V.A.G.-G.); (A.H.); (A.P.); (M.N.); (R.M.-M.); (J.M.P.); (Á.R.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
| | - Silvia López
- Department of Animal Medicine and Surgery, Facultad de Veterinaria, UCM, 28040 Madrid, Spain; (S.L.); (R.A.G.-F.)
| | - Ana Hernando
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (J.P.A.); (V.A.G.-G.); (A.H.); (A.P.); (M.N.); (R.M.-M.); (J.M.P.); (Á.R.)
- Bionomous Sàrl, PFL Innovation Park, Bâtiment, FCH-1015 Lausanne, Switzerland
| | - Angustias Page
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (J.P.A.); (V.A.G.-G.); (A.H.); (A.P.); (M.N.); (R.M.-M.); (J.M.P.); (Á.R.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Manuel Navarro
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (J.P.A.); (V.A.G.-G.); (A.H.); (A.P.); (M.N.); (R.M.-M.); (J.M.P.); (Á.R.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Rodolfo Moreno-Maldonado
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (J.P.A.); (V.A.G.-G.); (A.H.); (A.P.); (M.N.); (R.M.-M.); (J.M.P.); (Á.R.)
- Bio-innova Consulting, 28049 Madrid, Spain
| | - Jesús M. Paramio
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (J.P.A.); (V.A.G.-G.); (A.H.); (A.P.); (M.N.); (R.M.-M.); (J.M.P.); (Á.R.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ángel Ramírez
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (J.P.A.); (V.A.G.-G.); (A.H.); (A.P.); (M.N.); (R.M.-M.); (J.M.P.); (Á.R.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Rosa A. García-Fernández
- Department of Animal Medicine and Surgery, Facultad de Veterinaria, UCM, 28040 Madrid, Spain; (S.L.); (R.A.G.-F.)
| | - María Llanos Casanova
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (J.P.A.); (V.A.G.-G.); (A.H.); (A.P.); (M.N.); (R.M.-M.); (J.M.P.); (Á.R.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
14
|
Qin H, Wu T, Liu J, Wang G, Fan L, Wang B, Shen Y. MALT-1 inhibition attenuates the inflammatory response of ankylosing spondylitis by targeting NF-κB activation. Injury 2021; 52:1287-1293. [PMID: 33827774 DOI: 10.1016/j.injury.2021.03.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/11/2021] [Accepted: 03/24/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND The present study aimed to investigate the effects of mucosa-associated lymphoid tissue lymphoma translocation protein (MALT)-1 on ankylosing spondylitis and its underlying mechanisms. METHODS Proteoglycan induced ankylosing spondylitis (PGIA) mouse model was established and the expression patterns of MALT-1 were determined in joint tissue. Next, the mice were intraarticularly administrated with MALT-1 in the PGIA mouse model. Meanwhile, shRNA was intraarticularly administrated to PGIA mice. The incidence of arthritis and clinical score was evaluated. Besides, the levels of inflammatory cytokines and matrix metalloproteinases (MMPs) were measured. Protein expressions of full-length CYLD (FL-CYLD), C-terminal cleavage fragment (CYLD-CL), and nuclear factor (NF)-κB were determined. RESULTS The mRNA and protein levels of MALT1 were increased in the PGIA mouse model. The treatment of MALT-1 accelerated arthritis incidence and joint damage, whereas shMALT-1 suppressed arthritis symptoms in the PGIA mouse model. In addition, treatment of shMALT-1 suppressed the levels of inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β), MMP-3, and MMP-9. Furthermore, the treatment of shMALT-1 suppressed the levels of CYLD and NF-κB in the joint tissues in the PGIA mouse model. CONCLUSION The inhibition of MALT-1 suppressed the inflammatory response in ankylosing spondylitis in part by the regulation of CYLD and NF-κB.
Collapse
Affiliation(s)
- Hu Qin
- Department of Orthopedics, the Second Affiliated Hospital of Nanjing Medical University, No. 121 Jiangjiayuan, Nanjing 210011, Jiangsu, China
| | - Tao Wu
- Department of Orthopedics, the Second Affiliated Hospital of Nanjing Medical University, No. 121 Jiangjiayuan, Nanjing 210011, Jiangsu, China.
| | - Jun Liu
- Department of Orthopedics, the Second Affiliated Hospital of Nanjing Medical University, No. 121 Jiangjiayuan, Nanjing 210011, Jiangsu, China.
| | - Gang Wang
- Department of Orthopedics, the Second Affiliated Hospital of Nanjing Medical University, No. 121 Jiangjiayuan, Nanjing 210011, Jiangsu, China
| | - Lei Fan
- Department of Orthopedics, the Second Affiliated Hospital of Nanjing Medical University, No. 121 Jiangjiayuan, Nanjing 210011, Jiangsu, China
| | - Boyao Wang
- Department of Orthopedics, the Second Affiliated Hospital of Nanjing Medical University, No. 121 Jiangjiayuan, Nanjing 210011, Jiangsu, China
| | - Yajun Shen
- Department of Orthopedics, the Second Affiliated Hospital of Nanjing Medical University, No. 121 Jiangjiayuan, Nanjing 210011, Jiangsu, China
| |
Collapse
|
15
|
Schuster R, Rockel JS, Kapoor M, Hinz B. The inflammatory speech of fibroblasts. Immunol Rev 2021; 302:126-146. [PMID: 33987902 DOI: 10.1111/imr.12971] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Activation of fibroblasts is a key event during normal tissue repair after injury and the dysregulated repair processes that result in organ fibrosis. To most researchers, fibroblasts are rather unremarkable spindle-shaped cells embedded in the fibrous collagen matrix of connective tissues and/or deemed useful to perform mechanistic studies with adherent cells in culture. For more than a century, fibroblasts escaped thorough classification due to the lack of specific markers and were treated as the leftovers after all other cells have been identified from a tissue sample. With novel cell lineage tracing and single cell transcriptomics tools, bona fide fibroblasts emerge as only one heterogeneous sub-population of a much larger group of partly overlapping cell types, including mesenchymal stromal cells, fibro-adipogenic progenitor cells, pericytes, and/or perivascular cells. All these cells are activated to contribute to tissue repair after injury and/or chronic inflammation. "Activation" can entail various functions, such as enhanced proliferation, migration, instruction of inflammatory cells, secretion of extracellular matrix proteins and organizing enzymes, and acquisition of a contractile myofibroblast phenotype. We provide our view on the fibroblastic cell types and activation states playing a role during physiological and pathological repair and their crosstalk with inflammatory macrophages. Inflammation and fibrosis of the articular synovium during rheumatoid arthritis and osteoarthritis are used as specific examples to discuss inflammatory fibroblast phenotypes. Ultimately, delineating the precursors and functional roles of activated fibroblastic cells will contribute to better and more specific intervention strategies to treat fibroproliferative and fibrocontractive disorders.
Collapse
Affiliation(s)
- Ronen Schuster
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.,PhenomicAI, MaRS Centre, Toronto, ON, Canada
| | - Jason S Rockel
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Mohit Kapoor
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Ji M, Ryu HJ, Hong JH. Signalling and putative therapeutic molecules on the regulation of synoviocyte signalling in rheumatoid arthritis. Bone Joint Res 2021; 10:285-297. [PMID: 33890482 PMCID: PMC8077181 DOI: 10.1302/2046-3758.104.bjr-2020-0331.r1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by symmetrical and chronic polyarthritis. Fibroblast-like synoviocytes are mainly involved in joint inflammation and cartilage and bone destruction by inflammatory cytokines and matrix-degrading enzymes in RA. Approaches that induce various cellular growth alterations of synoviocytes are considered as potential strategies for treating RA. However, since synoviocytes play a critical role in RA, the mechanism and hyperplastic modulation of synoviocytes and their motility need to be addressed. In this review, we focus on the alteration of synoviocyte signalling and cell fate provided by signalling proteins, various antioxidant molecules, enzymes, compounds, clinical candidates, to understand the pathology of the synoviocytes, and finally to achieve developed therapeutic strategies of RA. Cite this article: Bone Joint Res 2021;10(4):285–297.
Collapse
Affiliation(s)
- Minjeong Ji
- Department of Physiology, College of Medicine, Gachon University, Lee Gil Ya Cancer and Diabetes Institute, Incheon, South Korea
| | - Hee Jung Ryu
- Department of Rheumatology, Gachon University Gil Medical Center, Incheon, South Korea
| | - Jeong Hee Hong
- Department of Physiology, College of Medicine, Gachon University, Lee Gil Ya Cancer and Diabetes Institute, Incheon, South Korea.,Department of Health Sciences and Technology, GAIHST, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea
| |
Collapse
|
17
|
Hongbo M, Yanjiao D, Shuo W, Kun S, Yanjie L, Mengmeng L. Podocyte RNF166 deficiency alleviates diabetic nephropathy by mitigating mitochondria impairment and apoptosis via regulation of CYLD signal. Biochem Biophys Res Commun 2021; 545:46-53. [PMID: 33545631 DOI: 10.1016/j.bbrc.2020.12.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/04/2020] [Indexed: 01/12/2023]
Abstract
Diabetic nephropathy (DN) is a major cause of renal failure in diabetic patients. RING-finger protein 166 (RNF166), composed of an N-terminal RING domain and C-terminal ubiquitin interaction motif, plays a critical role in mediating various cellular processes. However, its potential in DN has not been investigated. In the present study, we found that DN patients exhibited significantly increased expression of RNF166 in renal tissues compared with the normal individuals, and abundant RNF166 was detected in podocytes. We then showed that podocyte-conditional RNF166 knockout (RNF166cKO) markedly reduced blood glucose levels and ameliorated renal dysfunction in streptozotocin (STZ)-induced diabetic mice. Additionally, abnormal histological changes and podocyte injury were observed in STZ-induced diabetic mice, while being markedly ameliorated by RNF166cKO. Furthermore, podocyte-specific RNF166 deficiency considerably mitigated apoptosis and mitochondrial impairments in glomeruli podocytes of STZ-challenged mice through suppressing Caspase-3 cleavage and improving mitochondrial fission-associated molecules. In vitro studies further confirmed that high glucose (HG) induced mitochondrial dysfunction, along with enhanced releases of Cyto-c from mitochondria and elevated expression of cleaved Caspase-9, contributing to intrinsic apoptosis in podocytes. Intriguingly, these effects triggered by HG were dramatically ameliorated by RNF166 knockout. Mechanistically, we demonstrated that RNF166 directly interacted with cylindromatosis (CYLD), and negatively regulated CYLD expression. Notably, RNF166 knockout-attenuated mitochondrial damage and apoptosis were mainly through CYLD in podocytes upon HG stimulation. Together, all these findings provided new insights into the novel effects of RNF166 on maintaining mitochondrial function and apoptosis in podocytes during DN progression both in vivo and in vitro through interacting with CYLD, indicating that RNF166/CYLD may be an innovative therapeutic target for developing effective strategy against DN development.
Collapse
Affiliation(s)
- Ma Hongbo
- Department of Nephrology, Shandong Province Zibo Central Hospital, 54 West Communist Youth League Road, Zhangdian District, Zibo, Shandong, 255000, China.
| | - Dong Yanjiao
- Department of Nephrology, Shandong Province Zibo Central Hospital, 54 West Communist Youth League Road, Zhangdian District, Zibo, Shandong, 255000, China
| | - Wang Shuo
- Department of Nephrology, Shandong Province Zibo Central Hospital, 54 West Communist Youth League Road, Zhangdian District, Zibo, Shandong, 255000, China
| | - Sun Kun
- Department of Nephrology, Shandong Province Zibo Central Hospital, 54 West Communist Youth League Road, Zhangdian District, Zibo, Shandong, 255000, China
| | - Liu Yanjie
- Department of Nephrology, Shandong Province Zibo Central Hospital, 54 West Communist Youth League Road, Zhangdian District, Zibo, Shandong, 255000, China
| | - Li Mengmeng
- Department of Nephrology, Shandong Province Zibo Central Hospital, 54 West Communist Youth League Road, Zhangdian District, Zibo, Shandong, 255000, China
| |
Collapse
|
18
|
Fu YW, Li L, Wang XQ, Zhou Y, Zhu LF, Mei YM, Xu Y. The inhibitory effect of the deubiquitinase cylindromatosis (CYLD) on inflammatory responses in human gingival fibroblasts. Oral Dis 2020; 27:1487-1497. [PMID: 33031609 DOI: 10.1111/odi.13672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 09/22/2020] [Accepted: 10/01/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Experiments were performed to evaluate CYLD expression in human gingival tissue samples and to examine the effects of CYLD on inflammatory responses in lipopolysaccharide (LPS)- or TNF-α-stimulated human gingival fibroblasts (HGFs). METHODS Immunohistochemistry for CYLD and p65 expression was performed with healthy and inflamed gingival tissue samples. siRNA was used to knock down the expression of CYLD in HGFs. Upon LPS or TNF-α stimulation, NF-κB activation was detected in control and CYLD-knockdown HGFs. RT-PCR was applied to determine gene expression. Western blot analyses were employed to assess protein expression. Immunofluorescence staining was carried out to evaluate the nuclear translocation of p65. RESULTS Immunohistochemical staining showed the expression of CYLD in human gingival tissues. In addition, CYLD protein expression was reduced in inflamed gingival tissue samples compared with healthy tissue samples. CYLD knockdown greatly enhanced the mRNA expression of proinflammatory cytokines in LPS- or TNF-α-stimulated HGFs. Furthermore, knocking down CYLD expression increased LPS-stimulated NF-κB activation in HGFs. Unexpectedly, CYLD knockdown did not affect TNF-α-induced NF-κB activation. CONCLUSIONS Our results suggest that CYLD participates in periodontal inflammatory responses by negatively regulating LPS-induced NF-κB signalling.
Collapse
Affiliation(s)
- Yong-Wei Fu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China.,Department of Stomatology, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Qian Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Li-Fang Zhu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - You-Min Mei
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Chen Y, Lei Y, Lin J, Huang Y, Zhang J, Chen K, Sun S, Lin X. The LINC01260 Functions as a Tumor Suppressor via the miR-562/CYLD/NF-κB Pathway in Non-Small Cell Lung Cancer. Onco Targets Ther 2020; 13:10707-10719. [PMID: 33116647 PMCID: PMC7585791 DOI: 10.2147/ott.s253730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Recently, long noncoding RNAs (lncRNAs) have been identified as novel and potential therapeutic targets in various cancer types. Nonetheless, the levels and biological effects of lncRNAs in non-small cell lung cancer (NSCLC) remain largely unknown. In this study, we aimed to identify the effects of lncRNA-LINC01260 throughout the progression of NSCLC and explore the underlying mechanism. METHODS Quantitative real-time PCR (qRT-PCR) and Western blot were performed to measure LINC01260, miR-562, and CYLD expression and protein levels. Luciferase reporter assay was employed to investigate the relationship between LINC01260 and miR-562, and miR-562 and CYLD, respectively. The viability and migration of cells were evaluated using CCK-8, colony formation, and transwell assays. The effects of LINC01260 were identified through tumorigenesis in vivo. ELISA was performed to detect the activity of NF-κB and p65 expression. RESULTS In NSCLC tissues and cell lines, LINC01260 expression was downregulated, which corresponded to a lower survival rate of patients with NSCLC. Knockdown of LINC01260 accelerated the proliferation, colony formation, and migration of NSCLC cells. Moreover, downregulation of LINC01260 inhibited apoptosis of NSCLC cells by regulating the expression of Bcl-2 and Bax proteins in vitro. In vivo, the downregulation of LINC01260 promoted tumor growth. miR-562 was identified as the target gene of LINC01260, which was upregulated in NSCLC tumors. Furthermore, CYLD was identified as the target gene of miR-562. The effects of LINC01260 were exerted by regulating CYLD via sponging miR-562. ELISA confirmed that the upregulation of CYLD inhibited NF-κB activity; however, the co-transfection of sh-LINC01260 partly reversed the inhibition. Additionally, CYLD reduced p65 expression; however, downregulation of LINC01260 slightly increased the expression level. CONCLUSION This study revealed a novel LINC01260/miR-562/CYLD/NF-κB pathway in the pathogenesis of NSCLC and suggested a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Yangming Chen
- Department of Thoracic Surgery, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian350001, People’s Republic of China
| | - Yujie Lei
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, The International Cooperation Key Laboratory of Regional Tumor in High Altitude Area, Kunming, Yunnan650106, People’s Republic of China
| | - Jianbin Lin
- Department of Thoracic Surgery, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian350001, People’s Republic of China
| | - Yunchao Huang
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, The International Cooperation Key Laboratory of Regional Tumor in High Altitude Area, Kunming, Yunnan650106, People’s Republic of China
| | - Jiguang Zhang
- Department of Thoracic Surgery, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian350001, People’s Republic of China
| | - Kai Chen
- Department of Thoracic Surgery, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian350001, People’s Republic of China
| | - Shihui Sun
- Department of Thoracic Surgery, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian350001, People’s Republic of China
| | - Xing Lin
- Department of Thoracic Surgery, Shengli Clinical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian350001, People’s Republic of China
| |
Collapse
|
20
|
Kong L, Wang L, Zhao Q, Di G, Wu H. Rhodojaponin II inhibits TNF-α-induced inflammatory cytokine secretion in MH7A human rheumatoid arthritis fibroblast-like synoviocytes. J Biochem Mol Toxicol 2020; 34:e22551. [PMID: 32613688 DOI: 10.1002/jbt.22551] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/29/2020] [Accepted: 05/29/2020] [Indexed: 12/19/2022]
Abstract
Rhodojaponin II (R-II) has been shown to possess anti-inflammatory activity. Herein, we aimed to explore the effect of R-II on tumor necrosis factor-α (TNF-α)-induced inflammation in MH7A rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLSs). We found that R-II treatment at high concentration suppressed the viability of MH7A cells. R-II suppressed the levels of nitric oxide and prostaglandin E2, and inhibited messenger RNA expression and concentrations of interleukin-1β (IL-1β), IL-6 and matrix metalloproteinase-1 in TNF-α-stimulated RA-FLSs. Additionally, R-II repressed TNF-α-induced activation of the Akt, nuclear factor-κB (NF-κB), and toll-like receptor 4 (TLR4)/MyD88 pathways in MH7A cells. Inhibition of the Akt, NF-κB, and TLR4/MyD88 pathways by the corresponding inhibitors reinforced the inhibitory effect of R-II on TNF-α-induced inflammatory cytokine secretion in MH7A cells. R-II ameliorated the severity of collagen-induced arthritis in mice by inhibiting inflammation. In conclusion, R-II repressed TNF-α-induced inflammatory response in MH7A cells by inactivating the Akt, NF-κB, and TLR4/MyD88 pathways.
Collapse
Affiliation(s)
- Lingli Kong
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Laifang Wang
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Qing Zhao
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Guijuan Di
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Huiqiang Wu
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
21
|
Li Z, Qi F, Li F. Identification of drug targets and potential molecular mechanisms for Wantong Jingu Tablet extract in treatment of rheumatoid arthritis: bioinformatics analysis of fibroblast-like synoviocytes. Chin Med 2020; 15:59. [PMID: 32518584 PMCID: PMC7275334 DOI: 10.1186/s13020-020-00339-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/25/2020] [Indexed: 01/20/2023] Open
Abstract
Background Rheumatoid arthritis-fibroblast-like synoviocytes (RA-FLSs) play important roles in pathogenesis of rheumatoid arthritis (RA). Wantong Jingu Tablet (WJT), a mixture of traditional Chinese medicine, is a potentially effective therapy for RA, but its underlying mechanism is unclear. In this study, we explore the effects of WJT on human RA-FLSs and the underlying molecular mechanism. Methods The major components of WJT were determined using ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UHPLC-QTOF/MS). Cell proliferative ability was evaluated by CCK-8, colony formation assay, and EdU incorporation assay. Cell apoptotic capacity was examined by caspase-3 and caspase-9 activity test. Protein levels of Bax and Bcl-2 were investigated by western blotting. High-throughput sequencing and bioinformatics analysis were conducted to screen and identify targeted genes, followed by identification by qRT-PCR and western blotting. Results In this study, we have identified 346 compounds in WJT. Our results showed that WJT inhibited the RA-FLSs proliferation, and promoted apoptosis in a dose- and time-dependent manner. More importantly, 184 differentially expressed genes (DEGs) has been screened after WJT treatment based on DEGSeq2 and 278 DEGs was identified by DEGSeq2 combined with WGCNA. Then, 10 hub genes were identified based on two different analyses, while the expression levels of only SMC3, THOC1, BUB1, and STAG2 were decreased after WJT treatment, which was identical to the sequencing profiles. Conclusions WJT exerted its anti-proliferation and pro-apoptosis effects possibly through suppressing the expression of SMC3, THOC1, BUB1, and STAG2 in RA-FLSs. Thus, therapeutics targeting these genes may be a promising strategy for rescuing RA.
Collapse
Affiliation(s)
- Zhaodong Li
- Department of Pathogen Biology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, No. 126 Xinmin Street, Changchun, 130021 Jilin China
| | - Fangyuan Qi
- Department of Pathogen Biology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, No. 126 Xinmin Street, Changchun, 130021 Jilin China
| | - Fan Li
- Department of Pathogen Biology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, No. 126 Xinmin Street, Changchun, 130021 Jilin China.,The Key Laboratory for Bionics Engineering, Ministry of Education, China, Jilin University, Changchun, 130021 Jilin China.,Engineering Research Center for Medical Biomaterials of Jilin Province, Jilin University, Changchun, 130021 Jilin China.,Key Laboratory for Biomedical Materials of Jilin Province, Jilin University, Changchun, 130021 Jilin China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang China
| |
Collapse
|
22
|
Liang T, Song M, Xu K, Guo C, Xu H, Zhang H, Xu L. TRIM32 promotes inflammatory responses in rheumatoid arthritis fibroblast‐like synoviocytes. Scand J Immunol 2020; 91:e12876. [PMID: 32145086 DOI: 10.1111/sji.12876] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/11/2020] [Accepted: 03/02/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Tian Liang
- Department of Orthopedics Gansu Traditional Chinese Medicine University Lanzhou China
| | - Min Song
- Department of Orthopedics Gansu Traditional Chinese Medicine University Lanzhou China
| | - Kewu Xu
- Department of Orthopedics Affiliated Hospital of Gansu Traditional Chinese Medicine University Lanzhou China
| | - Chenglong Guo
- Department of Orthopedics Affiliated Hospital of Gansu Traditional Chinese Medicine University Lanzhou China
| | - Hongbin Xu
- Department of Orthopedics Affiliated Hospital of Gansu Traditional Chinese Medicine University Lanzhou China
| | - Hongwei Zhang
- Department of Orthopedics Affiliated Hospital of Gansu Traditional Chinese Medicine University Lanzhou China
| | - Lanping Xu
- Department of Dermatology Affiliated Hospital of Gansu Traditional Chinese Medicine University Lanzhou China
| |
Collapse
|
23
|
Dinesh P, Kalaiselvan S, Sujitha S, Rasool M. MiR-145-5p mitigates dysregulated Wnt1/β-catenin signaling pathway in rheumatoid arthritis. Int Immunopharmacol 2020; 82:106328. [PMID: 32088641 DOI: 10.1016/j.intimp.2020.106328] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/04/2020] [Accepted: 02/14/2020] [Indexed: 12/28/2022]
Abstract
Fibroblast-like synoviocytes (FLS) lining the arthritic synovial joint region have been implicated to be a key player in bone remodeling. The uncontrolled proliferation of this cell subtype is strictly regulated by various molecular elements including microRNAs (miRNAs). The Wnt1/β-catenin signaling pathway plays a crucial role in the survival of FLS cells. This study explores the underlying mechanism of miR-145-5p towards the Wnt1/β-catenin pathway. MiR-145-5p depicted a strong binding affinity towards frizzled class receptor 4 (FZD4) 3' UTR, a key receptor complex essential for recognizing circulating Wnt1 molecules. Adjuvant induced arthritic fibroblast-like synoviocytes (AA-FLS) isolated from rats stimulated with Wnt1 (10 ng/ml) elicited active Wnt1/β-catenin signaling. Transfection of miR-145-5p mimic (50 pmol) to AA-FLS stimulated with Wnt1 elicited reduced expression levels of various factors of Wnt1/β-catenin signaling including low-density lipoprotein receptor-related protein 5 (LRP5), dishevelled segment polarity protein 1 (Dvl1) and β-catenin transcription factor. Moreover, pro-inflammatory cytokines (TNFα, IL-1β, IL-6 and IL-23) were regulated compared to the diseased groups. Furthermore, miR-145-5p counterbalanced the levels of receptor activator of nuclear factor kappa B ligand (RANKL) and osteoprotegerin (OPG) at the cellular level, essential for bone remodeling. Hence, we suggest that miR-145-5p regulates the survival/proliferation of FLS cells in RA disease condition through attenuation of Wnt1/β-catenin signaling.
Collapse
Affiliation(s)
- Palani Dinesh
- Immunopathology Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632 014, Tamil Nadu, India
| | - Sowmiya Kalaiselvan
- Immunopathology Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632 014, Tamil Nadu, India
| | - Sali Sujitha
- Immunopathology Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632 014, Tamil Nadu, India
| | - Mahaboobkhan Rasool
- Immunopathology Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632 014, Tamil Nadu, India.
| |
Collapse
|
24
|
Wang T, Jia Q, Chen T, Yin H, Tian X, Lin X, Liu Y, Zhao Y, Wang Y, Shi Q, Huang C, Xu H, Liang Q. Alleviation of Synovial Inflammation of Juanbi-Tang on Collagen-Induced Arthritis and TNF-Tg Mice Model. Front Pharmacol 2020; 11:45. [PMID: 32116720 PMCID: PMC7033619 DOI: 10.3389/fphar.2020.00045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 01/14/2020] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that is primarily characterized by synovial inflammation. In this study, we found that a traditional Chinese decoction, Juanbi-Tang (JBT), JBT attenuated the symptoms of collagen-induced arthritis (CIA) mice and in tumor necrosis factor transgenic (TNF-Tg) mice by attenuating the arthritis index and hind paw thickness. According to histopathological staining of ankle sections, JBT significantly decreased the area of inflammation and reduced bone destruction of ankle joints in both these two types of mice. Moreover, decreased tartaric acid phosphatase-positive osteoclasts were observed in the JBT group compared with those found in the control group. We also revealed that JBT suppressed monocytes and T cells as well as the production of CCL2, CCR6, and CXCR3 ligands. We next used high-performance liquid chromatography to investigate the components and pharmacological properties of this classical herbal medicine in traditional Chinese medicine. Based on network pharmacology, we performed computational prediction simulation of the potential targets of JBT, which indicated the NF-kappa B pathway as its target, which was confirmed in vitro. JBT suppressed the production of pro-inflammatory cytokines including interleukin-6 (IL-6) and IL-8, and inhibited the expression of matrix metalloproteinase 1 in fibroblast-like synoviocytes derived from RA patients (MH7A cells). Furthermore, JBT also suppressed the phosphorylation of p38, JNK, and p65 in TNF-α-treated MH7A cells. In summary, this study proved that JBT could inhibit synovial inflammation and bone destruction, possibly by blocking the phosphorylation of NF-kappa B pathway-mediated production of proinflammatory effectors.
Collapse
Affiliation(s)
- Tengteng Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingyun Jia
- Second Ward of Trauma Surgery Department, Linyi People's Hospital, Linyi, China
| | - Tao Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Yin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoting Tian
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xi Lin
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY, United States
| | - Yang Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenggang Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
25
|
Komatsu K, Nam DH, Lee JY, Yoneda G, Yan C, Li JD. Vinpocetine Suppresses Streptococcus pneumoniae-Induced Inflammation via Inhibition of ERK1 by CYLD. THE JOURNAL OF IMMUNOLOGY 2020; 204:933-942. [PMID: 31900337 DOI: 10.4049/jimmunol.1901299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022]
Abstract
Otitis media (OM) is the most common bacterial infection in children. It remains a major health problem and a substantial socioeconomic burden. Streptococcus pneumoniae (S. pneumoniae) is one of the most common bacterial pathogens causing OM. Innate inflammatory response plays a critical role in host defense against bacterial pathogens. However, if excessive, it has a detrimental impact on the middle ear, leading to middle ear inflammation, a hallmark of OM. Currently, there has been limited success in developing effective therapeutic agents to suppress inflammation without serious side effects. In this study, we show that vinpocetine, an antistroke drug, suppressed S. pneumoniae-induced inflammatory response in cultured middle ear epithelial cells as well as in the middle ear of mice. Interestingly, vinpocetine inhibited S. pneumoniae-induced inflammation via upregulating a key negative regulator cylindromatosis (CYLD). Moreover, CYLD suppressed S. pneumoniae-induced inflammation via inhibiting the activation of ERK. Importantly, the postinfection administration of vinpocetine markedly inhibited middle ear inflammation induced by S. pneumoniae in a well-established mouse OM model. These studies provide insights into the molecular mechanisms underlying the tight regulation of inflammation via inhibition of ERK by CYLD and identified vinpocetine as a potential therapeutic agent for suppressing the inflammatory response in the pathogenesis of OM via upregulating negative regulator CYLD expression.
Collapse
Affiliation(s)
- Kensei Komatsu
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303
| | - Dae-Hwan Nam
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303
| | - Ji-Yun Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303.,College of Pharmacy, Chung-Ang University, Dongjak-gu, Seoul 06974, South Korea; and
| | - Go Yoneda
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303
| | - Chen Yan
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642
| | - Jian-Dong Li
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303;
| |
Collapse
|
26
|
Claudel M, Jouzeau JY, Cailotto F. Secreted Frizzled-related proteins (sFRPs) in osteo-articular diseases: much more than simple antagonists of Wnt signaling? FEBS J 2019; 286:4832-4851. [PMID: 31677330 DOI: 10.1111/febs.15119] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/10/2019] [Accepted: 10/31/2019] [Indexed: 12/30/2022]
Abstract
Osteo-articular diseases are characterized by a dysregulation of joint and/or bone homeostasis. These include diseases affecting the joints originally, such as osteoarthritis and rheumatoid arthritis, or the bone, such as osteoporosis. Inflammation and the involvement of Wingless-related integration site (Wnt) signaling pathways are key pathophysiological features of these diseases resulting in tissue degradation by matrix-degrading enzymes, namely matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinases with thrombospondin motifs (ADAMTs), secreted by the joint resident cells and/or by infiltrating immune cells. Activation of Wnt signaling pathways is modulated by different families of proteins, including Dickkopfs and the secreted Frizzled-related proteins (sFRPs). The sFRP family is composed of five secreted glycoproteins in mammals that regulate Wnt signaling in the extracellular compartment. Indeed, sFRPs are able to bind both to the soluble Wnt ligands and to their cell membrane receptors, the Frizzled proteins. Their expression profile is altered in osteo-articular diseases, suggesting that they could account for the abnormal activation of Wnt pathways. In the present article, we review how sFRPs are more than simple antagonists of the Wnt signaling pathways and discuss their pathophysiological relevance in the context of osteo-articular diseases. We detail their Wnt-dependent and their Wnt-independent roles, with a particular emphasis on their ability to modulate the inflammatory response and extracellular matrix (ECM) remodeling. We also discuss their potential therapeutic use with a focus on bone remodeling, osteo-articular cancers, and tissue engineering.
Collapse
Affiliation(s)
- Marion Claudel
- UMR 7365 CNRS-UL IMoPA, Biopôle de l'Université de Lorraine, Vandoeuvre-Les-Nancy, France
| | - Jean-Yves Jouzeau
- UMR 7365 CNRS-UL IMoPA, Biopôle de l'Université de Lorraine, Vandoeuvre-Les-Nancy, France
| | - Frédéric Cailotto
- UMR 7365 CNRS-UL IMoPA, Biopôle de l'Université de Lorraine, Vandoeuvre-Les-Nancy, France
| |
Collapse
|
27
|
Cai P, Lu Z, Jiang T, Wang Z, Yang Y, Zheng L, Zhao J. Syndecan-4 involves in the pathogenesis of rheumatoid arthritis by regulating the inflammatory response and apoptosis of fibroblast-like synoviocytes. J Cell Physiol 2019; 235:1746-1758. [PMID: 31309562 DOI: 10.1002/jcp.29093] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/27/2019] [Indexed: 12/28/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease, and the pathogenesis of RA is still unknown. Rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs) are of significance in the pathogenesis of RA. In this study, three microarray profiles (GSE55457, GSE55584, and GSE55235) of human joint FLSs from 33 RA patients and 20 normal controls were extracted from the Gene Expression Omnibus Dataset and analyzed to investigate the underlying pathogenesis of RA. As analyzed by the differently expressed genes, gene ontology, Kyoto Encyclopedia of Genes and Genomes pathway enrichment, and protein-protein interaction network analysis, syndecan-4 (SDC4), a receptor of multiple cytokines and chemokines, which played a key role in the regulation of inflammatory response, was found to be an essential regulator in RA. To further validate these results, the levels of SDC4, reactive oxygen species (ROS), nitric oxide (NO), inflammation, and apoptosis in RA-FLSs were examined. SDC4-silenced RA-FLSs were also used. The results demonstrated that SDC4 and the level of ROS, NO, and inflammation were highly expressed while the apoptosis was decreased in RA-FLSs compared with normal FLSs. SDC4 silencing significantly suppressed the levels of ROS, NO, and inflammation; elevated the expression of nuclear factor erythroid 2-related factor 2; and promoted the apoptosis of RA-FLSs. Collectively, our results demonstrated a new mechanism of SDC4 in initiating the inflammation and inhibiting the apoptosis of RA-FLSs and that a potential target for the diagnosis and treatment of RA in the clinic might be developed.
Collapse
Affiliation(s)
- Peian Cai
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhenhui Lu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tongmeng Jiang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zetao Wang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Yifeng Yang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|