1
|
Baito QN, Jaafar HM, Mohammad TAM. Piperine suppresses inflammatory fibroblast-like synoviocytes derived from rheumatoid arthritis patients Via NF-κB inhibition. Cell Immunol 2023; 391-392:104752. [PMID: 37536000 DOI: 10.1016/j.cellimm.2023.104752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/20/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023]
Abstract
Rheumatoid Arthritis (RA) is a common autoimmune disease recognized by hyperplasia of synoviocytes and chronic joint inflammation. Activation of fibroblast-like synoviocytes (FLSs) is one of the main features of RA which can trigger inflammation leading to articular cartilage and joint destruction. Aberrant activation of NF-κB signaling cascade was found to be responsible for the high proliferation and defective apoptosis of FLSs and subsequent inflammation in RA. Piperine is a principal constituent of piper species frequently used as antitumor and anti-inflammatory natural compound. In this study we aimed to assess the anti-inflammatory effect of piperine on RA-FLS through NF-κB inhibition. FLSs were isolated from 68 RA patients and 30 healthy controls and were exposed to piperine. The main assays were MTT assay, flow cytometric analysis, PI staining, reverse transcription-PCR (RT-PCR), and ELISA. Results showed that piperine can induce the apoptosis and reduce the proliferation of RA-FLSs in vitro. Moreover, piperine directly reduced the phosphorylation of NF-kB and the expression of NF-κB target genes related to RA-FLSs proliferation (c-Myc and Cycline D1), apoptosis inhibition (Bcl2 and Bcl-xl) and inflammation (COX2, IL-1β, TNF-α,IL-6, CCL5 and CXCL10) while increasing the expression of apoptosis related ones (Bax) in vitro. Piperine also reduced the protein levels of cytokines and chemokines secreted by FLSs as a result of NF-κB inhibition. In conclusion, our results provide evidence for the anti-inflammatory capacity of piperine through inhibition of NF-κB pathway in FLSs proposing this compound as a suitable alternative for chemical treatment of RA.
Collapse
Affiliation(s)
- Qoyama Noel Baito
- Hawler Medical University, College of Pharmacy, Department of Clinical Pharmacy, Kurdistan Region-Erbil, Iraq
| | - Halmat M Jaafar
- Hawler Medical University, College of Pharmacy, Department of Clinical Pharmacy, Kurdistan Region-Erbil, Iraq
| | - Talar Ahmad Merza Mohammad
- Hawler Medical University, College of Pharmacy, Department of Clinical Pharmacy, Kurdistan Region-Erbil, Iraq.
| |
Collapse
|
2
|
Wang S, Zhou Y, Huang J, Li H, Pang H, Niu D, Li G, Wang F, Zhou Z, Liu Z. Advances in experimental models of rheumatoid arthritis. Eur J Immunol 2023; 53:e2249962. [PMID: 36330559 DOI: 10.1002/eji.202249962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/16/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by persistent articular inflammation and joint damage. RA was first described over 200 years ago; however, its etiology and pathophysiology remain insufficiently understood. The current treatment of RA is mainly empirical or based on the current understanding of etiology with limited efficacy and/or substantial side effects. Thus, the development of safer and more potent therapeutics, validated and optimized in experimental models, is urgently required. To improve the transition from bench to bedside, researchers must carefully select the appropriate experimental models as well as draw the right conclusions. Here, we summarize the establishment, pathological features, potential mechanisms, advantages, and limitations of the currently available RA models. The aim of the review is to help researchers better understand available RA models; discuss future trends in RA model development, which can help highlight new translational and human-based avenues in RA research.
Collapse
Affiliation(s)
- Siwei Wang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Yanhua Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Jiangrong Huang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huilin Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huidan Pang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Dandan Niu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Guangyao Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Fei Wang
- Department of Experiment and Training, Hubei College of Chinese Medicine, Hubei Province, China
| | - Zushan Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Zhenzhen Liu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
3
|
Zhao T, Xie Z, Xi Y, Liu L, Li Z, Qin D. How to Model Rheumatoid Arthritis in Animals: From Rodents to Non-Human Primates. Front Immunol 2022; 13:887460. [PMID: 35693791 PMCID: PMC9174425 DOI: 10.3389/fimmu.2022.887460] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/28/2022] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease influenced by both genetic and environmental factors. At present, rodent models are primarily used to study the pathogenesis and treatment of RA. However, the genetic divergences between rodents and humans determine differences in the development of RA, which makes it necessary to explore the establishment of new models. Compared to rodents, non-human primates (NHPs) are much more closely related to humans in terms of the immune system, metabolic conditions, and genetic make-up. NHPs model provides a powerful tool to study the development of RA and potential complications, as well as preclinical studies in drug development. This review provides a brief overview of the RA animal models, emphasizes the replication methods, pros and cons, as well as evaluates the validity of the rodent and NHPs models.
Collapse
Affiliation(s)
- Ting Zhao
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhaohu Xie
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Yujiang Xi
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Li Liu
- Ge Jiu People’s Hospital, Yunnan Honghe Prefecture Central Hospital, Gejiu, China
| | - Zhaofu Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
4
|
Aarntzen EHJG, Noriega-Álvarez E, Artiko V, Dias AH, Gheysens O, Glaudemans AWJM, Lauri C, Treglia G, van den Wyngaert T, van Leeuwen FWB, Terry SYA. EANM recommendations based on systematic analysis of small animal radionuclide imaging in inflammatory musculoskeletal diseases. EJNMMI Res 2021; 11:85. [PMID: 34487263 PMCID: PMC8421483 DOI: 10.1186/s13550-021-00820-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/02/2021] [Indexed: 11/26/2022] Open
Abstract
Inflammatory musculoskeletal diseases represent a group of chronic and disabling conditions that evolve from a complex interplay between genetic and environmental factors that cause perturbations in innate and adaptive immune responses. Understanding the pathogenesis of inflammatory musculoskeletal diseases is, to a large extent, derived from preclinical and basic research experiments. In vivo molecular imaging enables us to study molecular targets and to measure biochemical processes non-invasively and longitudinally, providing information on disease processes and potential therapeutic strategies, e.g. efficacy of novel therapeutic interventions, which is of complementary value next to ex vivo (post mortem) histopathological analysis and molecular assays. Remarkably, the large body of preclinical imaging studies in inflammatory musculoskeletal disease is in contrast with the limited reports on molecular imaging in clinical practice and clinical guidelines. Therefore, in this EANM-endorsed position paper, we performed a systematic review of the preclinical studies in inflammatory musculoskeletal diseases that involve radionuclide imaging, with a detailed description of the animal models used. From these reflections, we provide recommendations on what future studies in this field should encompass to facilitate a greater impact of radionuclide imaging techniques on the translation to clinical settings.
Collapse
Affiliation(s)
- Erik H J G Aarntzen
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Medical Imaging, Radboud University Nijmegen Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Edel Noriega-Álvarez
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Nuclear Medicine, General University Hospital of Ciudad Real, Ciudad Real, Spain
| | - Vera Artiko
- Inflammation and Infection Committee EANM, Vienna, Austria
- Center for Nuclear Medicine Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000, Belgrade, Serbia
| | - André H Dias
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Olivier Gheysens
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc and Institute of Clinical and Experimental Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Andor W J M Glaudemans
- Inflammation and Infection Committee EANM, Vienna, Austria.
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen Medical Imaging Center, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Chiara Lauri
- Inflammation and Infection Committee EANM, Vienna, Austria
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Giorgio Treglia
- Inflammation and Infection Committee EANM, Vienna, Austria
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Faculty of Biology and Medicine, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Tim van den Wyngaert
- Bone and Joint Committee EANM, Vienna, Austria
- Antwerp University Hospital Belgium, Edegem, Belgium
- Molecular Imaging Center Antwerp (MICA) - IPPON, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Fijs W B van Leeuwen
- Translational Molecular Imaging and Therapy Committee EANM, Vienna, Austria
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - Samantha Y A Terry
- Inflammation and Infection Committee EANM, Vienna, Austria.
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
5
|
Dhote V, Dangi U, Mandloi AS, Soni M, Shukla DN, Kawadkar M, Ganeshpurkar A. Preferential cyclooxygenase inhibition by Jasminum sambac: A possible relationship with potent anti-arthritic activity. J Tradit Complement Med 2021; 11:217-227. [PMID: 34012868 PMCID: PMC8116718 DOI: 10.1016/j.jtcme.2020.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 11/26/2022] Open
Abstract
Traditional claim validation. Molecular target identification. In-vitro estimation. In-vivo anti-arthritic activity.
Collapse
Affiliation(s)
- Vipin Dhote
- Faculty of Pharmacy, VNS Group of Institutions, Bhopal, India
| | - Uma Dangi
- Faculty of Pharmacy, VNS Group of Institutions, Bhopal, India
| | | | - Maneesh Soni
- Faculty of Pharmacy, VNS Group of Institutions, Bhopal, India
| | | | | | | |
Collapse
|
6
|
Dimitrijević M, Arsenović-Ranin N, Bufan B, Nacka-Aleksić M, Kosec D, Pilipović I, Kotur-Stevuljević J, Simić L, Sopta J, Leposavić G. Sex-Based Differences in Monocytic Lineage Cells Contribute to More Severe Collagen-Induced Arthritis in Female Rats Compared with Male Rats. Inflammation 2020; 43:2312-2331. [PMID: 32857321 DOI: 10.1007/s10753-020-01302-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Monocytes' plasticity has an important role in the development of rheumatoid arthritis (RA), an autoimmune disease exhibiting greater prevalence in women. Contribution of this phenomenon to sex bias in RA severity was investigated in rat collagen-induced arthritis (CIA) model of RA. The greater severity of CIA in females (exhibiting signs of bone resorption) was accompanied by the higher blood level of advanced oxidation protein products and a more pro-oxidant profile. Consistently, in females, the greater density of giant multinuclear cells (monocytes/macrophages and osteoclasts) in inflamed joint tissue was found. This correlated with the higher frequencies of CCR2- and CX3CR1- expressing cells (precursors of inflammatory monocytes/macrophages and osteoclasts) among CD11b+ splenocytes. This in conjunction with the enhanced migratory capacity of CD11b+ monocytic cells in females compared with males could be linked with the higher frequencies of CCR2+CX3CR1-CD43lowCD11b+ and CCR2-CX3CR1+CD43hiCD11b+ cells (corresponding to "classical" and "non-classical" monocytes, respectively) and the greater density of CD68+ cells (monocytes/macrophages and osteoclast precursors/osteoclasts) in blood and inflamed paws from female rats, respectively. Consistently, the higher levels of GM-CSF, TNF-α and IL-6, IL-1β (driving Th17 cell differentiation), and IL-17 followed by the lower level of IL-10 were measured in inflamed paw cultures from female compared with male rats. To the greater IL-17 production (associated with enhanced monocyte immigration and differentiation into osteoclasts) most likely contributed augmented Th17 cell generation in the lymph nodes draining arthritic joints from female compared with male rats. Overall, the study suggests the sex-specific contribution of monocytic lineage cells to CIA, and possibly RA development.
Collapse
Affiliation(s)
- Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research "Siniša Stanković" National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, Belgrade, Serbia.
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, Serbia
| | - Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, Serbia
| | - Mirjana Nacka-Aleksić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Center "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Vojvode Stepe 458, Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Center "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Vojvode Stepe 458, Belgrade, Serbia
| | - Jelena Kotur-Stevuljević
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, Serbia
| | - Ljubica Simić
- Department for Pathology, Faculty of Medicine, University of Belgrade, Dr Subotića 4/2, Belgrade, Serbia
| | - Jelena Sopta
- Department for Pathology, Faculty of Medicine, University of Belgrade, Dr Subotića 4/2, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, Belgrade, Serbia.
| |
Collapse
|
7
|
Ashrafizadeh M, Ahmadi Z, Mohammadinejad R, Ghasemipour Afshar E. Tangeretin: a mechanistic review of its pharmacological and therapeutic effects. J Basic Clin Physiol Pharmacol 2020; 31:/j/jbcpp.ahead-of-print/jbcpp-2019-0191/jbcpp-2019-0191.xml. [PMID: 32329752 DOI: 10.1515/jbcpp-2019-0191] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/07/2019] [Indexed: 06/11/2023]
Abstract
To date, a large number of synthetic drugs have been developed for the treatment and prevention of different disorders, such as neurodegenerative diseases, diabetes mellitus, and cancer. However, these drugs suffer from a variety of drawbacks including side effects and low efficacy. In response to this problem, researchers have focused on the plant-derived natural products due to their valuable biological activities and low side effects. Flavonoids consist of a wide range of naturally occurring compounds exclusively found in fruits and vegetables and demonstrate a number of pharmacological and therapeutic effects. Tangeretin (TGN) is a key member of flavonoids that is extensively found in citrus peels. It has different favorable biological activities such as antioxidant, anti-inflammatory, antitumor, hepatoprotective, and neuroprotective effects. In the present review, we discuss the various pharmacological and therapeutic effects of TGN and then, demonstrate how this naturally occurring compound affects signaling pathways to exert its impacts.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran, Phone: +989032360639
| | - Zahra Ahmadi
- Department of Basic Science, Faculty of Veterinary Medicine, Islamic Azad Branch, University of Shushtar, Khuzestan, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
8
|
Opoku YK, Liu Z, Liu H, Afrifa J, Koranteng H, Ren G, Li D. Fibroblast Growth Factor–21 Ameliorates Rheumatoid Arthritis by Maintaining Articular Integrity. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09872-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
9
|
Effects of the Cathepsin K Inhibitor ONO-5334 and Concomitant Use of ONO-5334 with Methotrexate on Collagen-Induced Arthritis in Cynomolgus Monkeys. Int J Rheumatol 2019; 2019:5710340. [PMID: 30906325 PMCID: PMC6397998 DOI: 10.1155/2019/5710340] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/13/2022] Open
Abstract
We examined whether the cathepsin K inhibitor, ONO-5334, administered alone or in combination with methotrexate (MTX), could ameliorate joint destruction evoked by collagen-induced arthritis (CIA) in female cynomolgus monkeys. CIA was induced by immunizing with bovine type II collagen. ONO-5334 (30 mg/kg/day) was orally administered once daily and MTX (10 mg/body/day) twice weekly for 9 weeks. X-ray (evaluation of joint destruction) and swelling (inflammatory) scores of proximal interphalangeal (PIP), distal interphalangeal (DIP), and metacarpophalangeal (MP) joints were evaluated. Urinary concentrations of C-terminal telopeptide of type I collagen (CTX-I) and type II collagen (CTX-II) were measured. Arthritis, accompanied by bone and cartilage destruction, was successfully induced in this collagen-induced arthritis monkey model. ONO-5334 showed no suppressive effect on joint swelling, while the joint swelling scores in the MTX and combination (ONO-5334 + MTX) groups were less than 50% compared with the control group. ONO-5334 decreased X-ray score by a mean of 64% (p<0.05 vs the control group), and MTX also decreased in X-ray score by a mean of 46% but with no statistical significance. Combination of ONO-5334 and MTX further decreased the X-ray score by 28% over MTX group (74% reduction vs the control group, p<0.01). Maximum increase in CTX-I (10-fold) and CTX-II (7-fold) compared to baseline was observed at 7 and 3 weeks after the first sensitization, respectively. After treatment with ONO-5334 alone or in combination with MTX, concentrations were maintained near baseline for both markers. In conclusion, ONO-5334 prevented joint destruction but not joint inflammation in this monkey CIA model. Concomitant use of ONO-5334 with MTX reduced architectural joint destruction compared to MTX alone; therefore, ONO-5334 may help to prevent joint destruction in combination with MTX for the treatment of rheumatoid arthritis.
Collapse
|
10
|
Acton PD. Multimodality Preclinical Imaging in Inflammatory Diseases. IMAGE FUSION IN PRECLINICAL APPLICATIONS 2019:135-160. [DOI: 10.1007/978-3-030-02973-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Yu D, Ye X, Che R, Wu Q, Qi J, Song L, Guo X, Zhang S, Wu H, Ren G, Li D. FGF21 exerts comparable pharmacological efficacy with Adalimumab in ameliorating collagen-induced rheumatoid arthritis by regulating systematic inflammatory response. Biomed Pharmacother 2017; 89:751-760. [PMID: 28273637 DOI: 10.1016/j.biopha.2017.02.059] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/09/2017] [Indexed: 12/12/2022] Open
Abstract
Previous studies have reported that Fibroblast growth factor 21 (FGF21) can regulate inflammation and may play an important role in inflammatory and immune-mediated diseases, such as autoimmune diseases. Adalimumab is one of the clinically effective anti-rheumatoid arthritis (RA) drugs. The aim of this study was to compare the therapeutic efficacy of FGF21 and Adalimumab on collagen-induced arthritis (CIA) model mice. Mice with CIA were subcutaneously treated with FGF21 or Adalimumab at dose of 1mgkg-1d-1, respectively. Our results showed that FGF21 significantly alleviated the severity of arthritis by reducing cellular immune responses and exerted the similar anti-inflammatory effects with Adalimumab in decreasing the mRNA and protein expression levels of IL-2, IL-6 and IL-17. However, the expression levels of IL-1β, RANKL and IL-10 in the mice treated with FGF21 were decreased 2.2-fold, 2.5-fold and increased 4.3-fold compared with Adalimumab, respectively. However, the levels of TNF-α in the mice treated with Adalimumab were lower than those in the mice treated with FGF21. Western blotting results demonstrated that FGF21 displayed equivalent effects with Adalimumab by inhibiting NF-κB/IκBα signaling pathway. However, FGF21 could also regulate systematic inflammatory response and the mechanism maybe related to other signal pathway. In summary, FGF21 exerts comparable pharmacological efficacy with Adalimumab by regulating systematic inflammatory response, providing that FGF21 may be a promising therapeutic agent for RA patients.
Collapse
Affiliation(s)
- Dan Yu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xianlong Ye
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Ruixiang Che
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China
| | - Qiang Wu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China
| | - Jianying Qi
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Liying Song
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China
| | - Xiaochen Guo
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China
| | - Shengqi Zhang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China
| | - Hongsong Wu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China
| | - Guiping Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China.
| | - Deshan Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
12
|
Bevaart L, Aalbers CJ, Vierboom MPM, Broekstra N, Kondova I, Breedveld E, Hauck B, Wright JF, Tak PP, Vervoordeldonk MJ. Safety, Biodistribution, and Efficacy of an AAV-5 Vector Encoding Human Interferon-Beta (ART-I02) Delivered via Intra-Articular Injection in Rhesus Monkeys with Collagen-Induced Arthritis. HUM GENE THER CL DEV 2016; 26:103-12. [PMID: 26086763 DOI: 10.1089/humc.2015.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Preclinical studies to assess biodistribution, safety, and initial efficacy of ART-I02, an adeno-associated type 5 (rAAV5) vector expressing human interferon β (hIFN-β), were performed in a total of 24 rhesus monkeys with collagen-induced arthritis. All monkeys were naïve or showed limited neutralizing antibody (Nab) titers to AAV5 at the start of the study. Animals were injected with a single intra-articular dose of ART-I02 or placebo, consisting of 3.2×10(13) vg (Dose A=maximum feasible dose), 4.58×10(12) vg (Dose B), or placebo in the first affected finger joint, the ipsilateral knee, and ankle joint at the same time point. Animals were monitored for clinical parameters and well-being with a maximum of 4 weeks, with the option that the severity of arthritis could necessitate an earlier time point of sacrifice. No adverse events were noted after injection of ART-I02. No abnormalities were observed after histological evaluation of all organs. At both dose levels, immunohistochemical staining indicated expression of hIFN-β. In animals injected with Dose A, we observed stabilization or a reduction in swelling in the finger joint in which vector was administered. The highest copy numbers of vector DNA were detected in synovial tissue of the injected joint and the draining lymph node of the injected knee. High titers of Nab to rAAV5 were observed at the end of the study. Five monkeys developed an rAAV5-specific T-cell response. Two monkeys developed Nab to hIFN-β. In conclusion, intra-articular injection of ART-I02 was well-tolerated and did not induce adverse events. After administration of Dose A of ART-I02, we observed a beneficial effect on joint swelling, substantiated by decreased histological inflammation and bone erosion scores. A GMP vector for clinical application has been manufactured and is currently being tested in GLP rodent studies, with the aim to move forward to a clinical trial.
Collapse
Affiliation(s)
| | - Caroline J Aalbers
- 1 Arthrogen B.V., Amsterdam 1105 BA, The Netherlands .,2 Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam , 1105 AZ, The Netherlands
| | - Michel P M Vierboom
- 3 Department of Immunobiology, Biomedical Primate Research Centre , Rijswijk 2288 GH, The Netherlands
| | | | - Ivanela Kondova
- 3 Department of Immunobiology, Biomedical Primate Research Centre , Rijswijk 2288 GH, The Netherlands
| | - Elia Breedveld
- 3 Department of Immunobiology, Biomedical Primate Research Centre , Rijswijk 2288 GH, The Netherlands
| | - Bernd Hauck
- 4 Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia , Philadelphia, PA 19104
| | - J Fraser Wright
- 4 Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia , Philadelphia, PA 19104
| | - Paul Peter Tak
- 1 Arthrogen B.V., Amsterdam 1105 BA, The Netherlands .,2 Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam , 1105 AZ, The Netherlands
| | - Margriet J Vervoordeldonk
- 1 Arthrogen B.V., Amsterdam 1105 BA, The Netherlands .,2 Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam , 1105 AZ, The Netherlands
| |
Collapse
|
13
|
Joint Degradation in a Monkey Model of Collagen-Induced Arthritis: Role of Cathepsin K Based on Biochemical Markers and Histological Evaluation. Int J Rheumatol 2016; 2016:8938916. [PMID: 26949397 PMCID: PMC4754492 DOI: 10.1155/2016/8938916] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 12/24/2015] [Accepted: 01/06/2016] [Indexed: 01/29/2023] Open
Abstract
The role of cathepsin K in joint degradation in a model of collagen-induced arthritis (CIA) in cynomolgus monkey was examined using biochemical markers and histology. Joint swelling, urinary C-telopeptide of type II collagen (CTX-II), deoxypyridinoline (DPD), and N- and C-telopeptides of type I collagen (NTX and CTX-I, resp.) were analyzed. Immunohistochemistry of type II collagen, cathepsin K, and CTX-II were performed using joints. Joint swelling reached peak on day 42 and continued at this level. The CTX-II level peaked on day 28 and declined thereafter, while CTX-I, NTX, and DPD reached plateau on day 43. Joint swelling was positively correlated with CTX-II increases on days 20 and 42/43, with increases in CTX-I and NTX/Cr on days 42/43 and 84, and with DPD increases throughout the study period. Intense cathepsin K staining was observed in osteoclasts and in articular cartilage and synovial tissue in arthritic joints. CTX-II was present in the superficial layer of articular cartilage in CIA monkeys. Evidence from biochemical markers suggests that matrix degradation in the CIA model starts with degradation of cartilage, rather than bone resorption. Cathepsin K expressed in osteoclasts, articular cartilage, and synovial tissue may contribute to degradation of cartilage.
Collapse
|
14
|
Van Roy M, Ververken C, Beirnaert E, Hoefman S, Kolkman J, Vierboom M, Breedveld E, 't Hart B, Poelmans S, Bontinck L, Hemeryck A, Jacobs S, Baumeister J, Ulrichts H. The preclinical pharmacology of the high affinity anti-IL-6R Nanobody® ALX-0061 supports its clinical development in rheumatoid arthritis. Arthritis Res Ther 2015; 17:135. [PMID: 25994180 PMCID: PMC4476083 DOI: 10.1186/s13075-015-0651-0] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/11/2015] [Indexed: 12/21/2022] Open
Abstract
Introduction The pleiotropic cytokine interleukin-6 (IL-6) plays an important role in the pathogenesis of different diseases, including rheumatoid arthritis (RA). ALX-0061 is a bispecific Nanobody® with a high affinity and potency for IL-6 receptor (IL-6R), combined with an extended half-life by targeting human serum albumin. We describe here the relevant aspects of its in vitro and in vivo pharmacology. Methods ALX-0061 is composed of an affinity-matured IL-6R-targeting domain fused to an albumin-binding domain representing a minimized two-domain structure. A panel of different in vitro assays was used to characterize the biological activities of ALX-0061. The pharmacological properties of ALX-0061 were examined in cynomolgus monkeys, using plasma levels of total soluble (s)IL-6R as pharmacodynamic marker. Therapeutic effect was evaluated in a human IL-6-induced acute phase response model in the same species, and in a collagen-induced arthritis (CIA) model in rhesus monkeys, using tocilizumab as positive control. Results ALX-0061 was designed to confer the desired pharmacological properties. A 200-fold increase of target affinity was obtained through affinity maturation of the parental domain. The high affinity for sIL-6R (0.19 pM) translated to a concentration-dependent and complete neutralization of sIL-6R in vitro. In cynomolgus monkeys, ALX-0061 showed a dose-dependent and complete inhibition of hIL-6-induced inflammatory parameters, including plasma levels of C-reactive protein (CRP), fibrinogen and platelets. An apparent plasma half-life of 6.6 days was observed after a single intravenous administration of 10 mg/kg ALX-0061 in cynomolgus monkeys, similar to the estimated expected half-life of serum albumin. ALX-0061 and tocilizumab demonstrated a marked decrease in serum CRP levels in a non-human primate CIA model. Clinical effect was confirmed in animals with active drug exposure throughout the study duration. Conclusions ALX-0061 represents a minimized bispecific biotherapeutic of 26 kDa, nearly six times smaller than monoclonal antibodies. High in vitro affinity and potency was demonstrated. Albumin binding as a half-life extension technology resulted in describable and expected pharmacokinetics. Strong IL-6R engagement was shown to translate to in vivo effect in non-human primates, demonstrated via biomarker deregulation as well as clinical effect. Presented results on preclinical pharmacological properties of ALX-0061 are supportive of clinical development in RA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0651-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Els Beirnaert
- Ablynx NV, Technologiepark 21, 9052, Zwijnaarde, Belgium. .,VIB, Rijvisschestraat 120, 9052, Zwijnaarde, Belgium.
| | - Sven Hoefman
- Ablynx NV, Technologiepark 21, 9052, Zwijnaarde, Belgium.
| | - Joost Kolkman
- Ablynx NV, Technologiepark 21, 9052, Zwijnaarde, Belgium. .,Crucell, Archimedesweg 4-6, 2333, CA, Leiden, The Netherlands.
| | - Michel Vierboom
- Department of Immunobiology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands.
| | - Elia Breedveld
- Department of Immunobiology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands.
| | - Bert 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands. .,Department of Neuroscience, University of Groningen, University Medical Center, Hanzeplein 1, 9700 RB, Groningen, The Netherlands.
| | - Sofie Poelmans
- Ablynx NV, Technologiepark 21, 9052, Zwijnaarde, Belgium.
| | | | - Alex Hemeryck
- Ablynx NV, Technologiepark 21, 9052, Zwijnaarde, Belgium.
| | - Sandy Jacobs
- Ablynx NV, Technologiepark 21, 9052, Zwijnaarde, Belgium.
| | | | - Hans Ulrichts
- Ablynx NV, Technologiepark 21, 9052, Zwijnaarde, Belgium.
| |
Collapse
|
15
|
Bevaart L, Aalbers CC, Vierboom M, Broekstra N, Kondova I, Breedveld E, Hauck B, Wright F, Tak PP, Vervoordeldonk MJ. SAFETY, BIODISTRIBUTION, AND EFFICACY OF AN AAV-5 VECTOR ENCODING HUMAN INTERFERON-BETA (ART-I02) DELIVERED VIA INTRA-ARTICULAR INJECTION IN RHESUS MONKEYS WITH COLLAGEN-INDUCED ARTHRITIS. HUM GENE THER CL DEV 2015. [DOI: 10.1089/hum.2015.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
16
|
Yu Y, Li S, Liu Y, Tian G, Yuan Q, Bai F, Wang W, Zhang Z, Ren G, Zhang Y, Li D. Fibroblast growth factor 21 (FGF21) ameliorates collagen-induced arthritis through modulating oxidative stress and suppressing nuclear factor-kappa B pathway. Int Immunopharmacol 2015; 25:74-82. [PMID: 25601498 DOI: 10.1016/j.intimp.2015.01.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 12/16/2014] [Accepted: 01/06/2015] [Indexed: 01/09/2023]
Abstract
It has been demonstrated that circulating FGF21 levels are elevated in the serum and synovial fluid of patients with rheumatoid arthritis (RA). The aim of this study is to investigate efficacy of FGF21 for treatment of RA and the molecular mechanisms of the therapeutic effect on collagen-induced arthritis (CIA). Mice with CIA were subcutaneously administered with FGF21 (5, 2 or 1mg·kg(-1)·d(-1)), IL-1β antibody (5mg·kg(-1)·d(-1)), IL-17A antibody (5mg·kg(-1)·d(-1)) and dexamethasone (DEX) (1mg·kg(-1)·d(-1)), respectively. The effects of treatment were determined by arthritis severity score, histological damage and cytokine production. The activation of NF-κB was analyzed by Western blotting. We also detected the levels of oxidative stress parameters. Our results showed that FGF21 had beneficial effects on clinical symptom and histological lesion of CIA mice. Similar to antibody and DEX, FGF21 treatment alleviated the severity of arthritis by reducing humoral and cellular immune responses and down-regulating the expression of pro-inflammatory cytokines. FGF21 treatment also reduced the expression of TNF-α, IL-1β, IL-6, IFN-γ and MMP-3 and increased level of IL-10 in the spleen tissue or the plasma of CIA mice in a dose-dependent manner. Furthermore, FGF21 inhibited IκBα degradation and NF-κB p65 nuclear translocation and induced significant changes of oxidative stress parameters (MDA, SOD, CAT, GSH-PX and GSH) in the plasma. FGF21 exerts therapeutic efficacy for RA through antioxidant reaction and inhibiting NF-κB inflammatory pathway. This study provides evidence that FGF21 may be a promising therapeutic agent for RA patients.
Collapse
Affiliation(s)
- Yinhang Yu
- Bio-pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin 150030, China
| | - Siming Li
- Bio-pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin 150030, China; Harbin University of Commerce, Harbin 150028, China
| | - Yaonan Liu
- Bio-pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin 150030, China
| | - Guiyou Tian
- Bio-pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin 150030, China
| | - Qingyan Yuan
- Bio-pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin 150030, China
| | - Fuliang Bai
- Bio-pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin 150030, China
| | - Wenfei Wang
- Bio-pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Harbin 150030, China
| | - Zhiyi Zhang
- Harbin Medical University, The First Affiliated Hospital, Nangang District, 150001 Harbin, China
| | - Guiping Ren
- Bio-pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Harbin 150030, China.
| | - Yu Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150030, China
| | - Deshan Li
- Bio-pharmaceutical Lab, Life Science College, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Harbin 150030, China.
| |
Collapse
|
17
|
Woo J, Vierboom MPM, Kwon H, Chao D, Ye S, Li J, Lin K, Tang I, Belmar NA, Hartman T, Breedveld E, Vexler V, 't Hart BA, Law DA, Starling GC. PDL241, a novel humanized monoclonal antibody, reveals CD319 as a therapeutic target for rheumatoid arthritis. Arthritis Res Ther 2014; 15:R207. [PMID: 24299175 PMCID: PMC3978732 DOI: 10.1186/ar4400] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 11/19/2013] [Indexed: 12/11/2022] Open
Abstract
Introduction Targeting the CD20 antigen has been a successful therapeutic intervention in the treatment of rheumatoid arthritis (RA). However, in some patients with an inadequate response to anti-CD20 therapy, a persistence of CD20- plasmablasts is noted. The strong expression of CD319 on CD20- plasmablast and plasma cell populations in RA synovium led to the investigation of the potential of CD319 as a therapeutic target. Methods PDL241, a novel humanized IgG1 monoclonal antibody (mAb) to CD319, was generated and examined for its ability to inhibit immunoglobulin production from plasmablasts and plasma cells generated from peripheral blood mononuclear cells (PBMC) in the presence and absence of RA synovial fibroblasts (RA-SF). The in vivo activity of PDL241 was determined in a human PBMC transfer into NOD scid IL-2 gamma chain knockout (NSG) mouse model. Finally, the ability of PDL241 to ameliorate experimental arthritis was evaluated in a collagen-induced arthritis (CIA) model in rhesus monkeys. Results PDL241 bound to plasmablasts and plasma cells but not naïve B cells. Consistent with the binding profile, PDL241 inhibited the production of IgM from in vitro PBMC cultures by the depletion of CD319+ plasmablasts and plasma cells but not B cells. The activity of PDL241 was dependent on an intact Fc portion of the IgG1 and mediated predominantly by natural killer cells. Inhibition of IgM production was also observed in the human PBMC transfer to NSG mouse model. Treatment of rhesus monkeys in a CIA model with PDL241 led to a significant inhibition of anti-collagen IgG and IgM antibodies. A beneficial effect on joint related parameters, including bone remodeling, histopathology, and joint swelling was also observed. Conclusions The activity of PDL241 in both in vitro and in vivo models highlights the potential of CD319 as a therapeutic target in RA.
Collapse
|
18
|
Vinson A, Prongay K, Ferguson B. The value of extended pedigrees for next-generation analysis of complex disease in the rhesus macaque. ILAR J 2014; 54:91-105. [PMID: 24174435 DOI: 10.1093/ilar/ilt041] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Complex diseases (e.g., cardiovascular disease and type 2 diabetes, among many others) pose the biggest threat to human health worldwide and are among the most challenging to investigate. Susceptibility to complex disease may be caused by multiple genetic variants (GVs) and their interaction, by environmental factors, and by interaction between GVs and environment, and large study cohorts with substantial analytical power are typically required to elucidate these individual contributions. Here, we discuss the advantages of both power and feasibility afforded by the use of extended pedigrees of rhesus macaques (Macaca mulatta) for genetic studies of complex human disease based on next-generation sequence data. We present these advantages in the context of previous research conducted in rhesus macaques for several representative complex diseases. We also describe a single, multigeneration pedigree of Indian-origin rhesus macaques and a sample biobank we have developed for genetic analysis of complex disease, including power of this pedigree to detect causal GVs using either genetic linkage or association methods in a variance decomposition approach. Finally, we summarize findings of significant heritability for a number of quantitative traits that demonstrate that genetic contributions to risk factors for complex disease can be detected and measured in this pedigree. We conclude that the development and application of an extended pedigree to analysis of complex disease traits in the rhesus macaque have shown promising early success and that genome-wide genetic and higher order -omics studies in this pedigree are likely to yield useful insights into the architecture of complex human disease.
Collapse
|
19
|
Umar S, Umar K, Sarwar AHMG, Khan A, Ahmad N, Ahmad S, Katiyar CK, Husain SA, Khan HA. Boswellia serrata extract attenuates inflammatory mediators and oxidative stress in collagen induced arthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:847-856. [PMID: 24667331 DOI: 10.1016/j.phymed.2014.02.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 12/11/2013] [Accepted: 02/13/2014] [Indexed: 06/03/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease which leads to destruction of joints. Current treatment modalities for RA either produce symptomatic relief (NSAIDs) or modify the disease process (DMARDs). Though effective, their use is also limited by their side effects. As a result, the interest in alternative, well tolerated anti-inflammatory remedies has re-emerged. Our aim was to evaluate the antioxidant and antiarthritic activity of Boswellia serrata gum resin extract (BSE) in collagen induced arthritis. Arthritis was induced in male Wistar rats by collagen induced arthritis (CIA) method. BSE was administered at doses of 100 and 200mg/kg body weight once daily for 21 days. The effects of treatment in the rats were assessed by biochemical (articular elastase, MPO, LPO, GSH, catalase, SOD and NO), inflammatory mediators (IL-1β, IL-6, TNF-α, IL-10, IFN-γ and PGE2), and histological studies in joints. BSE was effective in bringing significant changes on all the parameters (articular elastase, MPO, LPO, GSH, catalase, SOD and NO) studied. Oral administration of BSE resulted in significantly reduced levels of inflammatory mediators (IL-1β, IL-6, TNF-α, IFN-γ and PGE2), and increased level of IL-10. The protective effects of BSE against RA were also evident from the decrease in arthritis scoring and bone histology. The abilities to inhibit proinflammatory cytokines and modulation of antioxidant status suggest that the protective effect of Boswellia serrata extract on arthritis in rats might be mediated via the modulation of immune system.
Collapse
Affiliation(s)
- Sadiq Umar
- Clinical Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India; Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India.
| | - Khalid Umar
- Department of Chemistry, Aligarh Muslim University, Aligarh, U.P. 200202, India
| | | | - Altaf Khan
- Research Centre, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Niyaz Ahmad
- Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Sayeed Ahmad
- Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | | | - Syed Akhtar Husain
- Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India
| | - Haider A Khan
- Clinical Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India.
| |
Collapse
|
20
|
Lee J, Kim Y, Yi H, Diecke S, Kim J, Jung H, Rim YA, Jung SM, Kim M, Kim YG, Park SH, Kim HY, Ju JH. Generation of disease-specific induced pluripotent stem cells from patients with rheumatoid arthritis and osteoarthritis. Arthritis Res Ther 2014; 16:R41. [PMID: 24490617 PMCID: PMC3978583 DOI: 10.1186/ar4470] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 01/15/2014] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Since the concept of reprogramming mature somatic cells to generate induced pluripotent stem cells (iPSCs) was demonstrated in 2006, iPSCs have become a potential substitute for embryonic stem cells (ESCs) given their pluripotency and "stemness" characteristics, which resemble those of ESCs. We investigated to reprogram fibroblast-like synoviocytes (FLSs) from patients with rheumatoid arthritis (RA) and osteoarthritis (OA) to generate iPSCs using a 4-in-1 lentiviral vector system. METHODS A 4-in-1 lentiviral vector containing Oct4, Sox2, Klf4, and c-Myc was transduced into RA and OA FLSs isolated from the synovia of two RA patients and two OA patients. Immunohistochemical staining and real-time PCR studies were performed to demonstrate the pluripotency of iPSCs. Chromosomal abnormalities were determined based on the karyotype. SCID-beige mice were injected with iPSCs and sacrificed to test for teratoma formation. RESULTS After 14 days of transduction using the 4-in-1 lentiviral vector, RA FLSs and OA FLSs were transformed into spherical shapes that resembled embryonic stem cell colonies. Colonies were picked and cultivated on matrigel plates to produce iPSC lines. Real-time PCR of RA and OA iPSCs detected positive markers of pluripotency. Immunohistochemical staining tests with Nanog, Oct4, Sox2, Tra-1-80, Tra-1-60, and SSEA-4 were also positive. Teratomas that comprised three compartments of ectoderm, mesoderm, and endoderm were formed at the injection sites of iPSCs. Established iPSCs were shown to be compatible by karyotyping. Finally, we confirmed that the patient-derived iPSCs were able to differentiate into osteoblast, which was shown by an osteoimage mineralization assay. CONCLUSION FLSs derived from RA and OA could be cell resources for iPSC reprogramming. Disease- and patient-specific iPSCs have the potential to be applied in clinical settings as source materials for molecular diagnosis and regenerative therapy.
Collapse
|
21
|
Jagessar SA, Vierboom M, Blezer ELA, Bauer J, Hart BA', Kap YS. Overview of models, methods, and reagents developed for translational autoimmunity research in the common marmoset (Callithrix jacchus). Exp Anim 2014; 62:159-71. [PMID: 23903050 PMCID: PMC4160941 DOI: 10.1538/expanim.62.159] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The common marmoset (Callithrix jacchus) is a small-bodied Neotropical
primate and a useful preclinical animal model for translational research into
autoimmune-mediated inflammatory diseases (AIMID), such as rheumatoid arthritis (RA) and
multiple sclerosis (MS). The animal model for MS established in marmosets has proven their
value for exploratory research into (etio) pathogenic mechanisms and for the evaluation of
new therapies that cannot be tested in lower species because of their specificity for
humans. Effective usage of the marmoset in preclinical immunological research has been
hampered by the limited availability of blood for immunological studies and of reagents
for profiling of cellular and humoral immune reactions. In this paper, we give a concise
overview of the procedures and reagents that were developed over the years in our
laboratory in marmoset models of the above-mentioned diseases.
Collapse
Affiliation(s)
- S Anwar Jagessar
- Department of Immunobiology, Biomedical Primate Research Centre, P.O. Box 3306, 2280 GH Rijswijk, The Netherlands.
| | | | | | | | | | | |
Collapse
|
22
|
Piperine ameliorates oxidative stress, inflammation and histological outcome in collagen induced arthritis. Cell Immunol 2013; 284:51-9. [PMID: 23921080 DOI: 10.1016/j.cellimm.2013.07.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/03/2013] [Accepted: 07/09/2013] [Indexed: 01/18/2023]
|
23
|
Thiberville SD, Moyen N, Dupuis-Maguiraga L, Nougairede A, Gould EA, Roques P, de Lamballerie X. Chikungunya fever: epidemiology, clinical syndrome, pathogenesis and therapy. Antiviral Res 2013; 99:345-70. [PMID: 23811281 PMCID: PMC7114207 DOI: 10.1016/j.antiviral.2013.06.009] [Citation(s) in RCA: 322] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 05/21/2013] [Accepted: 06/18/2013] [Indexed: 12/11/2022]
Abstract
Chikungunya fever is caused by a mosquito-borne alphavirus originating in East Africa. During the past 7 years, the disease has spread to islands of the Indian Ocean, Asia and Europe. Its spread has been facilitated by a mutation favouring replication in the mosquito Ae. albopictus. No vaccines or antiviral drugs are available to prevent or treat chikungunya fever. This paper provides an extensive review of the virus and disease, including Supplementary Tables.
Chikungunya virus (CHIKV) is the aetiological agent of the mosquito-borne disease chikungunya fever, a debilitating arthritic disease that, during the past 7 years, has caused immeasurable morbidity and some mortality in humans, including newborn babies, following its emergence and dispersal out of Africa to the Indian Ocean islands and Asia. Since the first reports of its existence in Africa in the 1950s, more than 1500 scientific publications on the different aspects of the disease and its causative agent have been produced. Analysis of these publications shows that, following a number of studies in the 1960s and 1970s, and in the absence of autochthonous cases in developed countries, the interest of the scientific community remained low. However, in 2005 chikungunya fever unexpectedly re-emerged in the form of devastating epidemics in and around the Indian Ocean. These outbreaks were associated with mutations in the viral genome that facilitated the replication of the virus in Aedes albopictus mosquitoes. Since then, nearly 1000 publications on chikungunya fever have been referenced in the PubMed database. This article provides a comprehensive review of chikungunya fever and CHIKV, including clinical data, epidemiological reports, therapeutic aspects and data relating to animal models for in vivo laboratory studies. It includes Supplementary Tables of all WHO outbreak bulletins, ProMED Mail alerts, viral sequences available on GenBank, and PubMed reports of clinical cases and seroprevalence studies.
Collapse
Affiliation(s)
- Simon-Djamel Thiberville
- UMR_D 190 "Emergence des Pathologies Virales" (Aix-Marseille Univ. IRD French Institute of Research for Development EHESP French School of Public Health), Marseille, France; University Hospital Institute for Infectious Disease and Tropical Medicine, Marseille, France.
| | | | | | | | | | | | | |
Collapse
|
24
|
Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 2013; 11:1087-101. [PMID: 23151166 DOI: 10.1586/erv.12.84] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In 2004, chikungunya virus (CHIKV) re-emerged from East Africa to cause devastating epidemics of debilitating and often chronic arthralgia that have affected millions of people in the Indian Ocean Basin and Asia. More limited epidemics initiated by travelers subsequently occurred in Italy and France, as well as human cases exported to most regions of the world, including the Americas where CHIKV could become endemic. Because CHIKV circulates during epidemics in an urban mosquito-human cycle, control of transmission relies on mosquito abatement, which is rarely effective. Furthermore, there is no antiviral treatment for CHIKV infection and no licensed vaccine to prevent disease. Here, we discuss the challenges to the development of a safe, effective and affordable chikungunya vaccine and recent progress toward this goal.
Collapse
Affiliation(s)
- Scott C Weaver
- Institute for Human Infections and Immunity, Sealy Center for Vaccine Development and Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
| | | | | | | | | |
Collapse
|
25
|
Vierboom M, Breedveld E, Kondova I, 't Hart BA. The significance of non-human primates as preclinical models of human arthritic disease. Expert Opin Drug Discov 2013; 3:299-310. [PMID: 23480265 DOI: 10.1517/17460441.3.3.299] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The broad immunological gap between inbred SPF-raised strains of mice and rats and the diverse rheumatoid arthritis (RA) patient population limits the predictive value of the existing disease models for clinical success of new therapies, in particular for those using highly specific biologicals. OBJECTIVE This review argues that because of their closer immunological and physiological proximity to patients, disease models in non-human primates (NHPs) may bridge this gap and help reduce the failure of many (± 80%) new therapies in clinical trials. In various research areas, NHPs are an accepted intermediate between disease models in rodents and the ultimate introduction for clinical use in patients. However, with the exception of transplantation, this is not the case for immune-mediated inflammatory disorders, such as RA, although useful preclinical models are being developed. METHOD The validity and use of the rhesus monkey model of collagen-induced arthritis as a preclinical RA model is reviewed. The discussion comprises present genetic and immunological aspects, biomarkers, and an overview of published preclinical therapy evaluations. CONCLUSION It is time to consider the use of NHPs with a greater evolutionary proximity to humans as models for preclinical evaluation of new human-specific drugs for arthritic disease.
Collapse
Affiliation(s)
- Michel Vierboom
- Biomedical Primate Research Centre, Department of Immunobiology, PO Box 3306, 2280 GH Rijswijk, The Netherlands +31 15 284 2500 ; +31 15 284 2600 ;
| | | | | | | |
Collapse
|
26
|
Tao Z, Yang H, Jia D, Wan L, Cheng J, Lu X. Molecular cloning, recombinant expression and characterization of GMCSF from the rhesus monkey, Macaca mulatta. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 40:69-77. [PMID: 23352624 DOI: 10.1016/j.dci.2013.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 01/07/2013] [Accepted: 01/07/2013] [Indexed: 02/05/2023]
Abstract
Recent studies have found that, in addition to hematopoiesis, granulocyte-macrophage colony-stimulating factor (GMCSF) plays pivotal roles in multiple immune disorders. The gene encoding Macaca mulatta GMCSF (mmGMCSF) was cloned from stimulated peripheral blood mononuclear cells (PBMCs). Concanavalin A (Con A) and mismatched allogeneic antigen-stimulation significantly increased the production of mmGMCSF by monkey PBMCs. The gene encoding mature mmGMCSF was first expressed as a soluble fusion protein in Escherichia coli, and native mmGMCSF was further prepared by protease cleavage. The recombinant mmGMCSF induced antigen-presenting dendritic cells from monkey PBMCs, suggesting a central role of mmGMCSF in the immune system of the rhesus monkey. Although the predicted mature mmGMCSF protein differs from human GMCSF (hGMCSF) at six amino acid residues, mmGMCSF showed a strong ability to support human TF-1 cell survival. Additional co-immunoprecipitation experiments revealed that mmGMCSF cross reacts with the hGMCSF receptor (hGMCSFR). In addition, the hGMCSF-neutralizing agents hGMCSFR-Fc and anti-hGMCSF antibody reduced the biological effects of mmGMCSF on TF-1 cells. These results suggest that recombinant mmGMCSF might be used for the in vitro evaluation of novel hGMCSF-neutralizing agents prior to the in vivo preclinical evaluation of these agents in the rhesus monkey model.
Collapse
Affiliation(s)
- Ze Tao
- Key Lab of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | |
Collapse
|
27
|
Jonker M, Wubben J, Haanstra K, Vierboom M, 't Hart B. Comparative analysis of inflammatory infiltrates in collagen-induced arthritis, kidney graft rejection and delayed-type hypersensitivity in non-human primates. Inflamm Res 2012; 62:181-94. [PMID: 23064655 DOI: 10.1007/s00011-012-0564-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/15/2012] [Accepted: 10/03/2012] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Non-human primates are immunologically closely related to humans providing relevant models of inflammatory disorders often used to evaluate new immunomodulating therapies. The aim of the study was to compare inflammatory infiltrates of acute graft rejection (AR) and collagen-induced arthritis (CIA) to delayed-type hypersensitivity (DTH) reactions as the latter model may serve as a less invasive animal model. MATERIALS AND METHODS Tissue samples of AR, CIA and DTH were obtained from rhesus monkeys used in several pre-clinical studies. The infiltrate composition was determined by immunohistochemical analysis. RESULTS The infiltrates in AR consisted of T cells, macrophages and B cells. The presence of lymphoid structures in AR suggested ongoing intragraft immune activation. The synovia of CIA contained predominantly macrophages and few T cells. The DTH infiltrates were dominated by T cells when the challenged was ovalbumin (OVA) and by macrophages when the challenge was tetanus toxoid (TT). CONCLUSIONS The histology of AR resembles aspects of DTH to OVA while that of CIA showed similarities of the DTH to TT. The DTH reaction could serve as a model to study immunomodulating drugs for acute rejection and the acute inflammatory phase of autoimmunity.
Collapse
Affiliation(s)
- Margreet Jonker
- Biomedical Primate Research Centre, PO BOX 3306, 2280 GH Rijswijk, Netherlands.
| | | | | | | | | |
Collapse
|
28
|
Umar S, Zargan J, Umar K, Ahmad S, Katiyar CK, Khan HA. Modulation of the oxidative stress and inflammatory cytokine response by thymoquinone in the collagen induced arthritis in Wistar rats. Chem Biol Interact 2012; 197:40-6. [PMID: 22450443 DOI: 10.1016/j.cbi.2012.03.003] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 02/22/2012] [Accepted: 03/08/2012] [Indexed: 10/28/2022]
Abstract
Thymoquinone (TQ) is the major active compound derived from Nigella sativa. Our aim of this work was to evaluate the antioxidant and antiarthritic activity of TQ in Wistar rat by collagen induced arthritis (CIA). TQ was administered at a dose of 5mgkg(-1) body weight once daily for 21days. The effects of treatment in the rats were assessed by biochemical (articular elastase, MPO, LPO, GSH, catalase, SOD and NO), inflammatory mediators (IL-1β, IL-6, TNF-α, IL-10, IFN-γ and PGE(2)) and histological studies in joints. TQ was effective in bringing significant changes on all the parameters (articular elastase, MPO, LPO, GSH, catalase, SOD and NO) studied. Oral administration of TQ resulted in significantly reduced the levels of pro-inflammatory mediators (IL-1β, IL-6, TNF-α, IFN-γ and PGE(2)) and increased level of IL-10. The protective effects of TQ against RA were also evident from the decrease in arthritis scoring and bone histology. In conclusion, the fact that TQ abolished a number of factors known to be involved in RA pathogenesis indicates that the administration of thymoquinone may have potential value in the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Sadiq Umar
- Clinical Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | | | | | | | | | | |
Collapse
|
29
|
Vierboom M, Breedveld E, 't Hart BA. New drug discovery strategies for rheumatoid arthritis: a niche for nonhuman primate models to address systemic complications in inflammatory arthritis. Expert Opin Drug Discov 2012; 7:315-25. [PMID: 22458503 DOI: 10.1517/17460441.2012.666523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Despite the tremendous advances made in the treatment of rheumatoid arthritis (RA), there is still excess mortality observed in RA patients, which is mainly caused by cardiovascular disease (CVD). Altered lipid metabolism plays a major role in the etiology of CVD. A second common complication observed in RA patients is anemia. Both conditions are serious, reduce quality of life and are undertreated. AREAS COVERED The authors postulate that there is a specific niche for nonhuman primate models of inflammatory arthritis to address these systemic complications that occur in RA. Furthermore, the authors postulate that these nonhuman primate models are a useful platform to unveil the mechanisms underlying dyslipidemia and anemia, which are responsible for the manifestation of these complications. EXPERT OPINION The presence of currently untreated systemic complications of RA, such as dyslipidemia and anemia, provides interesting opportunities to include these in the preclinical evaluation of new therapies. In the selection of relevant models for the evaluation of new treatments for RA or the identification of new targets for therapy, we postulate that nonhuman primates should be considered as a valid preclinical model. Because of their closer immunological and physiological proximity to humans, these models in nonhuman primates can be valuable for studying disease-related aspects that cannot be addressed in rodent models.
Collapse
Affiliation(s)
- Michel Vierboom
- Biomedical Primate Research Centre, Department of Immunobiology, PO BOX 3306, 2280 GH Rijswijk, The Netherlands.
| | | | | |
Collapse
|
30
|
Jia D, Yang H, Wan L, Cheng J, Lu X. Production of bioactive, SUMO-modified, and native-like TNF-α of the rhesus monkey, Macaca mulatta, in Escherichia coli. Appl Microbiol Biotechnol 2011; 93:2345-55. [DOI: 10.1007/s00253-011-3794-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 11/23/2011] [Accepted: 11/24/2011] [Indexed: 02/08/2023]
|
31
|
Zhu S, Liu S, Wan L, Yang G, Yang H, Cheng J, Lu X. Molecular cloning, expression and characterization of the functional domain of CTLA4 from the rhesus monkey, Macaca mulatta. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:736-744. [PMID: 21349284 DOI: 10.1016/j.dci.2011.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 02/15/2011] [Accepted: 02/16/2011] [Indexed: 05/30/2023]
Abstract
Cytotoxic T lymphocyte-associated antigen 4 (CTLA4) is a potent inhibitor of T cell activation. The genes encoding the membrane and soluble forms of Macaca mulatta CTLA4 (mmCTLA4) were isolated from peripheral blood mononuclear cells (PBMCs). The predicted mmCTLA4 protein is nearly identical to human CTLA4 (hCTLA4), with the exception of a serine instead of an asparagine residue at position 49 and a leucine instead of a methionine residue at position 141 of its extracellular domain. The fusion protein mmCTLA4Ig, containing the extracellular domain of mmCTLA4 and the constant region of the human IgG1 antibody, was expressed in Pichia pastoris. The mmCTLA4Ig produced by P. pastoris exhibited specific binding to human B7-positive Raji cells that could be inhibited by competitive binding of hCTLA4Ig. MmCTLA4Ig and hCTLA4Ig could comparably suppress the proliferation of lymphocytes derived from rhesus monkeys, humans, or mice that had been stimulated either by concanavalin A (Con A) or allogeneic cells. These results suggest that mmCTLA4 is a negative regulator of T cell activation and that mmCTLA4Ig may be useful for immunotherapy of immunologic diseases in the rhesus monkey.
Collapse
Affiliation(s)
- Shengyun Zhu
- Key Lab of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | | | |
Collapse
|
32
|
Mohammad Shahi M, Rashidi MR, Mahboob S, Haidari F, Rashidi B, Hanaee J. Protective effect of soy protein on collagen-induced arthritis in rat. Rheumatol Int 2011. [DOI: 10.10.1007/s00296-011-1979-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
33
|
Protective effect of soy protein on collagen-induced arthritis in rat. Rheumatol Int 2011; 32:2407-14. [PMID: 21681567 DOI: 10.1007/s00296-011-1979-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 06/02/2011] [Indexed: 01/09/2023]
Abstract
To evaluate preventive and therapeutic effects of soy protein on collagen-induced arthritis rats. Sprague-Dawley rats immunized with bovine type II collagen emulsified in adjuvant and treated with soy protein (7 g/kg), dexamethasone (1 mg/kg), and casein (in control groups) by daily gavages feedings for 30 days. Score of arthritis recorded every day for each paws of animal. Tumor necrosis factor-alpha, interleukin6, leptin, and adiponectin were measured in serums. Treatment with soy protein resulted in significant delay in time to onset of arthritis as well as significantly decreased arthritis incidence, clinical arthritis severity score, histopathological arthritis severity score, and in vivo cell-mediated immunity to collagen (P < 0.05). Administration of soy protein significantly suppressed the progression of collagen II-induced arthritis and inhibited the production of tumor necrosis factor-alpha, interleukin6, leptin, and adiponectin. Soy protein appeared to be a potent immunomodulatory inhibitor of collagen II-induced arthritis in rats. It could delay onset of RA and reduced cartilage erosion and synovitis inflammation. Therefore, it may be a useful protein in the prevention and treatment of rheumatoid arthritis patient.
Collapse
|
34
|
Vierboom MPM, Breedveld E, Kondova I, 't Hart BA. Collagen-induced arthritis in common marmosets: a new nonhuman primate model for chronic arthritis. Arthritis Res Ther 2010; 12:R200. [PMID: 20977720 PMCID: PMC2991037 DOI: 10.1186/ar3172] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 09/10/2010] [Accepted: 10/26/2010] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION There is an ever-increasing need for animal models to evaluate efficacy and safety of new therapeutics in the field of rheumatoid arthritis (RA). Particularly for the early preclinical evaluation of human-specific biologicals targeting the progressive phase of the disease, there is a need for relevant animal models. In response to this requirement we set out to develop a model of collagen-induced arthritis (CIA) in a small-sized nonhuman primate species (300 to 400 g at adult age); that is, the common marmoset (Callithrix jacchus). METHODS Twenty-two animals divided into three experiments were immunized with collagen type II (CII) of either bovine or chicken origin with different immunization strategies. The animals were analyzed for clinical manifestation of arthritis, hematology and clinical chemistry, immunological responses against CII and histopathological features of the arthritis. RESULTS Clinically manifest arthritis was observed in almost 100% (21 out of 22) of the animals. Fifty percent of the animals developed semi-acute CIA while the other 50% displayed a more chronic disease. Both cellular (CD3/CD4 and CD3/CD8) and humoral responses (IgM and IgG) against CII were involved in the development of the disease. Besides mild histopathological changes in bone and cartilage, severe inflammation in extraarticular tissues like periosteum and subcutaneous tissues was observed. CONCLUSIONS This new model in marmosets more closely resembles chronic RA with respect to the chronic disease course and pathomorphological presentation than the more acute monophasic and destructive CIA model in macaques. This model can therefore fill a niche in preclinical testing of new human specific therapeutics.
Collapse
Affiliation(s)
- Michel P M Vierboom
- Department of Immunobiology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, The Netherlands.
| | | | | | | |
Collapse
|
35
|
Xu HL, Wang YT, Cheng AC, Yao YF, Ni QY, Zeng W, Bi FJ, Yang ZX, Chen XY. [Polymorphism of MHC-DPB1 gene exon 2 in rhesus macaques (Macaca mulatta)]. YI CHUAN = HEREDITAS 2010; 32:588-98. [PMID: 20566463 DOI: 10.3724/sp.j.1005.2010.00588] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Rhesus macaque (Macaca mulatta) has long been used as an experimental model animal for biomedical research and was under the key state protection (class II) from Chinese government. In order to facilitate the use of Chinese rhesus macaques in biomedical research and their protection based on better understanding of the major mistocompability complex (MHC) genes in these macaques, the exon 2 of Mamu-DPB1 genes were determined in 106 wild rhesus macaques using DGGE, cloning and sequencing. A total of 21 Mamu-DPB1 alleles were obtained, of which 15 alleles were novel sequences that had not been documented previously. Mamu-DPB1 30 was the most frequent allele in the whole large population comprising all 106 rhesus macaque individuals (0.1120) and in Xiaojin population (0.1120), Mamu-DPB1 04 in Heishui (0.1702), -DPB1 32 in Bazhong (0.1613), -DPB1 30 in Hanyuan (0.1120), and -DPB1 04 in Jiulong (0.1139). The alignment of the amino acids sequences showed that 12 variable sites were species-specific, of which 9 sites occurred in the putative amino acids sequences of the 15 novel Mamu-DPB1 alleles. Trans-species polymorphism was observed on the phylogenetic tree based on the DPB1 alleles of rhesus macaques and cynomolgus (Macaca fascicularis). In addition, these results also demonstrated that significant genetic differentiation has occurred between Chinese and Indian rhesus macaque population.
Collapse
Affiliation(s)
- Huai-Liang Xu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Labadie K, Larcher T, Joubert C, Mannioui A, Delache B, Brochard P, Guigand L, Dubreil L, Lebon P, Verrier B, de Lamballerie X, Suhrbier A, Cherel Y, Le Grand R, Roques P. Chikungunya disease in nonhuman primates involves long-term viral persistence in macrophages. J Clin Invest 2010; 120:894-906. [PMID: 20179353 DOI: 10.1172/jci40104] [Citation(s) in RCA: 406] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 01/06/2010] [Indexed: 02/06/2023] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that induces in humans a disease characterized by fever, rash, and pain in muscles and joints. The recent emergence or reemergence of CHIKV in the Indian Ocean Islands and India has stressed the need to better understand the pathogenesis of this disease. Previous CHIKV disease models have used young or immunodeficient mice, but these do not recapitulate human disease patterns and are unsuitable for testing immune-based therapies. Herein, we describe what we believe to be a new model for CHIKV infection in adult, immunocompetent cynomolgus macaques. CHIKV infection in these animals recapitulated the viral, clinical, and pathological features observed in human disease. In the macaques, long-term CHIKV infection was observed in joints, muscles, lymphoid organs, and liver, which could explain the long-lasting CHIKV disease symptoms observed in humans. In addition, the study identified macrophages as the main cellular reservoirs during the late stages of CHIKV infection in vivo. This model of CHIKV physiopathology should allow the development of new therapeutic and/or prophylactic strategies.
Collapse
Affiliation(s)
- Karine Labadie
- CEA, Division of Immuno-Virology/Institute of Emerging Diseases and Innovative Therapies, Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kap YS, van Meurs M, van Driel N, Koopman G, Melief MJ, Brok HPM, Laman JD, 't Hart BA. A monoclonal antibody selection for immunohistochemical examination of lymphoid tissues from non-human primates. J Histochem Cytochem 2009; 57:1159-67. [PMID: 19729671 DOI: 10.1369/jhc.2009.954123] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Non-human primates (NHPs) offer valuable animal models for basic research into human diseases and for the preclinical validation of new therapeutics. Detailed in situ examination of the involved cell types using immunohistochemistry is often hampered by the lack of cross-reactive antibodies (Abs). In the current study, we have tested a large panel of monoclonal antibodies raised against human leukocyte differentiation and activation markers for cross-reactivity on cryosections of lymphoid tissue from six NHP species. In total, we have tested 130 Abs against 69 antigens expressed in tissues from one great ape species (chimpanzee/Pan troglodytes), two Old World species (rhesus macaque/Macaca mulatta and cynomolgus macaque/Macaca fascicularis), and three New World species (common marmoset/Callithrix jacchus, cotton-top tamarin/Saguinus oedipus, and owl monkey/Aotus triviogatus). We have found a large panel of cross-reactive Abs: 93 of 102 (91%) in chimpanzee, 97 of 125 (78%) in rhesus macaque, 70 of 109 (64%) in cynomolgus macaque, 69 of 116 (60%) in common marmoset, 40 of 81 (49%) in cotton-top tamarin, and 35 of 80 (44%) in owl monkey. The availability of a reliable panel of cross-reactive markers is important to gaining further insight into immunological processes in disease-affected tissues from NHP species.
Collapse
Affiliation(s)
- Yolanda S Kap
- Departments of Immunobiology, Biomedical Primate Research Center, Rijswijk, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Qiu CL, Yang GB, Yu K, Li Y, Li XL, Liu Q, Zhao H, Xing H, Shao Y. Characterization of the major histocompatibility complex class II DQB (MhcMamu-DQB1) alleles in a cohort of Chinese rhesus macaques (Macaca mulatta). Hum Immunol 2008; 69:513-21. [PMID: 18582516 DOI: 10.1016/j.humimm.2008.05.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 05/22/2008] [Accepted: 05/27/2008] [Indexed: 10/21/2022]
Abstract
Rhesus macaques have long been used in animal models for various human diseases, the susceptibility and/or resistance to some of which have been associated with the major histocompatibilty complex (MHC). To gain insight into the MHC background and to facilitate the experimental use of Chinese rhesus macaques, the second exon of MhcMamu-DQB1 genes in 105 rhesus macaques were characterized by cloning and sequencing. A total of 37 MhcMamu-DQB1 alleles were identified, illustrating a marked allelic polymorphism at DQB1 in these monkeys. In addition to 10 alleles were novel sequences that had not been documented in earlier reports, at least 14 alleles reported in earlier studies were not detected in this study. Most of the sequences (73%) observed in this study belong to DQB1 06 (13 alleles) and DQB1 18 (14 alleles) lineages, and the rest (27%) belong to DQB1 15, DQB1 16 and DQB1 17 lineages. The most frequent allele detected among these monkeys was MhcMamu-DQB1 06111 (22%), followed by DQB1 1503 (19%); and most of the novel alleles were present at a frequency of less than 2.5%. As for individual animals, 24 of 105 (23%) were homozygous whereas 81 of 105 (77%) were heterozygous at the MhcMamu-DQB1 locus. These data indicated significant differences in MhcMamu-DQB1 allele distribution between the Chinese rhesus macaques and the previously reported rhesus macaques, which were mostly of Indian origin. This information will not only promote the understanding of rhesus macaque MHC diversity and polymorphism but will also facilitate the use of Chinese rhesus macaques in human disease studies, especially those that may be associated with HLA-DQB genes.
Collapse
Affiliation(s)
- Chen-Li Qiu
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Beijing 100050, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Preclinical evaluation of anti-rheumatic drugs in a non-human primate model of arthritic disease. ACTA ACUST UNITED AC 2008. [DOI: 10.1016/j.ddmod.2008.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Anemia in monkey collagen-induced arthritis is correlated with serum IL-6, but not TNFα. Rheumatol Int 2008; 28:879-83. [DOI: 10.1007/s00296-008-0547-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Accepted: 02/10/2008] [Indexed: 10/22/2022]
|