1
|
Abdallah AM, Naiem AHA, Abdelraheim SR, Mohafez OM, Abdelghany HM, Elsayed SA, Gomaa W, Marey H. Pyrazole derivatives ameliorate synovial inflammation in collagen-induced arthritis mice model via targeting p38 MAPK and COX-2. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:819-832. [PMID: 39073417 PMCID: PMC11787049 DOI: 10.1007/s00210-024-03290-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
The type II collagen-induced arthritis (CIA) model and human rheumatoid arthritis exhibit similar characteristics. Both diseases involve the production of inflammatory cytokines and other mediators, triggering an inflammatory cascade linked to bone and cartilage damage. Recently, new pyrazole compounds with various pharmacological activities, including antimicrobial, anticancer, anti-inflammatory, and analgesic agents, have been reported. Our aim is to evaluate the therapeutic effectiveness of two newly synthesized pyrazole derivatives, M1E and M1G, in reducing inflammation and oxidative stress in a mouse model of collagen-induced arthritis. Arthritis was induced in DBA/1J mice, and the therapeutic effect of the M1E and M1G is assessed by measuring the arthritic index, quantifying the expression of inflammatory genes such as p38 MAPK, COX-2, IL1β, MMP3, and TNF-α using real-time PCR and analyzing protein expression using western blotting for phosphorylated p38 MAPK and COX-2. Oxidative stress markers and hind paws joint histopathology were also evaluated. Treatment with the two pyrazole derivatives significantly (p < 0.001) improved the arthritic score; downregulated the expression of inflammatory genes p38 MAPK, COX-2, IL1β, MMP3, and TNF-α; and reduced the protein expression of phosphorylated p3 MAPK and COX-2. In addition, both compounds ameliorated oxidative stress by increasing the activities of SOD and reducing the formation of MDA in the paw tissue homogenates. Both M1E and M1G significantly (p < 0.001) improved the pathological features of synovitis. The pyrazole derivatives, M1E and M1G, significantly reduced the arthritic score and the inflammatory cytokine expression, improved synovitis histopathology, and ameliorated oxidative stress in the CIA mice model.
Collapse
Affiliation(s)
- Ahlam M Abdallah
- Department of Medical Biochemistry, Faculty of Medicine, Minia University, Minia, 61511, Egypt
| | - Amany H Abdel Naiem
- Department of Medical Biochemistry, Faculty of Medicine, Minia University, Minia, 61511, Egypt
| | - Salama R Abdelraheim
- Department of Medical Biochemistry, Faculty of Medicine, Minia University, Minia, 61511, Egypt.
| | - Omar M Mohafez
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Hend M Abdelghany
- Department of Medical Biochemistry, Faculty of Medicine, Minia University, Minia, 61511, Egypt
| | - Sahar A Elsayed
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Sohag University, Sohag, 82524, Egypt
| | - Wafaey Gomaa
- Department of Pathology, Faculty of Medicine, Minia University, Minia, 61511, Egypt
| | - Heba Marey
- Department of Medical Biochemistry, Faculty of Medicine, Minia University, Minia, 61511, Egypt
| |
Collapse
|
2
|
Huang H, Wei X. Therapeutic potential of CD20/CD3 bispecific antibodies in the treatment of autoimmune diseases. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2024; 5:209-216. [PMID: 39802547 PMCID: PMC11720466 DOI: 10.1515/rir-2024-0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/08/2024] [Indexed: 01/16/2025]
Abstract
Autoimmune diseases arise from immune system dysfunction that immune cells mistakenly attack the body's own tissues, resulting in systemic disorders or localized lesions such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Autoreactive B cells play a critical role in the pathogenesis of many autoimmune diseases and B cell depletion using anti-CD20 monoclonal antibody (mAb) has been shown to effectively mitigate disease progression in both preclinical and clinical studies. Recently, bispecific antibody (bsAb) targeting CD20/CD3 have demonstrated substantial clinical benefits in the treatment of various hematologic malignancies. Given their similar B cell cytotoxic mechanism, CD20/CD3 bsAb therapy may offer significant improvements in the management of many autoimmune diseases, providing a novel therapeutic option for patients. This concise review aims to summarize recent findings on CD20/CD3 bsAbs and discuss their potential in treating autoimmune diseases.
Collapse
Affiliation(s)
- Hongpeng Huang
- Department of Bioactivity, SinoCellTech Ltd., Beijing100176, China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University; PekingChina
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing100191, China
| |
Collapse
|
3
|
Fukasawa T, Yoshizaki-Ogawa A, Enomoto A, Yamashita T, Miyagawa K, Sato S, Yoshizaki A. Single cell analysis in systemic sclerosis - A systematic review. Immunol Med 2024; 47:118-129. [PMID: 38818750 DOI: 10.1080/25785826.2024.2360690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
In recent years, rapid advances in research methods have made single cell analysis possible. Systemic sclerosis (SSc), a disease characterized by the triad of immune abnormalities, fibrosis, and vasculopathy, has also been the subject of various analyses. To summarize the results of single cell analysis in SSc accumulated to date and to deepen our understanding of SSc. Four databases were used to perform a database search on 23rd June 2023. Assessed Grading of Recommendations Assessment, Development and Evaluation certainty of evidence were performed according to PRISMA guidelines. The analysis was completed on July 2023. 17 studies with 358 SSc patients were included. Three studies used PBMCs, six used skin, nine used lung with SSc-interstitial lung diseases (ILDs), and one used lung with SSc-pulmonary arterial hypertension (PAH). The cells studied included immune cells such as T cells, natural killer cells, monocytes, macrophages, and dendritic cells, as well as endothelial cells, fibroblasts, keratinocytes, alveolar type I cells, basal epithelial cells, smooth muscle cells, mesothelial cells, etc. This systematic review revealed the results of single cell analysis, suggesting that PBMCs, skin, SSc-ILD, and SSc-PAH show activation and dysfunction of cells associated with immune-abnormalities, fibrosis, and vasculopathy, respectively.
Collapse
Affiliation(s)
- Takemichi Fukasawa
- Department of Dermatology, Systemic sclerosis center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Asako Yoshizaki-Ogawa
- Department of Dermatology, Systemic sclerosis center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Enomoto
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takashi Yamashita
- Department of Dermatology, Systemic sclerosis center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kiyoshi Miyagawa
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, Systemic sclerosis center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, Systemic sclerosis center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
4
|
Fukasawa T, Yoshizaki-Ogawa A, Sato S, Yoshizaki A. The role of B cells in systemic sclerosis. J Dermatol 2024; 51:904-913. [PMID: 38321641 DOI: 10.1111/1346-8138.17134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 02/08/2024]
Abstract
Systemic sclerosis (SSc) is a rare and refractory systemic disease characterized by fibrosis and vasculopathy in the presence of autoimmune abnormalities. While the exact cause of SSc is incompletely understood, the specific autoantibodies identified in SSc are closely linked to disease severity and prognosis, indicating a significant role of autoimmune abnormalities in the pathogenesis of SSc. Although the direct pathogenic mechanisms of autoantibodies in SSc are not fully elucidated, numerous prior investigations have demonstrated the involvement of B cells in the pathogenesis of SSc through various mechanisms. Additionally, several clinical trials have explored the efficacy of B-cell depletion therapy for SSc, with many reporting positive outcomes. However, the role of B cells in SSc pathogenesis is multifaceted, as they can both promote inflammation and exert inhibitory functions. This article provides an overview of the involvement of B cells in SSc development, incorporating the latest research findings.
Collapse
Affiliation(s)
- Takemichi Fukasawa
- Department of Dermatology, Systemic Sclerosis Center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Asako Yoshizaki-Ogawa
- Department of Dermatology, Systemic Sclerosis Center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, Systemic Sclerosis Center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, Systemic Sclerosis Center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Cheng S, Jiang D, Lan X, Liu K, Fan C. Voltage-gated potassium channel 1.3: A promising molecular target in multiple disease therapy. Biomed Pharmacother 2024; 175:116651. [PMID: 38692062 DOI: 10.1016/j.biopha.2024.116651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
Voltage-gated potassium channel 1.3 (Kv1.3) has emerged as a pivotal player in numerous biological processes and pathological conditions, sparking considerable interest as a potential therapeutic target across various diseases. In this review, we present a comprehensive examination of Kv1.3 channels, highlighting their fundamental characteristics and recent advancements in utilizing Kv1.3 inhibitors for treating autoimmune disorders, neuroinflammation, and cancers. Notably, Kv1.3 is prominently expressed in immune cells and implicated in immune responses and inflammation associated with autoimmune diseases and chronic inflammatory conditions. Moreover, its aberrant expression in certain tumors underscores its role in cancer progression. While preclinical studies have demonstrated the efficacy of Kv1.3 inhibitors, their clinical translation remains pending. Molecular imaging techniques offer promising avenues for tracking Kv1.3 inhibitors and assessing their therapeutic efficacy, thereby facilitating their development and clinical application. Challenges and future directions in Kv1.3 inhibitor research are also discussed, emphasizing the significant potential of targeting Kv1.3 as a promising therapeutic strategy across a spectrum of diseases.
Collapse
Affiliation(s)
- Sixuan Cheng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Kun Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Cheng Fan
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
6
|
Bandeira M, Dourado E, Lopes F, Tenazinha C, Barros R, Barreira SC. Reactive arthritis as a rare complication of intravesical bacillus Calmette-Guérin treatment: Report of two cases. Int J Rheum Dis 2024; 27:e14862. [PMID: 37578016 DOI: 10.1111/1756-185x.14862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/22/2023] [Accepted: 07/27/2023] [Indexed: 08/15/2023]
Abstract
Intravesical bacillus Calmette-Guérin (BCG) immunotherapy is recommended for non-muscle-invasive bladder cancer after transurethral resection. BCG-associated musculoskeletal adverse events are rare. We report two cases of BCG reactive arthritis that were unusually severe and refractory. These describe two male patients who presented with polyarthritis after BCG exposure. Ultrasonography-guided glucocorticoid injections, high-dose systemic glucocorticoids and the institution of sulfasalazine were required for achievement of remission. Bacillus Calmette-Guérin reactive arthritis can present as polyarthritis of small and medium joints or as mono-oligoarthritis of asymmetrical ankles and knees, frequently associated with tenosynovitis and enthesitis. The mechanism by which BCG promotes arthralgia and arthritis is poorly understood. The most well-accepted theory is that the BCG antigens migrate to different peripheral tissues, including the joints. There is also a lack of knowledge regarding risk factors, with possible genetic factors playing a role. As the two presented cases show, BCG-induced reactive arthritis should be considered in the differential diagnosis of arthritis and refractory tenosynovitis in BCG-exposed patients.
Collapse
Affiliation(s)
- Matilde Bandeira
- Serviço de Reumatologia, Centro Hospitalar Universitário Lisboa Norte (CHULN), Centro Académico de Medicina de Lisboa (CAML), Lisboa, Portugal
- Unidade de Investigação em Reumatologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, CAML, Lisboa, Portugal
| | - Eduardo Dourado
- Serviço de Reumatologia, Centro Hospitalar Universitário Lisboa Norte (CHULN), Centro Académico de Medicina de Lisboa (CAML), Lisboa, Portugal
- Unidade de Investigação em Reumatologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, CAML, Lisboa, Portugal
| | - Filipe Lopes
- Serviço de Urologia, Centro Hospitalar Universitário Lisboa Norte (CHULN), Centro Académico de Medicina de Lisboa (CAML), Lisboa, Portugal
| | - Catarina Tenazinha
- Serviço de Reumatologia, Centro Hospitalar Universitário Lisboa Norte (CHULN), Centro Académico de Medicina de Lisboa (CAML), Lisboa, Portugal
- Unidade de Investigação em Reumatologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, CAML, Lisboa, Portugal
| | - Rita Barros
- Serviço de Reumatologia, Centro Hospitalar Universitário Lisboa Norte (CHULN), Centro Académico de Medicina de Lisboa (CAML), Lisboa, Portugal
- Unidade de Investigação em Reumatologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, CAML, Lisboa, Portugal
| | - Sofia C Barreira
- Serviço de Reumatologia, Centro Hospitalar Universitário Lisboa Norte (CHULN), Centro Académico de Medicina de Lisboa (CAML), Lisboa, Portugal
- Serviço de Urologia, Centro Hospitalar Universitário Lisboa Norte (CHULN), Centro Académico de Medicina de Lisboa (CAML), Lisboa, Portugal
| |
Collapse
|
7
|
Jo M, Hwang S, Lee CG, Hong JE, Kang DH, Yoo SH, Kim WS, Yoo JY, Rhee KJ. Promotion of Colitis in B Cell-Deficient C57BL/6 Mice Infected with Enterotoxigenic Bacteroides fragilis. Int J Mol Sci 2023; 25:364. [PMID: 38203534 PMCID: PMC10778593 DOI: 10.3390/ijms25010364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Enterotoxigenic Bacteroides fragilis (ETBF) causes colitis and is implicated in inflammatory bowel diseases and colorectal cancer. The ETBF-secreted B. fragilis toxin (BFT) causes cleavage of the adherence junction, the E-cadherin, resulting in the large intestine showing IL-17A inflammation in wild-type (WT) mice. However, intestinal pathology by ETBF infection is not fully understood in B-cell-deficient mice. In this study, ETBF-mediated inflammation was characterized in B-cell-deficient mice (muMT). WT or muMT C57BL/6J mice were orally inoculated with ETBF and examined for intestinal inflammation. The indirect indicators for colitis (loss of body weight and cecum weight, as well as mortality) were increased in muMT mice compared to WT mice. Histopathology and inflammatory genes (Nos2, Il-1β, Tnf-α, and Cxcl1) were elevated and persisted in the large intestine of muMT mice compared with WT mice during chronic ETBF infection. However, intestinal IL-17A expression was comparable between WT and muMT mice during infection. Consistently, flow cytometry analysis applied to the mesenteric lymph nodes showed a similar Th17 immune response in both WT and muMT mice. Despite elevated ETBF colonization, the ETBF-infected muMT mice showed no histopathology or inflammation in the small intestine. In conclusion, B cells play a protective role in ETBF-induced colitis, and IL-17A inflammation is not attributed to prompted colitis in B-cell-deficient mice. Our data support the fact that B cells are required to ameliorate ETBF infection-induced colitis in the host.
Collapse
Affiliation(s)
- Minjeong Jo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Soonjae Hwang
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Chang-Gun Lee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Ju-Eun Hong
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Da-Hye Kang
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Sang-Hyeon Yoo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Woo-Seung Kim
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Jung-Yoon Yoo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| |
Collapse
|
8
|
Wang M, Liu H, Huang M, Huang Y, Ming Y, Chen W, Chen Y, Tang Z, Jia B. Immunomodulatory functions of microorganisms in tissue regenerative healing. Acta Biomater 2023; 172:38-52. [PMID: 37816417 DOI: 10.1016/j.actbio.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 10/12/2023]
Abstract
External pathogenic microorganisms and commensal microorganisms in the body have either harmful or beneficial impacts on the regenerative repair of tissues, and the immune system plays a crucial regulatory role in this process. This review summarises our current understanding of microorganism-immune system interactions, with a focus on how these interactions impact the renewal and repair ability of tissues, including skin, bone, gut, liver, and nerves. This review concludes with a discussion of the mechanisms by which microbes act on various types of immune cells to affect tissue regeneration, offers potential strategies for using microbial therapies to enhance the regenerative repair function of tissues, and suggest novel therapeutic approaches for regenerative medicine. STATEMENT OF SIGNIFICANCE: Microbiological communities have crucial impacts on human health and illness by participating in energy collection and storage and performing various metabolic processes. External pathogenic microorganisms and commensal microorganisms in the body have either harmful or beneficial impacts on the regenerative repair of tissues, and the immune system plays a critical regulatory role in this process. This study reviews the important correlation between microorganisms and the immune system and investigates the mechanism of various microorganism that participate in the regeneration and repair of tissues and organs by modulating immune system.
Collapse
Affiliation(s)
- Min Wang
- Department of Oral Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Hongyu Liu
- Department of Oral Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Mingshu Huang
- Department of Oral Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yisheng Huang
- Department of Oral Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yue Ming
- Department of Oral Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Weixing Chen
- Department of Oral Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yuanxin Chen
- Department of Oral Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Zhengming Tang
- Department of Oral Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Bo Jia
- Department of Oral Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Ansari MA, Nadeem A, Attia SM, Bakheet SA, Alasmari AF, Alomar HA, Al-Mazroua HA, Alhamed AS, Shahid M, Alqinyah M, Assiri MA, Al-Hamamah MA, Alassmrry YA, Ahmad SF. Rituximab exerts its anti-arthritic effects via inhibiting NF-κB/GM-CSF/iNOS signaling in B cells in a mouse model of collagen-induced arthritis. Heliyon 2023; 9:e16673. [PMID: 37274671 PMCID: PMC10238934 DOI: 10.1016/j.heliyon.2023.e16673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/06/2023] Open
Abstract
Rheumatoidarthritis (RA) is an autoimmune disease characterized by uncontrolled joint inflammation and damage to bone and cartilage. B cells are known to play a crucial role in the pathogenesis and development of arthritis. Previous studies have found that B cells may be a potential target for treating RA. Rituximab, a monoclonal antibody targeting B cells, has induced long-term clinical responses in RA. Collagen-induced arthritis (CIA) mouse model is a widely studied autoimmune model of RA. CIA mouse model was used to investigate the effect of rituximab on the RA severity in the mice. Following induction of CIA, animals were treated with rituximab (250 mg/kg/week) intraperitoneally on the days 28, 35, 42, 49, 56, and 63 after collagen induction. We investigated the effect of rituximab on NF-κB p65, IκBα, GM-CSF, MCP-1, iNOS, TNF-α, and IL-6 cells in splenic CD19+ and CD45R+ B cells using flow cytometry. We also assessed the effect of rituximab on NF-κB p65, GM-CSF, IκBα, MCP-1, iNOS, TNF-α, and IL-6 at mRNA levels using RT-PCR analyses of knee tissues. Rituximab treatment significantly decreased CD19+NF-κB p65+, CD45R+NF-κB p65+, CD19+GM-CSF+, CD45R+GM-CSF+, CD19+MCP-1+, CD45R+MCP-1+, CD19+TNF-α+, CD45R+TNF-α+, CD19+iNOS+, CD45R+iNOS+, CD19+IL-6+, and CD45R+IL-6+, and increased CD45R+IκBα+ in spleen cells of CIA mice. We further observed that rituximab treatment downregulated NF-κB p65, GM-CSF, MCP-1, iNOS, TNF-α, and IL-6, whereas it upregulated IκBα, mRNA level. All these findings suggest that rituximab may be a novel therapeutic target for the treatment of RA.
Collapse
Affiliation(s)
- Mushtaq A. Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M. Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A. Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hatun A. Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A. Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah S. Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Al-Hamamah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yasseen A. Alassmrry
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
10
|
Shirai T, Nakai A, Ando E, Fujimoto J, Leach S, Arimori T, Higo D, van Eerden FJ, Tulyeu J, Liu YC, Okuzaki D, Murayama MA, Miyata H, Nunomura K, Lin B, Tani A, Kumanogoh A, Ikawa M, Wing JB, Standley DM, Takagi J, Suzuki K. Celastrol suppresses humoral immune responses and autoimmunity by targeting the COMMD3/8 complex. Sci Immunol 2023; 8:eadc9324. [PMID: 37000855 DOI: 10.1126/sciimmunol.adc9324] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Celastrol, a bioactive molecule extracted from the
Tripterygium wilfordii
plant, has been shown to exhibit anti-inflammatory properties. However, its mechanism of action has not been fully elucidated. Here, we show that celastrol suppresses humoral immune responses and autoimmunity by disabling a protein complex consisting of copper metabolism MURR1 domain–containing (COMMD) 3 and COMMD8 (COMMD3/8 complex), a signaling adaptor for chemoattractant receptors. Having demonstrated the involvement of the COMMD3/8 complex in a mouse model of rheumatoid arthritis, we identified celastrol as a compound that covalently bound to and dissociated the COMMD3/8 complex. Celastrol inhibited B cell migration, reduced antibody responses, and blocked arthritis progression, recapitulating deficiency of the COMMD3/8 complex. These effects of celastrol were abolished in mice expressing a celastrol-resistant mutant of the COMMD3/8 complex. These findings establish that celastrol exerts immunosuppressive activity by targeting the COMMD3/8 complex. Our study suggests that the COMMD3/8 complex is a potentially druggable target in autoimmune diseases and points to celastrol as a lead pharmacologic candidate in this capacity.
Collapse
Affiliation(s)
- Taiichiro Shirai
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Akiko Nakai
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Immune Response Dynamics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Emiko Ando
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Jun Fujimoto
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Sarah Leach
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Takao Arimori
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Daisuke Higo
- Thermo Fisher Scientific K.K., Yokohama, Kanagawa, Japan
| | - Floris J. van Eerden
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Janyerkye Tulyeu
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Yu-Chen Liu
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Masanori A. Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| | - Haruhiko Miyata
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kazuto Nunomura
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Suita, Osaka, Japan
| | - Bangzhong Lin
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Suita, Osaka, Japan
| | - Akiyoshi Tani
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Masahito Ikawa
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - James B. Wing
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Daron M. Standley
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| | - Kazuhiro Suzuki
- Laboratory of Immune Response Dynamics, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immune Response Dynamics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
11
|
Lactic Acid Regulation: A Potential Therapeutic Option in Rheumatoid Arthritis. J Immunol Res 2022; 2022:2280973. [PMID: 36061305 PMCID: PMC9433259 DOI: 10.1155/2022/2280973] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/16/2022] [Indexed: 11/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, persistent autoimmune disease that causes severe joint tissue damage and irreversible disability. Cumulative evidence suggests that patients suffering from RA for long durations are at risk of functional damage to cardiovascular, kidney, lung, and other tissues. This seriously affects the quality of work and life of patients. To date, no clear etiology of RA has been found. Recent studies have revealed that the massive proliferation of synoviocytes and immune cells requires a large amount of energy supply. Rapid energy supply depends on the anaerobic glucose metabolic pathway in both RA animal models and clinical patients. Anaerobic glycolysis can increase intracellular lactic acid (LA) content. LA induces the overexpression of monocarboxylate transporters (MCTs) in cell membranes. MCTs rapidly transport LA from the intracellular to the intercellular or articular cavity. Hence, a relatively high accumulation of LA could be formed in the intercellular and articular cavities of inflammatory joints. Moreover, LA contributes to the migration and activation of immune cells. Immune cells proliferate and secrete interleukins (IL) including IL-1, IL-2, IL-13, IL-17, and other inflammatory factors. These inflammatory factors enhance the immune inflammatory response of the body and aggravate the condition of RA patients. In this paper, the effects of LA on RA pathogenesis will be summarized from the perspective of the production, transport, and metabolism of synoviocytes and immune cells. Additionally, the drugs involved in the production, transport, and metabolism of LA are highlighted.
Collapse
|
12
|
Khanfar E, Olasz K, Gajdócsi E, Jia X, Berki T, Balogh P, Boldizsár F. Splenectomy modulates the immune response but does not prevent joint inflammation in a mouse model of RA. Clin Exp Immunol 2022; 209:201-214. [PMID: 35576510 PMCID: PMC9390846 DOI: 10.1093/cei/uxac052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/04/2022] [Accepted: 05/24/2022] [Indexed: 11/14/2022] Open
Abstract
The spleen is the largest secondary lymphoid organ which is involved in the development of B cells and also in systemic (auto)immune responses. Using the recombinant human G1 domain-induced arthritis (GIA) model in splenectomized and control BALB/c mice, we investigated the role of the spleen in the induction and pathogenesis of autoimmune arthritis. Splenectomized mice developed GIA with a similar clinical picture to the control group. However, we observed significant alterations in the humoral and cellular immune responses in splenectomized mice. In the sera of the splenectomized mice, we found lower pro-inflammatory cytokine and anti-rhG1 IgM levels, but higher IL-4, anti-rhG1 IgG1 and anti-CCP and RF antibodies. The arthritis induction in the splenectomized group was associated with a significant expansion of activated helper T cells and an increase in the proportion of the circulating B1 and marginal zone B cell subsets. Importantly, immunization of the splenectomized mice with rhG1 induced the formation of germinal centers in the inguinal- and mesenteric lymph nodes (i/mLNs) which showed an active immune response to rhG1. Finally, both B and T cells from the mLNs of the splenectomized mice showed decreased intracellular Ca2+ signaling than those of the control group. Collectively, these findings indicate that the presence of the spleen is not critical for the induction of GIA, and in its absence the autoimmune arthritis is most likely promoted through the compensatory activity of the i/mLNs. However, our data implies the immunological role of the spleen in arthritis which could be further assessed in human RA.
Collapse
Affiliation(s)
- Esam Khanfar
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Hungary
| | - Katalin Olasz
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Hungary
| | - Erzsébet Gajdócsi
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Hungary
| | - Xinkai Jia
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Hungary
- Lymphoid Organogenesis Research Group, Szentagothai Research Center, University of Pécs, Hungary
| | - Timea Berki
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Hungary
- Lymphoid Organogenesis Research Group, Szentagothai Research Center, University of Pécs, Hungary
| | - Ferenc Boldizsár
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Hungary
| |
Collapse
|
13
|
Yoshizaki A, Fukasawa T, Ebata S, Yoshizaki-Ogawa A, Sato S. Involvement of B cells in the development of systemic sclerosis. Front Immunol 2022; 13:938785. [PMID: 35967355 PMCID: PMC9365989 DOI: 10.3389/fimmu.2022.938785] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare intractable systemic disease that causes fibrosis and vasculopathy against a background of autoimmune abnormalities. Although the etiology is not yet fully understood, the type of autoantibodies detected in SSc is closely associated with disease severity and prognosis, supporting that those autoimmune abnormalities play an important role in the pathogenesis of SSc. Although the direct pathogenicity of autoantibodies found in SSc is unknown, many previous studies have shown that B cells are involved in the development of SSc through a variety of functions. Furthermore, a number of clinical studies have been conducted in which B-cell depletion therapy has been tried for SSc, and many of these studies have found B-cell depletion therapy to be effective for SSc. However, the involvement of B cells in pathogenesis is complex, as they not only promote inflammation but also play an inhibitory role. This article outlines the role of B cells in the development of SSc, including the latest research.
Collapse
|
14
|
Al-Hamamah MA, Alotaibi MR, Ahmad SF, Nadeem A, Attia MSM, Ansari MA, Bakheet SA, Alanazi MM, Attia SM. Treatment with the anti-CD20 monoclonal antibody rituximab mitigates gonadal disruptions in the collagen-induced arthritis in male DBA/1 J mouse model. Mutat Res 2022; 825:111799. [PMID: 36108541 DOI: 10.1016/j.mrfmmm.2022.111799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/08/2022] [Accepted: 09/04/2022] [Indexed: 06/15/2023]
Abstract
Rheumatoid arthritis (RA), which is driven by persistent activation of the immune system, primarily affects the joints. Several reports have estimated the risk of gonadal disruptions in arthritic patients, with potential attributable risk factors such as treatments with the disease-modifying antirheumatic drugs and the influence of the disease itself. The FDA approved rituximab, a therapy for non-Hodgkin's lymphoma, for management of RA in February 2006. However, the influence of repeated treatment with rituximab on gonadal function in RA has not been reported yet. Thus, the aim of the presents study is to evaluate whether repeated treatment with the clinically relevant dose of rituximab may change the gonadal disruptions in collagen-induced arthritis in male DBA/1 J mouse, a model of RA. Testicular disruptions, as determined by the sperm DNA strand breaks, spermatocyte chromosomal analysis and spermiogram examination have been conducted by the use of standard techniques. Additionally, we aimed to test whether the anti-rheumatic effect of rituximab also decreases the cellular oxidant-antioxidant imbalance in arthritic male DBA/1 J mice. Repeated treatment of naïve control DBA/1 J mice with rituximab did not exhibit any significant deleterious effects. Moreover, repeated administration of rituximab to the arthritic DBA/1 J mice suppressed disease severity and decreased testicular disruptions. Rituximab treatment also diminished gonadal oxidative stress, through decreasing reactive oxygen species generation and restoring the reduced glutathione level in arthritic DBA/1 J mice. In conclusion, rituximab is a safe therapeutic agent and can mitigate gonadal disruptions induced by arthritis, which insinuates the importance for arthritic patients especially at reproductive age.
Collapse
Affiliation(s)
- Mohammed A Al-Hamamah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Moureq R Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Mohamed S M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
15
|
Zhang X, Sun Y, Wang N, Zhang Y, Xia Y, Liu Y. Immunomodulatory Treatment Strategies Targeting B Cells for Heart Failure. Front Pharmacol 2022; 13:854592. [PMID: 35350762 PMCID: PMC8957947 DOI: 10.3389/fphar.2022.854592] [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: 01/14/2022] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Cardio-oncology, a nascent specialty, has evolved as a concerted strategy to address the cardiovascular complications of cancer therapies. On the other hand, emerging evidence has shown that some anti-tumor drugs, such as CD20-targeted rotuximab, also have markedly cardioprotective effects in addition to treating cancers. Rituximab is a CD20-targeted monoclonal antibody and kill tumor B-cells through antibody-mediated and antibody-independent pathways, indicating that B cells participate and promote the progression of cardiovascular diseases. In this review, we mainly present the evidence that B cells contribute to the development of hypertrophy, inflammation, and maladaptive tissue remodeling, with the aim of proposing novel immunomodulatory therapeutic strategies targeting B cells and their products for the treatment of heart failure.
Collapse
Affiliation(s)
- Xinxin Zhang
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuxi Sun
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ning Wang
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanli Zhang
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunlong Xia
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Liu
- Heart Failure and Structural Cardiology Division, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
16
|
Park JS, Lee D, Yang S, Jeong HY, Na HS, Cho KH, Choi J, Koo H, Cho ML, Park SH. Methotrexate-loaded nanoparticles ameliorate experimental model of autoimmune arthritis by regulating the balance of interleukin-17-producing T cells and regulatory T cells. J Transl Med 2022; 20:85. [PMID: 35148758 PMCID: PMC8840785 DOI: 10.1186/s12967-022-03267-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/18/2022] [Indexed: 11/23/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a progressive systemic autoimmune disease that is characterized by infiltration of inflammatory cells into the hyperplastic synovial tissue, resulting in subsequent destruction of adjacent articular cartilage and bone. Methotrexate (MTX), the first conventional disease-modifying antirheumatic drug (DMARD), could alleviate articular damage in RA and is implicated in humoral and cellular immune responses. However, MTX has several side effects, so efficient delivery of low-dose MTX is important. Methods To investigate the efficacy of MTX-loaded nanoparticles (MTX-NPs) against experimental model of RA, free MTX or MTX-NPs were administered as subcutaneous route to mice with collagen-induced arthritis (CIA) at 3 weeks after CII immunization. The levels of inflammatory factors in tissues were determined by immunohistochemistry, confocal microscopy, real-time PCR, and flow cytometry. Results MTX-NPs ameliorated arthritic severity and joint destruction in collagen-induced arthritis (CIA) mice compared to free MTX-treated CIA mice. The levels of inflammatory cytokines, including interleukin (IL)-1β, tumor necrosis factor-α, and vascular endothelial growth factor, were reduced in MTX-NPs-treated mice. Number of CD4 + IL-17 + cells decreased whereas the number of CD4 + CD25 + Foxp3 + cells increased in spleens from MTX- NPs-treated CIA mice compared to MTX-treated CIA mice. The frequency of CD19 + CD25 + Foxp3 + regulatory B cells increased in ex vivo splenocytes from MTX-loaded NPs-treated CIA mice compared to MTX-treated CIA mice. Conclusion The results suggest that MTX-loaded NPs have therapeutic potential for RA.
Collapse
Affiliation(s)
- Jin-Sil Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Donghyun Lee
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - SeungCheon Yang
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ha Yeon Jeong
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Sik Na
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Keun-Hyung Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - JeongWon Choi
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, 222 Banpo-Daero, Seocho-gu, Seoul, 137-701, South Korea.
| |
Collapse
|
17
|
Bounia CA, Liossis SNC. B cell depletion treatment decreases Th17 cells in patients with rheumatoid arthritis. Clin Immunol 2021; 233:108877. [PMID: 34740842 DOI: 10.1016/j.clim.2021.108877] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 10/30/2021] [Accepted: 10/30/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION We aimed to evaluate for any possible effects of treatment with rituximab (RTX) on the peripheral Th17 and Treg subpopulations in patients with rheumatoid arthritis (RA). PATIENTS AND METHODS We analyzed 16 patients with RA initiating RTX treatment, 11 patients with RA initiating abatacept treatment, 11 patients with RA treated with other medications, 8 patients with other autoimmune rheumatic diseases initiating RTX, and 14 healthy volunteers. Th17 cells (CD4+IL23R+IL17A+) and Treg cells (CD4+CD25hiFoxP3+) were evaluated flow-cytometrically. RESULTS Th17 cells from patients treated with RTX decreased significantly at weeks 8 and 16 (mean ± SEΜ: 3.01% ± 0.54℅ CD4+ cells at week 0 vs. 1.53% ± 0.24℅ at week 8 vs 1.10% ± 0.20℅ at week 16, p = 0.0004). Reductions of Th17 cells were evident in clinical responders (DAS28 score ≤ 3.2), ACPA (+) and RF (-) patients; circulating Tregs remained stable. Th17 and Tregs were not affected by ABA treatment or by changes in disease activity. Tregs, but not Th17 cells, decreased following treatment with RTX in patients with other autoimmune diseases (0.75% ± 0.16% at week 0 vs. 0.43% ± 0.16% at week 8, p = 0.033). CONCLUSION RTX-induced B cell depletion results in a significant reduction of circulating Th17 cell percentages, whereas it has no effect on Tregs of patients with RA. This reduction of Th17 cells was evident particularly in responders to RTX treatment, ACPA+ and RF (-) patients with RA.
Collapse
Affiliation(s)
- Constantina A Bounia
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, Rion, Patras, Greece
| | - Stamatis-Nick C Liossis
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, Rion, Patras, Greece; Department of Medicine, University of Patras Medical School, Rion, Patras, Greece.
| |
Collapse
|
18
|
Sustained microglial activation in the area postrema of collagen-induced arthritis mice. Arthritis Res Ther 2021; 23:273. [PMID: 34715926 PMCID: PMC8556992 DOI: 10.1186/s13075-021-02657-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023] Open
Abstract
Background Central nervous system (CNS)-mediated symptoms, such as fatigue, depression, and hyperalgesia, are common complications among patients with rheumatoid arthritis (RA). However, it remains unclear how the peripheral pathology of RA spreads to the brain. Accumulated evidence showing an association between serum cytokine levels and aberrant CNS function suggests that humoral factors participate in this mechanism. In contrast to the well-known early responses of microglia (CNS-resident immune cells) in the area postrema [AP; a brain region lacking a blood–brain barrier (BBB)] to experimental inflammation, microglial alterations in the AP during chronic inflammation like RA remain unclear. Therefore, to determine whether microglia in the AP can react to persistent autoimmune-arthritis conditions, we analyzed these cells in a mouse model of collagen-induced arthritis (CIA). Methods Microglial number and morphology were analyzed in the AP of CIA and control mice (administered Freund’s adjuvant or saline). Immunostaining for ionized calcium-binding adaptor molecule-1 was performed at various disease phases: “pre-onset” [post-immunization day (PID) 21], “establishment” (PID 35), and “chronic” (PID 56 and 84). Quantitative analyses of microglial number and morphology were performed, with principal component analysis used to classify microglia. Interleukin-1β (IL-1β) mRNA expression was analyzed by multiple fluorescent in situ hybridization and real-time polymerase chain reaction. Behavioral changes were assessed by sucrose preference test. Results Microglia in the AP significantly increased in density and exhibited changes in morphology during the establishment and chronic phases, but not the pre-onset phase. Non-subjective clustering classification of cell morphology (CIA, 1,256 cells; saline, 852 cells) showed that the proportion of highly activated microglia increased in the CIA group during establishment and chronic phases. Moreover, the density of IL-1β-positive microglia, a hallmark of functional activation, was increased in the AP. Sucrose preferences in CIA mice negatively correlated with IL-1β expression in brain regions containing the AP. Conclusions Our findings demonstrate that microglia in the AP can sustain their activated state during persistent autoimmune arthritis, which suggests that chronic inflammation, such as RA, may affect microglia in brain regions lacking a BBB and have various neural consequences. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02657-x.
Collapse
|
19
|
Ben Nasr M, Usuelli V, Seelam AJ, D'Addio F, Abdi R, Markmann JF, Fiorina P. Regulatory B Cells in Autoimmune Diabetes. THE JOURNAL OF IMMUNOLOGY 2021; 206:1117-1125. [PMID: 33685919 DOI: 10.4049/jimmunol.2001127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/11/2020] [Indexed: 12/24/2022]
Abstract
Since they were discovered almost three decades ago, a subset of B cells denoted as regulatory B cells (Bregs) have elicited interest throughout the immunology community. Many investigators have sought to characterize their phenotype and to understand their function and immunosuppressive mechanisms. Indeed, studies in murine models have demonstrated that Bregs possess varied phenotypic markers and could be classified into different subsets whose action and pivotal role depend on the pathological condition or stimuli. Similar conclusions were drawn in clinical settings delineating an analogous Breg population phenotypically resembling the murine Bregs that ultimately may be associated with a state of tolerance. Recent studies suggested that Bregs may play a role in the onset of autoimmune diabetes. This review will focus on deciphering the different subclasses of Bregs, their emerging role in autoimmune diabetes, and their potential use as a cell-based therapeutic.
Collapse
Affiliation(s)
- Moufida Ben Nasr
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115.,International Center for T1D, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Science "L. Sacco," University of Milan, 20157 Milan, Italy.,Transplantation Research Center, Nephrology Division, Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Vera Usuelli
- International Center for T1D, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Science "L. Sacco," University of Milan, 20157 Milan, Italy
| | - Andy Joe Seelam
- International Center for T1D, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Science "L. Sacco," University of Milan, 20157 Milan, Italy
| | - Francesca D'Addio
- International Center for T1D, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Science "L. Sacco," University of Milan, 20157 Milan, Italy
| | - Reza Abdi
- Transplantation Research Center, Nephrology Division, Children's Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - James F Markmann
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114; and
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115; .,International Center for T1D, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Science "L. Sacco," University of Milan, 20157 Milan, Italy.,Division of Endocrinology, ASST Fatebenefratelli Sacco, 20157 Milan, Italy
| |
Collapse
|
20
|
Feng C, Li L, Zhou L, Li D, Liu M, Han S, Zheng B. Critical roles of the E3 ubiquitin ligase FBW7 in B-cell response and the pathogenesis of experimental autoimmune arthritis. Immunology 2021; 164:617-636. [PMID: 34351636 DOI: 10.1111/imm.13398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 01/04/2023] Open
Abstract
Proper regulation of B-cell function is essential for effective humoral immunity and maintenance of immune tolerance. Here, we found that FBW7 (F-box/WD40 repeat-containing protein 7) is highly expressed in germinal centre B and B1 cells, and confirmed that it has an intrinsic role in maintaining homeostasis of mature B cells and B-1 cells. FBW7 deletion led to an impairment of antibody response, and although germinal centre formation was not affected, antibody class-switch recombination and affinity maturation processes were defective. Likewise, memory immune response was severely impaired. Moreover, FBW7 ablation ameliorated the pathogenesis of an autoimmune disease model, collagen-induced arthritis, by reducing the production of anti-collagen II autoantibodies. Taken together, these data suggest that FBW7 may be an attractive target for developing new therapeutics for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Chunlei Feng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Lingyun Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Lei Zhou
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Shuhua Han
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Biao Zheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
21
|
Azizov V, Zaiss MM. Alcohol Consumption in Rheumatoid Arthritis: A Path through the Immune System. Nutrients 2021; 13:1324. [PMID: 33923766 PMCID: PMC8072698 DOI: 10.3390/nu13041324] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/27/2022] Open
Abstract
Benefits and harms of different components of human diet have been known for hundreds of years. Alcohol is one the highest consumed, abused, and addictive substances worldwide. Consequences of alcohol abuse are increased risks for diseases of the cardiovascular system, liver, and nervous system, as well as reduced immune system function. Paradoxically, alcohol has also been a consistent protective factor against the development of autoimmune diseases such as type 1 diabetes, multiple sclerosis, systemic lupus erythematosus, and rheumatoid arthritis (RA). Here, we focused on summarizing current findings on the effects of alcohol, as well as of its metabolites, acetaldehyde and acetate, on the immune system and RA. Heavy or moderate alcohol consumption can affect intestinal barrier integrity, as well as the microbiome, possibly contributing to RA. Additionally, systemic increase in acetate negatively affects humoral immune response, diminishing TFH cell as well as professional antigen-presenting cell (APC) function. Hence, alcohol consumption has profound effects on the efficacy of vaccinations, but also elicits protection against autoimmune diseases. The mechanism of alcohol's negative effects on the immune system is multivariate. Future studies addressing alcohol and its metabolite acetate's effect on individual components of the immune system remains crucial for our understanding and development of novel therapeutic pathways.
Collapse
Affiliation(s)
- Vugar Azizov
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany;
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Mario M. Zaiss
- Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany;
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
22
|
Neuraminidase Inhibitor Zanamivir Ameliorates Collagen-Induced Arthritis. Int J Mol Sci 2021; 22:ijms22031428. [PMID: 33572654 PMCID: PMC7867009 DOI: 10.3390/ijms22031428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/27/2021] [Accepted: 01/27/2021] [Indexed: 01/20/2023] Open
Abstract
Altered sialylation patterns play a role in chronic autoimmune diseases such as rheumatoid arthritis (RA). Recent studies have shown the pro-inflammatory activities of immunoglobulins (Igs) with desialylated sugar moieties. The role of neuraminidases (NEUs), enzymes which are responsible for the cleavage of terminal sialic acids (SA) from sialoglycoconjugates, is not fully understood in RA. We investigated the impact of zanamivir, an inhibitor of the influenza virus neuraminidase, and mammalian NEU2/3 on clinical outcomes in experimental arthritides studies. The severity of arthritis was monitored and IgG titers were measured by ELISA. (2,6)-linked SA was determined on IgG by ELISA and on cell surfaces by flow cytometry. Zanamivir at a dose of 100 mg/kg (zana-100) significantly ameliorated collagen-induced arthritis (CIA), whereas zana-100 was ineffective in serum transfer-induced arthritis. Systemic zana-100 treatment reduced the number of splenic CD138+/TACI+ plasma cells and CD19+ B cells, which was associated with lower IgG levels and an increased sialylation status of IgG compared to controls. Our data reveal the contribution of NEU2/3 in CIA. Zanamivir down-modulated the T and B cell-dependent humoral immune response and induced an anti-inflammatory milieu by inhibiting sialic acid degradation. We suggest that neuraminidases might represent a promising therapeutic target for RA and possibly also for other antibody-mediated autoimmune diseases.
Collapse
|
23
|
Mohd Jaya FN, Liu Z, Chan GCF. Early Treatment of Interleukin-33 can Attenuate Lupus Development in Young NZB/W F1 Mice. Cells 2020; 9:cells9112448. [PMID: 33182616 PMCID: PMC7696801 DOI: 10.3390/cells9112448] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 01/12/2023] Open
Abstract
Interleukin-33 (IL-33), a member of the IL-1 cytokine family, has been recently associated with the development of autoimmune diseases, including systemic lupus erythematosus (SLE). IL-33 is an alarmin and a pleiotropic cytokine that affects various types of immune cells via binding to its receptor, ST2. In this study, we determine the impact of intraperitoneal IL-33 treatments in young lupus, NZB/W F1 mice. Mice were treated from the age of 6 to 11 weeks. We then assessed the proteinuria level, renal damage, survival rate, and anti-dsDNA antibodies. The induction of regulatory B (Breg) cells, changes in the level of autoantibodies, and gene expression were also examined. In comparison to the control group, young NZB/W F1 mice administered with IL-33 had a better survival rate as well as reduced proteinuria level and lupus nephritis. IL-33 treatments significantly increased the level of IgM anti-dsDNA antibodies, IL-10 expressing Breg cells, and alternatively-induced M2 macrophage gene signatures. These results imply that IL-33 exhibits a regulatory role during lupus onset via the expansion of protective IgM anti-dsDNA as well as regulatory cells such as Breg cells and M2 macrophages.
Collapse
|
24
|
Antigen presentation, autoantibody production, and therapeutic targets in autoimmune liver disease. Cell Mol Immunol 2020; 18:92-111. [PMID: 33110250 PMCID: PMC7852534 DOI: 10.1038/s41423-020-00568-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023] Open
Abstract
The liver is an important immunological organ that controls systemic tolerance. The liver harbors professional and unconventional antigen-presenting cells that are crucial for tolerance induction and maintenance. Orchestrating the immune response in homeostasis depends on a healthy and well-toned immunological liver microenvironment, which is maintained by the crosstalk of liver-resident antigen-presenting cells and intrahepatic and liver-infiltrating leukocytes. In response to pathogens or autoantigens, tolerance is disrupted by unknown mechanisms. Intrahepatic parenchymal and nonparenchymal cells exhibit unique antigen-presenting properties. The presentation of microbial and endogenous lipid-, metabolite- and peptide-derived antigens from the gut via conventional and nonconventional mechanisms can educate intrahepatic immune cells and elicit effector responses or tolerance. Perturbation of this balance results in autoimmune liver diseases, such as autoimmune hepatitis, primary biliary cholangitis, and primary sclerosing cholangitis. Although the exact etiologies of these autoimmune liver diseases are unknown, it is thought that the disruption of tolerance towards self-antigens and microbial metabolites and lipids, as well as alterations in bile acid composition, may result in changes in effector cell activation and polarization and may reduce or impair protective anti-inflammatory regulatory T and B cell responses. Additionally, the canonical and noncanonical transmission of antigens and antigen:MHC complexes via trogocytosis or extracellular vesicles between different (non) immune cells in the liver may play a role in the induction of hepatic inflammation and tolerance. Here, we summarize emerging aspects of antigen presentation, autoantibody production, and the application of novel therapeutic approaches in the characterization and treatment of autoimmune liver diseases.
Collapse
|
25
|
Decker DA, Higgins P, Hayes K, Bollinger C, Becker P, Wright D. Repository corticotropin injection attenuates collagen-induced arthritic joint structural damage and has enhanced effects in combination with etanercept. BMC Musculoskelet Disord 2020; 21:586. [PMID: 32867752 PMCID: PMC7460755 DOI: 10.1186/s12891-020-03609-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 08/24/2020] [Indexed: 12/29/2022] Open
Abstract
Background Melanocortin receptor (MCR) agonists have anti-inflammatory and immunomodulatory properties mediated by receptors expressed on cells relevant to arthritis. Repository corticotropin injection (RCI; Acthar® Gel), an MCR agonist preparation, is approved as adjunctive therapy for rheumatoid arthritis (RA), but its mechanism of action in RA is unclear. This study explored the efficacy of RCI as monotherapy or adjunctive therapy with etanercept (ETN) in an established animal model of collagen-induced arthritis (CIA). Methods After induction of CIA, rats (n = 10 per group) were randomized to receive subcutaneous RCI (40, 160, or 400 U/kg twice daily) alone or in combination with ETN (10 mg/kg 3 times daily), ETN alone, or vehicle (on days 13 through 19). Inflammation was assessed via changes in paw edema. Bone damage was determined by microfocal computed tomography histopathology, and immunohistochemistry. Statistical analyses were performed using a 2-way analysis of variance (ANOVA) followed by the Newman-Keuls, Dunn’s, or Dunnett’s multiple comparisons test or a 1-way ANOVA followed by the Dunnett’s or Holm-Sidak multiple comparisons test. Results RCI administration resulted in dose-dependent decreases in ankle edema and histopathologic measures of inflammation, pannus formation, cartilage damage, bone resorption, and periosteal bone formation. RCI and ETN showed combined benefits on all parameters measured. Radiographic evidence of bone damage was significantly reduced in rats that received RCI alone or in combination with ETN. This reduction in bone density loss correlated with decreases in the number of CD68-positive macrophages and cathepsin K–positive osteoclasts within the lesions. Conclusions As monotherapy or adjunctive therapy with ETN, RCI attenuated CIA-induced joint structural damage in rats. These data support the clinical efficacy of RCI as adjunctive therapy for patients with RA.
Collapse
Affiliation(s)
- Dima A Decker
- Former employee of Mallinckrodt Pharmaceuticals, Bedminster, NJ, USA
| | - Paul Higgins
- Former employee of Mallinckrodt Pharmaceuticals, Bedminster, NJ, USA
| | - Kyle Hayes
- Mallinckrodt Pharmaceuticals, 675 McDonnell Blvd, Bedminster, NJ, 63042, USA
| | - Chris Bollinger
- Mallinckrodt Pharmaceuticals, 675 McDonnell Blvd, Bedminster, NJ, 63042, USA
| | - Patrice Becker
- Former employee of Mallinckrodt Pharmaceuticals, Bedminster, NJ, USA
| | - Dale Wright
- Mallinckrodt Pharmaceuticals, 675 McDonnell Blvd, Bedminster, NJ, 63042, USA.
| |
Collapse
|
26
|
Lewis BJ, Branch DR. Mouse Models of Rheumatoid Arthritis for Studies on Immunopathogenesis and Preclinical Testing of Fc Receptor-Targeting Biologics. Pharmacology 2020; 105:618-629. [DOI: 10.1159/000508239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/27/2020] [Indexed: 11/19/2022]
Abstract
<b><i>Background:</i></b> Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes inflammation, swelling, and pain in the joints and involves systemic complications. Mouse models of RA have been extensively used to model the pathogenesis of RA and to develop effective therapies. Although many components of the immune system have been studied in these models, the role of crystallizable fragment (Fc) gamma receptors (FcγRs) in RA has been sorely neglected. The aim of this review was to introduce the different mouse models of RA and to describe the different drug development strategies that have been tested in these models to target FcγR function, with the focus being on drugs that have been made from the Fc of immunoglobulin G (IgG). <b><i>Summary:</i></b> Evidence suggests that FcγRs play a major role in immune complex-induced inflammation in autoimmune diseases, such as RA. However, there is limited knowledge on the importance of FcγRs in the human disease even though there has been extensive work in mouse models of RA. Numerous mouse models of RA are available, with each model depicting certain aspects of the disease. Induced models of RA have nonspecific immune activation with cartilage-directed autoimmunity, whereas spontaneous models of RA develop without immunization, which results in a more chronic form of arthritis. These models have been used to test FcγR-targeting monoclonal antibodies, intravenous immunoglobulin (IVIg), subcutaneously administered IVIg, and recombinant Fcs for their ability to interact with and modify FcγR function. Recombinant Fcs avidly bind FcγRs and exhibit enhanced therapeutic efficacy in mouse models of RA. <b><i>Key Message:</i></b> The therapeutic utility of targeting FcγRs with recombinant Fcs is great and should be explored in human clinical trials for autoimmune diseases, such as RA.
Collapse
|
27
|
Wang J, Li Y, Li L, Yang J, Kopeček J. Exploration and Evaluation of Therapeutic Efficacy of Drug-Free Macromolecular Therapeutics in Collagen-Induced Rheumatoid Arthritis Mouse Model. Macromol Biosci 2020; 20:e1900445. [PMID: 32196951 PMCID: PMC7549750 DOI: 10.1002/mabi.201900445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 12/21/2022]
Abstract
Monoclonal antibodies (mAbs) against B cell antigens are extensively used in the treatment of rheumatoid arthritis (RA). The B cell depletion therapy prevents RA symptoms and/or alleviates existing inflammation. The previously established two-step drug-free macromolecular therapeutics (DFMT) is applied in the treatment of collagen-induced rheumatoid arthritis in a collagen-induced rheumatoid arthritis mouse model. DFMT is a B cell depletion strategy utilizing Fab' fragment of anti-CD20 mAb for biorecognition and receptor crosslinking to induce B cell apoptosis. DFMT is composed from two nanoconjugates: 1) bispecific engager, Fab'-MORF1 (anti-CD20 Fab' fragment conjugated with morpholino oligonucleotide MORF1), and 2) a crosslinking (effector) component P-(MORF2)X (N-(2-hydroxypropyl)methacrylamide copolymer grafted with multiple copies of complementary morpholino oligonucleotide MORF2). The absence of Fc fragment has the potential to avoid development of resistance and infusion-related reactions. DFMT produces B cell depletion, keeps the RA score low for more than 100 days, and shows minimal cartilage and bone erosion and inflammatory cell infiltration. Further improvements will be explored to optimize DFMT strategy in autoimmune disease treatment.
Collapse
Affiliation(s)
- Jiawei Wang
- Department of Pharmaceutics and Pharmaceutical Chemistry/Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, USA
| | - Yachao Li
- Department of Pharmaceutics and Pharmaceutical Chemistry/Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, USA
| | - Lian Li
- Department of Pharmaceutics and Pharmaceutical Chemistry/Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, USA
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry/Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, Utah 84112, USA
| | - Jindřich Kopeček
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
28
|
Dimitrijević M, Arsenović-Ranin N, Kosec D, Bufan B, Nacka-Aleksić M, Pilipović I, Leposavić G. Sex differences in Tfh cell help to B cells contribute to sexual dimorphism in severity of rat collagen-induced arthritis. Sci Rep 2020; 10:1214. [PMID: 31988383 PMCID: PMC6985112 DOI: 10.1038/s41598-020-58127-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/06/2020] [Indexed: 12/03/2022] Open
Abstract
The study examined germinal centre (GC) reaction in lymph nodes draining inflamed joints and adjacent tissues (dLNs) in male and female Dark Agouti rat collagen type II (CII)-induced arthritis (CIA) model of rheumatoid arthritis. Female rats exhibiting the greater susceptibility to CIA mounted stronger serum CII-specific IgG response than their male counterparts. This correlated with the higher frequency of GC B cells in female compared with male dLNs. Consistently, the frequency of activated/proliferating Ki-67+ cells among dLN B cells was higher in females than in males. This correlated with the shift in dLN T follicular regulatory (Tfr)/T follicular helper (Tfh) cell ratio towards Tfh cells in females, and greater densities of CD40L and CD40 on their dLN T and B cells, respectively. The higher Tfh cell frequency in females was consistent with the greater dLN expression of mRNA for IL-21/27, the key cytokines involved in Tfh cell generation and their help to B cells. Additionally, in CII-stimulated female rat dLN cell cultures IFN-γ/IL-4 production ratio was shifted towards IFN-γ. Consistently, the serum IgG2a(b)/IgG1 CII-specific antibody ratio was shifted towards an IgG2a(b) response in females. Thus, targeting T-/B-cell interactions should be considered in putative further sex-based translational pharmacology research.
Collapse
Affiliation(s)
- Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research "Siniša Stanković", 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
| | - Duško Kosec
- Immunology Research Center "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Vojvode Stepe, 458, 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
| | - Ivan Pilipović
- Immunology Research Center "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Vojvode Stepe, 458, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe, 450, Belgrade, Serbia.
| |
Collapse
|
29
|
IL-10 producing B cells rescue mouse fetuses from inflammation-driven fetal death and are able to modulate T cell immune responses. Sci Rep 2019; 9:9335. [PMID: 31249364 PMCID: PMC6597542 DOI: 10.1038/s41598-019-45860-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/11/2019] [Indexed: 11/26/2022] Open
Abstract
Understanding the mechanisms leading to fetal death following maternal subclinical infections is crucial to develop new therapeutic strategies. Here we addressed the relevance of IL-10 secreting B cells (B10) in the maintenance of the immune balance during gestation. µMT females lacking mature B cells presented normal pregnancies, although their fetuses were smaller and their Treg pool did not expand as in B cell sufficient controls. Pregnant µMT females were more susceptible to LPS despite having less Treg; their fetuses died at doses compatible with pregnancy in WT animals. Adoptive transfer of IL-10 negative B effector cells or B cells from IL-10 deficient mice did not modify this outcome. The transfer of B10 cells or application of recombinant murine IL-10 reduced the fetal loss, associated with a normalization of Treg numbers and cytokine modulation at the feto-maternal interface. B cell-derived IL-10 suppressed the production of IL-17A and IL-6 by T cells and promoted the conversion of naïve cells into Treg. B10 cells are required to restore the immune balance at the feto-maternal interface when perturbed by inflammatory signals. Our data position B cells in a central role in the maintenance of the balance between immunity and tolerance during pregnancy.
Collapse
|
30
|
Collagen-induced arthritis in Dark Agouti rats as a model for study of immunological sexual dimorphisms in the human disease. Exp Mol Pathol 2018; 105:10-22. [DOI: 10.1016/j.yexmp.2018.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/05/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022]
|
31
|
Yao Y, Huang W, Li X, Li X, Qian J, Han H, Sun H, An X, Lu L, Zhao H. Tespa1 Deficiency Dampens Thymus-Dependent B-Cell Activation and Attenuates Collagen-Induced Arthritis in Mice. Front Immunol 2018; 9:965. [PMID: 29867947 PMCID: PMC5960706 DOI: 10.3389/fimmu.2018.00965] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022] Open
Abstract
Thymocyte-expressed, positive selection-associated 1 (Tespa1) plays an important role in both T cell receptor (TCR)-driven thymocyte development and in the FcεRI-mediated activation of mast cells. Herein, we show that lack of Tespa1 does not impair B cell development but dampens the in vitro activation and proliferation of B cells induced by T cell-dependent (TD) antigens, significantly reduces serum antibody concentrations in vivo, and impairs germinal center formation in both aged and TD antigen-immunized mice. We also provide evidence that dysregulated signaling in Tespa1-deficient B cells may be linked to CD40-induced TRAF6 degradation, and subsequent effects on 1-phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2 (PLCγ2) phosphorylation, MAPK activation, and calcium influx. Furthermore, we demonstrate that Tespa1 plays a critical role in pathogenic B cells, since Tespa1-deficient chimeric mice showed a lower incidence and clinical disease severity of collagen-induced arthritis. Overall, our study demonstrates that Tespa1 is essential for TD B cell responses, and suggests an important role for Tespa1 during the development of autoimmune arthritis.
Collapse
Affiliation(s)
- Yunliang Yao
- Program in Molecular and Translational Medicine (PMTM), School of Medicine, Huzhou University, Huzhou, China
| | - Wei Huang
- The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Li
- Program in Molecular and Translational Medicine (PMTM), School of Medicine, Huzhou University, Huzhou, China
| | - Xiawei Li
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jin Qian
- Program in Molecular and Translational Medicine (PMTM), School of Medicine, Huzhou University, Huzhou, China
| | - Hui Han
- First Affiliated Hospital, Huzhou University, Huzhou, China
| | - Hui Sun
- First Affiliated Hospital, Huzhou University, Huzhou, China
| | - Xiangli An
- Program in Molecular and Translational Medicine (PMTM), School of Medicine, Huzhou University, Huzhou, China
| | - Linrong Lu
- School of Medicine, Institute of Immunology, Zhejiang University, Hangzhou, China
| | - Hongxing Zhao
- First Affiliated Hospital, Huzhou University, Huzhou, China
| |
Collapse
|
32
|
Roser-Page S, Vikulina T, Weiss D, Habib MM, Beck GR, Pacifici R, Lane TF, Weitzmann MN. CTLA-4Ig (abatacept) balances bone anabolic effects of T cells and Wnt-10b with antianabolic effects of osteoblastic sclerostin. Ann N Y Acad Sci 2018; 1415:21-33. [PMID: 29500936 DOI: 10.1111/nyas.13643] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 12/12/2022]
Abstract
Activated lymphocytes promote inflammation and bone destruction in rheumatoid arthritis (RA), making T cells and B cells therapeutic targets. Indeed, pharmacological blockade of CD28 costimulation using CTLA-4Ig (abatacept), approved for amelioration of RA, renders T cells dormant (anergic). CTLA-4Ig also promotes bone accretion in healthy mice; surprisingly, however, this effect is driven exclusively through upregulation of bone formation, rather than anti-inflammatory effects on resorption. In the study presented here, we utilized T cell receptor β gene and Wnt-10b gene knockout mice to investigate the roles of T cells and Wnt-10b in CTLA-4Ig-induced bone anabolism. Ablation of either T cells or Wnt-10b not only abolished CTLA-4Ig-induced bone anabolism but also, paradoxically, suppressed bone formation leading to bone loss. Stalled bone formation was accompanied by bone marrow stromal cell expression of the Wnt pathway inhibitor sclerostin. Our data suggest that an immunoskeletal pivot may promote or suppress bone formation, depending on the net outcome of CTLA-4Ig action directed independently on T cells and osteoblast-linage cells that counter Wnt-10b-induced bone anabolism, by secretion of sclerostin. While CTLA-4Ig action is tipped in favor of bone formation under physiological conditions, pathological immunodeficiency may lead to suppressed bone formation and skeletal damage.
Collapse
Affiliation(s)
| | - Tatyana Vikulina
- Atlanta VA Medical Center, Decatur, Georgia.,Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Daiana Weiss
- Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Mark M Habib
- Atlanta VA Medical Center, Decatur, Georgia.,Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - George R Beck
- Atlanta VA Medical Center, Decatur, Georgia.,Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Emory Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Roberto Pacifici
- Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia
| | - Timothy F Lane
- Departments of Obstetrics and Gynecology and Biological Chemistry, and Orthopedic Hospital Research Center, University of California Los Angeles, Los Angeles, California
| | - M Neale Weitzmann
- Atlanta VA Medical Center, Decatur, Georgia.,Division of Endocrinology and Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Emory Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
33
|
Dahdah A, Habir K, Nandakumar KS, Saxena A, Xu B, Holmdahl R, Malin S. Germinal Center B Cells Are Essential for Collagen-Induced Arthritis. Arthritis Rheumatol 2018; 70:193-203. [PMID: 29045049 DOI: 10.1002/art.40354] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/12/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is considered to be a prototypical autoimmune disorder. Several mechanisms have been proposed for the known pathologic function of B cells in RA, including antigen presentation, cytokine secretion, and humoral immunity. The aim of this study was to address the function of B lymphocytes in experimental arthritis. METHODS We mapped the adaptive immune response following collagen-induced arthritis (CIA). We subsequently monitored these responses and disease outcomes in genetically modified mouse strains that lack mature B cell or germinal center (GC) functionality in a B cell-intrinsic manner. RESULTS Following primary immunization, the draining lymph nodes broadly reacted against type II collagen (CII) with the formation of GCs and T cell activation. Mice that lacked mature B cell function were fully protected against CIA and had a severely attenuated ability to mount isotype-switched humoral immune responses against CII. Almost identical results were observed in mice that were selectively deficient in GC responses. Importantly, GC-deficient mice were fully susceptible to collagen antibody-induced arthritis. CONCLUSION We identified GC formation and anticollagen antibody production as the key pathogenic functions of B cells in CIA. The role of B cells in RA is likely to be more complex. However, targeting the GC reaction could allow for therapeutic interventions that are more refined than general B cell depletion.
Collapse
Affiliation(s)
- Albert Dahdah
- Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Katrin Habir
- Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Kutty Selva Nandakumar
- Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden, and Southern Medical University, Guangzhou, China
| | - Amit Saxena
- Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Bingze Xu
- Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Rikard Holmdahl
- Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Stephen Malin
- Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Gillooly KM, Pulicicchio C, Pattoli MA, Cheng L, Skala S, Heimrich EM, McIntyre KW, Taylor TL, Kukral DW, Dudhgaonkar S, Nagar J, Banas D, Watterson SH, Tino JA, Fura A, Burke JR. Bruton's tyrosine kinase inhibitor BMS-986142 in experimental models of rheumatoid arthritis enhances efficacy of agents representing clinical standard-of-care. PLoS One 2017; 12:e0181782. [PMID: 28742141 PMCID: PMC5524405 DOI: 10.1371/journal.pone.0181782] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/06/2017] [Indexed: 12/17/2022] Open
Abstract
Bruton’s tyrosine kinase (BTK) regulates critical signal transduction pathways involved in the pathobiology of rheumatoid arthritis (RA) and other autoimmune disorders. BMS-986142 is a potent and highly selective reversible small molecule inhibitor of BTK currently being investigated in clinical trials for the treatment of both RA and primary Sjögren’s syndrome. In the present report, we detail the in vitro and in vivo pharmacology of BMS-986142 and show this agent provides potent and selective inhibition of BTK (IC50 = 0.5 nM), blocks antigen receptor-dependent signaling and functional endpoints (cytokine production, co-stimulatory molecule expression, and proliferation) in human B cells (IC50 ≤ 5 nM), inhibits Fcγ receptor-dependent cytokine production from peripheral blood mononuclear cells, and blocks RANK-L-induced osteoclastogenesis. Through the benefits of impacting these important drivers of autoimmunity, BMS-986142 demonstrated robust efficacy in murine models of rheumatoid arthritis (RA), including collagen-induced arthritis (CIA) and collagen antibody-induced arthritis (CAIA). In both models, robust efficacy was observed without continuous, complete inhibition of BTK. When a suboptimal dose of BMS-986142 was combined with other agents representing the current standard of care for RA (e.g., methotrexate, the TNFα antagonist etanercept, or the murine form of CTLA4-Ig) in the CIA model, improved efficacy compared to either agent alone was observed. The results suggest BMS-986142 represents a potential therapeutic for clinical investigation in RA, as monotherapy or co-administered with agents with complementary mechanisms of action.
Collapse
Affiliation(s)
- Kathleen M. Gillooly
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Claudine Pulicicchio
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Mark A. Pattoli
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Lihong Cheng
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Stacey Skala
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Elizabeth M. Heimrich
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Kim W. McIntyre
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Tracy L. Taylor
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Daniel W. Kukral
- Exploratory Clinical and Translational Research, Imaging, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Shailesh Dudhgaonkar
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb Research Center, Syngene International Limited, Bangalore, India
| | - Jignesh Nagar
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb Research Center, Syngene International Limited, Bangalore, India
| | - Dana Banas
- Discovery Translational Sciences, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Scott H. Watterson
- Immunosciences Discovery Chemistry, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Joseph A. Tino
- Immunosciences Discovery Chemistry, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - Aberra Fura
- Department of Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
| | - James R. Burke
- Immunosciences Discovery Biology, Bristol-Myers Squibb Research & Development, Princeton, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
35
|
Yamashita M, Matsumoto K, Endo T, Ukibe K, Hosoya T, Matsubara Y, Nakagawa H, Sakai F, Miyazaki T. Preventive Effect of Lactobacillus helveticus SBT2171 on Collagen-Induced Arthritis in Mice. Front Microbiol 2017; 8:1159. [PMID: 28680422 PMCID: PMC5478730 DOI: 10.3389/fmicb.2017.01159] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/07/2017] [Indexed: 12/18/2022] Open
Abstract
We recently reported that the intraperitoneal inoculation of Lactobacillus helveticus SBT2171 inhibited the development of collagen-induced arthritis (CIA), a murine model of rheumatoid arthritis (RA). In the present study, we evaluated the effect of the oral administration of L. helveticus SBT2171 on CIA development and on the regulation of antigen-specific antibody production and inflammatory immune cells, which have been implicated in the development of RA. Both oral administration and intraperitoneal inoculation of L. helveticus SBT2171 reduced joint swelling, body weight loss, and the serum level of bovine type II collagen (CII)-specific antibodies in the CIA mouse model. The intraperitoneal inoculation also decreased the arthritis incidence, joint damage, and serum level of interleukin (IL)-6. In addition, the numbers of total immune cells, total B cells, germinal center B cells, and CD4+ T cells in the draining lymph nodes were decreased following intraperitoneal inoculation of L. helveticus SBT2171. These findings demonstrate the ability of L. helveticus SBT2171 to downregulate the abundance of immune cells and the subsequent production of CII-specific antibodies and IL-6, thereby suppressing the CIA symptoms, indicating its potential for use in the prevention of RA.
Collapse
Affiliation(s)
- Maya Yamashita
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd.Saitama, Japan
| | - Kurumi Matsumoto
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd.Saitama, Japan
| | - Tsutomu Endo
- Department of Orthopedic Surgery, Graduate School of Medicine, Hokkaido UniversitySapporo, Japan
| | - Ken Ukibe
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd.Saitama, Japan
| | - Tomohiro Hosoya
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd.Saitama, Japan
| | - Yumi Matsubara
- Department of Probiotics Immunology, Institute for Genetic Medicine, Hokkaido UniversitySapporo, Japan
| | - Hisako Nakagawa
- Department of Probiotics Immunology, Institute for Genetic Medicine, Hokkaido UniversitySapporo, Japan
| | - Fumihiko Sakai
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd.Saitama, Japan
| | - Tadaaki Miyazaki
- Department of Probiotics Immunology, Institute for Genetic Medicine, Hokkaido UniversitySapporo, Japan
| |
Collapse
|
36
|
Behrens M, Luckey D, Luthra H, David C, Taneja V. B cells influence sex specificity of arthritis via myeloid suppressors and chemokines in humanized mice. Clin Immunol 2017; 178:10-19. [DOI: 10.1016/j.clim.2015.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/15/2015] [Accepted: 05/31/2015] [Indexed: 12/15/2022]
|
37
|
Kim SJ, Chen Z, Essani AB, Elshabrawy HA, Volin MV, Fantuzzi G, McInnes IB, Baker JF, Finn P, Kondos G, Volkov S, Swedler W, Arami S, Sweiss N, Shahrara S. Differential impact of obesity on the pathogenesis of RA or preclinical models is contingent on the disease status. Ann Rheum Dis 2017; 76:731-739. [PMID: 27797749 PMCID: PMC10026536 DOI: 10.1136/annrheumdis-2016-209206] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 07/15/2016] [Accepted: 09/04/2016] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Studies were performed to uncover the significance of obesity in rheumatoid arthritis (RA) and preclinical models. METHODS Preclinical arthritis models were used to examine the impact of obesity on disease onset and remission. Conditioned media from RA adipose tissues were used to investigate the mechanism contributing to joint neutrophil influx and M1 macrophage differentiation observed in early and remission phases of arthritis. RESULTS We report that mice fed with high fat diet (HFD) have an earlier onset of collagen-induced arthritis (CIA) compared with mice on regular diet. However, the differences in CIA joint swelling between the two diet groups are lost once disease is established. We found that early arthritis triggered by obesity is due to elevated joint MIP2/interleukin-8 levels detected in CIA as well as in the RA and mouse adipose tissues and the effect of this chemokine on neutrophil recruitment. Although active disease progression is similarly affected in both diet groups, arthritis resolution is accelerated in lean mice while joint inflammation is sustained in obese mice. We document that HFD can prolong toll-like receptor (TLR)4-induced arthritis by increasing joint monocyte migration and further remodelling the recruited cells into M1 macrophages. Consistently, we show that adipose condition media can transform RA and wild-type naïve myeloid cells into M1 macrophages; however, this function is impaired by TLR4 blockade or deficiency. CONCLUSIONS We conclude that despite established disease being unaffected by obesity, the early and the resolution phases of RA are impacted by obesity through different mechanisms.
Collapse
Affiliation(s)
- Seung-jae Kim
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, IL 60612
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Zhenlong Chen
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, IL 60612
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Abdul B. Essani
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, IL 60612
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Hatem A. Elshabrawy
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, IL 60612
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Michael V. Volin
- Department of Microbiology & Immunology, Midwestern University, Downers Grove, IL 60515
| | - Giamila Fantuzzi
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, IL 60612
| | - Iain B. McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Joshua F. Baker
- Philadelphia VA medical center and Division of Rheumatology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104
| | - Patricia Finn
- Division of Pulmonary, University of Illinois at Chicago, IL 60612
| | - George Kondos
- Division of Cardiology, University of Illinois at Chicago, IL 60612
| | - Suncica Volkov
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - William Swedler
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, IL 60612
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Shiva Arami
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, IL 60612
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| | - Nadera Sweiss
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, IL 60612
| | - Shiva Shahrara
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, IL 60612
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612
| |
Collapse
|
38
|
He M, Harms AC, van Wijk E, Wang M, Berger R, Koval S, Hankemeier T, van der Greef J. Role of amino acids in rheumatoid arthritis studied by metabolomics. Int J Rheum Dis 2017; 22:38-46. [PMID: 28328075 DOI: 10.1111/1756-185x.13062] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a complex, chronic autoimmune disease characterized by various inflammatory symptoms, including joint swelling, joint pain, and both structural and functional joint damage. The most commonly used animal model for studying RA is mice with collagen-induced arthritis (CIA); the wide use of this model is due primarily to many similarities with RA in human patients. Metabolomics is used increasingly in biological studies for diagnosing disease and for predicting and evaluating drug interventions, as a large number of disease-associated metabolites can be analyzed and interpreted from a biological perspective. AIM To profile free amino acids and their biogenic metabolites in CIA mice plasma. METHOD Ultra-high-performance liquid chromatography/tandem mass spectrometry coupled with multiple reaction monitoring (MRM) was used for metabolomics study. RESULTS Profile of 45 amine metabolites, including free amino acids and their biogenic metabolites in plasma was obtained from CIA mice. We found that the plasma levels of 20 amine metabolites were significantly decreased in the CIA group. CONCLUSION The results suggest that a disordered amine response is linked to RA-associated muscle wasting and energy expenditure.
Collapse
Affiliation(s)
- Min He
- Analytical BioSciences, LACDR, Leiden University, Leiden, the Netherlands.,Sino-Dutch Center for Preventive and Personalized Medicine, Leiden University, Leiden, the Netherlands
| | - Amy C Harms
- Analytical BioSciences, LACDR, Leiden University, Leiden, the Netherlands
| | - Eduard van Wijk
- Analytical BioSciences, LACDR, Leiden University, Leiden, the Netherlands.,Sino-Dutch Center for Preventive and Personalized Medicine, Leiden University, Leiden, the Netherlands
| | - Mei Wang
- Analytical BioSciences, LACDR, Leiden University, Leiden, the Netherlands.,Sino-Dutch Center for Preventive and Personalized Medicine, Leiden University, Leiden, the Netherlands.,SU Biomedicine, Zeist, the Netherlands
| | - Ruud Berger
- Analytical BioSciences, LACDR, Leiden University, Leiden, the Netherlands
| | - Slavik Koval
- Analytical BioSciences, LACDR, Leiden University, Leiden, the Netherlands
| | - Thomas Hankemeier
- Analytical BioSciences, LACDR, Leiden University, Leiden, the Netherlands.,Sino-Dutch Center for Preventive and Personalized Medicine, Leiden University, Leiden, the Netherlands
| | - Jan van der Greef
- Analytical BioSciences, LACDR, Leiden University, Leiden, the Netherlands.,Sino-Dutch Center for Preventive and Personalized Medicine, Leiden University, Leiden, the Netherlands.,TNO, Zeist, the Netherlands
| |
Collapse
|
39
|
Levels MJ, Van Tok MN, Cantaert T, Cañete JD, Kroese FGM, Germar K, Spits H, Baeten DLP, Yeremenko NG. The Transcriptional Coactivator Bob1 Is Associated With Pathologic B Cell Responses in Autoimmune Tissue Inflammation. Arthritis Rheumatol 2017; 69:750-762. [DOI: 10.1002/art.39993] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 11/08/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Maria J. Levels
- Academic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Melissa N. Van Tok
- Academic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Tineke Cantaert
- Academic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Juan D. Cañete
- Hospital Clinic of Barcelona and Institut d'Investigacions Biomèdiques August Pi i SunyerBarcelona Spain
| | | | - Kristine Germar
- Academic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Hergen Spits
- Academic Medical Center, University of Amsterdam and AIMM TherapeuticsAmsterdam The Netherlands
| | | | | |
Collapse
|
40
|
|
41
|
Franks SE, Getahun A, Hogarth PM, Cambier JC. Targeting B cells in treatment of autoimmunity. Curr Opin Immunol 2016; 43:39-45. [PMID: 27718447 DOI: 10.1016/j.coi.2016.09.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/10/2016] [Accepted: 09/13/2016] [Indexed: 01/06/2023]
Abstract
B cells have emerged as effective targets for therapeutic intervention in autoimmunities in which the ultimate effectors are antibodies, as well as those in which T cells are primary drivers of inflammation. Proof of this principle has come primarily from studies of the efficacy of Rituximab, an anti-CD20 mAb that depletes B cells, in various autoimmune settings. These successes have inspired efforts to develop more effective anti-CD20s tailored for specific needs, as well as biologicals and small molecules that suppress B cell function without the risks inherent in B cell depletion. Here we review the current status of B cell-targeted therapies for autoimmunity.
Collapse
Affiliation(s)
- S Elizabeth Franks
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Denver, CO, USA
| | - Andrew Getahun
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Denver, CO, USA; Department of Biomedical Research, National Jewish Health, Denver, CO, USA
| | - P Mark Hogarth
- Centre for Biomedicine, Burnet Institute, Melbourne, Vic., Australia; Department of Immunology, Monash University, Melbourne, Vic., Australia; Department of Pathology, University of Melbourne, Melbourne, Vic., Australia
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Denver, CO, USA; Department of Biomedical Research, National Jewish Health, Denver, CO, USA.
| |
Collapse
|
42
|
Lino AC, Dörner T, Bar-Or A, Fillatreau S. Cytokine-producing B cells: a translational view on their roles in human and mouse autoimmune diseases. Immunol Rev 2016; 269:130-44. [PMID: 26683150 DOI: 10.1111/imr.12374] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
B-cell depletion therapy has beneficial effects in autoimmune diseases. This is only partly explained by an elimination of autoantibodies. How does B-cell depletion improve disease? Here, we review preclinical studies showing that B cells can propagate autoimmune disorders through cytokine production. We also highlight clinical observations indicating the relevance of these B-cell functions in human autoimmunity. Abnormalities in B-cell cytokine production have been observed in rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, and systemic lupus erythematosus. In the first two diseases, B-cell depletion erases these abnormalities, and improves disease progression, suggesting a causative role for defective B-cell cytokine expression in disease pathogenesis. However, in the last two disorders, the pathogenic role of B cells and the effect of B-cell depletion on cytokine-producing B cells remain to be clarified. A better characterization of cytokine-expressing human B-cell subsets, and their modulation by B cell-targeted therapies might help understanding both the successes and failures of current B cell-targeted approaches. This may even lead to the development of novel strategies to deplete or amplify selectively pathogenic or protective subsets, respectively, which might be more effective than global depletion of the B-cell compartment.
Collapse
Affiliation(s)
- Andreia C Lino
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany
| | - Thomas Dörner
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany.,CC12, Department of Medicine/Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Amit Bar-Or
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, 3801 University, Montreal, QC, Canada
| | - Simon Fillatreau
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany.,Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants Malades, Paris, France
| |
Collapse
|
43
|
Abstract
Osteoporosis develops when the rate of osteoclastic bone breakdown (resorption) exceeds that of osteoblastic bone formation, which leads to loss of BMD and deterioration of bone structure and strength. Osteoporosis increases the risk of fragility fractures, a cause of substantial morbidity and mortality, especially in elderly patients. This imbalance between bone formation and bone resorption is brought about by natural ageing processes, but is frequently exacerbated by a number of pathological conditions. Of importance to the aetiology of osteoporosis are findings over the past two decades attesting to a deep integration of the skeletal system with the immune system (the immuno-skeletal interface (ISI)). Although protective of the skeleton under physiological conditions, the ISI might contribute to bone destruction in a growing number of pathophysiological states. Although numerous research groups have investigated how the immune system affects basal and pathological osteoclastic bone resorption, recent findings suggest that the reach of the adaptive immune response extends to the regulation of osteoblastic bone formation. This Review examines the evolution of the field of osteoimmunology and how advances in our understanding of the ISI might lead to novel approaches to prevent and treat bone loss, and avert fractures.
Collapse
Affiliation(s)
- M Neale Weitzmann
- The Atlanta Department of Veterans Affairs Medical Center, 1670 Clairmont Road, Decatur, Georgia, 30033, USA
- Department of Medicine, Division of Endocrinology and Metabolism and Lipids, Emory University School of Medicine, 101 Woodruff Circle, 1305 WMB, Atlanta, Georgia 30322, USA
| | - Ighovwerha Ofotokun
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, 49 Jesse Hill Jr Drive, Atlanta, Georgia 30303, USA
- Grady Healthcare System, 80 Jesse Hill Jr Drive SE, Atlanta, Georgia, 30303, USA
| |
Collapse
|
44
|
Iglesias M, Augustin JJ, Alvarez P, Santiuste I, Postigo J, Merino J, Merino R. Selective Impairment of TH17-Differentiation and Protection against Autoimmune Arthritis after Overexpression of BCL2A1 in T Lymphocytes. PLoS One 2016; 11:e0159714. [PMID: 27433938 PMCID: PMC4951111 DOI: 10.1371/journal.pone.0159714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/06/2016] [Indexed: 11/29/2022] Open
Abstract
The inhibition of apoptotic cell death in T cells through the dysregulated expression of BCL2 family members has been associated with the protection against the development of different autoimmune diseases. However, multiple mechanisms were proposed to be responsible for such protective effect. The purpose of this study was to explore the effect of the T-cell overexpression of BCL2A1, an anti-apoptotic BCL2 family member without an effect on cell cycle progression, in the development of collagen-induced arthritis. Our results demonstrated an attenuated development of arthritis in these transgenic mice. The protective effect was unrelated to the suppressive activity of regulatory T cells but it was associated with a defective activation of p38 mitogen-activated protein kinase in CD4+ cells after in vitro TCR stimulation. In addition, the in vitro and in vivo TH17 differentiation were impaired in BCL2A1 transgenic mice. Taken together, we demonstrated here a previously unknown role for BCL2A1 controlling the activation of CD4+ cells and their differentiation into pathogenic proinflammatory TH17 cells and identified BCL2A1 as a potential target in the control of autoimmune/inflammatory diseases.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Apoptosis/immunology
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Autoimmunity
- CD4 Antigens/genetics
- CD4 Antigens/immunology
- Cell Differentiation
- Cytokines/genetics
- Cytokines/immunology
- Gene Expression Regulation
- Lymphocyte Activation
- Mice
- Mice, Transgenic
- Minor Histocompatibility Antigens/genetics
- Minor Histocompatibility Antigens/immunology
- Protective Factors
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Th17 Cells/immunology
- Th17 Cells/pathology
- p38 Mitogen-Activated Protein Kinases/genetics
- p38 Mitogen-Activated Protein Kinases/immunology
Collapse
Affiliation(s)
- Marcos Iglesias
- Departamento de Biología Molecular-IDIVAL Universidad de Cantabria, Santander, Spain
| | - Juan Jesús Augustin
- Departamento de Biología Molecular-IDIVAL Universidad de Cantabria, Santander, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas-Universidad de Cantabria, Santander, Spain
| | - Pilar Alvarez
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas-Universidad de Cantabria, Santander, Spain
| | - Inés Santiuste
- Departamento de Biología Molecular-IDIVAL Universidad de Cantabria, Santander, Spain
| | - Jorge Postigo
- Departamento de Biología Molecular-IDIVAL Universidad de Cantabria, Santander, Spain
| | - Jesús Merino
- Departamento de Biología Molecular-IDIVAL Universidad de Cantabria, Santander, Spain
| | - Ramón Merino
- Departamento de Biología Molecular-IDIVAL Universidad de Cantabria, Santander, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, Consejo Superior de Investigaciones Científicas-Universidad de Cantabria, Santander, Spain
| |
Collapse
|
45
|
Yoshizaki A. B cell abnormalities and therapeutic strategies in systemic sclerosis. ACTA ACUST UNITED AC 2016; 39:197-206. [PMID: 27320935 DOI: 10.2177/jsci.39.197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
One of the major roles of B cells is to generate antibodies to specific antigens. Secreted antibodies are the principal molecules involved in humoral immunity, with the capacity not only to neutralize pathogens, but also to enhance their elimination by activating phagocytosis or complement proteins. Recently, it has been highlighted that B cells exert important regulatory roles independent of their antibody producing function. These roles include efficiently presenting antigens to the T cells, secreting cytokines, and inducing immune cell differentiation. Thus, B cells have emerged as cells that play crucial roles in immune systems in addition to producing antibodies. Systemic sclerosis (SSc) is characterized by autoimmunity and tissue fibrosis of several organs. Although the pathogenic relationship between systemic autoimmunity and the clinical manifestations of SSc remains unknown, SSc patients display a variety of abnormal immune activation including the production of disease-specific autoantibodies. Previous studies have demonstrated that immune cells, mainly including B cells, play a critical role in systemic autoimmunity and disease expression, though the role of autoimmunity in generating the clinical and pathologic phenotype in SSc remains uncertain. SSc patients have B cell abnormalities characterized by chronic hyper-reactivity of memory B cells. Although distinct subsets of autoantibodies do not have a proven pathogenic role, they are selectively associated with unique disease manifestations. To date, the treatment of SSc has largely relied on cytotoxic immunosuppressants and corticosteroids. Although this has resulted in improved disease survival, these patients may still suffer severe adverse events and refractory disease to conventional immunosuppressive therapies. Recently, clinical trials involving the chimeric monoclonal antibody rituximab have raised high expectations. B cell depleting therapy with rituximab offers a promising treatment for the rheumatic autoimmune diseases including SSc. This article reviews the current knowledge of B cell biology and pathogenesis in SSc as well as the therapeutic approaches focusing on the targeting of B cell specific surface molecules and on the blocking of B cell activation and survival.
Collapse
Affiliation(s)
- Ayumi Yoshizaki
- Department of Dermatology, The University of Tokyo Graduate School of Medicine
| |
Collapse
|
46
|
Andersson SEM, Eneljung T, Tengvall S, Jirholt P, Stern A, Henningsson L, Liang B, Thorarinsdottir K, Kihlberg J, Holmdahl R, Mårtensson IL, Gustafsson K, Gjertsson I. Collagen epitope expression on B cells is sufficient to confer tolerance to collagen-induced arthritis. Arthritis Res Ther 2016; 18:140. [PMID: 27301320 PMCID: PMC4908726 DOI: 10.1186/s13075-016-1037-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/27/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mechanisms underlying tolerance induction and maintenance in autoimmune arthritis remain elusive. In a mouse model of rheumatoid arthritis, collagen type II (CII)-induced arthritis, we explore the contribution of B cells to antigen-specific tolerance. METHODS To generate expression of the CII-peptide specifically on B-cell major histocompatibility complex type II, lentiviral-based gene therapy including a B-cell-specific Igk promoter was used. RESULTS Presentation of the CII-peptide on B cells significantly reduced the frequency and severity of arthritis as well as the serum levels of CII -specific IgG antibodies. Further, both frequency and suppressive function of regulatory T cells were increased in tolerized mice. Adoptive transfer of regulatory T cells from tolerized mice to naïve mice ameliorated the development of CII-induced arthritis. CONCLUSION Our data suggest that endogenous presentation of the CII-peptide on B cells is one of the key contributors to arthritis tolerance induction and maintenance.
Collapse
Affiliation(s)
- Sofia E M Andersson
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, SE 405 30, Gothenburg, Sweden
| | - Tove Eneljung
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, SE 405 30, Gothenburg, Sweden
- Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sara Tengvall
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, SE 405 30, Gothenburg, Sweden
- Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pernilla Jirholt
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, SE 405 30, Gothenburg, Sweden
| | - Anna Stern
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, SE 405 30, Gothenburg, Sweden
| | - Louise Henningsson
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, SE 405 30, Gothenburg, Sweden
| | - Bibo Liang
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Southern Medical University, Guangzhou, People's Republic China
| | - Katrin Thorarinsdottir
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, SE 405 30, Gothenburg, Sweden
- Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Kihlberg
- Department of Chemistry, BMC, Uppsala University, Uppsala, Sweden
| | - Rikard Holmdahl
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Southern Medical University, Guangzhou, People's Republic China
| | - Inga-Lill Mårtensson
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, SE 405 30, Gothenburg, Sweden
| | - Kenth Gustafsson
- Molecular Immunology Unit, UCL Institute of Child Health, London, UK
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Box 480, SE 405 30, Gothenburg, Sweden.
- Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
47
|
Tengvall S, Eneljung T, Jirholt P, Turesson O, Wing K, Holmdahl R, Kihlberg J, Stern A, Mårtensson IL, Henningsson L, Gustafsson K, Gjertsson I. Gene Therapy Induces Antigen-Specific Tolerance in Experimental Collagen-Induced Arthritis. PLoS One 2016; 11:e0154630. [PMID: 27159398 PMCID: PMC4861286 DOI: 10.1371/journal.pone.0154630] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 04/16/2016] [Indexed: 12/22/2022] Open
Abstract
Here, we investigate induction of immunological tolerance by lentiviral based gene therapy in a mouse model of rheumatoid arthritis, collagen II-induced arthritis (CIA). Targeting the expression of the collagen type II (CII) to antigen presenting cells (APCs) induced antigen-specific tolerance, where only 5% of the mice developed arthritis as compared with 95% of the control mice. In the CII-tolerized mice, the proportion of Tregs as well as mRNA expression of SOCS1 (suppressors of cytokine signaling 1) increased at day 3 after CII immunization. Transfer of B cells or non-B cell APC, as well as T cells, from tolerized to naïve mice all mediated a certain degree of tolerance. Thus, sustainable tolerance is established very early during the course of arthritis and is mediated by both B and non-B cells as APCs. This novel approach for inducing tolerance to disease specific antigens can be used for studying tolerance mechanisms, not only in CIA but also in other autoimmune diseases.
Collapse
Affiliation(s)
- Sara Tengvall
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Tove Eneljung
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pernilla Jirholt
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Olof Turesson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Kajsa Wing
- Medical Inflammation Research, Dept of medical Biochemistry and biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rikard Holmdahl
- Medical Inflammation Research, Dept of medical Biochemistry and biophysics, Karolinska Institutet, Stockholm, Sweden
- Southern Medical University, Guangzhou, PR China
| | - Jan Kihlberg
- Department of Chemistry, BMC, Uppsala University, Uppsala, Sweden
| | - Anna Stern
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Inga-Lill Mårtensson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Louise Henningsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Kenth Gustafsson
- Molecular and Cellular Immunology Section, UCL Institute of Child Health, London, United Kingdom
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska University Hospital, Gothenburg, Sweden
- * E-mail:
| |
Collapse
|
48
|
Head-to-head comparison of protocol modifications for the generation of collagen-induced arthritis in a specific-pathogen free facility using DBA/1 mice. Biotechniques 2016; 60:119-28. [PMID: 26956089 DOI: 10.2144/000114388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/16/2015] [Indexed: 11/23/2022] Open
Abstract
Collagen-induced arthritis (CIA) is a widely used mouse model for studying inflammatory arthritis (IA). However, CIA induction protocols differ between laboratories, and direct comparison between protocol variations has not been reported. To address this issue, DBA/1 mice housed in conventional and specific-pathogen free (SPF) facilities were administered various combinations of two doses of collagen type II (CII) in complete (CFA) or incomplete Freund's adjuvant (IFA); some mice were also injected with lipopolysaccharide (LPS) and/or additional CII at specific intervals. Mice were evaluated for IA over the subsequent 2 months. Depending directly on the combination of CII, CFA, IFA, and LPS used, the incidence of IA ranged between 20%-100%, and severity extended from mild to severe even in an SPF environment. Our results demonstrate for the first time in head-to-head comparisons that specific variations in the use of CII, CFA, IFA, and LPS can induce a range of arthritic disease intensity and severity in an SPF facility. Thus, distinct experimental settings can be designed for robust assessment of factors that either exacerbate or inhibit arthritis pathogenesis. Furthermore, by achieving 100% incidence in an SPF facility, the protocols provide a practical and humane benefit by reducing the number of mice necessary for experimental assessment.
Collapse
|
49
|
Pérez-Verdaguer M, Capera J, Serrano-Novillo C, Estadella I, Sastre D, Felipe A. The voltage-gated potassium channel Kv1.3 is a promising multitherapeutic target against human pathologies. Expert Opin Ther Targets 2015; 20:577-91. [DOI: 10.1517/14728222.2016.1112792] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
50
|
Endo T, Ito K, Morimoto J, Kanayama M, Ota D, Ikesue M, Kon S, Takahashi D, Onodera T, Iwasaki N, Uede T. Syndecan 4 Regulation of the Development of Autoimmune Arthritis in Mice by Modulating B Cell Migration and Germinal Center Formation. Arthritis Rheumatol 2015; 67:2512-22. [PMID: 25989265 DOI: 10.1002/art.39193] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 05/07/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Syndecan 4 has been implicated as a critical mediator of inflammatory responses because of its functions as a coreceptor and reservoir for growth factors and chemokines. Although syndecan 4 is known to be expressed on B cells, its role in immune responses remains unclear. The purpose of this study was to investigate the contribution of syndecan 4 to the development of immune arthritis in murine models. METHODS The clinical severity of 3 immunopathologically distinct models, collagen-induced arthritis (CIA), antigen-induced arthritis (AIA), and collagen antibody-induced arthritis (CAIA), was evaluated in wild-type (WT) mice and in syndecan 4-deficient (Sdc4(-/-) ) mice. Germinal center (GC) formation, B cell profiles, humoral immune responses, and B cell migration were analyzed during the course of CIA. RESULTS Sdc4(-/-) mice were resistant to the induction of CIA, which is T cell and B cell dependent, but AIA and CAIA, which are T cell dependent and T cell independent, respectively, occurred with equal frequency in WT mice and Sdc4(-/-) mice. Furthermore, Sdc4(-/-) mice had reduced numbers of B cells and deficient GC formation in draining lymph nodes (DLNs) during the course of CIA, resulting in reduced production of collagen-specific autoantibodies. Compared with B cells from WT mice, B cells from Sdc4(-/-) mice showed reduced chemotactic migration toward stromal cell-derived factor 1 (SDF-1) and reduced SDF-1-mediated Akt phosphorylation. Consistent with these findings, in vivo transfer experiments showed that fewer Sdc4(-/-) B cells than WT B cells were found in and around the follicles in the DLNs. CONCLUSION Our findings suggest that syndecan 4 contributes to the development of CIA by promoting GC formation and autoantibody production through its regulation of SDF-1-mediated B cell migration.
Collapse
Affiliation(s)
- Tsutomu Endo
- Institute for Genetic Medicine and Hokkaido University, Graduate School of Medicine, Sapporo, Japan
| | - Koyu Ito
- Institute for Genetic Medicine and Hokkaido University, Sapporo, Japan
| | - Junko Morimoto
- Institute for Genetic Medicine and Hokkaido University, Sapporo, Japan
| | - Masashi Kanayama
- Institute for Genetic Medicine and Hokkaido University, Sapporo, Japan
| | - Daichi Ota
- Institute for Genetic Medicine and Hokkaido University, Sapporo, Japan
| | - Masahiro Ikesue
- Institute for Genetic Medicine and Hokkaido University, Sapporo, Japan
| | - Shigeyuki Kon
- Hokkaido University, Graduate School of Pharmaceutical Sciences, Sapporo, Japan
| | | | - Tomohiro Onodera
- Hokkaido University, Graduate School of Medicine, Sapporo, Japan
| | - Norimasa Iwasaki
- Hokkaido University, Graduate School of Medicine, Sapporo, Japan
| | - Toshimitsu Uede
- Institute for Genetic Medicine and Hokkaido University, Sapporo, Japan
| |
Collapse
|