1
|
Zhang W, Sun J, Li J, Wang Y, Liu W, Xue W, Yuan Y, Wang X. Comparison of Angio PLanewave UltraSensitive and Power Doppler Ultrasound in Detecting Synovial Blood Flow in Wrist and Finger Joints of Rheumatoid Arthritis Patients. Acad Radiol 2024; 31:1528-1537. [PMID: 37777427 DOI: 10.1016/j.acra.2023.09.006] [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: 07/23/2023] [Revised: 08/19/2023] [Accepted: 09/03/2023] [Indexed: 10/02/2023]
Abstract
RATIONALE AND OBJECTIVES The purpose of this study is to conduct a comparison between the newly introduced Angio PLanewave UltraSensitive (AngioPLUS) method and the power Doppler ultrasound (PDUS) technique, evaluating the efficacy of these two methods in detecting synovial blood flow in wrist and finger joints of rheumatoid arthritis (RA) patients. Furthermore, the study aimed to investigate the potential associations between the observed blood flow patterns and various symptoms and indicators associated with RA. MATERIALS AND METHODS A cohort of 101 patients diagnosed with RA was included and subsequently categorized into two groups: 20 male participants (19.80%) and 81 female participants (80.20%). Their grayscale ultrasound, PDUS, and AngioPLUS were utilized to acquire data, and subsequent scoring was conducted. Serological tests of the patients were also performed, and DAS28 scores were calculated. The McNemar and Wilcoxon tests were used to compare the blood flow display rate and grading of PDUS as well as AngioPLUS, respectively. RESULTS AngioPLUS blood was significantly improved compared to PDUS. In all joints, the proportion of slight and significant improvement in wrist joints was the highest (14.11% and 1.98%, respectively). AngioPLUS was moderately correlated with C-reactive Protein (CRP), Disease Activity Score that includes 28-joint counts, and swollen joint counts and weakly correlated with platelet, hemoglobin, tender joint counts, and CRP before and after treatment. CONCLUSION Compared to PDUS, AngioPLUS has a better auxiliary diagnostic role in evaluating disease activity and can provide a reference to improve the management of RA further.
Collapse
Affiliation(s)
- Wuyue Zhang
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, NanGang District, Harbin, 150086, China (W.Z., J.S., J.L., Y.W., W.L., W.X., X.W.)
| | - Jiawei Sun
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, NanGang District, Harbin, 150086, China (W.Z., J.S., J.L., Y.W., W.L., W.X., X.W.)
| | - Jinyao Li
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, NanGang District, Harbin, 150086, China (W.Z., J.S., J.L., Y.W., W.L., W.X., X.W.)
| | - Yanyan Wang
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, NanGang District, Harbin, 150086, China (W.Z., J.S., J.L., Y.W., W.L., W.X., X.W.)
| | - Weiyao Liu
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, NanGang District, Harbin, 150086, China (W.Z., J.S., J.L., Y.W., W.L., W.X., X.W.)
| | - Weili Xue
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, NanGang District, Harbin, 150086, China (W.Z., J.S., J.L., Y.W., W.L., W.X., X.W.)
| | - Yan Yuan
- Ultrasound Department, Heilongjiang Red Cross Sengong General Hospital, Harbin, China (Y.Y.)
| | - Xiaolei Wang
- In-Patient Ultrasound Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, NanGang District, Harbin, 150086, China (W.Z., J.S., J.L., Y.W., W.L., W.X., X.W.).
| |
Collapse
|
2
|
Duan Z, Jin C, Deng Y, Liu J, Gu C, Wang J, Cai X, Li S, Zhou Y. Exploring the chondroprotective effect of Chaenomeles speciosa on Glucose-6-Phosphate Isomerase model mice using an integrated approach of network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116553. [PMID: 37178981 DOI: 10.1016/j.jep.2023.116553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) has been used in China for a long time and is gradually gaining more and more recognition worldwide. Chaenomeles speciosa (CSP) (Chinese Pinyin: mugua) is a medicinal and food herb that has long been used as a folk medicine for rheumatic diseases, yet its bioactive components and therapeutic mechanisms are not clear. AIM OF THE STUDY Exploring anti-inflammatory and chondroprotective effects of CSP on rheumatoid arthritis (RA) and its possible targets of action. MATERIALS AND METHODS In this study, we performed an integrated approach of network pharmacology, molecular docking and experimental studies to explore the potential mechanism of action of CSP in the treatment of cartilage damage in RA. RESULTS Studies have shown that Quercetin, ent-Epicatechin and Mairin may be the main active compounds of CSP in the treatment of RA, while AKT1, VEGFA, IL-1β, IL-6, MMP9 etc. are considered as core target proteins to which the main active compounds in CSP bind, as further confirmed by molecular docking. In addition, the potential molecular mechanism of CSP for the treatment of cartilage damage in RA predicted by network pharmacology analysis was validated by in vivo experiments. CSP was found to downregulate the expression of AKT1, VEGFA, IL-1β, IL-6, MMP9, ICAM1, VCAM1, MMP3, MMP13 and TNF-α and increase the expression of COL-2 in the joint tissue of Glucose-6-Phosphate Isomerase (G6PI) model mice. Thus CSP contributes to the treatment of rheumatoid arthritis cartilage destruction. CONCLUSION This study showed that CSP has multi-component, multi-target and multi-pathway characteristics in treating cartilage damage in RA, which can achieve the effect of treating RA by inhibiting the expression of inflammatory factors, reducing neovascularization and alleviating the damage to cartilage caused by the diffusion of synovial vascular opacities, and reducing the degradation of cartilage by MMPs to play a protective role in RA cartilage damage. In conclusion, this study indicates that CSP is a candidate Chinese medicine for further research in treating cartilage damage in RA.
Collapse
Affiliation(s)
- Zhihao Duan
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Can Jin
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Ying Deng
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Jinlang Liu
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China
| | - Chengyi Gu
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China
| | - Jie Wang
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China
| | - Xiangquan Cai
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China
| | - Shigang Li
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China.
| | - You Zhou
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443001, Hubei, China.
| |
Collapse
|
3
|
Comprehensive Bioinformatics Analysis Reveals Hub Genes and Inflammation State of Rheumatoid Arthritis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6943103. [PMID: 32802866 PMCID: PMC7424395 DOI: 10.1155/2020/6943103] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/16/2020] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by erosive arthritis, which has not been thoroughly cured yet, and standardized treatment is helpful for alleviating clinical symptoms. Here, various bioinformatics analysis tools were comprehensively utilized, aiming to identify critical biomarkers and possible pathogenesis of RA. Three gene expression datasets profiled by microarray were obtained from GEO database. Dataset GSE55235 and GSE55457 were merged for subsequent analyses. We identified differentially expressed genes (DEGs) in RStudio with limma package, performing functional enrichment analysis based on GSEA software and clusterProfiler package. Next, protein-protein interaction (PPI) network was set up through STRING database and Cytoscape. Moreover, CIBERSORT website was used to assess the inflammatory state of RA. Finally, we validated the candidate hub genes with dataset GSE77298. As a result, we identified 106 DEGs (72 upregulated and 34 downregulated genes). Through GO, KEGG, and GSEA analysis, we found that DEGs were mainly involved in immune response and inflammatory signaling pathway. With the help of Cytoscape software and MCODE plug-in, the most prominent subnetwork was screened out, containing 14 genes and 45 edges. For ROC curve analysis, eight genes with AUC >0.80 were considered as hub genes of RA. In conclusion, compared with healthy controls, the DEGs and their closely related biological functions were analyzed, and we held that chemokines and immune cells infiltration promote the progression of rheumatoid arthritis. Targeting the eight biomarkers we identified may be useful for the diagnosis and treatment of rheumatoid arthritis.
Collapse
|
4
|
Zeng W, Zhu JF, Liu JY, Li YL, Dong X, Huang H, Shan L. miR-133b inhibits cell proliferation, migration and invasion of esophageal squamous cell carcinoma by targeting EGFR. Biomed Pharmacother 2018; 111:476-484. [PMID: 30594787 DOI: 10.1016/j.biopha.2018.12.057] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is an aggressive tumor entity characterized by early metastasis and late diagnosis. MicroRNA-133b (miR-133b) has been considered as a tumor suppressor in many human cancers by regulating epidermal growth factor receptor (EGFR). However, the specific effects of miR-133b and EGFR on ESCC remain unclear. METHODS qRT-PCR and western blotting were applied for measuring expression of mRNA and protein. Flow cytometry was used for detecting cell cycle and apoptosis. Cell proliferation, migration and invasion were detected by colony formation and transwell assays. Luciferase reporter assay was used to confirm the interaction between miR-133b and EGFR. RESULTS Low expression of miR-133b and high expression of EGFR were identified in ESCC cells and tissues. Overexpression of miR-133b or knockdown of EGFR suppressed the cell proliferation, migration, and invasion of ESCC cells, and raised the percentage of G1 phase cells. The apoptosis of ESCC cells were promoted by increasing miR-133b and decreasing EGFR expression. Luciferase reporter assay confirmed EGFR as the target of miR-133b in ESCC cells. Overexpression of miR-133b significantly decreased the phosphorylation of PI3K, ERK and AKT by directly down-regulating EGFR. Higher expression of E-cadherin and CK-18 and lower expression of Vimentin and N-cadherin were observed after the transfection of miR-133b mimics or shEGFR. CONCLUSION Overexpression of miR-133b could suppress proliferation, migration and invasion of ESCC cells by inhibiting MAPK/ERK and PI3K/AKT signaling pathways through targeting EGFR, indicating that miR-133b might be a potential therapeutic target for the treatment of ESCC.
Collapse
Affiliation(s)
- Wei Zeng
- First Department of Lung Cancer Chemotherapy, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830011, PR China; Department of Hematology and Oncology, Shenzhen University General Hospital, Shenzhen 518055, PR China
| | - Jin-Feng Zhu
- Department of Gastrointestinal Surgery, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830011, PR China
| | - Jun-Yuan Liu
- First Department of Lung Cancer Chemotherapy, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830011, PR China
| | - Ying-Long Li
- First Department of Lung Cancer Chemotherapy, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830011, PR China
| | - Xiang Dong
- Institute of Cancer Prevention and Treatment, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830011, PR China
| | - He Huang
- Department of Histology and Embryology, Xinjiang Medical University, Urumqi 830011, PR China; Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha 410013, PR China.
| | - Li Shan
- First Department of Lung Cancer Chemotherapy, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi 830011, PR China.
| |
Collapse
|
5
|
Li T, Kang G, Wang T, Huang H. Tumor angiogenesis and anti-angiogenic gene therapy for cancer. Oncol Lett 2018; 16:687-702. [PMID: 29963134 PMCID: PMC6019900 DOI: 10.3892/ol.2018.8733] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 07/11/2017] [Indexed: 12/22/2022] Open
Abstract
When Folkman first suggested a theory about the association between angiogenesis and tumor growth in 1971, the hypothesis of targeting angiogenesis to treat cancer was formed. Since then, various studies conducted across the world have additionally confirmed the theory of Folkman, and numerous efforts have been made to explore the possibilities of curing cancer by targeting angiogenesis. Among them, anti-angiogenic gene therapy has received attention due to its apparent advantages. Although specific problems remain prior to cancer being fully curable using anti-angiogenic gene therapy, several methods have been explored, and progress has been made in pre-clinical and clinical settings over previous decades. The present review aimed to provide up-to-date information concerning tumor angiogenesis and gene delivery systems in anti-angiogenic gene therapy, with a focus on recent developments in the study and application of the most commonly studied and newly identified anti-angiogenic candidates for anti-angiogenesis gene therapy, including interleukin-12, angiostatin, endostatin, tumstatin, anti-angiogenic metargidin peptide and endoglin silencing.
Collapse
Affiliation(s)
- Tinglu Li
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P.R. China
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, P.R. China
- Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, P.R. China
| | - Guangbo Kang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P.R. China
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, P.R. China
- Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, P.R. China
| | - Tingyue Wang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P.R. China
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, P.R. China
- Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, P.R. China
| | - He Huang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P.R. China
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, P.R. China
- Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, P.R. China
| |
Collapse
|
6
|
Jia W, Wu W, Yang D, Xiao C, Huang M, Long F, Su Z, Qin M, Liu X, Zhu YZ. GATA4 regulates angiogenesis and persistence of inflammation in rheumatoid arthritis. Cell Death Dis 2018; 9:503. [PMID: 29717129 PMCID: PMC5931571 DOI: 10.1038/s41419-018-0570-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/26/2018] [Accepted: 03/29/2018] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by abnormal inflammation, angiogenesis, and cartilage destruction. In RA, neoangiogenesis is an early and crucial event to promote the formation of pannus, causing further inflammatory cell infiltration. The transcription factor GATA4 is a critical regulator of cardiac differentiation-specific gene expression. We find that a higher level of GATA4 exists in synovium of rheumatoid arthritis (RA) patients, but the function of GATA4 in RA remains unclear. In the present study, IL-1β induces inflammation in fibroblast-like synoviocytes (FLS) MH7A, which is accompanied with the increased expression of GATA4 and VEGF production. Through application of GATA4 loss-of-function assays, we confirm the requirement of GATA4 expression for inflammation induced by IL-1β in FLS. In addition, we demonstrate for the first time that GATA4 plays key roles in regulating VEGF secretion from RA FLS to promote cellular proliferation, induce cell migration, and angiogenic tube formation of endothelial cells. GATA4 induces the angiogenic factors VEGFA and VEGFC, by directly binding to the promoter and enhancing transcription. The knockdown of GATA4 attenuates the development of collagen-induced arthritis (CIA) and prevents RA-augmented angiogenesis in vivo, which are accompanied with decreased VEGF level. These results reveal a previously unrecognized function for GATA4 as a regulator of RA angiogenesis and we provide experimental data validating the therapeutic target of GATA4 in RA mice.
Collapse
Affiliation(s)
- Wanwan Jia
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| | - Weijun Wu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Di Yang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Chenxi Xiao
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Mengwei Huang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Fen Long
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhenghua Su
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Ming Qin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xinhua Liu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Yi Zhun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China. .,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
7
|
Al-Soudi A, Kaaij MH, Tas SW. Endothelial cells: From innocent bystanders to active participants in immune responses. Autoimmun Rev 2017; 16:951-962. [PMID: 28698091 DOI: 10.1016/j.autrev.2017.07.008] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 06/05/2017] [Indexed: 02/07/2023]
Abstract
The endothelium is crucially important for the delivery of oxygen and nutrients throughout the body under homeostatic conditions. However, it also contributes to pathology, including the initiation and perpetuation of inflammation. Understanding the function of endothelial cells (ECs) in inflammatory diseases and molecular mechanisms involved may lead to novel approaches to dampen inflammation and restore homeostasis. In this article, we discuss the various functions of ECs in inflammation with a focus on pathological angiogenesis, attraction of immune cells, antigen presentation, immunoregulatory properties and endothelial-to-mesenchymal transition (EndMT). We also review the current literature on approaches to target these processes in ECs to modulate immune responses and advance anti-inflammatory therapies.
Collapse
Affiliation(s)
- A Al-Soudi
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology & Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - M H Kaaij
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology & Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - S W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology & Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
8
|
Antiangiogenic Effects of Nerve Growth Factor Loop 4 Monomeric Dipeptide Mimetic. Bull Exp Biol Med 2017; 163:49-53. [PMID: 28580522 DOI: 10.1007/s10517-017-3735-9] [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: 12/28/2015] [Indexed: 10/19/2022]
Abstract
The effects of GK-1, a monomeric dipeptide mimetic of nerve growth factor (NGF) loop 4, on angiogenesis were studied in vitro and in vivo. Experiments on human umbilical vein endothelial cells HUVEC showed that the test compound did not affect tubulogenesis (initial stage of angiogenesis) and prevented realization of the angiogenic effect of NGF and its dimeric dipeptide mimetic GK-2. Experiments on rat hind limb ischemia model demonstrated that GK-1 (1 mg/kg/day intraperitoneally over 14 days) significantly reduced the density of the capillary network in ischemic tissue and increased the number and area of Zenker necrosis in comparison with the control. These data suggest that GK-1 exhibits a pronounced antiangiogenic activity.
Collapse
|
9
|
Leblond A, Allanore Y, Avouac J. Targeting synovial neoangiogenesis in rheumatoid arthritis. Autoimmun Rev 2017; 16:594-601. [PMID: 28414154 DOI: 10.1016/j.autrev.2017.04.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/03/2017] [Indexed: 12/18/2022]
Abstract
In Rheumatoid arthritis (RA), neoangiogenesis is an early and crucial event to promote the development of the hyperplasic proliferative pathologic synovium. Endothelial cells are critical for the formation of new blood vessels since they highly contribute to angiogenesis and vasculogenesis. Current therapies in RA target the inflammatory consequences of autoimmune activation and despite major improvements these last years still refractory patients or incomplete responders may be seen raising the point of the need to identify complementary additive and innovative therapies. This review resumes the mechanisms of synovial neoangiogenesis in RA, including recent insights on the implication of vasculogenesis, and the regulation of synovial neoangiogenesis by angiogenic and inflammatory mediators. In line with the recent development of vascular-targeted therapies used in cancer and beyond, we also discuss possible therapeutic implications in RA, in particular the combination of targeted immunotherapies with anti-angiogenic molecules.
Collapse
Affiliation(s)
- Agathe Leblond
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France
| | - Yannick Allanore
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Service de Rhumatologie A, Hôpital Cochin, Paris, France
| | - Jérôme Avouac
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Service de Rhumatologie A, Hôpital Cochin, Paris, France.
| |
Collapse
|
10
|
Imberti C, Terry SYA, Cullinane C, Clarke F, Cornish GH, Ramakrishnan NK, Roselt P, Cope AP, Hicks RJ, Blower PJ, Ma MT. Enhancing PET Signal at Target Tissue in Vivo: Dendritic and Multimeric Tris(hydroxypyridinone) Conjugates for Molecular Imaging of α vβ 3 Integrin Expression with Gallium-68. Bioconjug Chem 2017; 28:481-495. [PMID: 27966893 PMCID: PMC5314429 DOI: 10.1021/acs.bioconjchem.6b00621] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 11/23/2016] [Indexed: 12/12/2022]
Abstract
Tris(hydroxypyridinone) chelators conjugated to peptides can rapidly complex the positron-emitting isotope gallium-68 (68Ga) under mild conditions, and the resulting radiotracers can delineate peptide receptor expression at sites of diseased tissue in vivo. We have synthesized a dendritic bifunctional chelator containing nine 1,6-dimethyl-3-hydroxypyridin-4-one groups (SCN-HP9) that can coordinate up to three Ga3+ ions. This derivative has been conjugated to a trimeric peptide (RGD3) containing three peptide groups that target the αvβ3 integrin receptor. The resulting dendritic compound, HP9-RGD3, can be radiolabeled in 97% radiochemical yield at a 3-fold higher specific activity than its homologues HP3-RGD and HP3-RGD3 that contain only a single metal binding site. PET scanning and biodistribution studies show that [68Ga(HP9-RGD3)] demonstrates higher receptor-mediated tumor uptake in animals bearing U87MG tumors that overexpress αvβ3 integrin than [68Ga(HP3-RGD)] and [68Ga(HP3-RGD3)]. However, concomitant nontarget organ retention of [68Ga(HP9-RGD3)] results in low tumor to nontarget organ contrast in PET images. On the other hand, the trimeric peptide homologue containing a single tris(hydroxypyridinone) chelator, [68Ga(HP3-RGD3)], clears nontarget organs and exhibits receptor-mediated uptake in mice bearing tumors and in mice with induced rheumatoid arthritis. PET imaging with [68Ga(HP3-RGD3)] enables clear delineation of αvβ3 integrin receptor expression in vivo.
Collapse
Affiliation(s)
- Cinzia Imberti
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Samantha Y. A. Terry
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Carleen Cullinane
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Fiona Clarke
- King’s College
London, Academic Department of Rheumatology,
Centre for Molecular and Cellular Biology of Inflammation, Faculty
of Life Sciences and Medicine, London SE1 1UL, United Kingdom
| | - Georgina H. Cornish
- King’s College
London, Academic Department of Rheumatology,
Centre for Molecular and Cellular Biology of Inflammation, Faculty
of Life Sciences and Medicine, London SE1 1UL, United Kingdom
| | - Nisha K. Ramakrishnan
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Peter Roselt
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Andrew P. Cope
- King’s College
London, Academic Department of Rheumatology,
Centre for Molecular and Cellular Biology of Inflammation, Faculty
of Life Sciences and Medicine, London SE1 1UL, United Kingdom
| | - Rodney J. Hicks
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Philip J. Blower
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| | - Michelle T. Ma
- King’s College
London, Division of Imaging
Sciences and Biomedical Engineering, Fourth
Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
11
|
Maracle CX, Kucharzewska P, Helder B, van der Horst C, Correa de Sampaio P, Noort AR, van Zoest K, Griffioen AW, Olsson H, Tas SW. Targeting non-canonical nuclear factor-κB signalling attenuates neovascularization in a novel 3D model of rheumatoid arthritis synovial angiogenesis. Rheumatology (Oxford) 2017; 56:294-302. [PMID: 27864565 DOI: 10.1093/rheumatology/kew393] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 10/03/2016] [Indexed: 12/16/2023] Open
Abstract
OBJECTIVE Angiogenesis is crucial in RA disease progression. Lymphotoxin β receptor (LTβR)-induced activation of the non-canonical nuclear factor-κB (NF-κB) pathway via NF-κB-inducing kinase (NIK) has been implicated in this process. Consequently, inhibition of this pathway may hold therapeutic potential in RA. We describe a novel three-dimensional (3D) model of synovial angiogenesis incorporating endothelial cells (ECs), RA fibroblast-like synoviocytes (RAFLSs) and RA synovial fluid (RASF) to further investigate the contributions of NF-κB in this process. METHODS Spheroids consisting of RAFLSs and ECs were stimulated with RASF, the LTβR ligands LTβ and LIGHT, or growth factor bFGF and VEGF, followed by quantification of EC sprouting using confocal microscopy and digital image analysis. Next, the effects of anginex, NIK-targeting siRNA (siNIK), LTβR-Ig fusion protein (baminercept) and a novel pharmacological NIK inhibitor were investigated. RESULTS RASF significantly promoted sprout formation, which was blocked by the established angiogenesis inhibitor anginex (P < 0.05). LTβ and LIGHT induced significant sprouting (P < 0.05), as did bFGF/VEGF (P < 0.01). siNIK pre-treatment of ECs led to reductions in LTβR-induced vessel formation (P < 0.05). LTβR-Ig not only blocked LTβ- or LIGHT-induced sprouting, but also RASF-induced sprouting (P < 0.05). The NIK inhibitor blocked angiogenesis induced by LTβ, LIGHT, growth factors (P < 0.05) and RASF (P < 0.01). CONCLUSION We present a novel 3D model of synovial angiogenesis incorporating RAFLSs, ECs and RASF that mimics the in vivo situation. Using this system, we demonstrate that non-canonical NF-κB signalling promotes neovascularization and show that this model is useful for dissecting relative contributions of signalling pathways in specific cell types to angiogenic responses and for testing pharmacological inhibitors of angiogenesis.
Collapse
Affiliation(s)
- Chrissta X Maracle
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Boy Helder
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Corine van der Horst
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Ae-Ri Noort
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katinka van Zoest
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Sander W Tas
- Amsterdam Rheumatology and immunology Center
- Department of Clinical Immunology & Rheumatology
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Liu CT, Bi KW, Huang CC, Wu HT, Ho HY, S Pang JH, Huang ST. Davallia bilabiata exhibits anti-angiogenic effect with modified MMP-2/TIMP-2 secretion and inhibited VEGF ligand/receptors expression in vascular endothelial cells. JOURNAL OF ETHNOPHARMACOLOGY 2017; 196:213-224. [PMID: 27993633 DOI: 10.1016/j.jep.2016.12.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 12/01/2016] [Accepted: 12/10/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Davallia bilabiata Hosokawa (D. bilabiata), also called GuSuiBu, is popularly used as a substitute for Drynaria fortunei J. Sm for rheumatoid and degenerative arthritis in traditional Chinese medicine. Little is known about the underlying mechanisms of anti-angiogenesis responsible for arthritis in D. bilabiata which needs to be elucidated. AIM OF THE STUDY The present study is intended to investigate the anti-angiogenic effect of D. bilabiata associated with the modulation of matrix metalloproteinases (MMPs) and down regulation of vascular endothelial growth factor (VEGF) ligand/receptors both in vivo and in vitro. MATERIALS AND METHODS We investigated the potential anti-angiogenic effect of D. bilabiata by the in vivo neovascularization of chick chorioallantoic membranes (CAM) assay, and the in vitro migration and matrix-induced tube formation assay using human umbilical vascular endothelial cells (HUVECs). The expressions of MMP-2, TIMP-2, RECK and VEGF/VEGFR were analyzed by real-time RT-PCR or Western blot method. RESULTS One major compound from water extract of D. bilabiata was identified as Epicatechin 3-O-β-D-allopyranoside. D. bilabiata was confirmed to inhibit in vivo angiogenesis by CAM assay. D. bilabiata also exhibited in vitro anti-angiogenic and anti-regrowth effects as demonstrated by tube formation assay, transwell migration assay and wound healing assay. The mRNA expressions of MMP-2, and MMP-14 were decreased. On the contrary, tissue inhibitor of metalloproteinase-2 (TIMP-2), reversion-inducing cysteine-rich protein with kazal motifs (RECK) were increased by D. bilabiata. The extracellular MMP-2 activity was found to be reduced both in vitro and in vivo by D. bilabiata as determined by gelatin zymography. Results from western blot analysis and ELISA further demonstrated the decrease of MMP-2 and increase of TIMP-2 secretion after D. bilabiata treatment. The gene expressions of VEGF-A, -B, -C, -D and VEGFR-1, -2, -3 were all inhibited by D. bilabiata. CONCLUSION We concluded that the anti-angiogenic effect of D. bilabiata was associated with the decreased MMP-2 activity mediated by the upregulation of TIMP-2 and RECK, and the suppression of VEGF/VEGFRs expression.
Collapse
Affiliation(s)
- Chun-Ting Liu
- Department of Chinese Medicine and Mitochondrial Research Unit, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kuo-Wei Bi
- Department of Chinese Medicine and Mitochondrial Research Unit, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chao-Chun Huang
- Division of General Surgery, Ministry of Health and Welfare Pingtung Hospital, Pingtung, Taiwan
| | - Hsiao-Ting Wu
- Department of Chinese Medicine and Mitochondrial Research Unit, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hui-Ya Ho
- Jen Li Biotech Company Ltd., Taiping District, Taichung 41143, Taiwan
| | - Jong-Hwei S Pang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Tao-Yuan,Taiwan; Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Sheng-Teng Huang
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan; School of Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
13
|
Glucose-6-Phosphate Isomerase (G6PI) Mediates Hypoxia-Induced Angiogenesis in Rheumatoid Arthritis. Sci Rep 2017; 7:40274. [PMID: 28067317 PMCID: PMC5220294 DOI: 10.1038/srep40274] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/05/2016] [Indexed: 12/22/2022] Open
Abstract
The higher level of Glucose-6-phosphate isomerase (G6PI) has been found in both synovial tissue and synovial fluid of rheumatoid arthritis (RA) patients, while the function of G6PI in RA remains unclear. Herein we found the enrichment of G6PI in microvascular endothelial cells of synovial tissue in RA patients, where a 3% O2 hypoxia environment has been identified. In order to determine the correlation between the high G6PI level and the low oxygen concentration in RA, a hypoxia condition (~3% O2) in vitro was applied to mimic the RA environment in vivo. Hypoxia promoted cellular proliferation of rheumatoid arthritis synovial fibroblasts (RASFs), and induced cell migration and angiogenic tube formation of human dermal microvascular endothelial cells (HDMECs), which were accompanied with the increased expression of G6PI and HIF-1α. Through application of G6PI loss-of-function assays, we confirmed the requirement of G6PI expression for those hypoxia-induced phenotype in RA. In addition, we demonstrated for the first time that G6PI plays key roles in regulating VEGF secretion from RASFs to regulate the hypoxia-induced angiogenesis in RA. Taken together, we demonstrated a novel pathway regulating hypoxia-induced angiogenesis in RA mediated by G6PI.
Collapse
|
14
|
Pascart T, Herbaux C, Lemaire A, Soncin F, Hachulla E, Hatron PY, Terriou L. Coexistence of rheumatoid arthritis and TEMPI syndrome: New insight in microangiogenic-related diseases. Joint Bone Spine 2016; 83:587-8. [DOI: 10.1016/j.jbspin.2015.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/05/2015] [Indexed: 11/16/2022]
|
15
|
Bhattaram P, Chandrasekharan U. The joint synovium: A critical determinant of articular cartilage fate in inflammatory joint diseases. Semin Cell Dev Biol 2016; 62:86-93. [PMID: 27212252 DOI: 10.1016/j.semcdb.2016.05.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 11/26/2022]
Abstract
The synovium constitutes the envelope of articular joints and is a critical provider of synovial fluid components and articular cartilage nutrients. Its inflammation is a predominant feature and cause of joint degeneration in diseases as diverse as rheumatoid, psoriatic, juvenile and idiopathic arthritis, and lupus, gout and lyme disease. These inflammatory joint diseases (IJDs) are due to a wide variety of genetic, epigenetic and environmental factors that trigger, promote, and perpetuate joint destabilization. In spite of this variety of causes, IJDs share main pathological features, namely inflammation of the joint synovium (synovitis) and progressive degeneration of articular cartilage. In addition to being a driving force behind the destruction of articular cartilage in IJD, synovitis is also increasingly being recognized as a significant contributor of articular cartilage degeneration in osteoarthritis, a disease primarily due to aging- or trauma-related wear and tear of cartilage surfaces. In view of this important role of the synovium in determining the fate of articular cartilage, this review focuses on its underlying mechanisms in the pathology of IJD. We address the roles of synovial fibroblasts, macrophages and endothelial cells in the maintenance of joint health and in the destruction of articular cartilage integrity during IJD. Molecular mechanisms that have been recently shown to govern the pathological activities of the resident synovial cells are highlighted. Finally, advantages and disadvantages of targeting these new molecular mechanisms for preventing cartilage degeneration due to chronic inflammation are also discussed.
Collapse
Affiliation(s)
- Pallavi Bhattaram
- Department of Cellular & Molecular Medicine, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, NC-10, Cleveland, OH, 44195, USA.
| | - Unnikrishnan Chandrasekharan
- Department of Cellular & Molecular Medicine, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, NC-10, Cleveland, OH, 44195, USA.
| |
Collapse
|
16
|
Olfert IM. Physiological Capillary Regression is not Dependent on Reducing VEGF Expression. Microcirculation 2016; 23:146-56. [PMID: 26660949 PMCID: PMC4744091 DOI: 10.1111/micc.12263] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/03/2015] [Indexed: 01/04/2023]
Abstract
Investigations into physiologically controlled capillary regression report the provocative finding that microvessel regression occurs in the face of persistent elevation of skeletal muscle VEGF expression. TSP-1, a negative angiogenic regulator, is increasingly being observed to temporally correlate with capillary regression, suggesting that increased TSP-1 (and not reduction in VEGF per se) is needed to initiate, and likely regulate, capillary regression. Based on evidence being gleaned from physiologically mediated regression of capillaries, it needs to be recognized that capillary regression (and perhaps capillary rarefaction with disease) is not simply the reversal of factors used to stimulate angiogenesis. Rather, the conceptual understanding that angiogenesis and capillary regression each have specific and unique requirements that are biologically constrained to opposite sides of the balance between positive and negative angioregulatory factors may shed light on why anti-VEGF therapies have not lived up to the promise in reversing angiogenesis and providing the cure that many had hoped toward fighting cancer. Emerging evidence from physiological controlled angiogenesis suggest that cases involving excessive or uncontrolled capillary expansion may be best treated by therapies designed to increase expression of negative angiogenic regulators, whereas those involving capillary rarefaction may benefit from inhibiting negative regulators (like TSP-1).
Collapse
Affiliation(s)
- I Mark Olfert
- Division of Exercise Physiology, Center for Cardiovascular and Respiratory Sciences, Mary Babb Randolph Cancer Center, West Virginia Clinical and Translational Science Institute, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| |
Collapse
|
17
|
Tas SW, Maracle CX, Balogh E, Szekanecz Z. Targeting of proangiogenic signalling pathways in chronic inflammation. Nat Rev Rheumatol 2015; 12:111-22. [PMID: 26633288 DOI: 10.1038/nrrheum.2015.164] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Angiogenesis is de novo capillary outgrowth from pre-existing blood vessels. This process not only is crucial for normal development, but also has an important role in supplying oxygen and nutrients to inflamed tissues, as well as in facilitating the migration of inflammatory cells to the synovium in rheumatoid arthritis, spondyloarthritis and other systemic autoimmune diseases. Neovascularization is dependent on the balance of proangiogenic and antiangiogenic mediators, including growth factors, cytokines, chemokines, cell adhesion molecules and matrix metalloproteinases. This Review describes the various intracellular signalling pathways that govern these angiogenic processes and discusses potential approaches to interfere with pathological angiogenesis, and thereby ameliorate inflammatory disease, by targeting these pathways.
Collapse
Affiliation(s)
- Sander W Tas
- Amsterdam Rheumatology &Immunology Centre, Department of Experimental Immunology, Academic Medical Centre and University of Amsterdam, EULAR &FOCIS (Federation of Clinical Immunology Societies) Centre of Excellence, Meibergdreef 9, F4-105, 1105 AZ Amsterdam, Netherlands
| | - Chrissta X Maracle
- Amsterdam Rheumatology &Immunology Centre, Department of Experimental Immunology, Academic Medical Centre and University of Amsterdam, EULAR &FOCIS (Federation of Clinical Immunology Societies) Centre of Excellence, Meibergdreef 9, F4-105, 1105 AZ Amsterdam, Netherlands
| | - Emese Balogh
- Department of Rheumatology, Institute of Medicine, University of Debrecen, Faculty of Medicine, Nagyerdei Str. 98, Debrecen 4032, Hungary
| | - Zoltán Szekanecz
- Department of Rheumatology, Institute of Medicine, University of Debrecen, Faculty of Medicine, Nagyerdei Str. 98, Debrecen 4032, Hungary
| |
Collapse
|
18
|
Szekanecz Z, Koch AE. Successes and failures of chemokine-pathway targeting in rheumatoid arthritis. Nat Rev Rheumatol 2015; 12:5-13. [PMID: 26607389 DOI: 10.1038/nrrheum.2015.157] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chemokines and chemokine receptors are involved in leukocyte recruitment and angiogenesis underlying the pathogenesis of rheumatoid arthritis (RA) and other inflammatory rheumatic diseases. Numerous chemokines, along with both conventional and atypical cell-surface chemokine receptors, are found in inflamed synovia. Preclinical studies carried out in animal models of arthritis involving agents targeting chemokines and chemokine receptors have yielded promising results. However, most human trials of treatment of RA with antibodies and synthetic compounds targeting chemokine signalling have failed to show clinical improvements. Chemokines can have overlapping actions, and their activities can be altered by chemical modification or proteolytic degradation. Effective targeting of chemokine pathways must take acount of these properties, and can also require high levels of receptor occupancy by therapeutic agents to prevent signalling. CCR1 is a promising target for chemokine-receptor blockade.
Collapse
Affiliation(s)
- Zoltán Szekanecz
- Department of Rheumatology, Institute of Medicine, University of Debrecen Faculty of Medicine, Nagyerdei Str 98, Debrecen, H-4004, Hungary
| | - Alisa E Koch
- University of Michigan Health System, Department of Internal Medicine, Division of Rheumatology, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
| |
Collapse
|