1
|
Jeyaraman N, Migliorini F, Ramkumar V, Ramasubramanian S, Patro BP, Maffulli N, Jeyaraman M. Angioembolization in the management of joint pain: current concepts. ORTHOPADIE (HEIDELBERG, GERMANY) 2024; 53:688-697. [PMID: 39060549 DOI: 10.1007/s00132-024-04531-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/19/2024] [Indexed: 07/28/2024]
Abstract
Joint pain is a common complaint owing to its origin in inflammatory and degenerative joint diseases. Recent research has helped narrow down inadequate angiogenesis as one of the causes. Angioembolization has emerged as a treatment option for this condition when it is refractory to conservative treatment. This review describes angioembolization by elaborating on the mechanism, safety, efficacy, comparative analysis of treatment and the road ahead, in addition to summarizing the existing data on this treatment. The inferences from this review further consolidate transcatheter arterial embolization as one of the prime options for managing joint pain when it is refractory to conservative treatment and label it as one of the most exciting prospects. A limitation of this review is that most of the data were from open label case series or case reports.
Collapse
Affiliation(s)
- Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, 600077, Chennai, Tamil Nadu, India
| | - Filippo Migliorini
- Department of Orthopaedic and Trauma Surgery, Academic Hospital of Bolzano (SABES-ASDAA), Via Lorenz Bohler 5, 39100, Bolzano, Italy
- Department of Life Sciences, Health, and Health Professions, Link Campus University, 00165, Rome, Italy
| | - Varunkumar Ramkumar
- Department of Orthopaedics, Madurai Medical College and Hospital, 625020, Madurai, Tamil Nadu, India
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, 600002, Chennai, Tamil Nadu, India
| | - Bishnu Prasad Patro
- Department of Orthopaedics, All India Institute of Medical Sciences, 751019, Bhubaneswar, Odisha, India
| | - Nicola Maffulli
- Department of Trauma and Orthopaedic Surgery, Faculty of Medicine and Psychology, University La Sapienza, 00185, Roma, Italy.
- School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, ST4 7QB, Stoke on Trent, UK.
- Centre for Sports and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Mile End Hospital, Queen Mary University of London, E1 4DG, London, UK.
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, 600077, Chennai, Tamil Nadu, India
| |
Collapse
|
2
|
Gao C, Song XD, Chen FH, Wei GL, Guo CY. The protective effect of natural medicines in rheumatoid arthritis via inhibit angiogenesis. Front Pharmacol 2024; 15:1380098. [PMID: 38881875 PMCID: PMC11176484 DOI: 10.3389/fphar.2024.1380098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/10/2024] [Indexed: 06/18/2024] Open
Abstract
Rheumatoid arthritis is a chronic immunological disease leading to the progressive bone and joint destruction. Angiogenesis, accompanied by synovial hyperplasia and inflammation underlies joint destruction. Delaying or even blocking synovial angiogenesis has emerged as an important target of RA treatment. Natural medicines has a long history of treating RA, and numerous reports have suggested that natural medicines have a strong inhibitory activity on synovial angiogenesis, thereby improving the progression of RA. Natural medicines could regulate the following signaling pathways: HIF/VEGF/ANG, PI3K/Akt pathway, MAPKs pathway, NF-κB pathway, PPARγ pathway, JAK2/STAT3 pathway, etc., thereby inhibiting angiogenesis. Tripterygium wilfordii Hook. f. (TwHF), sinomenine, and total glucoside of Paeonia lactiflora Pall. Are currently the most representative of all natural products worthy of development and utilization. In this paper, the main factors affecting angiogenesis were discussed and different types of natural medicines that inhibit angiogenesis were systematically summarized. Their specific anti-angiogenesis mechanisms are also reviewed which aiming to provide new perspective and options for the management of RA by targeting angiogenesis.
Collapse
Affiliation(s)
- Chang Gao
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Xiao-Di Song
- Gannan Medical University, Jiangxi, Ganzhou, China
| | - Fang-Hui Chen
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Gui-Lin Wei
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| | - Chun-Yu Guo
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Jiangxi, Ganzhou, China
| |
Collapse
|
3
|
Yang X, Fu Q, Zhang W, An Q, Zhang Z, Li H, Chen X, Chen Z, Cheng Y, Chen S, Man C, Du L, Chen Q, Wang F. Overexpression of Pasteurella multocida OmpA induces transcriptional changes and its possible implications for the macrophage polarization. Microb Pathog 2023; 183:106212. [PMID: 37353176 DOI: 10.1016/j.micpath.2023.106212] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Pasteurella multocida (P. multocida) is a highly infectious, zoonotic pathogen. Outer membrane protein A (OmpA) is an important virulence component of the outer membrane of P. multocida. OmpA mediates bacterial biofilm formation, eukaryotic cell infection, and immunomodulation. It is unclear how OmpA affects the host immune response. We estimated the role of OmpA in the pathogenesis of P. multocida by investigating the effect of OmpA on the immune cell transcriptome. Changes in the transcriptome of rat alveolar macrophages (NR8383) upon overexpression of P. multocida OmpA were demonstrated. A model cell line for stable transcription of OmpA was constructed by infecting NR8383 cells with OmpA-expressing lentivirus. RNA was extracted from cells and sequenced on an Illumina HiSeq platform. Key gene analysis of genes in the RNA-seq dataset were performed using various bioinformatics methods, such as gene ontology enrichment analysis, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, Gene Set Enrichment Analysis, and Protein-Protein Interaction Analysis. Our findings revealed 1340 differentially expressed genes. Immune-related pathways that were significantly altered in rat alveolar macrophages under the effect of OmpA included focal adhesion, extracellular matrix and vascular endothelial growth factor signaling pathways, antigen processing and presentation, nucleotide oligomerization domain-like receptor and Toll-like receptor signaling pathways, and cytokine-cytokine receptor interaction. The key genes screened were Vegfa, Igf2r, Fabp5, P2rx1, C5ar1, Nedd4l, Gas6, Cxcl1, Pf4, Pdgfb, Thbs1, Col7a1, Vwf, Ccl9, and Arg1. Data of associated pathways and altered gene expression indicated that OmpA might cause the conversion of rat alveolar macrophages to M2-like. The related pathways and key genes can serve as a reference for OmpA of P. multitocida and host interaction mechanism studies.
Collapse
Affiliation(s)
- Xiaohong Yang
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Qiaoyu Fu
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Wencan Zhang
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Qi An
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Zhenxing Zhang
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Hong Li
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Xiangying Chen
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Zhen Chen
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Yiwen Cheng
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Si Chen
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Churiga Man
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China
| | - Li Du
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China.
| | - Qiaoling Chen
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China.
| | - Fengyang Wang
- Hainan Key Lab of Tropical Animal Reproduction, Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou, School of Animal Science and Technology, Hainan University, Haikou, Hainan, China.
| |
Collapse
|
4
|
Meng M, Yao J, Zhang Y, Sun H, Liu M. Potential Anti-Rheumatoid Arthritis Activities and Mechanisms of Ganoderma lucidum Polysaccharides. Molecules 2023; 28:molecules28062483. [PMID: 36985456 PMCID: PMC10052150 DOI: 10.3390/molecules28062483] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic and autoimmune disease characterized by inflammation, autoimmune dysfunction, and cartilage and bone destruction. In this review, we summarized the available reports on the protective effects of Ganoderma lucidum polysaccharides (GLP) on RA in terms of anti-inflammatory, immunomodulatory, anti-angiogenic and osteoprotective effects. Firstly, GLP inhibits RA synovial fibroblast (RASF) proliferation and migration, modulates pro- and anti-inflammatory cytokines and reduces synovial inflammation. Secondly, GLP regulates the proliferation and differentiation of antigen-presenting cells such as dendritic cells, inhibits phagocytosis by mononuclear macrophages and nature killer (NK) cells and regulates the ratio of M1, M2 and related inflammatory cytokines. In addition, GLP produced activities in balancing humoral and cellular immunity, such as regulating immunoglobulin production, modulating T and B lymphocyte proliferative responses and cytokine release, exhibiting immunomodulatory effects. Thirdly, GLP inhibits angiogenesis through the direct inhibition of vascular endothelial cell proliferation and induction of cell death and the indirect inhibition of vascular endothelial growth factor (VEGF) production in the cells. Finally, GLP can inhibit the production of matrix metalloproteinases and promote osteoblast formation, exerting protective effects on bone and articular cartilage. It is suggested that GLP may be a promising agent for the treatment of RA.
Collapse
Affiliation(s)
- Meng Meng
- Department of Orthopaedics, First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Jialin Yao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116041, China
| | - Yukun Zhang
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing 404120, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116041, China
| | - Mozhen Liu
- Department of Orthopaedics, First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| |
Collapse
|
5
|
PAR2 promotes tumor-associated angiogenesis in lung adenocarcinoma through activating EGFR pathway. Tissue Cell 2022; 79:101918. [DOI: 10.1016/j.tice.2022.101918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/23/2022]
|
6
|
Deng H, Liu H, Yang Z, Bao M, Lin X, Han J, Qu C. Progress of Selenium Deficiency in the Pathogenesis of Arthropathies and Selenium Supplement for Their Treatment. Biol Trace Elem Res 2022; 200:4238-4249. [PMID: 34779998 DOI: 10.1007/s12011-021-03022-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/08/2021] [Indexed: 11/26/2022]
Abstract
Selenium, an essential trace element for human health, exerts an indispensable effect in maintaining physiological homeostasis and functions in the body. Selenium deficiency is associated with arthropathies, such as Kashin-Beck disease, rheumatoid arthritis, osteoarthritis, and osteoporosis. Selenium deficiency mainly affects the normal physiological state of bone and cartilage through oxidative stress reaction and immune reaction. This review aims to explore the role of selenium deficiency and its mechanisms existed in the pathogenesis of arthropathies. Meanwhile, this review also summarized various experiments to highlight the crucial functions of selenium in maintaining the homeostasis of bone and cartilage.
Collapse
Affiliation(s)
- Huan Deng
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Haobiao Liu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Zhihao Yang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Miaoye Bao
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Xue Lin
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Jing Han
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China.
| | - Chengjuan Qu
- Department of Odontology, Umeå University, 90187, Umeå, Sweden
| |
Collapse
|
7
|
Hou R, Guo D, Fan M, Hou Y, Zhao J, Wu X. Screening and Analysis of Potential Critical Gene in Acute Myocardial Infarction Based on a miRNA-mRNA Regulatory Network. Int J Gen Med 2022; 15:2847-2860. [PMID: 35300139 PMCID: PMC8923688 DOI: 10.2147/ijgm.s354641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/03/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Ruirui Hou
- Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Dong Guo
- Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
- Correspondence: Dong Guo, Email
| | - Maoxia Fan
- Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Yawei Hou
- Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Jisen Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Xiaoqi Wu
- Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| |
Collapse
|
8
|
Michallek F, Ulas ST, Poddubnyy D, Proft F, Schneider U, Hermann KGA, Dewey M, Diekhoff T. Fractal analysis of perfusion imaging in synovitis: a novel imaging biomarker for grading inflammatory activity based on assessing angiogenesis. RMD Open 2022; 8:rmdopen-2021-002078. [PMID: 35149603 PMCID: PMC8845323 DOI: 10.1136/rmdopen-2021-002078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/17/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives The mutual and intertwined dependence of inflammation and angiogenesis in synovitis is widely acknowledged. However, no clinically established tool for objective and quantitative assessment of angiogenesis is routinely available. This study establishes fractal analysis as a novel method to quantitatively assess inflammatory activity based on angiogenesis in synovitis. Methods First, we established a pathophysiological framework for synovitis including fractal analysis of software perfusion phantoms, which allowed to derive explainability with a known and controllable reference standard for vascular structure. Second, we acquired MRI datasets of patients with suspected rheumatoid arthritis of the hand, and three imaging experts independently assessed synovitis analogue to Rheumatoid Arthritis MRI Scoring (RAMRIS) criteria. Finally, we performed fractal analysis of dynamic first-pass perfusion MRI in vivo to evaluate angiogenesis in relation to inflammatory activity with RAMRIS as reference standard. Results Fractal dimension (FD) achieved highly significant discriminability for different degrees of inflammatory activity (p<0.01) in software phantoms with known ground-truth of angiogenic structure. FD indicated increasingly chaotic perfusion patterns with increasing grades of inflammatory activity (Spearman’s ρ=0.94, p<0.001). In 36 clinical patients, fractal analysis quantitatively and objectively discriminated individual RAMRIS scores (p≤0.05). Area under the receiver-operating curve was 0.84 (95% CI 0.7 to 0.89) for fractal analysis when considering RAMRIS as ground-truth. Fractal analysis additionally identified angiogenesis in cases where RAMRIS underestimated inflammatory activity. Conclusions Based on angiogenesis and perfusion pathophysiology, fractal analysis non-invasively enables comprehensive, objective and quantitative characterisation of inflammatory angiogenesis with subjective and qualitative RAMRIS as reference standard. Further studies are required to establish the clinical value of fractal analysis for diagnosis, prognostication and therapy monitoring in inflammatory arthritis.
Collapse
Affiliation(s)
- Florian Michallek
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Sevtap Tugce Ulas
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Denis Poddubnyy
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Udo Schneider
- Department of Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Kay-Geert A Hermann
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Marc Dewey
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| | - Torsten Diekhoff
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Berlin, Germany
| |
Collapse
|
9
|
Co-treatment of Nimbolide augmented the anti-arthritic effects of methotrexate while protecting against organ toxicities. Life Sci 2022; 295:120372. [PMID: 35143824 DOI: 10.1016/j.lfs.2022.120372] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/20/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Prolonged exposure to the pharmacological doses of disease-modifying anti-rheumatic drugs (DMARDs) often results in major organ toxicities resulting in poor patient compliance. Methotrexate (MTX) is one of the commonly prescribed DMARDs for the treatment of arthritis, which results in vital organ dysfunction. To retain the anti-arthritic activity of MTX with the reduction in toxicities, combination therapies are warranted. Nimbolide (NMB) is a potent anticancer, anti-inflammatory and anti-fibrotic agent whose potential has been demonstrated in various pre-clinical models. Monoarthritis was developed with Complete Freund's Adjuvant in the knees of Wistar rats and treatment was given with either NMB (3 mg/kg/day) or MTX (2 mg/kg/week) alone or combination therapy (NMB + MTX). The anti-arthritic effects were evaluated by arthritic scoring, radiological imaging, synovial tissue proteins analysis, and histopathological staining. While hepato-renal toxicity was assessed in serum by evaluating the kidney and liver functional parameters, in tissues by oxidative-nitrosative stress markers, and pro-inflammatory cytokines levels. Histopathological analysis was performed to study the extent of tissue damage. Molecular studies like immunoblotting and immunohistochemistry were performed to understand the effect of combination therapy. We thereby report that monotherapy with either NMB or MTX exhibited significant anti-arthritic effects, while combination therapy resulted in augmented anti-arthritic effects with significant reduction in hepato-renal toxicity produced by MTX probably through anti-inflammatory and anti-oxidant effects. Therefore, our proposed combination of NMB and MTX may serve as a potential strategy for the effective management of arthritis.
Collapse
|
10
|
Gouveia VM, Rizzello L, Vidal B, Nunes C, Poma A, Lopez‐Vasquez C, Scarpa E, Brandner S, Oliveira A, Fonseca JE, Reis S, Battaglia G. Targeting Macrophages and Synoviocytes Intracellular Milieu to Augment Anti‐Inflammatory Drug Potency. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Virgínia M. Gouveia
- Department of Chemistry University College London London WC1H 0AJ UK
- Institute of Physics of Living Systems University College London London WC1H 0AJ UK
- SomaServe Ltd Babraham Research Campus Cambridge CB22 3AT UK
- LAQV REQUIMTE Department of Chemical Sciences Faculty of Pharmacy University of Porto Porto 4050‐313 Portugal
- Abel Salazar Biomedical Sciences Institute University of Porto Porto 4050‐313 Portugal
| | - Loris Rizzello
- Department of Chemistry University College London London WC1H 0AJ UK
- Institute for Bioengineering of Catalonia (IBEC) The Barcelona Institute of Science and Technology Barcelona 08028 Spain
- Department of Pharmaceutical Sciences University of Milan Milan 20133 Italy
- National Institute of Molecular Genetics (INGM) Milan 20122 Italy
| | - Bruno Vidal
- Rheumatology Research Unit Institute of Molecular Medicine – IMM João Lobo Antunes Faculty of Medicine University of Lisbon Lisbon 1649‐028 Portugal
| | - Claudia Nunes
- LAQV REQUIMTE Department of Chemical Sciences Faculty of Pharmacy University of Porto Porto 4050‐313 Portugal
| | - Alessandro Poma
- Department of Chemistry University College London London WC1H 0AJ UK
- Division of Biomaterials and Tissue Engineering Eastman Dental Institute Royal Free Hospital UCL Medical School London NW3 2PF UK
| | - Ciro Lopez‐Vasquez
- Department of Chemistry University College London London WC1H 0AJ UK
- Institute of Physics of Living Systems University College London London WC1H 0AJ UK
| | - Edoardo Scarpa
- Department of Chemistry University College London London WC1H 0AJ UK
- Department of Pharmaceutical Sciences University of Milan Milan 20133 Italy
- National Institute of Molecular Genetics (INGM) Milan 20122 Italy
| | - Sebastian Brandner
- Department of Neurodegenerative Disease Queen Square Institute of Neurology University College London London WC1N 3BG UK
| | - António Oliveira
- Abel Salazar Biomedical Sciences Institute University of Porto Porto 4050‐313 Portugal
| | - João E. Fonseca
- Rheumatology Research Unit Institute of Molecular Medicine – IMM João Lobo Antunes Faculty of Medicine University of Lisbon Lisbon 1649‐028 Portugal
- Serviço de Reumatologia Centro Hospitalar Universitário Lisboa Norte Centro Academico de Medicina de Lisboa Lisbon 1649‐028 Portugal
| | - Salette Reis
- LAQV REQUIMTE Department of Chemical Sciences Faculty of Pharmacy University of Porto Porto 4050‐313 Portugal
| | - Giuseppe Battaglia
- Department of Chemistry University College London London WC1H 0AJ UK
- Institute of Physics of Living Systems University College London London WC1H 0AJ UK
- Institute for Bioengineering of Catalonia (IBEC) The Barcelona Institute of Science and Technology Barcelona 08028 Spain
- Catalan Institution for Research and Advanced Studies (ICREA) Barcelona 08010 Spain
| |
Collapse
|
11
|
Gold Nanoparticles: Multifaceted Roles in the Management of Autoimmune Disorders. Biomolecules 2021; 11:biom11091289. [PMID: 34572503 PMCID: PMC8470500 DOI: 10.3390/biom11091289] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/12/2021] [Accepted: 08/24/2021] [Indexed: 12/29/2022] Open
Abstract
Gold nanoparticles (GNPs) have been recently applied for various diagnostic and therapeutic purposes. The unique properties of these nanoparticles (NPs), such as relative ease of synthesis in various sizes, shapes and charges, stability, high drug-loading capacity and relative availability for modification accompanied by non-cytotoxicity and biocompatibility, make them an ideal field of research in bio-nanotechnology. Moreover, their potential to alleviate various inflammatory factors, nitrite species, and reactive oxygen production and the capacity to deliver therapeutic agents has attracted attention for further studies in inflammatory and autoimmune disorders. Furthermore, the characteristics of GNPs and surface modification can modulate their toxicity, biodistribution, biocompatibility, and effects. This review discusses in vitro and in vivo effects of GNPs and their functionalized forms in managing various autoimmune disorders (Ads) such as rheumatoid arthritis, type 1 diabetes, and multiple sclerosis.
Collapse
|
12
|
Xin PL, Jie LF, Cheng Q, Bin DY, Dan CW. Pathogenesis and Function of Interleukin-35 in Rheumatoid Arthritis. Front Pharmacol 2021; 12:655114. [PMID: 34054534 PMCID: PMC8155723 DOI: 10.3389/fphar.2021.655114] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 01/06/2023] Open
Abstract
It is well known that RA (Rheumatoid arthritis) is an autoimmune disease characterized by multiple and symmetric arthropathy. The main pathological features of RA are synovial hyperplasia, angiogenesis, pannus formation, inflammatory cell infiltration, articular cartilage, bone destruction, and ultimately joint dysfunction, even deformity. IL-35 (Interleukin-35) is a new member of the IL-12 (Interleukin-12) family, which is an immunosuppressive and anti-inflammatory cytokine secreted mainly by Treg (T regulatory cells). There is evidence suggested that IL-35 can attenuate the progression of RA through influencing the immune and pathological process. It suggests that IL-35 played an important role in the pathogenesis of RA, and can be used as a potential target for the future treatment of RA. This review summarizes the recent advances of IL-35 in the pathological roles and the therapeutic potential roles in RA.
Collapse
Affiliation(s)
- Pan Lin Xin
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Li Fan Jie
- Department of Orthopedic, Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qian Cheng
- Research and Experimental Center of Anhui Medical University, Hefei, China
| | - Du Yi Bin
- Department of Orthopedic, Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cheng Wen Dan
- Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Haque M, Siegel RJ, Fox DA, Ahmed S. Interferon-stimulated GTPases in autoimmune and inflammatory diseases: promising role for the guanylate-binding protein (GBP) family. Rheumatology (Oxford) 2021; 60:494-506. [PMID: 33159795 DOI: 10.1093/rheumatology/keaa609] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/16/2020] [Accepted: 08/23/2020] [Indexed: 12/14/2022] Open
Abstract
Human IFNs are secreted cytokines shown to stimulate the expression of over one thousand genes. These IFN-inducible genes primarily encode four major protein families, known as IFN-stimulated GTPases (ISGs), namely myxovirus-resistance proteins, guanylate-binding proteins (GBPs), p47 immunity-related GTPases and very large inducible guanosine triphosphate hydrolases (GTPases). These families respond specifically to type I or II IFNs and are well reported in coordinating immunity against some well known as well as newly discovered viral, bacterial and parasitic infections. A growing body of evidence highlights the potential contributory and regulatory roles of ISGs in dysregulated inflammation and autoimmune diseases. Our focus was to draw attention to studies that demonstrate increased expression of ISGs in the serum and affected tissues of patients with RA, SS, lupus, IBD and psoriasis. In this review, we analysed emerging literature describing the potential roles of ISGs, particularly the GBP family, in the context of autoimmunity. We also highlighted the promise and implications for therapeutically targeting IFNs and GBPs in the treatment of rheumatic diseases.
Collapse
Affiliation(s)
- Mahamudul Haque
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, WA, USA
| | - Ruby J Siegel
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, WA, USA
| | - David A Fox
- Division of Rheumatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Salahuddin Ahmed
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, WA, USA.,Division of Rheumatology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
14
|
Aslanalp Z, Tikiz C, Ulusoy A, Orguc Ş, Bİlgİ Yedekcİ A, Ulman C. The Relationship Between Serum Angiogenic Factor Levels and Disease Activity in Rheumatoid Arthritis. Arch Rheumatol 2021; 35:416-425. [PMID: 33458666 PMCID: PMC7788655 DOI: 10.46497/archrheumatol.2020.7416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 09/04/2019] [Indexed: 12/03/2022] Open
Abstract
Objectives
This study aims to evaluate the relationship between serum angiogenic factor levels and disease activity in patients with rheumatoid arthritis (RA) using both clinical and dynamic wrist magnetic resonance imaging (MRI) data. Patients and methods
Simultaneous serum angiogenesis markers [vascular endothelial growth factor (VEGF), angiopoietin-1 (ANG1), ANG2, and tyrosine-protein kinase receptor for angiopoietin (Tie-2)] were studied in 40 patients with RA (13 males, 27 females; mean age 51.1±10.8 years; range, 23 to 69 years) and 20 healthy controls (11 males, 9 females; mean age 47.3±12.8 years; range, 29 to 69 years) and dynamic contrast-enhanced wrist MRI was performed in 40 RA patients and seven controls. Rate of early in 55th second (REE) and Relative enhancement (REt) values were calculated from the signal time curve values obtained from the analysis of images. In clinical assessment, duration of morning stiffness, patient pain assessment [visual analog scale (VAS)], physician and patient global assessments (VAS) were recorded. The number of tender joints and swollen joints were determined. Disease activity score 28 and Ritchie scores were calculated. Health assessment questionnaire was used for functional evaluation. Anti-cyclic citrullinated peptide, rheumatoid factor, erythrocyte sedimentation rate and high sensitive C-reactive protein analyses were performed. Results
Serum VEGF, REE and REt values were significantly higher in RA patients than healthy controls (p=0.002, p=0.00, p=0.00, respectively). There was no significant correlation between serum angiogenesis markers and clinical parameters or REE and REt (p>0.05). VEGF value correlated positively with disease duration (p=0.024). Conclusion Serum VEGF was higher in RA patients. While its level was associated with disease duration, no significant correlation was found with disease activity. As a diagnostic test, dynamic contrast-enhanced MRI was a valuable method for showing disease activity.
Collapse
Affiliation(s)
- Zahide Aslanalp
- Department of Physical Medicine and Rehabilitation, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Canan Tikiz
- Department of Physical Medicine and Rehabilitation, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Aslıhan Ulusoy
- Department of Physical Medicine and Rehabilitation, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Şebnem Orguc
- Department of Radiodiagnostic, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Aysun Bİlgİ Yedekcİ
- Department of Biochemistry, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Cevval Ulman
- Department of Biochemistry, Manisa Celal Bayar University Faculty of Medicine, Manisa, Turkey
| |
Collapse
|
15
|
Zhu Z, Liao H, Scheetz J, Zhang J, He J, Wang W. The Association between Retinopathy and Arthritis: Findings from a US National Survey 2005-2008. Curr Eye Res 2020; 45:1543-1549. [PMID: 32323599 DOI: 10.1080/02713683.2020.1760306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Objective To explore the association between retinopathy and arthritis in a representative sample in the United States. Methods National Health and Nutrition Examination Survey (NHANES) is a series of biannual surveys in a nationally representative non-institutionalized population of the United States. Two waves of NHANES (2005-2008) were merged in the current analysis. Arthritis status and types of arthritis were obtained from questionnaires. Non-mydriatic fundus photographs were collected among participants aged 40 years and older and retinopathy status was assessed using the Airlie House classification scheme. The association between arthritis and retinopathy was evaluated using logistic regression models. Results From 6,797 eligible participants, 4,901 had information in terms of arthritis status and gradable fundus photographs of at least 1 eye. Retinopathy was less prevalent in patients with osteoarthritis compared with those without osteoarthritis (11.9% vs 17.5%, P < .05). Participants who had osteoarthritis had 43% reduced relative risk of retinopathy compared to those without, after adjusting for covariates (Odds Ratio [OR] = 0.57, 95% Confidence Interval [CI]: 0.33-0.98). This significant association persisted among diabetic participants (OR = 0.43, 95% CI: 0.19-1.00). However, no significant associations between any arthritis, rheumatoid arthritis, other types of arthritis and retinopathy were noted. Conclusions Patients with osteoarthritis were less likely to have retinopathy when compared with those without. Further studies investigating the possible protective effects of arthritis physiology and/or arthritis treatment in retinopathy are needed.
Collapse
Affiliation(s)
- Zhuoting Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China.,Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science
| | - Huan Liao
- Neural Regeneration Group, Institute of Reconstructive Neurobiology, University of Bonn , Bonn, Germany
| | - Jane Scheetz
- Centre for Eye Research Australia, Royal Victorian Eye & Ear Hospital , Melbourne, Australia
| | - Jian Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China
| | - Jingyi He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University , Guangzhou, China
| |
Collapse
|
16
|
Naito H, Iba T, Takakura N. Mechanisms of new blood-vessel formation and proliferative heterogeneity of endothelial cells. Int Immunol 2020; 32:295-305. [DOI: 10.1093/intimm/dxaa008] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/27/2020] [Indexed: 12/26/2022] Open
Abstract
Abstract
The vast blood-vessel network of the circulatory system is crucial for maintaining bodily homeostasis, delivering essential molecules and blood cells, and removing waste products. Blood-vessel dysfunction and dysregulation of new blood-vessel formation are related to the onset and progression of many diseases including cancer, ischemic disease, inflammation and immune disorders. Endothelial cells (ECs) are fundamental components of blood vessels and their proliferation is essential for new vessel formation, making them good therapeutic targets for regulating the latter. New blood-vessel formation occurs by vasculogenesis and angiogenesis during development. Induction of ECs termed tip, stalk and phalanx cells by interactions between vascular endothelial growth factor A (VEGF-A) and its receptors (VEGFR1–3) and between Notch and Delta-like Notch ligands (DLLs) is crucial for regulation of angiogenesis. Although the importance of angiogenesis is unequivocal in the adult, vasculogenesis effected by endothelial progenitor cells (EPCs) may also contribute to post-natal vessel formation. However, the definition of these cells is ambiguous and they include several distinct cell types under the simple classification of ‘EPC’. Furthermore, recent evidence indicates that ECs within the intima show clonal expansion in some situations and that they may harbor vascular-resident endothelial stem cells. In this article, we summarize recent knowledge on vascular development and new blood-vessel formation in the adult. We also introduce concepts of EC heterogeneity and EC clonal expansion, referring to our own recent findings.
Collapse
Affiliation(s)
- Hisamichi Naito
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Tomohiro Iba
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Signal Transduction, World Premier Institute Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
17
|
Baihu Jia Guizhi Decoction Improves Rheumatoid Arthritis Inflammation by Regulating Succinate/SUCNR1 Metabolic Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3258572. [PMID: 31949465 PMCID: PMC6948314 DOI: 10.1155/2019/3258572] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 11/04/2019] [Accepted: 11/26/2019] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovitis. Succinate is an inflammatory metabolic signal that exacerbates RA synovitis by activating succinate receptor 1 (SUCNR1) to amplify the release of IL-1β. Thus, inhibition of succinate activation of SUCRN1 could be an effective method to inhibit the inflammation of RA. Baihu Jia Guizhi decoction (BHGZ), which is composed of Gypsum Fibrosum, Anemarrhena asphodeloides Bge., Cinnamomum cassia Presl., Glycyrrhiza uralensis Fisch., and Oryza sativa L., is a Traditional Chinese Medicine (TCM) prescription used to treat RA in clinic. In addition, TCM believes that damp and heat environment is one of the causes of RA. In this study, we tested the role of damp and heat environments in exacerbating RA inflammation and the anti-inflammatory effect of BHGZ, based on succinate/SUCNR1/IL-1β pathway in the adjuvant arthritis (AA) model with damp and heat environment (AA + DHE). Results showed that paw swelling and synovial pathology were significantly increased in AA rats, and these results were aggravated by stimulation in damp and heat environment. BHGZ improved AA + DHE rats' paw swelling, synovial hyperplasia, and inflammatory cell infiltration and reduced IL-1β. In addition, AA rats significantly increased the expression of SUCNR1, and the stimulation of damp and heat environment not only increased the expression of SUCNR1 but also promoted the accumulation of succinate. BHGZ simultaneously reduced the concentration of succinate and the expression of SUCNR1. Finally, SDH activity was decreased in AA rats and AA + DHE rats, while BHGZ increased SDH activity and then reduced succinate concentration. Therefore, we prove that damp and heat environment deteriorated the inflammation of RA which is the activation of succinate/SUCNR1 pathway, while BHGZ regulates SDH activity to reduce the accumulation of succinate and inhibit the activation of SUCNR1 that is the underlying mechanism of its treatment of RA.
Collapse
|
18
|
Role of Interleukin 37 as a Novel Proangiogenic Factor in Juvenile Idiopathic Arthritis. J Clin Rheumatol 2019; 25:85-90. [PMID: 29683837 DOI: 10.1097/rhu.0000000000000779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The aim of this study was to investigate interleukin 37 (IL-37) levels in the serum and synovial fluid of patients with juvenile idiopathic arthritis (JIA), its expression in peripheral blood mononuclear cells, and correlation with disease activity and angiogenesis. METHODS Seventy JIA patients and 50 control subjects were examined. The Juvenile Arthritis Disease Activity Score in 27 joints (JADAS-27) was calculated. Immunoassays were used to measure the serum and synovial fluid levels of IL-37, vascular endothelial growth factor (VEGF), soluble VEGF receptor 1 (sVEGF-R1), and sVEGF-R2. Relative expression of IL-37 mRNA in peripheral blood mononuclear cells and the power Doppler ultrasound score of the affected joint were measured. RESULTS Patients with JIA were subdivided as 20 systemic-onset, 20 polyarticular, and 30 oligoarticular (10 persistent, 20 extended) cases. Serum levels of IL-37, VEGF, VEGF-R1, and VEGF-R2 and relative IL-37 mRNA expression were significantly higher in JIA patients when compared with the control subjects (p < 0.001). These concentrations were significantly higher in systemic-onset JIA compared with those in polyarticular and oligoarticular JIA, and in polyarticular JIA when compared with oligoarticular JIA (p < 0.001). Serum, synovial, and mRNA expression levels of IL-37 were positively correlated with C-reactive protein, erythrocyte sedimentation rate, Juvenile Arthritis Disease Activity Score in 27 joints, power Doppler ultrasound score (p < 0.001), and the serum and synovial VEGF and VEGF-RI and -R2 levels (p < 0.05). CONCLUSIONS Our results demonstrate that IL-37 levels and mRNA expression were significantly increased in JIA patients, and their levels were positively correlated with disease activity and markers of angiogenesis (VEGF and VEGF receptors), suggesting that IL-37 may be correlated with angiogenesis.
Collapse
|
19
|
Rai MF, Pan H, Yan H, Sandell LJ, Pham CTN, Wickline SA. Applications of RNA interference in the treatment of arthritis. Transl Res 2019; 214:1-16. [PMID: 31351032 PMCID: PMC6848781 DOI: 10.1016/j.trsl.2019.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/02/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022]
Abstract
RNA interference (RNAi) is a cellular mechanism for post-transcriptional gene regulation mediated by small interfering RNA (siRNA) and microRNA. siRNA-based therapy holds significant promise for the treatment of a wide-range of arthritic diseases. siRNA selectively suppresses the expression of a gene product and can thus achieve the specificity that is lacking in small molecule inhibitors. The potential use of siRNA-based therapy in arthritis, however, has not progressed to clinical trials despite ample evidence for efficacy in preclinical studies. One of the main challenges to clinical translation is the lack of a suitable delivery vehicle to efficiently and safely access diverse pathologies. Moreover, the ideal targets in treatment of arthritides remain elusive given the complexity and heterogeneity of these disease pathogeneses. Herein, we review recent preclinical studies that use RNAi-based drug delivery systems to mitigate inflammation in models of rheumatoid arthritis and osteoarthritis. We discuss a self-assembling peptide-based nanostructure that demonstrates the potential of overcoming many of the critical barriers preventing the translation of this technology to the clinic.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, Missouri; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Hua Pan
- Department of Cardiovascular Sciences, University of South Florida Health Heart Institute, Morsani School of Medicine, Tampa, Florida
| | - Huimin Yan
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Linda J Sandell
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, Missouri; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Christine T N Pham
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri.
| | - Samuel A Wickline
- Department of Cardiovascular Sciences, University of South Florida Health Heart Institute, Morsani School of Medicine, Tampa, Florida
| |
Collapse
|
20
|
Dolcino M, Tinazzi E, Puccetti A, Lunardi C. Long Non-Coding RNAs Target Pathogenetically Relevant Genes and Pathways in Rheumatoid Arthritis. Cells 2019; 8:cells8080816. [PMID: 31382516 PMCID: PMC6721587 DOI: 10.3390/cells8080816] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/16/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease driven by genetic, environmental and epigenetic factors. Long non-coding RNAs (LncRNAs) are a key component of the epigenetic mechanisms and are known to be involved in the development of autoimmune diseases. In this work we aimed to identify significantly differentially expressed LncRNAs (DE-LncRNAs) that are functionally connected to modulated genes strictly associated with RA. In total, 542,500 transcripts have been profiled in peripheral blood mononuclear cells (PBMCs) from four patients with early onset RA prior any treatment and four healthy donors using Clariom D arrays. Results were confirmed by real-time PCR in 20 patients and 20 controls. Six DE-LncRNAs target experimentally validated miRNAs able to regulate differentially expressed genes (DEGs) in RA; among them, only FTX, HNRNPU-AS1 and RP11-498C9.15 targeted a large number of DEGs. Most importantly, RP11-498C9.15 targeted the largest number of signalling pathways that were found to be enriched by the global amount of RA-DEGs and that have already been associated with RA and RA-synoviocytes. Moreover, RP11-498C9.15 targeted the most highly connected genes in the RA interactome, thus suggesting its involvement in crucial gene regulation. These results indicate that, by modulating both microRNAs and gene expression, RP11-498C9.15 may play a pivotal role in RA pathogenesis.
Collapse
Affiliation(s)
- Marzia Dolcino
- Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Elisa Tinazzi
- Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Antonio Puccetti
- Department of Experimental Medicine-Section of Histology, University of Genova, 16132 Genova, Italy
| | - Claudio Lunardi
- Department of Medicine, University of Verona, 37134 Verona, Italy.
| |
Collapse
|
21
|
Abstract
The formation and remodeling of a functional circulatory system is critical for sustaining prenatal and postnatal life. During embryogenesis, newly differentiated endothelial cells require further specification to create the unique features of distinct vessel subtypes needed to support tissue morphogenesis. In this review, we explore signaling pathways and transcriptional regulators that modulate endothelial cell differentiation and specification, as well as applications of these processes to stem cell biology and regenerative medicine. We also summarize recent technical advances, including the growing utilization of single-cell sequencing to study vascular heterogeneity and development.
Collapse
Affiliation(s)
- Jingyao Qiu
- From the Department of Genetics (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Department of Medicine (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Yale Cardiovascular Research Center (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT
| | - Karen K Hirschi
- From the Department of Genetics (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Department of Medicine (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Yale Cardiovascular Research Center (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT.,Vascular Biology and Therapeutics Program (J.Q., K.K.H.), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
22
|
Du Y, Lu C, Morgan RL, Stinson WA, Campbell PL, Cealey E, Fu W, Lepore NJ, Hervoso JL, Cui H, Urquhart AG, Lawton JN, Chung KC, Fox DA, Amin MA. Angiogenic and Arthritogenic Properties of the Soluble Form of CD13. THE JOURNAL OF IMMUNOLOGY 2019; 203:360-369. [PMID: 31189572 DOI: 10.4049/jimmunol.1801276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/15/2019] [Indexed: 11/19/2022]
Abstract
Aminopeptidase N/CD13 is expressed by fibroblast-like synoviocytes (FLS) and monocytes (MNs) in inflamed human synovial tissue (ST). This study examined the role of soluble CD13 (sCD13) in angiogenesis, MN migration, phosphorylation of signaling molecules, and induction of arthritis. The contribution of sCD13 was examined in angiogenesis and MN migration using sCD13 and CD13-depleted rheumatoid arthritis (RA) synovial fluids (SFs). An enzymatically inactive mutant CD13 and intact wild-type (WT) CD13 were used to determine whether its enzymatic activity contributes to the arthritis-related functions. CD13-induced phosphorylation of signaling molecules was determined by Western blotting. The effect of sCD13 on cytokine secretion from RA ST and RA FLS was evaluated. sCD13 was injected into C57BL/6 mouse knees to assess its arthritogenicity. sCD13 induced angiogenesis and was a potent chemoattractant for MNs and U937 cells. Inhibitors of Erk1/2, Src, NF-κB, Jnk, and pertussis toxin, a G protein-coupled receptor inhibitor, decreased sCD13-stimulated chemotaxis. CD13-depleted RA SF induced significantly less MN migration than sham-depleted SF, and addition of mutant or WT CD13 to CD13-depleted RA SF equally restored MN migration. sCD13 and recombinant WT or mutant CD13 had similar effects on signaling molecule phosphorylation, indicating that the enzymatic activity of CD13 had no role in these functions. CD13 increased the expression of proinflammatory cytokines by RA FLS, and a CD13 neutralizing Ab inhibited cytokine secretion from RA ST organ culture. Mouse knee joints injected with CD13 exhibited increased circumference and proinflammatory mediator expression. These data support the concept that sCD13 plays a pivotal role in RA and acute inflammatory arthritis.
Collapse
Affiliation(s)
- Yuxuan Du
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109.,Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,National Center for Clinical Laboratories/Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Chenyang Lu
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Rachel L Morgan
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - William A Stinson
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Phillip L Campbell
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Ellen Cealey
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Wenyi Fu
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109.,Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, China; and
| | - Nicholas J Lepore
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Jonatan L Hervoso
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Huadong Cui
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109.,Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, China; and
| | - Andrew G Urquhart
- Department of Orthopaedic Surgery, University of Michigan Health System, A. Alfred Taubman Health Care Center, Ann Arbor, MI 48109
| | - Jeffrey N Lawton
- Department of Orthopaedic Surgery, University of Michigan Health System, A. Alfred Taubman Health Care Center, Ann Arbor, MI 48109
| | - Kevin C Chung
- Department of Orthopaedic Surgery, University of Michigan Health System, A. Alfred Taubman Health Care Center, Ann Arbor, MI 48109
| | - David A Fox
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109;
| | - Mohammad A Amin
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
23
|
Wang JW, Chen SS, Zhang YM, Guan J, Su GY, Ding M, Li W, Zhao YQ. Anti-inflammatory and analgesic activity based on polymorphism of cedrol in mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 68:13-18. [PMID: 30852303 DOI: 10.1016/j.etap.2019.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/19/2019] [Accepted: 02/13/2019] [Indexed: 06/09/2023]
Abstract
Alternate forms of drug crystals display different physicochemical properties. These include stability, dissolution rate, bioavailability and solubility, which can affect pharmacokinetics and pharmacodynamics. It is therefore important to compare the crystal forms of cedrol to obtain optimal anti-inflammatory and analgesic effects. This study, for the first time, obtained and reports three novel forms (I-III) of cedrol polymorphs. The three forms of cedrol were recrystallized from seven organic solvents by slow cooling or volatilization and identified by thermal analysis, fourier transform infrared spectroscopy, scanning electron microscopy and powder X-ray diffraction analysis. Form I originated from acetone and cyclohexane. Form II was obtained from ethanol, ethyl acetate, acetonitrile and n-hexane. Form III was recrystallized from methanol. The anti-inflammatory and analgesic activities of the three crystalline forms were evaluated by acetic acid induced writhing in mice, the hot plate method, carrageenan induced mouse paw edema models, Xylene-induced mouse ear edema models and cotton pellet-induced mouse granuloma models. Experimental results revealed that the highest performance was achieved from Form I. These findings are of great significance during the early research study of cedrol polymorphs.
Collapse
Affiliation(s)
- Jie-Wen Wang
- Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Liaoning Xinzhong Modern Medicine Co. Ltd, Shenyang 110041, People's Republic of China
| | - Shan-Shan Chen
- Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yu-Meng Zhang
- Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Jian Guan
- Liaoning Xinzhong Modern Medicine Co. Ltd, Shenyang 110041, People's Republic of China
| | - Guang-Yue Su
- Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Meng Ding
- Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Wei Li
- Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Key Laboratory of Structure-based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yu-Qing Zhao
- Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China; Key Laboratory of Structure-based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| |
Collapse
|
24
|
Feng ZT, Yang T, Hou XQ, Wu HY, Feng JT, Ou BJ, Cai SJ, Li J, Mei ZG. Sinomenine mitigates collagen-induced arthritis mice by inhibiting angiogenesis. Biomed Pharmacother 2019; 113:108759. [PMID: 30856539 DOI: 10.1016/j.biopha.2019.108759] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE The objective of the present study is to investigate the inhibitory effects of sinomenine (SIN) on angiogenesis in a collagen-induced arthritis (CIA) mouse model. METHODS Arthritis assessments for all mice were recorded. The histopathological assessments were performed following haematoxylin and eosin (HE) staining. Immunohistochemistry and enzyme-linked immunosorbent assay (ELISA) analyses were used to detect the expression of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and angiopoietin 1 (ANG-1) in the serum and in the membrane. Immunohistochemistry was employed to detect the synovium microvessel density (MVD). RESULTS Compared with the CIA model group, SIN significantly ameliorated swelling and erythema extension, decreased the arthritis index, reduced inflammation, cartilage damage and bone erosion, and lessened the number of CD31 positive cells on the synovium. Moreover, the levels of HIF-1α, VEGF and ANG-1 in the synovium and in the peripheral serum were increased in the untreated CIA model group but were significantly reduced in the 30 mg/kg, 100 mg/kg and 300 mg/kg SIN treatment groups. CONCLUSION SIN could mitigate CIA by inhibiting angiogenesis, and the mechanism may associate with the HIF-1α-VEGF-ANG-1 axis. Additionally, our study provides a referable experimental basis for the use of SIN for the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Zhi-Tao Feng
- 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; Shenzhen Institute of Geriatrics, Shenzhen, Guangdong, 518020, China; The Institute of Rheumatology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, 443003, China
| | - Tong Yang
- 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
| | - Xiao-Qiang Hou
- The Institute of Rheumatology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei, 443003, China
| | - Han-Yu Wu
- 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
| | - Jia-Teng Feng
- 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
| | - Bing-Jin Ou
- 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
| | - San-Jin Cai
- 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
| | - Juan Li
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China; Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Zhi-Gang Mei
- 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.
| |
Collapse
|
25
|
Ross CL, Ang DC, Almeida-Porada G. Targeting Mesenchymal Stromal Cells/Pericytes (MSCs) With Pulsed Electromagnetic Field (PEMF) Has the Potential to Treat Rheumatoid Arthritis. Front Immunol 2019; 10:266. [PMID: 30886614 PMCID: PMC6409305 DOI: 10.3389/fimmu.2019.00266] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/31/2019] [Indexed: 01/14/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of synovium (synovitis), with inflammatory/immune cells and resident fibroblast-like synoviocytes (FLS) acting as major players in the pathogenesis of this disease. The resulting inflammatory response poses considerable risks as loss of bone and cartilage progresses, destroying the joint surface, causing joint damage, joint failure, articular dysfunction, and pre-mature death if left untreated. At the cellular level, early changes in RA synovium include inflammatory cell infiltration, synovial hyperplasia, and stimulation of angiogenesis to the site of injury. Different angiogenic factors promote this disease, making the role of anti-angiogenic therapy a focus of RA treatment. To control angiogenesis, mesenchymal stromal cells/pericytes (MSCs) in synovial tissue play a vital role in tissue repair. While recent evidence reports that MSCs found in joint tissues can differentiate to repair damaged tissue, this repair function can be repressed by the inflammatory milieu. Extremely-low frequency pulsed electromagnetic field (PEMF), a biophysical form of stimulation, has an anti-inflammatory effect by causing differentiation of MSCs. PEMF has also been reported to increase the functional activity of MSCs to improve differentiation to chondrocytes and osteocytes. Moreover, PEMF has been demonstrated to accelerate cell differentiation, increase deposition of collagen, and potentially return vascular dysfunction back to homeostasis. The aim of this report is to review the effects of PEMF on MSC modulation of cytokines, growth factors, and angiogenesis, and describe its effect on MSC regeneration of synovial tissue to further understand its potential role in the treatment of RA.
Collapse
Affiliation(s)
- Christina L Ross
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States.,Wake Forest Center for Integrative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Dennis C Ang
- Department of Rheumatology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| |
Collapse
|
26
|
Li Y, Liu Y, Wang C, Xia WR, Zheng JY, Yang J, Liu B, Liu JQ, Liu LF. Succinate induces synovial angiogenesis in rheumatoid arthritis through metabolic remodeling and HIF-1α/VEGF axis. Free Radic Biol Med 2018; 126:1-14. [PMID: 30030103 DOI: 10.1016/j.freeradbiomed.2018.07.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/02/2018] [Accepted: 07/16/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND PURPOSE In response to hypoxic succinate accumulates in arthritis synovium, however, the implication is little known. This study aims to investigate whether succinate could act as a metabolic signal linking metabolic alternation with angiogenesis in arthritis synovium. EXPERIMENTAL APPROACH The interaction between elevated succinate and VEGF production was examined in endothelial cells. Succinate production, HIF-1α induction and angiogenesis in the hypoxic synovium of collagen-induced arthritis rats were also investigated. KEY RESULTS Intracellular succinate promoted VEGF production and induced angiogenic response dependent on HIF-1α induction in endothelial cells. Luciferase reporter assay showed that succinate increased VEGF expression through gene promoter activation dependent on HIF-1α induction. Intracellular succinate released into intercellular space, where extracellular succinate activated succinate receptor G-protein-coupled receptor 91 (GPR91) and induced VEGF production, further exacerbating angiogenesis. In addition, TGF-β1 treatment increased succinate production due to the reversal of succinate dehydrogenase (SDH) activation, and consistently, SDH inhibitor dimethyl malonate reduced angiogenesis in the arthritis synovium. CONCLUSION AND IMPLICATIONS More than an intermediate, succinate functioned as a signaling molecule to link metabolic reprograming with angiogenesis. Intracellular succinate induced angiogenesis through HIF-1α induction, while extracellular succinate acted on GPR91 activation, working together to disturb energy metabolism and exacerbate inflammation and angiogenesis in arthritis synovium. Our work suggested that suppression of SDH could prevent succinate accumulation and inhibit angiogenesis via blocking HIF-1α/VEGF axis. This finding not only provides a novel insight into angiogenesis, but also reveals a potential therapeutical strategy to attenuate revascularization in arthritis.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/genetics
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Disease Models, Animal
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Rats
- Receptors, G-Protein-Coupled/genetics
- Signal Transduction/genetics
- Succinate Dehydrogenase/genetics
- Succinic Acid/metabolism
- Synovial Fluid/metabolism
- Transforming Growth Factor beta1/genetics
- Vascular Endothelial Growth Factor A/genetics
Collapse
Affiliation(s)
- Yi Li
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wen-Rui Xia
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jia-Yi Zheng
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Yang
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Baolin Liu
- State Key Laboratory of Natural Medicines, Department of Complex Prescription of TCM, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jian-Qun Liu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Li-Fang Liu
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
27
|
Wang ZZ, Liu F, Gong YF, Huang TY, Zhang XM, Huang XY. Antiarthritic Effects of Sorafenib in Rats with Adjuvant-Induced Arthritis. Anat Rec (Hoboken) 2018; 301:1519-1526. [DOI: 10.1002/ar.23856] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/25/2018] [Accepted: 03/01/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Zhen-Zhen Wang
- Department of Anatomy; Anhui Medical University; Hefei China
| | - Fei Liu
- Department of Anatomy; Anhui Medical University; Hefei China
| | - Yong-Fang Gong
- Department of Anatomy; Bengbu Medical College; Bengbu China
| | - Tian-Yu Huang
- Grade 2016, The First Department of Clinical Medicine; Bengbu Medical College; Bengbu China
| | - Xiao-Ming Zhang
- Department of Anatomy; Anhui Medical University; Hefei China
| | - Xue-Ying Huang
- Department of Anatomy; Anhui Medical University; Hefei China
| |
Collapse
|
28
|
O'Brien MJ, Shu Q, Stinson WA, Tsou PS, Ruth JH, Isozaki T, Campbell PL, Ohara RA, Koch AE, Fox DA, Amin MA. A unique role for galectin-9 in angiogenesis and inflammatory arthritis. Arthritis Res Ther 2018; 20:31. [PMID: 29433546 PMCID: PMC5809993 DOI: 10.1186/s13075-018-1519-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 01/17/2018] [Indexed: 01/25/2023] Open
Abstract
Background Galectin-9 (Gal-9) is a mammalian lectin secreted by endothelial cells that is highly expressed in rheumatoid arthritis synovial tissues and synovial fluid. Roles have been proposed for galectins in the regulation of inflammation and angiogenesis. Therefore, we examined the contribution of Gal-9 to angiogenesis and inflammation in arthritis. Methods To determine the role of Gal-9 in angiogenesis, we performed human dermal microvascular endothelial cell (HMVEC) chemotaxis, Matrigel tube formation, and mouse Matrigel plug angiogenesis assays. We also examined the role of signaling molecules in Gal-9-induced angiogenesis by using signaling inhibitors and small interfering RNA (siRNA). We performed monocyte (MN) migration assays in a modified Boyden chamber and assessed the arthritogenicity of Gal-9 by injecting Gal-9 into mouse knees. Results Gal-9 significantly increased HMVEC migration, which was decreased by inhibitors of extracellular signal-regulating kinases 1/2 (Erk1/2), p38, Janus kinase (Jnk), and phosphatidylinositol 3-kinase. Gal-9 HMVEC-induced tube formation was reduced by Erk1/2, p38, and Jnk inhibitors, and this was confirmed by siRNA knockdown. In mouse Matrigel plug assays, plugs containing Gal-9 induced significantly higher angiogenesis, which was attenuated by a Jnk inhibitor. Gal-9 also induced MN migration, and there was a marked increase in MN ingress when C57BL/6 mouse knees were injected with Gal-9 compared with the control, pointing to a proinflammatory role for Gal-9. Conclusions Gal-9 mediates angiogenesis, increases MN migration in vitro, and induces acute inflammatory arthritis in mice, suggesting a novel role for Gal-9 in angiogenesis, joint inflammation, and possibly other inflammatory diseases.
Collapse
Affiliation(s)
- Martin J O'Brien
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Qiang Shu
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA.,Shenzhen Research Institute of Shandong University, Shenzhen, China.,Rheumatology Department, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - W Alexander Stinson
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Pei-Suen Tsou
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Jeffrey H Ruth
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Takeo Isozaki
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Phillip L Campbell
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Ray A Ohara
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Alisa E Koch
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA.,Department of Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - David A Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - M Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
29
|
Hao J, Wu X, Setrerrahmane S, Qian K, Hou Y, Yu L, Lin C, Wu Q, Xu H. Combination Therapy of PEG-HM-3 and Methotrexate Retards Adjuvant-Induced Arthritis. Int J Mol Sci 2017; 18:E1538. [PMID: 28754008 PMCID: PMC5536026 DOI: 10.3390/ijms18071538] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 01/24/2023] Open
Abstract
At present, the early phenomenon of inflammatory angiogenesis is rarely studied in Rheumatoid arthritis (RA). Previous research found that PEG-HM-3, an integrin inhibitor, possessed anti-angiogenesis and anti-rheumatic activity. In this study, the advantages of inhibiting angiogenesis and immune cell adhesion and migration, as well as the benefits of anti-arthritis effects, were evaluated using a combination of PEG-HM-3 and methotrexate (MTX). In vitro, spleen cell proliferation and the levels of tumor necrosis factor α (TNF-α) in macrophage supernatant were assessed. Hind paw edema, arthritis index, clinical score, body weight and immunohistochemistry (IHC) of the spleen, thymus, and joint cavity were evaluated in vivo in adjuvant-induced arthritis rats. Joints of the left hind paws were imaged by X-ray. The expression of the toll-like receptor 4 (TLR-4) protein was assessed in lipopolysaccharide (LPS)-induced synoviocytes. PEG-HM-3 combined with MTX significantly reduced primary and secondary swelling of the hind paws, the arthritis index, the clinical score and bone erosion. The results of IHC showed that the levels of interleukin-6 (IL-6) in spleens and the levels of TNF-α, CD31 (cluster of differentiation 31), and CD105 in the joint cavity were decreased. The body weight of rats was maintained during combination therapy. Ankle cavity integrity, and bone erosion and deformity were improved in combination treatment. The expression of TLR-4 was significantly reduced with combination treatment in rat synoviocytes. Co-suppression of both inflammation and angiogenesis in arthritis was achieved in this design with combination therapy. The activity of nuclear transcription factor (NF-κB) and the expression of inflammatory factors were down regulated via integrin αvβ₃ and TLR-4 signaling pathways. In the future, the application of this combination can be a candidate in early and mid-term RA therapy.
Collapse
Affiliation(s)
- Jingchao Hao
- The Engineering Research Centre of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.
- School of Pharmaceutical Sciences & Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China.
| | - Xiaodong Wu
- The Engineering Research Centre of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.
| | - Sarra Setrerrahmane
- The Engineering Research Centre of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.
| | - Kun Qian
- School of Life Science, Huzhou University, Huzhou 313000, China.
| | - Yueying Hou
- XiangYa School of Medicine, Central South University, Changsha 410013, China.
| | - Liting Yu
- The Engineering Research Centre of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.
| | - Chenyu Lin
- The Engineering Research Centre of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.
| | - Qianqian Wu
- The Engineering Research Centre of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.
| | - Hanmei Xu
- The Engineering Research Centre of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 210009, China.
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
30
|
Ma B, Whiteford JR, Nourshargh S, Woodfin A. Underlying chronic inflammation alters the profile and mechanisms of acute neutrophil recruitment. J Pathol 2017; 240:291-303. [PMID: 27477524 PMCID: PMC5082550 DOI: 10.1002/path.4776] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/13/2016] [Accepted: 07/20/2016] [Indexed: 01/15/2023]
Abstract
Chronically inflamed tissues show altered characteristics that include persistent populations of inflammatory leukocytes and remodelling of the vascular network. As the majority of studies on leukocyte recruitment have been carried out in normal healthy tissues, the impact of underlying chronic inflammation on ongoing leukocyte recruitment is largely unknown. Here, we investigate the profile and mechanisms of acute inflammatory responses in chronically inflamed and angiogenic tissues, and consider the implications for chronic inflammatory disorders. We have developed a novel model of chronic ischaemia of the mouse cremaster muscle that is characterized by a persistent population of monocyte‐derived cells (MDCs), and capillary angiogenesis. These tissues also show elevated acute neutrophil recruitment in response to locally administered inflammatory stimuli. We determined that Gr1lowMDCs, which are widely considered to have anti‐inflammatory and reparative functions, amplified acute inflammatory reactions via the generation of additional proinflammatory signals, changing both the profile and magnitude of the tissue response. Similar vascular and inflammatory responses, including activation of MDCs by transient ischaemia–reperfusion, were observed in mouse hindlimbs subjected to chronic ischaemia. This response demonstrates the relevance of the findings to peripheral arterial disease, in which patients experience transient exercise‐induced ischaemia known as claudication.These findings demonstrate that chronically inflamed tissues show an altered profile and altered mechanisms of acute inflammatory responses, and identify tissue‐resident MDCs as potential therapeutic targets. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Bin Ma
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Cardiovascular Division, King's College London, London, UK
| | - James R Whiteford
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sussan Nourshargh
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Abigail Woodfin
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK. .,Cardiovascular Division, King's College London, London, UK.
| |
Collapse
|
31
|
Wu Y, Zhang G, Wang X, Zhao Z, Wang T, Wang X, Li XF. Early detection of rheumatoid arthritis in rats and humans with 99mTc-3PRGD2 scintigraphy: imaging synovial neoangiogenesis. Oncotarget 2017; 8:5753-5760. [PMID: 27992368 PMCID: PMC5351586 DOI: 10.18632/oncotarget.13953] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 12/08/2016] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES To validate 99mTc-labeled arginylglycylaspartic acid (99mTc-3PRGD2) scintigraphy as a means to image synovial neoangiogenesis in joints afflicted by rheumatoid arthritis and to investigate its potential in the early detection and management of rheumatoid arthritis. METHODS Rheumatoid arthritis and osteoarthritis were generated in Sprague Dawley rats by type II collagen immunization and papain injection, respectively. Rats were imaged with 99mTc-3PRGD2 and 99mTc- methyl diphosphonate (99mTc MDP). X-ray images were also obtained and assessed by a radiologist. Immunohistochemistry of αvβ3 and CD31confirmed the onset of synovial neoangiogenesis. The effect of bevacizumab on rheumatoid arthritis was followed with 99mTc-3PRGD2 scintigraphy. A patient with rheumatoid arthritis and a healthy volunteer were scanned with 99mTc-3PRGD2. RESULTS Two weeks after immunization, a significant increase in 99mTc-3PRGD2 was observed in the joints of the rheumatoid arthritis model though uptake in osteoarthritis model and untreated controls was low. 99mTc-MDP whole body scans failed to distinguish early rheumatoid arthritis joints from healthy controls. The expression of αvβ3 and CD31was significantly higher in the joints of rheumatoid arthritis rats compared to normal controls. In serial 99mTc-3PRGD2 scintigraphy studies, 99mTc-3PRGD2 uptake increased in parallel with disease progression. Bevacizumab anti-angiogenetic therapy both improved the symptoms of the rheumatoid arthritis rats and significantly decreased 99mTc-3PRGD2 uptake. Significantly higher 99mTc-3PRGD2 accumulation was also observed in rheumatoid arthritis joints in the patient. CONCLUSIONS Our findings indicate that 99mTc-3PRGD2 scintigraphy could detect early rheumatoid arthritis by imaging the associated synovial neoangiogenesis, and may be useful in disease management.
Collapse
Affiliation(s)
- Yu Wu
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated People’s Hospital, Hohhot, Inner Mongolia, China
| | - Guojian Zhang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
| | - Xiangcheng Wang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
| | - Zhenfang Zhao
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
| | - Tao Wang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
| | - Xuemei Wang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
| | - Xiao-Feng Li
- PET/CT/MRI Center, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Radiology, University of Louisville, Louisville KY, USA
| |
Collapse
|
32
|
Hu Z, Zhang H, Mordovanakis A, Paulus YM, Liu Q, Wang X, Yang X. High-precision, non-invasive anti-microvascular approach via concurrent ultrasound and laser irradiation. Sci Rep 2017; 7:40243. [PMID: 28074839 PMCID: PMC5225605 DOI: 10.1038/srep40243] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/05/2016] [Indexed: 11/09/2022] Open
Abstract
Antivascular therapy represents a proven strategy to treat angiogenesis. By applying synchronized ultrasound bursts and nanosecond laser irradiation, we developed a novel, selective, non-invasive, localized antivascular method, termed photo-mediated ultrasound therapy (PUT). PUT takes advantage of the high native optical contrast among biological tissues and can treat microvessels without causing collateral damage to the surrounding tissue. In a chicken yolk sac membrane model, under the same ultrasound parameters (1 MHz at 0.45 MPa and 10 Hz with 10% duty cycle), PUT with 4 mJ/cm2 and 6 mJ/cm2 laser fluence induced 51% (p = 0.001) and 37% (p = 0.018) vessel diameter reductions respectively. With 8 mJ/cm2 laser fluence, PUT would yield vessel disruption (90%, p < 0.01). Selectivity of PUT was demonstrated by utilizing laser wavelengths at 578 nm or 650 nm, where PUT selectively shrank veins or occluded arteries. In a rabbit ear model, PUT induced a 68.5% reduction in blood perfusion after 7 days (p < 0.001) without damaging the surrounding cells. In vitro experiments in human blood suggested that cavitation may play a role in PUT. In conclusion, PUT holds significant promise as a novel non-invasive antivascular method with the capability to precisely target blood vessels.
Collapse
Affiliation(s)
- Zizhong Hu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA.,Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Haonan Zhang
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA.,Institute of Acoustics, Tongji University, Shanghai, P.R. China
| | - Aghapi Mordovanakis
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Yannis M Paulus
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Qinghuai Liu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Xueding Wang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Xinmai Yang
- Bioengineering Research Center and Department of Mechanical Engineering, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
33
|
Maijer KI, van der Leij C, de Hair MJH, Tas SW, Maas M, Gerlag DM, Tak PP, Lavini C. Dynamic Contrast-Enhanced Magnetic Resonance Imaging Using Pharmacokinetic Modeling: Initial Experience in Patients With Early Arthritis. Arthritis Rheumatol 2016; 68:587-96. [PMID: 26473331 DOI: 10.1002/art.39469] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 10/01/2015] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Analysis of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) using pharmacokinetic modeling (PKM) provides quantitative measures that mirror microvessel integrity and can be used as an objective marker of the level of synovial inflammation. The aim of this study was to investigate the PKM parameters K(trans) , kep , and ve in a prospective cohort of disease-modifying antirheumatic drug (DMARD)-naive patients with early arthritis, and to validate the results by assessing their correlation with the number of synovial endothelial cells (ECs). METHODS Forty-seven patients with early arthritis (arthritis duration <1 year, DMARD naive; comprising 14 patients with rheumatoid arthritis, 22 with unclassified arthritis, 6 with spondyloarthritis [SpA], and 5 with other arthritides) were included. At baseline, DCE-MRI was performed on an inflamed knee joint of each patient. These images were used to calculate the K(trans) (volume transfer constant between the plasma and extracellular extravascular space [EES]), the kep (transfer constant between the EES and plasma), and the ve (fractional volume of the EES). Second, markers of disease activity were collected. Finally, vascularity was evaluated by immunohistochemical analysis of synovial tissue samples obtained from the inflamed knee joints, using antibodies to detect von Willebrand factor (vWF), a marker of ECs. RESULTS The 3 PKM parameters differed significantly between diagnostic groups at baseline, with the highest K(trans) value being observed in patients with SpA (median 0.050/minute, interquartile range [IQR] 0.041- 0.069). Furthermore, the K(trans) , kep , and ve values correlated significantly with markers of disease activity. Finally, the PKM parameters K(trans) and kep , but not ve , correlated significantly with synovial expression of vWF (r = 0.647, P = 0.004 for K(trans) ; r = 0.614, P = 0.007 for kep ; r = 0.398, P = 0.102 for ve ). CONCLUSION These results suggest that the K(trans) , kep , and ve can be used to detect synovial inflammation in patients with early arthritis, and these PKM parameters may be helpful in differential diagnosis. This approach may also be useful in translational research analyzing tissue microcirculation and angiogenesis.
Collapse
Affiliation(s)
- Karen I Maijer
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Maria J H de Hair
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sander W Tas
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mario Maas
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Daniëlle M Gerlag
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul P Tak
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Cristina Lavini
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Whiteford JR, De Rossi G, Woodfin A. Mutually Supportive Mechanisms of Inflammation and Vascular Remodeling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:201-78. [PMID: 27572130 DOI: 10.1016/bs.ircmb.2016.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic inflammation is often accompanied by angiogenesis, the development of new blood vessels from existing ones. This vascular response is a response to chronic hypoxia and/or ischemia, but is also contributory to the progression of disorders including atherosclerosis, arthritis, and tumor growth. Proinflammatory and proangiogenic mediators and signaling pathways form a complex and interrelated network in these conditions, and many factors exert multiple effects. Inflammation drives angiogenesis by direct and indirect mechanisms, promoting endothelial proliferation, migration, and vessel sprouting, but also by mediating extracellular matrix remodeling and release of sequestered growth factors, and recruitment of proangiogenic leukocyte subsets. The role of inflammation in promoting angiogenesis is well documented, but by facilitating greater infiltration of leukocytes and plasma proteins into inflamed tissues, angiogenesis can also propagate chronic inflammation. This review examines the mutually supportive relationship between angiogenesis and inflammation, and considers how these interactions might be exploited to promote resolution of chronic inflammatory or angiogenic disorders.
Collapse
Affiliation(s)
- J R Whiteford
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - G De Rossi
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - A Woodfin
- Cardiovascular Division, King's College, University of London, London, United Kingdom.
| |
Collapse
|
35
|
CdGAP/ARHGAP31, a Cdc42/Rac1 GTPase regulator, is critical for vascular development and VEGF-mediated angiogenesis. Sci Rep 2016; 6:27485. [PMID: 27270835 PMCID: PMC4895392 DOI: 10.1038/srep27485] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023] Open
Abstract
Mutations in the CdGAP/ARHGAP31 gene, which encodes a GTPase-activating protein for Rac1 and Cdc42, have been reported causative in the Adams-Oliver developmental syndrome often associated with vascular defects. However, despite its abundant expression in endothelial cells, CdGAP function in the vasculature remains unknown. Here, we show that vascular development is impaired in CdGAP-deficient mouse embryos at E15.5. This is associated with superficial vessel defects and subcutaneous edema, resulting in 44% embryonic/perinatal lethality. VEGF-driven angiogenesis is defective in CdGAP(-/-) mice, showing reduced capillary sprouting from aortic ring explants. Similarly, VEGF-dependent endothelial cell migration and capillary formation are inhibited upon CdGAP knockdown. Mechanistically, CdGAP associates with VEGF receptor-2 and controls VEGF-dependent signaling. Consequently, CdGAP depletion results in impaired VEGF-mediated Rac1 activation and reduced phosphorylation of critical intracellular mediators including Gab1, Akt, PLCγ and SHP2. These findings are the first to demonstrate the importance of CdGAP in embryonic vascular development and VEGF-induced signaling, and highlight CdGAP as a potential therapeutic target to treat pathological angiogenesis and vascular dysfunction.
Collapse
|
36
|
Hypoxia, mitochondrial dysfunction and synovial invasiveness in rheumatoid arthritis. Nat Rev Rheumatol 2016; 12:385-97. [DOI: 10.1038/nrrheum.2016.69] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Utilization of nanoparticle technology in rheumatoid arthritis treatment. Biomed Pharmacother 2016; 80:30-41. [PMID: 27133037 DOI: 10.1016/j.biopha.2016.03.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/03/2016] [Accepted: 03/03/2016] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is one of the common and severe autoimmune diseases related to joints. This chronic autoimmune inflammatory disease, leads to functional limitation and reduced quality of life, since as there is bone and cartilage destruction, joint swelling and pain. Current advances and new treatment approaches have considerably postponed disease progression and improved the quality of life for many patients. In spite of major advances in therapeutic options, restrictions on the routes of administration and the necessity for frequent and long-term dosing often result in systemic adverse effects and patient non-compliance. Unlike usual drugs, nanoparticle systems are planned to deliver therapeutic agents especially to inflamed synovium, so avoiding systemic and unpleasant effects. The present review discusses about some of the most successful drugs in RA therapy and their side effects and also focuses on key design parameters of RA-targeted nanotechnology-based strategies for improving RA therapies.
Collapse
|
38
|
Su CM, Huang CY, Tang CH. Characteristics of resistin in rheumatoid arthritis angiogenesis. Biomark Med 2016; 10:651-60. [PMID: 26867862 DOI: 10.2217/bmm.15.125] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adipokines have been reported to be involved in the regulation of various physiological processes, including the immune response. Rheumatoid arthritis (RA) is an example of a systemic immune disease that causes chronic inflammation of the synovium and bone destruction in the joint. Recent therapeutic strategies based on the understanding of the role of cytokines and cellular mechanisms in RA have improved our understanding of angiogenesis. On the other hand, endogenous endothelial progenitor cells, which are a population isolated from peripheral blood monocytes have recently been identified as a homing target for pro-angiogeneic factor and vessel formation. In this review, we summarize the effects of common adipokines, such as adiponectin, leptin and resistin in RA pathogenesis and discuss other potential mechanisms of relevance for the therapeutic treatment of RA.
Collapse
Affiliation(s)
- Chen-Ming Su
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China.,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chun-Yin Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
39
|
Macrophage Migration Inhibitory Factor Secretion Is Induced by Ionizing Radiation and Oxidative Stress in Cancer Cells. PLoS One 2016; 11:e0146482. [PMID: 26741693 PMCID: PMC4704778 DOI: 10.1371/journal.pone.0146482] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 12/17/2015] [Indexed: 12/22/2022] Open
Abstract
The macrophage migration inhibitory factor (MIF) has been increasingly implicated in cancer development and progression by promoting inflammation, angiogenesis, tumor cell survival and immune suppression. MIF is overexpressed in a variety of solid tumor types in part due to its responsiveness to hypoxia inducible factor (HIF) driven transcriptional activation. MIF secretion, however, is a poorly understood process owing to the fact that MIF is a leaderless polypeptide that follows a non-classical secretory pathway. Better understanding of MIF processing and release could have therapeutic implications. Here, we have discovered that ionizing radiation (IR) and other DNA damaging stresses can induce robust MIF secretion in several cancer cell lines. MIF secretion by IR appears independent of ABCA1, a cholesterol efflux pump that has been implicated previously in MIF secretion. However, MIF secretion is robustly induced by oxidative stress. Importantly, MIF secretion can be observed both in cell culture models as well as in tumors in mice in vivo. Rapid depletion of MIF from tumor cells observed immunohistochemically is coincident with elevated circulating MIF detected in the blood sera of irradiated mice. Given the robust tumor promoting activities of MIF, our results suggest that an innate host response to genotoxic stress may mitigate the beneficial effects of cancer therapy, and that MIF inhibition may improve therapeutic responses.
Collapse
|
40
|
Sánchez-Zamora YI, Juarez-Avelar I, Vazquez-Mendoza A, Hiriart M, Rodriguez-Sosa M. Altered Macrophage and Dendritic Cell Response in Mif-/- Mice Reveals a Role of Mif for Inflammatory-Th1 Response in Type 1 Diabetes. J Diabetes Res 2016; 2016:7053963. [PMID: 27699180 PMCID: PMC5028830 DOI: 10.1155/2016/7053963] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/10/2016] [Indexed: 12/13/2022] Open
Abstract
Macrophage migration inhibitory factor (Mif) is highly expressed in type 1 diabetes mellitus (T1DM). However, there is limited information about how Mif influences the activation of macrophages (Mφ) and dendritic cells (DC) in T1DM. To address this issue, we induced T1DM by administering multiple low doses of streptozotocin (STZ) to Mif-/- or wild-type (Wt) BALB/c mice. We found that Mif-/- mice treated with STZ (Mif-/-STZ) developed lower levels of hyperglycemia, inflammatory cytokines, and specific pancreatic islet antigen- (PIAg-) IgG and displayed reduced cellular infiltration into the pancreatic islets compared to Wt mice treated with STZ (WtSTZ). Moreover, Mφ and DC from Mif-/-STZ displayed lower expression of MHC-II, costimulatory molecules CD80, CD86, and CD40, Toll-like receptor- (TLR-) 2, and TLR-4 than WtSTZ. These changes were associated with a reduced capacity of Mφ and DC from Mif-/-STZ to induce proliferation in ovalbumin-specific T cells. All the deficiencies observed in Mif-/-STZ were recovered by exogenous administration of recombinant Mif. These findings suggest that Mif plays a role in the molecular mechanisms of Mφ and DC activation and drives T cell responses involved in the pathology of T1DM. Therefore, Mif is a potential therapeutic target to reduce the pathology of T1DM.
Collapse
Affiliation(s)
- Yuriko Itzel Sánchez-Zamora
- Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), 54090 Tlalnepantla, MEX, Mexico
| | - Imelda Juarez-Avelar
- Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), 54090 Tlalnepantla, MEX, Mexico
| | | | - Marcia Hiriart
- Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, UNAM, 04510 Coyoacán, MEX, Mexico
| | - Miriam Rodriguez-Sosa
- Unidad de Biomedicina, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), 54090 Tlalnepantla, MEX, Mexico
- *Miriam Rodriguez-Sosa:
| |
Collapse
|
41
|
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
|
42
|
Deng Q, Bai S, Gao W, Tong L. Pristimerin inhibits angiogenesis in adjuvant-induced arthritic rats by suppressing VEGFR2 signaling pathways. Int Immunopharmacol 2015; 29:302-313. [PMID: 26548348 DOI: 10.1016/j.intimp.2015.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 10/24/2015] [Accepted: 11/02/2015] [Indexed: 12/25/2022]
Abstract
Rheumatoid arthritis (RA) is a progressive, inflammatory autoimmune disease. As RA progresses, the hyperplastic synovial pannus creates a hypoxic, inflammatory environment that induces angiogenesis. Further vascularization of the synovial tissue promotes pannus growth and continued infiltration of inflammatory leukocytes, thus perpetuating the disease. Pristimerin inhibits inflammation and tumor angiogenesis. The present study focused on the inhibition of angiogenesis by Pristimerin in adjuvant-induced arthritic rats and the underlying molecular mechanisms. Our results clearly demonstrate for the first time that Pristimerin significantly reduces vessel density in synovial membrane tissues of inflamed joints and reduces the expression of pro-angiogenic factors in sera, including TNF-α, Ang-1, and MMP-9. Pristimerin also decreased the expression of VEGF and p-VEGFR2 in the synovial membrane, whereas the total amount of VEGFR2 remained unchanged. Pristimerin suppressed the sprouting vessels of the aortic ring and inhibited VEGF-induced HFLS-RA migration in vitro. Pristimerin also inhibited VEGF-induced proliferation, migration and tube formation by HUVECs, blocked the autophosphorylation of VEGF-induced VEGFR2 and consequently downregulated the signaling pathways of activated PI3K, AKT, mTOR, ERK1/2, JNK, and p38 in VEGF-induced HUVECs. Our results indicate that Pristimerin suppressed synovial angiogenesis in our rat model and in vitro by interrupting the targeting of VEGFR2 activation. Therefore, Pristimerin has potential as an angiogenesis inhibitor in the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Qiudi Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Shutong Bai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Wanjiao Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Li Tong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
43
|
Ferrari M, Onuoha SC, Pitzalis C. Going with the flow: harnessing the power of the vasculature for targeted therapy in rheumatoid arthritis. Drug Discov Today 2015; 21:172-179. [PMID: 26523772 DOI: 10.1016/j.drudis.2015.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/30/2015] [Accepted: 10/16/2015] [Indexed: 12/17/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic, autoimmune disease that leads to excessive joint inflammation and is associated with significant morbidity and mortality. Although much is still to be learned about the aetiology RA, a growing body of evidence suggests that an altered vascular environment is an important aspect of its pathophysiology. In this context, RA shares many similarities with cancer, and it is expected that several angiogenic targets in cancer might be relevant to the treatment of RA. Here, we discuss how these targets can be combined with advances in drug development to generate the next generation of RA therapeutics.
Collapse
Affiliation(s)
- Mathieu Ferrari
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Shimobi C Onuoha
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
44
|
Hu K, Tang X, Tang G, Yao S, Yao B, Wang H, Nie D, Liang X, Tang C, He S. 18F-FP-PEG2-β-Glu-RGD2: A Symmetric Integrin αvβ3-Targeting Radiotracer for Tumor PET Imaging. PLoS One 2015; 10:e0138675. [PMID: 26397833 PMCID: PMC4580323 DOI: 10.1371/journal.pone.0138675] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 09/02/2015] [Indexed: 12/14/2022] Open
Abstract
Radiolabeled cyclic arginine-glycine-aspartic (RGD) peptides can be used for noninvasive determination of integrin αvβ3 expression in tumors. In this study, we performed radiosynthesis and biological evaluation of a new 18F-labeled RGD homodimeric peptide with one 8-amino-3,6-dioxaoctanoic acid (PEG2) linker on the glutamate β-amino group (18F-FP-PEG2-β-Glu-RGD2) as a symmetric PET tracer for tumor imaging. Biodistribution studies showed that radioactivity of 18F-FP-PEG2-β-Glu-RGD2 was rapidly cleared from blood by predominately renal excretion. MicroPET-CT imaging with 18F-FP-PEG2-β-Glu-RGD2 revealed high tumor contrast and low background in A549 human lung adenocarcinoma-bearing mouse models, PC-3 prostate cancer-bearing mouse models, and orthotopic transplanted C6 brain glioma models. 18F-FP-PEG2-β-Glu-RGD2 exhibited good stability in vitro and in vivo. The results suggest that this tracer is a potential PET tracer for tumor imaging.
Collapse
Affiliation(s)
- Kongzhen Hu
- Department of Nuclear Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiaolan Tang
- College of Materials and Energy, Southern China Agricultural University, Guangzhou, 510642, China
| | - Ganghua Tang
- Department of Nuclear Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
- * E-mail: (GT); (DN)
| | - Shaobo Yao
- Department of Nuclear Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Baoguo Yao
- Department of Nuclear Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hongliang Wang
- Department of Nuclear Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Dahong Nie
- Department of Nuclear Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
- * E-mail: (GT); (DN)
| | - Xiang Liang
- Department of Nuclear Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Caihua Tang
- Department of Nuclear Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Shanzhen He
- Department of Nuclear Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| |
Collapse
|
45
|
Maijer KI, Noort AR, de Hair MJH, van der Leij C, van Zoest KPM, Choi IY, Gerlag DM, Maas M, Tak PP, Tas SW. Nuclear Factor-κB-inducing Kinase Is Expressed in Synovial Endothelial Cells in Patients with Early Arthritis and Correlates with Markers of Inflammation: A Prospective Cohort Study. J Rheumatol 2015; 42:1573-81. [PMID: 26178280 DOI: 10.3899/jrheum.150245] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2015] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The nuclear factor-κB (NF-κB) family of transcription factors is strongly involved in synovial inflammation. We have previously shown that NF-κB-inducing kinase (NIK) is a key regulator of inflammation-induced angiogenesis in rheumatoid arthritis (RA) synovial tissue (ST). Here, we investigated synovial NIK expression in patients with early arthritis and in autoantibody-positive individuals at risk of developing RA. METHODS ST biopsies were obtained by arthroscopy from 154 patients with early arthritis (duration < 1 yr) with various diagnoses and 54 IgM rheumatoid factor-positive and/or anticitrullinated protein antibodies-positive individuals without evidence of arthritis. ST was stained for NIK and endothelial cell (EC) markers. Additionally, measures of disease activity were collected and contrast-enhanced magnetic resonance imaging (MRI) was performed in a subset of these patients. RESULTS In patients with early arthritis, NIK was predominantly expressed in EC of small blood vessels. Further, NIK expression correlated with erythrocyte sedimentation rate (r 0.184, p = 0.024), C-reactive protein (r 0.194, p = 0.017), joint swelling (r 0.297, p < 0.001), synovial immune cell markers (lining r 0.585, p < 0.001; sublining macrophages r 0.728, p < 0.001; T cells r 0.733, p < 0.001; and B cells r 0.264, p = 0.040), MRI effusion (r 0.665, p < 0.001), MRI synovitis (r 0.632, p < 0.001), and MRI total score (r 0.569, p < 0.001). In 18.5% of autoantibody-positive individuals, ST NIK(+)EC were present, but this was not predictive of the development of arthritis. CONCLUSION NIK(+)EC are present in the earliest phase of synovial inflammation and may be indicative of high angiogenic activity in the inflamed ST. Therefore, NIK(+)EC may play an important role in the persistence of synovitis. Collectively, our data underscore the importance of angiogenesis in synovial inflammation and identify NIK as a potential therapeutic target in arthritis.
Collapse
Affiliation(s)
- Karen I Maijer
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Ae Ri Noort
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Maria J H de Hair
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Christiaan van der Leij
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Katinka P M van Zoest
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Ivy Y Choi
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Daniëlle M Gerlag
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Mario Maas
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Paul P Tak
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam
| | - Sander W Tas
- From the Division of Clinical Immunology and Rheumatology, the Department of Experimental Immunology, and the Department of Radiology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands; GlaxoSmithKline, Stevenage; University of Cambridge, Cambridge, UK.K.I. Maijer, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; A.R. Noort, MSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; M.J. de Hair, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; C. van der Leij, MD, Department of Radiology, Academic Medical Center/University of Amsterdam; K.P. van Zoest, BSc, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam; I.Y. Choi, MD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam; D.M. Gerlag, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline; M. Maas, MD, PhD, Department of Radiology, Academic Medical Center/University of Amsterdam; P.P. Tak, MD, PhD, Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, and GlaxoSmithKline, Stevenage, and University of Cambridge; S.W. Tas, MD, PhD, Division of Clinical Immunology and Rheumatology, and Department of Experimental Immunology, Academic Medical Center/University of Amsterdam.
| |
Collapse
|
46
|
Angiogenesis in glaucoma filtration surgery and neovascular glaucoma: A review. Surv Ophthalmol 2015; 60:524-35. [PMID: 25980779 DOI: 10.1016/j.survophthal.2015.04.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 12/25/2022]
Abstract
Angiogenesis may pose a clinical challenge in glaucoma, for example, during the wound healing phase after glaucoma filtration surgery and in the severe secondary glaucoma called neovascular glaucoma (NVG). Upregulation of vascular endothelial growth factor (VEGF), a key mediator of angiogenesis, occurs in eyes that have undergone glaucoma filtration surgery, as well as those with NVG. This has led investigation of the ability of anti-vascular endothelial growth factor therapy to improve outcomes, and we examine the findings with respect to the safety and efficacy of anti-vascular endothelial growth factor agents, mainly bevacizumab and ranibizumab, in eyes that have undergone glaucoma filtration surgery or have NVG. Combining conventional therapies-such as antimetabolites after filtration surgery and panretinal photocoagulation in NVG-and anti-vascular endothelial growth factor drugs may produce a synergetic effect, although further studies are required to evaluate the long-term efficacy of combination treatments.
Collapse
|
47
|
Su CM, Hsu CJ, Tsai CH, Huang CY, Wang SW, Tang CH. Resistin Promotes Angiogenesis in Endothelial Progenitor Cells Through Inhibition of MicroRNA206: Potential Implications for Rheumatoid Arthritis. Stem Cells 2015; 33:2243-55. [PMID: 25828083 DOI: 10.1002/stem.2024] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 03/11/2015] [Indexed: 12/15/2022]
Abstract
Endothelial progenitor cells (EPCs) promote angiogenesis and are therefore key contributors to a wide variety of angiogenesis-related autoimmune diseases such as rheumatoid arthritis (RA). However, the signaling mechanisms through which these progenitor cells influence RA pathogenesis remain unknown. The aim of this study was to examine whether resistin plays a role in the pathogenesis of and angiogenesis associated with RA by circulating EPCs. We found that levels of resistin in synovial fluid and tissue from patients with RA and from mice with collagen-induced arthritis were overexpressed and promoted the homing of EPCs into the synovium, thereby inducing angiogenesis. EPCs isolated from healthy donors were used to investigate the signal transduction pathway underlying EPC migration and tube formation after treatment with resistin. We found that resistin directly induced a significant increase in expression of vascular endothelial growth factor (VEGF) in EPCs. We also found that the expression of microRNA-206 (miR-206) was negatively correlated with the expression of resistin during EPC-mediated angiogenesis. Notably, the increased expression of VEGF was associated with decreased binding of miR-206 to the VEGF-A 3' untranslated region through protein kinase C delta-dependent AMP-activated protein kinase signaling pathway. Moreover, blockade of resistin reduced EPC homing into synovial fluid and angiogenesis in vivo. Taken together, our study is the first to demonstrate that resistin promotes EPCs homing into the synovium during RA angiogenesis via a signal transduction pathway that involves VEGF expression in primary EPCs. These findings provide support for resistin as a therapeutic target for the patients with RA. Stem Cells 2015;33:2243-2255.
Collapse
Affiliation(s)
- Chen-Ming Su
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chin-Jung Hsu
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University, Taichung, Taiwan
| | - Chun-Yin Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
48
|
Abstract
The contact system, also named as plasma kallikrein-kinin system, consists of three serine proteinases: coagulation factors XII (FXII) and XI (FXI), and plasma prekallikrein (PK), and the nonenzymatic cofactor high molecular weight kininogen (HK). This system has been investigated actively for more than 50 years. The components of this system and their interactions have been elucidated from in vitro experiments, which indicates that this system is prothrombotic by activating intrinsic pathway, and proinflammatory by producing bioactive peptide bradykinin. Although the activation of the contact system have been implicated in various types of human disease, in only a few instances is its role clearly defined. In the last 10 years, our understanding of the contact system, particularly its biology and (patho)physiology has greatly increased through investigations using gene-modified animal models. In this review we will describe a revitalized view of the contact system as a critical (patho)physiologic mediator of coagulation and inflammation.
Collapse
Affiliation(s)
- Yi Wu
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3420 N. Broad Street, Philadelphia, PA 19140 USA
| |
Collapse
|
49
|
Serum Angiogenesis Markers and Their Correlation with Ultrasound-Detected Synovitis in Juvenile Idiopathic Arthritis. J Immunol Res 2015; 2015:741457. [PMID: 26065004 PMCID: PMC4434192 DOI: 10.1155/2015/741457] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 04/13/2015] [Indexed: 11/21/2022] Open
Abstract
Synovial angiogenesis is considered to be an important early step in the pathogenesis of juvenile idiopathic arthritis (JIA). In this study we assessed levels of angiogenic markers in serum or synovial fluid and their possible relevance to disease activity or degree of ultrasound signs of synovial inflammation and angiogenesis in early JIA. The concentration of vascular endothelial growth factor (VEGF), its soluble receptors 1 and 2 (sVEGF-R1, sVEGF-R2), and angiopoietins 1 and 2 (ANG-1, ANG-2) were evaluated in 43 JIA patients and 23 healthy controls. Synovial angiogenesis was assessed by means of Power-Doppler Ultrasonography (PDUS), according to the fourth-grade vascularity scale. VEGF and its receptors' (sVEGF-R1, sVEGF-R2) serum levels were significantly higher in JIA patients (p = 0.002). We found large variation in serum ANG-1 and ANG-2 levels. The PDUS imaging identified increased synovial microvascular blood flow in 15 (35.7%) examined JIA children. Intensity of joint vascularization correlated with higher serum VEGF and its levels was lowest in grade 0 and highest in grade 3 (p < 0.007 and p < 0.001, resp.). In conclusion, the high correlation between synovial microvascular blood flow, serum angiogenic proteins, and symptoms of synovitis may indicate its important role in pathogenesis of JIA.
Collapse
|
50
|
Araujo AP, Giorgio S. Immunohistochemical evidence of stress and inflammatory markers in mouse models of cutaneous leishmaniosis. Arch Dermatol Res 2015; 307:671-82. [PMID: 25896942 DOI: 10.1007/s00403-015-1564-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/06/2015] [Accepted: 04/11/2015] [Indexed: 01/20/2023]
Abstract
Leishmanioses are chronic parasitic diseases and host responses are associated with pro- or anti-inflammatory cytokines involved, respectively, in the control or exacerbation of infection. The relevance of other inflammatory mediators and stress markers has not been widely studied and there is a need to search for biomarkers to leishmaniasis. In this work, the stress and inflammatory molecules p38 mitogen-activated protein kinase, cyclooxygenase-2, migration inhibitory factor, macrophage inflammatory protein 2, heat shock protein 70 kDa, vascular endothelial factor (VEGF), hypoxia-inducible factors (HIF-1α and HIF-2α), heme oxygenase and galectin-3 expression were assessed immunohistochemically in self-controlled lesions in C57BL/6 mice and severe lesions in Balb/c mice infected with Leishmania amazonensis. The results indicated that the majority of molecules were expressed in the cutaneous lesions of both C57BL/6 and Balb/c mice during various phases of infection, suggesting no obvious correlation between the stress and inflammatory molecule expression and the control/exacerbation of leishmanial lesions. However, the cytokine VEGF was only detected in C57BL/6 footpad lesions and small lesions in Balb/c mice treated with antimonial pentavalent. These findings suggest that VEGF expression could be a predictive factor for murine leishmanial control, a hypothesis that should be tested in human leishmaniosis.
Collapse
Affiliation(s)
- Alexandra Paiva Araujo
- Department of Animal Biology, Biology Institute, Universidade Estadual de Campinas, Caixa Postal 6109, Campinas, São Paulo, 13083-970, Brazil
| | - Selma Giorgio
- Department of Animal Biology, Biology Institute, Universidade Estadual de Campinas, Caixa Postal 6109, Campinas, São Paulo, 13083-970, Brazil.
| |
Collapse
|