1
|
Li J, Dan Y, Su W, Zhao M, Chen Z, Zhao Z. Immune-related biomarkers in the neuromyelitis optica spectrum disorder; pathogenesis and therapeutic approaches. Exp Eye Res 2025:110395. [PMID: 40274184 DOI: 10.1016/j.exer.2025.110395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system that mostly affects the optic nerves and spinal cord. About eighty percent of patients have antibodies that are directed against the water channel aquaporin-4 (AQP4)-IgG, which is expressed on astrocytes. This protein was shown to be both a biomarker and a pathogenic cause of NMOSD. Researchers have discovered that antibodies against myelin oligodendrocyte glycoprotein (MOG) IgG can serve as a biomarker for a distinct condition known as MOG antibody-associated disease (MOGAD). This condition shares some similarities with AQP4-IgG-positive NMOSD, but it has distinct differences in terms of its underlying causes, clinical characteristics, response to treatment, and prognosis. Identifying AQP4 antibodies in the blood serum confirms the diagnosis of seropositive NMOSD. Nevertheless, it remains uncertain if there is a correlation between AQP4-IgG levels and disease activity, severity, responsiveness to medication, or long-term effects. Furthermore, there is still a need to establish and confirm biomarkers specifically for patients diagnosed with seronegative NMOSD. This study primarily examines the immunological aspects of NMOSD, which might have significant consequences for clinical practice. These implications include the possible use of new biomarkers to aid in the early and correct diagnosis of NMOSD, as well as the development of current treatment options to enhance the long-term prognosis of NMOSD patients.
Collapse
Affiliation(s)
- Jingyong Li
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Ya Dan
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Wei Su
- Department of Medical Imaging, the Second People's Hospital of Yaan, 625000, China
| | - Mingjun Zhao
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China
| | - Zhiguo Chen
- People's Liberation Army Joint Logistic Support Force rehabilitation center, Da Lian, Yaan, 625000, China
| | - Zhuyang Zhao
- Department of Laboratory Pathology and Blood Transfusion, People's Liberation Army Joint Logistic Support Force 945th Hospital, Yaan, 625000, China.
| |
Collapse
|
2
|
He L, Li X, Jiang S, Ou Y, Wang S, Shi N, Yang Z, Yuan JL, Silverman G, Niu H. The influence of the gut microbiota on B cells in autoimmune diseases. Mol Med 2025; 31:149. [PMID: 40264032 PMCID: PMC12016346 DOI: 10.1186/s10020-025-01195-5] [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: 11/07/2023] [Accepted: 04/01/2025] [Indexed: 04/24/2025] Open
Abstract
Mounting evidence shows that gut microbiota communities and the human immune system coexist and influence each other, and there are a number of reports of a correlation between specific changes in gut microbiota and the occurrence of autoimmune diseases. B lymphocytes play a central role in the regulation of both gut microbiota communities and in autoimmune diseases. Here, we summarize evidence of the influence of gut microbiota-B cell pathways on autoimmune diseases and how B cells regulate microorganisms, which provides mechanistic insights with relevance for identification of potential therapeutic targets and related fields.
Collapse
Affiliation(s)
- Lun He
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xin Li
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shan Jiang
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yanhua Ou
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shanshan Wang
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Na Shi
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhongshan Yang
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, China
| | - Jia-Li Yuan
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, China.
| | - Gregg Silverman
- Division of Rheumatology, New York University School of Medicine, New York, NY, 10016, USA.
| | - Haitao Niu
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education; Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, China.
| |
Collapse
|
3
|
Cosău DE, Costache Enache II, Costache AD, Tudorancea I, Ancuța C, Șerban DN, Bădescu CM, Loghin C, Șerban IL. From Joints to the Heart: An Integrated Perspective on Systemic Inflammation. Life (Basel) 2025; 15:629. [PMID: 40283183 PMCID: PMC12028888 DOI: 10.3390/life15040629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune inflammatory disease which predominantly affects joints, but it can also lead to significant extra-articular complications, particularly cardiovascular disease (CVD). Chronic systemic inflammation promotes endothelial dysfunction and accelerates atherosclerosis, increasing cardiovascular risk. METHODS Current data were analyzed to explore the mechanisms between RA and CVD, focusing on systemic inflammation, pro-inflammatory cytokine patways (IL-1, IL-6, TNF, and JAK-STAT), and their interactions with traditional cardiovascular risk factors. Recent studies and clinical guidelines were reviewed to highlight gaps and advances in risk assessment and management. RESULTS Persistent disease activity and the presence of autoantibodies significantly increase cardiovascular risk in RA contributing to atherosclerosis and major cardiovascular events. Data also suggest that anti-inflammatory treatments, including methotrexate and biologic agents, may lower this risk. CONCLUSION This review highlights the pathophysiological mechanisms between RA and CVD, and the need for early diagnosis and active monitoring to identify and assess cardiovascular risk. A multidisciplinary approach, involving rheumatologists and cardiologists is essential for optimizing cardiovascular risk management and improving patient outcomes. Optimization of cardiovascular risk management strategies in patients with RA should be an essential component of current medical practice, with the main goal of reducing morbidity and mortality from cardiovascular complications.
Collapse
Affiliation(s)
- Diana Elena Cosău
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.E.C.); (I.I.C.E.); (I.T.); (C.A.); (D.N.Ș.); (C.M.B.); (I.L.Ș.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Irina Iuliana Costache Enache
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.E.C.); (I.I.C.E.); (I.T.); (C.A.); (D.N.Ș.); (C.M.B.); (I.L.Ș.)
- “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Alexandru Dan Costache
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.E.C.); (I.I.C.E.); (I.T.); (C.A.); (D.N.Ș.); (C.M.B.); (I.L.Ș.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ionuț Tudorancea
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.E.C.); (I.I.C.E.); (I.T.); (C.A.); (D.N.Ș.); (C.M.B.); (I.L.Ș.)
- “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Codrina Ancuța
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.E.C.); (I.I.C.E.); (I.T.); (C.A.); (D.N.Ș.); (C.M.B.); (I.L.Ș.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Dragomir Nicolae Șerban
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.E.C.); (I.I.C.E.); (I.T.); (C.A.); (D.N.Ș.); (C.M.B.); (I.L.Ș.)
| | - Codruța Minerva Bădescu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.E.C.); (I.I.C.E.); (I.T.); (C.A.); (D.N.Ș.); (C.M.B.); (I.L.Ș.)
- “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Cătălin Loghin
- Department of Internal Medicine, Cardiology Division, University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Ionela Lăcrămioara Șerban
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.E.C.); (I.I.C.E.); (I.T.); (C.A.); (D.N.Ș.); (C.M.B.); (I.L.Ș.)
| |
Collapse
|
4
|
Ma Y, Li Y, Yin Z, Huang JJ, Ye Z, Chen X, Du J, Huang Z. Gadd45γ alleviates collagen-induced arthritis by increasing IL-10 level and suppressing JNK activity. Int Immunopharmacol 2025; 151:114329. [PMID: 40007379 DOI: 10.1016/j.intimp.2025.114329] [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: 10/07/2024] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
It has been shown that Gadd45β alleviated K/BxN serum-induced arthritis, but in collagen-induced arthritis (CIA), it exacerbated joint inflammatory response, clinical signs, and symptoms. So far, the function of Gadd45γ in arthritis remains to be explored. This study aimed to investigate the role and immune regulatory mechanism of Gadd45γ in arthritis by intra-articular injection of a lentiviral vector encoding the Gadd45γ gene (LV-Gadd45γ) or lentiviral vectors (LV). The experiments showed that CIA increased the level of Gadd45γ, overexpression of Gadd45γ inhibited the symptoms and articular destruction of CIA, reduced the pro-inflammatory cytokine IL-1β, IL-6, and matrix metalloproteinases-13 (MMP-13), and increased the anti-inflammatory cytokine IL-10. In turn, high levels of IL-10 elevated Gadd45γ in CIA mice, human rheumatoid synovial fibroblasts (HRSF), and RAW cell lines. Furthermore, the increasing expression of Gadd45γ dramatically raised IL-10 and diminished the phosphorylation of c-Jun N-terminal kinase (JNK) in CIA mice and HRSF. Mechanistic analysis showed that the attenuating effect of Gadd45γ on CIA may be attributed to the mutual enhancement of the expression of Gadd45γ and IL-10 and the inhibition of JNK activity through downregulating IL-1β and IL-6. The results indicate that Gadd45γ can act as an inflammatory suppressor and joint damage attenuator in CIA mice. Increased IL-10 levels through a positive feedback circuit between Gadd45γ and IL-10 and the inhibitory effect of Gadd45γ on JNK activity endows this protein with the ability to inhibit CIA.
Collapse
Affiliation(s)
- Yanmei Ma
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen 518040, China; Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen 518060, China; Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen 518060, China
| | - Yanqun Li
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Zhihua Yin
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen 518040, China; Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen 518060, China
| | - Jennifer Jin Huang
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Parkway Norman, OK 73019-5251, USA
| | - Zhizhong Ye
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen 518040, China; Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen 518060, China
| | - Xinpeng Chen
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen 518040, China; Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen 518060, China.
| | - Jing Du
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Zhong Huang
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen 518060, China; Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen 518060, China.
| |
Collapse
|
5
|
Yu Q, Tang X, Hart T, Homer R, Belperron AA, Bockenstedt LK, Ring A, Nakamura A, Fikrig E. Secretory leukocyte protease inhibitor influences periarticular joint inflammation in B. burgdorferi-infected mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.24.625079. [PMID: 39651186 PMCID: PMC11623497 DOI: 10.1101/2024.11.24.625079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Lyme disease, caused by Borrelia burgdorferi, is the most common tick-borne infection in the United States. Arthritis is a major clinical manifestation of infection, and synovial tissue damage has been attributed to the excessive pro-inflammatory responses. The secretory leukocyte protease inhibitor (SLPI) promotes tissue repair and exerts anti-inflammatory effects. The role of SLPI in the development of Lyme arthritis in C57BL/6 mice, which can be infected with B. burgdorferi, but only develop mild joint inflammation, was therefore examined. SLPI-deficient C57BL/6 mice challenged with B. burgdorferi had a higher infection load in the tibiotarsal joints and marked periarticular swelling, compared to infected wild type control mice. The ankle joint tissues of B. burgdorferi-infected SLPI-deficient mice contained significantly higher percentages of infiltrating neutrophils and macrophages. B. burgdorferi-infected SLPI-deficient mice also exhibited elevated serum levels of IL-6, neutrophil elastase, and MMP-8. Moreover, using a recently developed BASEHIT (BActerial Selection to Elucidate Host-microbe Interactions in high Throughput) library, we found that SLPI directly interacts with B. burgdorferi. These data demonstrate the importance of SLPI in suppressing periarticular joint inflammation in Lyme disease.
Collapse
Affiliation(s)
- Qian Yu
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Xiaotian Tang
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Thomas Hart
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Robert Homer
- Department of Pathology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Alexia A. Belperron
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Linda K. Bockenstedt
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Aaron Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Akira Nakamura
- Divisions of Immunology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
6
|
Formánek T, Mladá K, Mohr P, Lim MF, Olejárová M, Pavelka K, Winkler P, Osimo EF, Jones PB, Hušáková M. Psychiatric morbidity in people with autoimmune arthritides as a model of inflammatory mechanisms in mental disorders. BMJ MENTAL HEALTH 2025; 28:e301506. [PMID: 40121009 PMCID: PMC11931917 DOI: 10.1136/bmjment-2024-301506] [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: 12/05/2024] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) and axial spondyloarthritis (axSpA) are autoimmune illnesses characterised by chronic inflammation demonstrating differential associations with psychiatric conditions. OBJECTIVE In this matched-cohort study, we aimed to investigate whether the associations between these inflammatory illnesses and mental disorders are predominantly the consequence of the burden of the former or whether common causes might underpin the susceptibility to both. METHODS Using Czech national inpatient care data, we identified individuals with RA or axSpA during the years 1999-2012. We investigated the occurrence of psychiatric outcomes up to 2017 using stratified Cox proportional hazards models. In evidence triangulation, we assessed the potential moderation by age at inflammatory illness, the associations relative to counterparts with other similarly burdensome chronic illnesses and the temporal ordering of conditions. FINDINGS Both RA and axSpA were associated with mood and anxiety disorders and behavioural syndromes. In evidence triangulation, the associations with depression showed a decreasing age-at-inflammatory-illness gradient in RA; the association between RA and depression was stronger than that between other chronic illnesses and depression; and excluding prevalent depression attenuated the RA-depression association. RA showed consistent inverse associations with schizophrenia and Alzheimer's disease. CONCLUSIONS Common aetiologies might be involved in increasing the risk of developing both RA and depression. The consistent inverse associations between RA and schizophrenia and between RA and Alzheimer's disease suggest that at least part of these associations might also be a consequence of shared aetiologies as well as potential medication effects. CLINICAL IMPLICATIONS People with autoimmune arthritides are more likely to experience mood and anxiety disorders, even relative to counterparts with other similarly burdensome chronic illnesses.
Collapse
Affiliation(s)
- Tomáš Formánek
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Public Mental Health, National Institute of Mental Health, Klecany, Czechia
- Department of Clinical Epidemiology, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - Karolína Mladá
- Department of Public Mental Health, National Institute of Mental Health, Klecany, Czechia
- Department of Psychiatry, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Pavel Mohr
- Clinical Center, National Institute of Mental Health, Klecany, Czechia
- Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Mao Fong Lim
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Marta Olejárová
- Institute of Rheumatology, Prague, Czechia
- Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Karel Pavelka
- Institute of Rheumatology, Prague, Czechia
- Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Petr Winkler
- Department of Public Mental Health, National Institute of Mental Health, Klecany, Czechia
- Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Social Work, Faculty of Arts, Charles University, Prague, Czechia
| | - Emanuele Felice Osimo
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
- Institute of Clinical Sciences and MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Peter B Jones
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Markéta Hušáková
- Institute of Rheumatology, Prague, Czechia
- Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
7
|
Stauber T, Moschini G, Hussien AA, Jaeger PK, De Bock K, Snedeker JG. Il-6 signaling exacerbates hallmarks of chronic tendon disease by stimulating reparative fibroblasts. eLife 2025; 12:RP87092. [PMID: 39918402 PMCID: PMC11805502 DOI: 10.7554/elife.87092] [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] [Indexed: 02/09/2025] Open
Abstract
Tendinopathies are debilitating diseases currently increasing in prevalence and associated costs. There is a need to deepen our understanding of the underlying cell signaling pathways to unlock effective treatments. In this work, we screen cell signaling pathways in human tendinopathies and find positively enriched IL-6/JAK/STAT signaling alongside signatures of cell populations typically activated by IL-6 in other tissues. In human tendinopathic tendons, we also confirm the strong presence and co-localization of IL-6, IL-6R, and CD90, an established marker of reparative fibroblasts. To dissect the underlying causalities, we combine IL-6 knock-out mice with an explant-based assembloid model of tendon damage to successfully connect IL-6 signaling to reparative fibroblast activation and recruitment. Vice versa, we show that these reparative fibroblasts promote the development of tendinopathy hallmarks in the damaged explant upon IL-6 activation. We conclude that IL-6 activates tendon fibroblast populations which then initiate and deteriorate tendinopathy hallmarks.
Collapse
Affiliation(s)
- Tino Stauber
- Laboratory for Orthopedic Biomechanics, University Hospital Balgrist and ETH ZurichZurichSwitzerland
| | - Greta Moschini
- Laboratory for Orthopedic Biomechanics, University Hospital Balgrist and ETH ZurichZurichSwitzerland
- Laboratory of Exercise and Health Department of Health Sciences and Technology (D-HEST) ETH Zurich, Swiss Federal Institute of TechnologyZurichSwitzerland
| | - Amro A Hussien
- Laboratory for Orthopedic Biomechanics, University Hospital Balgrist and ETH ZurichZurichSwitzerland
| | - Patrick Klaus Jaeger
- Laboratory for Orthopedic Biomechanics, University Hospital Balgrist and ETH ZurichZurichSwitzerland
| | - Katrien De Bock
- Laboratory of Exercise and Health Department of Health Sciences and Technology (D-HEST) ETH Zurich, Swiss Federal Institute of TechnologyZurichSwitzerland
| | - Jess G Snedeker
- Laboratory for Orthopedic Biomechanics, University Hospital Balgrist and ETH ZurichZurichSwitzerland
| |
Collapse
|
8
|
Emerson D, Merriman E, Yachi PP. Rheumatoid arthritis associated cytokines and therapeutics modulate immune checkpoint receptor expression on T cells. Front Immunol 2025; 16:1534462. [PMID: 39981237 PMCID: PMC11840260 DOI: 10.3389/fimmu.2025.1534462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction We investigated the impact of rheumatoid arthritis (RA) associated cytokines and standard of care (SOC) RA therapeutics on immune checkpoint receptor (IR) expression on T cells to gain insights to disease pathology and therapeutic avenues. Methods We assessed IR expression by flow cytometry on T cell receptor activated T cells cultured in the presence of exogenously added single cytokines or RA patient synovial fluid. We also assessed RA synovial fluid stimulated samples in the presence of various single cytokine neutralizing antibodies or SOC therapeutics, including glucocorticoids, TNF, IL-6 receptor and JAK inhibitors. In addition to IR expression, we measured the impact on cytokine secretion profiles. Results RA-associated cytokines modulated IR expression, suggesting a role for these cytokines in regulation of disease pathology. By dissecting the influence of various inflammatory drivers within the RA inflammatory milieu, we discovered distinct regulation of IR expression by various cytokines including IL-10, IFNα/β, and TNF. Specifically, increased expression of TIM-3, PD-1, LAG-3 and CD28 in response to RA synovial fluid was driven by key cytokines including IL-6, IL-10, IL-12, IFNs, and TNF. In addition, SOC RA therapeutics such as glucocorticoids and TNF inhibitors modulated IR and cytokine expression in the presence of RA synovial fluid. Conclusions This study points to an important and intricate relationship between cytokines and IRs in shaping immune responses in autoimmune pathology. The modulation of IR expression by RA-associated cytokines and SOC therapeutics provides new insights for the use of targeted treatments in managing RA pathology.
Collapse
Affiliation(s)
| | | | - Pia P. Yachi
- Immunology Discovery Research, Lilly Research Laboratories, Lilly Biotechnology
Center, San Diego, CA, United States
| |
Collapse
|
9
|
Korzeniowska A, Bryl E. Infectious agents in the pathogenesis of autoimmune rheumatic diseases. Transl Res 2025; 276:39-45. [PMID: 39742962 DOI: 10.1016/j.trsl.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
Autoimmune rheumatic diseases (AIRDs) are diseases with complex outset and courses, in which both genetic and environmental factors participate. Many environmental factors can be committed to AIRDs outset and development. The most popular of them, with confirmed impact, are smoking, age, gender, and microorganisms. In light of recent research an assumption about the importance of various microorganisms in the pathogenesis of AIRDs is growing in popularity. The human immune system has various protective mechanisms against infectious antigens which in normal cases let organism manage potential infection faster and more effectively. Unfortunately in some situations, specific errors in those mechanisms can cause an autoreactive response despite mitigation of infection. Viruses including EBV, CMV, and even SARS-CoV2 can cause these errors. This in combination with genetic factors can lead to rheumatic disease development. This research aims to provide a brief review of the role of viruses in the outset and development of AIRDs.
Collapse
Affiliation(s)
| | - Ewa Bryl
- Department of Physiopathology, Faculty of Medicine, Medical University of Gdansk, Poland.
| |
Collapse
|
10
|
Scheffler JM, Drevinge C, Lindholm C, Gjertsson I, Lend K, Lund Hetland M, Østergaard M, Uhlig T, Schrumpf Heiberg M, Haavardsholm EA, Nurmohamed MT, Lampa J, Sokka‐Isler T, Nordström D, Hørslev‐Petersen K, Gudbjornsson B, Gröndal G, van Vollenhoven R, Carlsten H, Lorentzon M, Hultgård Ekwall A, Rudin A, Islander U. Circulating Baseline CXCR3 +Th2 and Th17 Cell Proportions Correlate With Trabecular Bone Loss After 48 Weeks of Biological Treatment in Early Rheumatoid Arthritis. ACR Open Rheumatol 2025; 7:e11742. [PMID: 39411912 PMCID: PMC11667770 DOI: 10.1002/acr2.11742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 12/25/2024] Open
Abstract
OBJECTIVE The high prevalence of osteoporosis in rheumatoid arthritis (RA) is due to inflammation that stimulates differentiation of osteoclasts, a process involving circulating monocytes and T cell-derived factors. The aim of this study was to evaluate relations between circulating monocytes, T cell subsets, and changes in bone characteristics before and after treatment with biological disease-modifying antirheumatic drugs (bDMARDs) in RA. METHODS Thirty patients with untreated early RA who met the American College of Rheumatology/EULAR 2010 criteria were included. Data were collected before and 48 weeks after treatment with methotrexate (MTX) together with one of three bDMARDs (abatacept, tocilizumab, or certolizumab pegol). Disease activity was measured using the Clinical Disease Activity Index, swollen or tender joint counts, C-reactive protein levels, and erythrocyte sedimentation rates. Proportions of monocyte and CD4+ T cell subsets in blood samples were analyzed by flow cytometry. Bone densitometry was performed using high-resolution peripheral quantitative computed tomography (HR-pQCT). RESULTS HR-pQCT revealed an overall decrease in cortical (P = 0.009) and trabecular (P = 0.034) bone mineral density, although a subset of patients showed no bone loss after 48 weeks of treatment. The overall bone loss was not associated with age, body mass index, sex, intraarticular glucocorticoid injections, or baseline disease activity. Loss of trabecular bone volume fraction correlated with high proportions of circulating CXCR3+Th2 cells (r = -0.38, P = 0.04) and CXCR3+Th17 cells (r = -0.36, P = 0.05) at baseline. Similarly, no loss of trabecular bone volume fraction correlated with high proportions of regulatory T cells (r = 0.4, P = 0.03) at baseline. However, the associations were not significant when corrected for confounders and multiple testing. CONCLUSION MTX together with bDMARDs efficiently reduce disease activity but only prevent bone loss in a subset of patients with RA after 48 weeks of treatment. The correlations of circulating baseline T helper cell and regulatory T cell populations with trabecular bone changes suggest a potential novel role for these cells in systemic bone homeostasis during early RA.
Collapse
Affiliation(s)
| | | | - Catharina Lindholm
- University of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Inger Gjertsson
- University of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Kristina Lend
- Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden, and Amsterdam University Medical CenterAmsterdamthe Netherlands
| | - Merete Lund Hetland
- Rigshospitalet, Glostrup, Denmark, and University of CopenhagenCopenhagenDenmark
| | - Mikkel Østergaard
- Rigshospitalet, Glostrup, Denmark, and University of CopenhagenCopenhagenDenmark
| | | | | | | | - Michael T. Nurmohamed
- Amsterdam Rheumatology and Immunology Center, Reade, the Netherlands, and Amsterdam University Medical CenterAmsterdamthe Netherlands
| | - Jon Lampa
- Karolinska Institute, Karolinska University HospitalStockholmSweden
| | | | - Dan Nordström
- Helsinki University and University HospitalHelsinkiFinland
| | - Kim Hørslev‐Petersen
- Danish Hospital for Rheumatic Diseases, University Hospital of Southern Denmark, Sønderborg, Denmark, and University of Southern DenmarkOdenseDenmark
| | - Bjorn Gudbjornsson
- Landspitali National University Hospital of Iceland and University of IcelandReykjavikIceland
| | - Gerdur Gröndal
- Landspitali National University Hospital of Iceland and University of IcelandReykjavikIceland
| | - Ronald van Vollenhoven
- Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden, and Amsterdam University Medical CenterAmsterdamthe Netherlands
| | - Hans Carlsten
- University of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Mattias Lorentzon
- University of Gothenburg, Gothenburg, Sweden and Australian Catholic UniversityMelbourneAustralia
| | | | - Anna Rudin
- University of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | | |
Collapse
|
11
|
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
|
12
|
Thangadurai M, Sethuraman S, Subramanian A. Drug Delivery Approaches for Rheumatoid Arthritis: Recent Advances and Clinical Translation Aspects. Crit Rev Ther Drug Carrier Syst 2025; 42:1-54. [PMID: 40084516 DOI: 10.1615/critrevtherdrugcarriersyst.v42.i3.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Rheumatoid arthritis (RA) is a multifactorial autoimmune disease characterized with symmetrical progression of joint deformity that is often diagnosed at a chronic condition with other associated pathological conditions such as pericarditis, keratitis, pulmonary granuloma. Despite the understanding of RA pathophysiology in disease progression, current clinical treatment options such as disease-modifying anti-rheumatic drugs (DMARDs), biologics, steroids, and non-steroidal anti-inflammatory drugs (NSAIDs) provide only palliative therapy while causing adverse side effects such as off-target multi-organ toxicity and risk of infections. Further, available drug delivery strategies to treat RA pathogenicity does not successfully reach the site of action due to various barriers such as phagocytosis and first pass effect in addition to the disease complexity and unknown etiology, thereby leading to the development of irreversible joint dysfunction. Therefore, novel and effective strategies remain an unmet need to control the disease progression and to maintain the balance between pro- and anti-inflammatory cytokines. This review provides a comprehensive outlook on the RA pathophysiology and its corresponding disease progression. Contributions of synoviocytes such as macrophages, fibroblast-like cells in increasing invasiveness to exacerbate joint damage is also outlined in this review, which could be a potential future therapeutic target to complement the existing treatment regimens in controlling RA pathogenesis. Further, various smart drug delivery approaches under research to achieve maximum therapeutic efficacy with minimal adverse side effects have been discussed, which in turn emphasize the unmet challenges and future perspectives in addressing RA complications.
Collapse
Affiliation(s)
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Laboratory, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| | - Anuradha Subramanian
- Tissue Engineering & Additive Manufacturing (TEAM) Laboratory, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| |
Collapse
|
13
|
Pareek A, Mehlawat K, Tripathi K, Pareek A, Chaudhary S, Ratan Y, Apostolopoulos V, Chuturgoon A. Melittin as a therapeutic agent for rheumatoid arthritis: mechanistic insights, advanced delivery systems, and future perspectives. Front Immunol 2024; 15:1510693. [PMID: 39759520 PMCID: PMC11695321 DOI: 10.3389/fimmu.2024.1510693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Rheumatoid arthritis (RA), a condition characterized by joint deterioration through the action of matrix metalloproteinases (MMPs), is prevalent worldwide. Bee venom (BV) has traditionally been used in Chinese medicine for pain, arthritis, rheumatism, skin diseases, etc. BV is enriched with active substances, notably melittin and phospholipase A2 (PLA2), offering significant therapeutic potential. Hence, the review summarizes current insights into BV's composition, antiarthritic mechanism and pharmacological benefits, focusing on melittin. Constituting 50-60% of BV, melittin notably downregulates nuclear factor Kappa B (NF-κB) activity, inhibits MMP-1 and MMP-8, and diminishes tumor necrosis factor (TNF-α), all of which contribute to the mitigation of type 2 collagen degradation. Despite its potential, melittin exhibits hemolytic activity and can significantly affect cell membranes, limiting its application, which poses a challenge to its therapeutic use. To overcome these challenges, delivery techniques utilizing nanocarriers and modifications in amino acid sequencing have been developed. Recent advancements in delivery systems, including nanocarriers, transdermal patches, and nanoemulsions, aim to minimize toxicity, expanding its therapeutic utility for RA. This article explores these novel strategies, underlining the evolving role of melittin in RA management.
Collapse
Affiliation(s)
- Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | | | | | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | | | - Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Vasso Apostolopoulos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Anil Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
14
|
Karnam S, Jindal AB, Paul AT. Quality by design-based optimization of teriflunomide and quercetin combinational topical transferosomes for the treatment of rheumatoid arthritis. Int J Pharm 2024; 666:124829. [PMID: 39406305 DOI: 10.1016/j.ijpharm.2024.124829] [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: 06/24/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Rheumatoid arthritis (RA) is an immune-mediated inflammatory disease. Combination therapy is anticipated to surpass monotherapy by targeting multiple pathways involved in RA progression. The present aim is to develop a combination of Teriflunomide (TFD) and Quercetin (QCN) loaded transferosomal gel to enhance permeability and achieve localized delivery to joint tissues. TFD or QCN transferosomes were optimized employing a 3-level, 3-factorial design Box-Behnken design (BBD). The transferosomes exhibited sustained in-vitro drug release. The topical combination gel underwent thorough evaluation of rheology, and also ex-vivo studies showed enhanced permeability through rat skin. The synergistic combination of TFD and QCN effectively suppressed NO, TNF-α and IL-6 levels in in-vitro RAW 264.7 cells. The cytotoxicity in HaCaT cell lines indicates non-toxicity of the gel, further confirmed by skin irritation study conducted in rats. The in-vivo anti-arthritic activity was evaluated in complete freund's adjuvant induced rat paw edema model illustrates the effectiveness of the combination transferosomal gel compared to other treatment groups. In conclusion, the topical delivery of TFD and QCN combination transferosomal gel demonstrated anti-arthritic activity through localized delivery whichallows for dose reduction, thereby may reduce the systemic drug exposure and mitigate the side effects associated with oral administration of TFD.
Collapse
Affiliation(s)
- Sriravali Karnam
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Pilani Campus, Rajasthan 333031, India
| | - Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Pilani Campus, Rajasthan 333031, India
| | - Atish T Paul
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
15
|
Bhuvaneshwari S, Venkataraman K, Sankaranarayanan K. Exploring potential ion channel targets for rheumatoid arthritis: combination of network analysis and gene expression analysis. Biotechnol Appl Biochem 2024; 71:1405-1427. [PMID: 39049164 DOI: 10.1002/bab.2638] [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: 12/28/2023] [Accepted: 06/29/2024] [Indexed: 07/27/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the synovial membrane that leads to the destruction of cartilage and bone. Currently, pharmacological targeting of ion channels is being increasingly recognized as an attractive and feasible strategy for the treatment of RA. The present work employs a network analysis approach to predict the most promising ion channel target for potential RA-treating drugs. A protein-protein interaction map was generated for 343 genes associated with inflammation in RA and ion channel genes using Search Tool for the Retrieval of Interacting Genes and visualized using Cytoscape. Based on the betweenness centrality and traffic values as key topological parameters, 17 hub nodes were identified, including FOS (9800.85), tumor necrosis factor (3654.60), TGFB1 (3305.75), and VEGFA (3052.88). The backbone network constructed with these 17 hub genes was intensely analyzed to identify the most promising ion channel target using network analyzer. Calcium permeating ion channels, especially store-operated calcium entry channels, and their associated regulatory proteins were found to highly interact with RA inflammatory hub genes. This significant ion channel target for RA identified by theoretical and statistical studies was further validated by a pilot case-control gene expression study. Experimental verification of the above findings in 75 RA cases and 25 controls showed increased ORAI1 expression. Thus, with a combination of network analysis approach and gene expression studies, we have explored potential targets for RA treatment.
Collapse
Affiliation(s)
- Sampath Bhuvaneshwari
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| | | | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| |
Collapse
|
16
|
Heikkinen J, Palosaari S, Lehenkari P. Cigarette smoke extract decreases human bone marrow mesenchymal stromal cell adipogenic differentiation. Toxicol In Vitro 2024; 101:105949. [PMID: 39343071 DOI: 10.1016/j.tiv.2024.105949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Smoking and nicotine impose detrimental health effects including adipose tissue dysfunction. Despite extensive physiological evidence, the cellular mechanisms remain poorly understood, with few studies examining the effects of cigarette smoke extract (CSE) or nicotine on adipocyte differentiation. METHODS Primary human bone marrow-derived mesenchymal stromal cells (MSCs) were exposed to CSE or nicotine (50-500 ng/ml) during adipogenic differentiation. Cell viability and metabolic activity were assessed via MTT assay. Lipid droplet accumulation was evaluated using Sudan III staining and quantitative image analysis. Adiponectin, IL6, and IL8 concentrations were measured after 35 days using ELISA. RESULTS At these doses, CSE and nicotine do not immediately affect cell viability but inhibit undifferentiated cell proliferation. Notably, both agents at 50 ng/ml significantly increased lipid accumulation during adipogenesis, while higher CSE doses nearly completely inhibited this process. Additionally, CSE dose-dependently decreased adiponectin secretion and increased IL6 and IL8, indicating a shift towards an inflammatory state. Nicotine alone primarily increased IL6 secretion with less pronounced effects. CONCLUSION The study highlights the complex impact of CSE and nicotine on adipocyte function during early differentiation from MSCs. Dose-dependent changes in lipid accumulation, cytokine, and adiponectin secretion induced by CSE and nicotine can partly explain smoking-related adipose tissue dysfunction.
Collapse
Affiliation(s)
- Janne Heikkinen
- Medical Faculty, Translational Medicine Research Unit, University of Oulu, Oulu, Finland.
| | - Sanna Palosaari
- Medical Faculty, Translational Medicine Research Unit, University of Oulu, Oulu, Finland; Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland.
| | - Petri Lehenkari
- Medical Faculty, Translational Medicine Research Unit, University of Oulu, Oulu, Finland; Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland; Division of Orthopaedic Surgery, Oulu University Hospital, Oulu, Finland.
| |
Collapse
|
17
|
Kadam R, Gupta M, Lazarov O, Prabhakar BS. Brain-immune interactions: implication for cognitive impairments in Alzheimer's disease and autoimmune disorders. J Leukoc Biol 2024; 116:1269-1290. [PMID: 38869088 DOI: 10.1093/jleuko/qiae134] [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: 11/13/2023] [Revised: 05/09/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024] Open
Abstract
Progressive memory loss and cognitive dysfunction, encompassing deficits in learning, memory, problem solving, spatial reasoning, and verbal expression, are characteristics of Alzheimer's disease and related dementia. A wealth of studies has described multiple roles of the immune system in the development or exacerbation of dementia. Individuals with autoimmune disorders can also develop cognitive dysfunction, a phenomenon termed "autoimmune dementia." Together, these findings underscore the pivotal role of the neuroimmune axis in both Alzheimer's disease and related dementia and autoimmune dementia. The dynamic interplay between adaptive and innate immunity, both in and outside the brain, significantly affects the etiology and progression of these conditions. Multidisciplinary research shows that cognitive dysfunction arises from a bidirectional relationship between the nervous and immune systems, though the specific mechanisms that drive cognitive impairments are not fully understood. Intriguingly, this reciprocal regulation occurs at multiple levels, where neuronal signals can modulate immune responses, and immune system-related processes can influence neuronal viability and function. In this review, we consider the implications of autoimmune responses in various autoimmune disorders and Alzheimer's disease and explore their effects on brain function. We also discuss the diverse cellular and molecular crosstalk between the brain and the immune system, as they may shed light on potential triggers of peripheral inflammation, their effect on the integrity of the blood-brain barrier, and brain function. Additionally, we assess challenges and possibilities associated with developing immune-based therapies for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Rashmi Kadam
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| |
Collapse
|
18
|
Vanegas SO, Zaki A, Dealy CN, Kinsey SG. The Minor Phytocannabinoid Delta-8-Tetrahydrocannabinol Attenuates Collagen-Induced Arthritic Inflammation and Pain-Depressed Behaviors. J Pharmacol Exp Ther 2024; 391:222-230. [PMID: 38834355 PMCID: PMC11493440 DOI: 10.1124/jpet.124.002189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/04/2024] [Accepted: 04/11/2024] [Indexed: 06/06/2024] Open
Abstract
Patients with arthritis report using cannabis for pain management, and the major cannabinoid delta-9-tetrahydrocannabinol (Δ9-THC) has anti-inflammatory properties, yet the effects of minor cannabinoids on arthritis are largely unknown. The goal of the present study was to determine the antiarthritic potential of the minor cannabinoid delta-8-tetrahydrocannabinol (Δ8-THC) using the collagen-induced arthritis (CIA) mouse model. Adult male DBA/1J mice were immunized and boosted 21 days later with an emulsion of collagen and complete Freund's adjuvant. Beginning on the day of the booster, mice were administered twice-daily injections of Δ8-THC (3 or 30 mg/kg), the steroid dexamethasone (2 mg/kg), or vehicle for two weeks. Dorsal-ventral paw thickness and qualitative measures of arthritis were recorded daily, and latency to fall from an inverted grid was measured on alternating days, to determine arthritis severity and functional impairment. On the final day of testing, spontaneous wire-climbing behavior and temperature preference in a thermal gradient ring were measured to assess CIA-depressed behavior. The Δ8-THC treatment (30 mg/kg) reduced paw swelling and qualitative signs of arthritis. Δ8-THC also blocked CIA-depressed climbing and CIA-induced preference for a heated floor without producing locomotor effects but did not affect latency to fall from a wire grid. In alignment with the morphologic and behavioral assessments in vivo, histology revealed that Δ8-THC reduced synovial inflammation, proteoglycan loss and cartilage and bone erosion in the foot joints in a dose-dependent manner. Together, these findings suggest that Δ8-THC not only blocked morphologic changes but also prevented functional loss caused by collagen-induced arthritis. SIGNIFICANCE STATEMENT: Despite increasing use of cannabis products, the potential effects of minor cannabinoids are largely unknown. Here, the minor cannabinoid delta-8-tetrahydrocannabinol blocked the development of experimentally induced arthritis by preventing both pathophysiological as well as functional effects of the disease model. These data support the development of novel cannabinoid treatments for inflammatory arthritis.
Collapse
Affiliation(s)
- S Olivia Vanegas
- School of Nursing (S.O.V., S.G.K.) and Department of Psychological Sciences (S.O.V.), University of Connecticut, Storrs, Connecticut; Department of Orthodontics (A.Z., C.N.D.) and Department of Biomedical Engineering (C.N.D.), School of Dental Medicine, University of Connecticut, Farmington, Connecticut; and Department of Orthopedic Surgery and Department of Cell Biology, School of Medicine, University of Connecticut, Farmington, Connecticut (C.N.D.)
| | - Arsalan Zaki
- School of Nursing (S.O.V., S.G.K.) and Department of Psychological Sciences (S.O.V.), University of Connecticut, Storrs, Connecticut; Department of Orthodontics (A.Z., C.N.D.) and Department of Biomedical Engineering (C.N.D.), School of Dental Medicine, University of Connecticut, Farmington, Connecticut; and Department of Orthopedic Surgery and Department of Cell Biology, School of Medicine, University of Connecticut, Farmington, Connecticut (C.N.D.)
| | - Caroline N Dealy
- School of Nursing (S.O.V., S.G.K.) and Department of Psychological Sciences (S.O.V.), University of Connecticut, Storrs, Connecticut; Department of Orthodontics (A.Z., C.N.D.) and Department of Biomedical Engineering (C.N.D.), School of Dental Medicine, University of Connecticut, Farmington, Connecticut; and Department of Orthopedic Surgery and Department of Cell Biology, School of Medicine, University of Connecticut, Farmington, Connecticut (C.N.D.)
| | - Steven G Kinsey
- School of Nursing (S.O.V., S.G.K.) and Department of Psychological Sciences (S.O.V.), University of Connecticut, Storrs, Connecticut; Department of Orthodontics (A.Z., C.N.D.) and Department of Biomedical Engineering (C.N.D.), School of Dental Medicine, University of Connecticut, Farmington, Connecticut; and Department of Orthopedic Surgery and Department of Cell Biology, School of Medicine, University of Connecticut, Farmington, Connecticut (C.N.D.)
| |
Collapse
|
19
|
Zeb S, Khan Z, Ashraf, Javaid M, Rumman, Swati MAA, Javaid Z, Luqman M. Relationship Between Serum Interleukin-6 Levels, Systemic Immune-Inflammation Index, and Other Biomarkers Across Different Rheumatoid Arthritis Severity Levels. Cureus 2024; 16:e72334. [PMID: 39469275 PMCID: PMC11516189 DOI: 10.7759/cureus.72334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 10/30/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by joint inflammation, pain, and progressive disability. Identifying biomarkers that accurately reflect disease severity is crucial for effective management. Interleukin-6 (IL-6) is a pro-inflammatory cytokine involved in the pathogenesis of RA, and the systemic immune-inflammation index (SII) is emerging as a useful marker of systemic inflammation. This study aims to explore the relationship between serum IL-6 levels, SII, and various biomarkers to better predict disease severity in RA patients. Objective To determine the relationship between serum IL-6 levels and the SII, along with various biomarkers, across different severity levels for predicting the severity of RA in patients. Methods This cross-sectional, observational study was conducted at the Mardan Medical Complex from January 2024 to August 2024, involving 67 RA patients. Clinical assessments included demographic data, disease activity (DAS28), pain (VAS), joint damage (Larsen score), and functional status (HAQ-DI). Serum IL-6 levels, along with other biomarkers such as C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), and the SII, were measured through fasting blood samples. Statistical analyses, including density plots, scatter plots, boxplots with ANOVA, and random forest models, were performed to explore associations between IL-6 and all other variables. Significance was set at p < 0.05. Results The study included 67 RA patients (mean age: 41.79 ± 10.51 years, 53.73% male). Elevated IL-6 levels (mean: 80.28 ± 35.27 pg/mL) were strongly associated with disease severity. Patients with DAS28 > 5.5 had IL-6 levels over 100 pg/mL, while those in remission had around 40 pg/mL. IL-6 levels correlated with joint damage (100 pg/mL in severe cases) and pain (over 120 pg/mL for severe pain). Patients with metabolic and cardiovascular comorbidities had the highest IL-6 levels, particularly with diabetes and hypertension (98.6 pg/mL) or cardiovascular disease (119.3 pg/mL). IL-6 correlated strongly with CRP (r = 0.65), ESR (r = 0.51), and SII (r = 0.62). Regression confirmed IL-6 as an independent predictor of severity (p < 0.001), with comorbidities being key predictors. Conclusion Elevated IL-6 and SII levels serve as critical markers for predicting the severity of RA. Addressing these markers may lead to more targeted and effective therapeutic strategies for managing disease progression.
Collapse
Affiliation(s)
- Shah Zeb
- Internal Medicine, Bacha Khan Medical College, Mardan, PAK
| | - Zahir Khan
- Orthopaedic Surgery, Medical Teaching Institution Mardan Medical Complex, Bacha Khan Medical College, Mardan, PAK
| | - Ashraf
- Research and Development, Pro-Gene Diagnostics and Research Laboratory, Mardan, PAK
| | - Mustafa Javaid
- Internal Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, GBR
| | - Rumman
- Active Drug Safety Monitoring and Management (aDSM), Global Fund, Combined Management Unit (CMU), Mardan, PAK
- Pulmonology, Medical Teaching Institution Mardan Medical Complex, Mardan, PAK
| | | | - Zenab Javaid
- General Medicine, Peshawar General Hospital, Peshawar, PAK
| | - Muhammad Luqman
- Pharmacology and Therapeutics, Peshawar Medical and Dental College, Peshawar, PAK
| |
Collapse
|
20
|
Kao CJ, Charmsaz S, Alden SL, Brancati M, Li HL, Balaji A, Munjal K, Howe K, Mitchell S, Leatherman J, Griffin E, Nakazawa M, Tsai HL, Danilova L, Thoburn C, Gizzi J, Gross NE, Hernandez A, Coyne EM, Shin SM, Babu JS, Apostol GW, Durham J, Christmas BJ, Konig MF, Lipson EJ, Naidoo J, Cappelli LC, Pabani A, Ged Y, Baretti M, Brahmer J, Hoffman-Censits J, Seiwert TY, Garonce-Hediger R, Guha A, Bansal S, Tang L, Jaffee EM, Chandler GS, Mohindra R, Ho WJ, Yarchoan M. Immune-related events in individuals with solid tumors on immunotherapy associate with Th17 and Th2 signatures. J Clin Invest 2024; 134:e176567. [PMID: 39403935 PMCID: PMC11473156 DOI: 10.1172/jci176567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUNDImmune-related adverse events (irAEs) and their associated morbidity/mortality are a key concern for patients receiving immune checkpoint inhibitors (ICIs). Prospective evaluation of the drivers of irAEs in a diverse pan-tumor cohort is needed to identify patients at greatest risk and to develop rational treatment and interception strategies.METHODSIn an observational study, we prospectively collected blood samples and performed regular clinical evaluations for irAEs in patients receiving ICI therapy as standard of care for solid tumors. We performed in-parallel analysis of cytokines by Luminex immunoassay and circulating immune cells by cytometry by time-of-flight (CyTOF) at baseline and on treatment to investigate mechanisms of irAEs.RESULTSWe enrolled 111 patients, of whom 40.5% developed a symptomatic irAE (grade ≥ 2). Development of a grade ≥ 2 irAE was positively associated with the use of combination ICI and a history of an autoimmune disorder. Early changes in T helper 17 (Th17) (IL-6, IL-17f), type 2 (IL-5, IL-13, IL-25), and type 1 (TNF-α) cytokine signatures and congruent on-treatment expansions of Th17 and Th2 effector memory (Th2EM) T cell populations in peripheral blood were positively associated with the development of grade ≥2 irAEs. IL-6 levels were also associated with inferior cancer-specific survival and overall survival.CONCLUSIONSIn a diverse, prospective pan-tumor cohort, Th17 and Th2 skewing during early ICI treatment was associated with the development of clinically relevant irAEs but not antitumor responses, providing possible targets for monitoring and therapeutic interventions.FUNDINGJohns Hopkins Bloomberg-Kimmel Institute for Cancer Immunotherapy, the NCI SPORE in Gastrointestinal Cancers (P50 CA062924), NCI grant (R50CA243627 to LD), the NIH Center Core Grant (P30 CA006973), Swim Across America (to MY), NIAMS (K23AR075872 to LC), and imCORE-Genentech grant 137515 (to Johns Hopkins Medicine on behalf of MY).
Collapse
Affiliation(s)
- Chester J. Kao
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Soren Charmsaz
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | | | - Madelena Brancati
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Howard L. Li
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aanika Balaji
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kabeer Munjal
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Kathryn Howe
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Sarah Mitchell
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - James Leatherman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Ervin Griffin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Mari Nakazawa
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Hua-Ling Tsai
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ludmila Danilova
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chris Thoburn
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Gizzi
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicole E. Gross
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Alexei Hernandez
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Erin M. Coyne
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Sarah M. Shin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Jayalaxmi Suresh Babu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - George W. Apostol
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Jennifer Durham
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Brian J. Christmas
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
| | - Maximilian F. Konig
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Evan J. Lipson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jarushka Naidoo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Beaumont Hospital, Dublin, Ireland
- RCSI University of Health Sciences, Dublin, Ireland
| | - Laura C. Cappelli
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aliyah Pabani
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yasser Ged
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marina Baretti
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julie Brahmer
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jean Hoffman-Censits
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tanguy Y. Seiwert
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Aditi Guha
- Genentech Inc., a member of the imCORE network, South San Francisco, California, USA
| | - Sanjay Bansal
- Genentech Inc., a member of the imCORE network, South San Francisco, California, USA
| | - Laura Tang
- Genentech Inc., a member of the imCORE network, South San Francisco, California, USA
| | - Elizabeth M. Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| | - G. Scott Chandler
- F. Hoffmann-La Roche Ltd., a member of the imCORE network, Basel, Switzerland
| | - Rajat Mohindra
- F. Hoffmann-La Roche Ltd., a member of the imCORE network, Basel, Switzerland
| | - Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
| | - Mark Yarchoan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, a member of the imCORE network, Baltimore, Maryland, USA
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Convergence Institute and
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Banerjee K, Chandrasekar B, Sathish S, Sohn H, Madhavan T. Computational drug repositioning for IL6 triggered JAK3 in rheumatoid arthritis using FDA database. Mol Divers 2024:10.1007/s11030-024-10958-x. [PMID: 39141207 DOI: 10.1007/s11030-024-10958-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 08/02/2024] [Indexed: 08/15/2024]
Abstract
Rheumatoid Arthritis (RA) is a persistent autoimmune disease affecting approximately 0.5-1 percent of the world population. RA prevalence is higher in woman aged between 35 and 50 years than in age matched men, though this difference is less evident among elderly patients. The profound immune specific effects of disrupted JAK 3 (Janus kinase 3) signaling highlight the possibility of therapeutic targeting of JAK3 as a highly specific mode of immune system suppression. To address the above problem which is unendurable to patients and in the hope to cater some respite to such suffering we have targeted JAK 3 protein and JAK/STAT signaling pathway with compounds downloaded from FDA database, and performed screening of all available compounds docked against JAK3 protein. The difference between the target protein and other proteins of the same family was studied using cross docking and the compounds having higher binding affinity to JAK3 protein also showed more selectivity towards the particular protein. Density functional theory and molecular dynamics simulation study was done to study the compounds at their atomic level to know more about their drug likeliness. At the end of the study and based on our analysis we have come up with three FDA approved drugs that can be proposed as a treatment option for Rheumatoid Arthritis.
Collapse
Affiliation(s)
- Kaushani Banerjee
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India
| | - Bavya Chandrasekar
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India
| | - Sruthy Sathish
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India
| | - Honglae Sohn
- Department of Chemistry and Department of Carbon Materials, Chosun University, Gwangju, South Korea
| | - Thirumurthy Madhavan
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Potheri, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
22
|
Kerkis I, da Silva ÁP, Araldi RP. The impact of interleukin-6 (IL-6) and mesenchymal stem cell-derived IL-6 on neurological conditions. Front Immunol 2024; 15:1400533. [PMID: 39015561 PMCID: PMC11249726 DOI: 10.3389/fimmu.2024.1400533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/04/2024] [Indexed: 07/18/2024] Open
Abstract
Interleukin-6 (IL-6) is a versatile cytokine crucial for immune response modulation, inflammation regulation, and various physiological processes in the body. Its wide-ranging functions underscore its importance in maintaining health. Dysregulated IL-6 is closely associated with many diseases, making it a key research and therapeutic target. Elevated IL-6 levels in the central nervous system worsen neuroinflammation in neurodegenerative diseases by activating microglia and astrocytes and releasing pro-inflammatory cytokines and neurotoxic molecules. Moreover, dysregulated IL-6 weakens the blood-brain barrier, exacerbating neuroinflammation and neuronal damage by allowing peripheral immune cells and inflammatory mediators to enter the brain. Mesenchymal stem cells (MSCs) show promise in modulating neuroinflammation by regulating IL-6 levels. They effectively suppress pro-inflammatory cytokines, including IL-6, while promoting anti-inflammatory factors. This therapeutic approach highlights the importance of targeting IL-6 and other inflammatory mediators to alleviate neuroinflammation and its adverse effects on neurological disorders. This review provides a comprehensive overview of IL-6's involvement in neurological disorders, examining endogenous IL-6 and IL-6 derived from MSCs. We explore IL-6's mechanisms affecting neuronal function, survival, and immune modulation in the central nervous system. Additionally, we discuss the potential of MSC-derived IL-6 in neuroregeneration and neuroprotection. By elucidating IL-6's interplay with neurological pathologies, this review offers insights into novel therapeutic strategies targeting IL-6 signaling pathways for neurological disorders.
Collapse
Affiliation(s)
- Irina Kerkis
- Genetics Laboratory, Center of Development and Innovation, Butantan Institute, São Paulo, Brazil
| | - Álvaro Prieto da Silva
- Genetics Laboratory, Center of Development and Innovation, Butantan Institute, São Paulo, Brazil
| | - Rodrigo Pinheiro Araldi
- BioDecision Analytics Ltda., São Paulo, Brazil
- Post-graduation Program in Structural and Functional Biology, Paulista School of Medicine Escola Paulista de Medicina (EPM), Federal University of São Paulo Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
23
|
Alshehri KM, Abdella EM. Galloyl-oligochitosan nano-vehicles for effective and controlled propolis delivery targeting upgrading its antioxidant and antiproliferative potential. Int J Biol Macromol 2024; 270:132283. [PMID: 38735605 DOI: 10.1016/j.ijbiomac.2024.132283] [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: 03/25/2024] [Revised: 05/04/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
A new conjugate, galloyl-oligochitosan nanoparticles (GOCNPs), was fabricated and used as nano-vehicle for effective and controlled delivery of propolis extract (PE) in the form of PE#GOCNPs, targeting improving its pharmaceutical potential. H-bonding interactions between the carboxyl, amino, and hydroxyl groups of the GOCNPs and PE resulted in successful encapsulation, with an entrapment efficacy of 97.3 %. The PE#GOCNPs formulation also exhibited excellent physicochemical stability and time-triggered drug release characteristics under physiological conditions. Furthermore, PE#GOCNPs showed significant activity against MCF-7 and HEPG2 carcinoma cells by scavenging free oxygen radicals and upregulating antioxidant enzymes. Additionally, PE#GOCNPs displayed anti-inflammatory properties by increasing IL10 and reducing pro-inflammatory cytokines more effectively than celecoxib. Furthermore, PE#GOCNPs reduced the expression of epidermal growth factor receptor (EGFR) and survivin genes. Furthermore, the encapsulated PE demonstrated significant activity in suppressing sonic hedgehog protein (SHH). The use of GOCNPs in combination with propolis presents a promising new strategy for chemotherapy with reduced toxicity and enhanced biocompatibility. This novel approach has the potential to revolutionize the field of chemotherapy. Future studies should focus on the application of the encapsulated PE in various cancer cell lines, distinct gene expression factors, and cell cycles.
Collapse
Affiliation(s)
| | - Ehab M Abdella
- Department of Biology, Al-Baha University, Saudi Arabia; Zoology department faculty of science Beni-Suef University, Beni-Suef, Egypt.
| |
Collapse
|
24
|
Warjukar PR, Mohabey AV, Jain PB, Bandre GR. Decoding the Correlation Between Inflammatory Response Marker Interleukin-6 (IL-6) and C-reactive Protein (CRP) With Disease Activity in Rheumatoid Arthritis. Cureus 2024; 16:e62954. [PMID: 39050325 PMCID: PMC11265957 DOI: 10.7759/cureus.62954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/23/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Excessive interleukin-6 (IL-6) production in rheumatoid arthritis (RA) leads to joint destruction, inflammation, and systemic symptoms. IL-6 inhibitors alleviate symptoms. C-reactive protein (CRP), an inflammation biomarker, correlates with RA activity. In this study, we assess IL-6 and CRP levels in RA patients to understand their association with disease activity. MATERIALS AND METHODS This cross-sectional study was conducted at a tertiary care hospital in central India for 15 months, from July 2022 to September 2023. The study involved 75 participants diagnosed with RA and receiving outpatient treatment. Exclusion criteria included anti-IL-6 drug treatment, bedridden individuals, proxy patients, and those without consent. Disease activity was assessed using the 28-joint disease activity score (DAS28), while IL-6 and CRP levels were measured following the standard procedures. RESULTS The average CRP levels were found to be 51.67 ± 47.49 mg/L, while IL-6 levels averaged 65.16 ± 43.67 pg/ml. The results revealed a substantial positive correlation between IL-6 levels and DAS28 (r = 0.603, p value < 0.001), indicating a significant association. Additionally, a moderate correlation between CRP levels and DAS28 (r = 0.493, p value < 0.001) highlighted a significant relationship between these variables. CONCLUSIONS The analysis showed that higher IL-6 levels were associated with increased disease activity and suggested IL-6 as a valuable indicator for assessing RA severity. Also, CRP levels had a moderate correlation with disease activity. Overall, IL-6 is a better marker for disease activity when compared to CRP levels in patients with RA.
Collapse
Affiliation(s)
- Prajakta R Warjukar
- Biochemistry, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research, Nagpur, IND
| | - Ankush V Mohabey
- Orthopedics, All India Institute of Medical Sciences, Nagpur, Nagpur, IND
| | - Pradeep B Jain
- Biochemistry, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research, Nagpur, IND
| | - Gulshan R Bandre
- Microbiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
25
|
Kars ME, Wu Y, Stenson PD, Cooper DN, Burisch J, Peter I, Itan Y. The landscape of rare genetic variation associated with inflammatory bowel disease and Parkinson's disease comorbidity. Genome Med 2024; 16:66. [PMID: 38741190 PMCID: PMC11092054 DOI: 10.1186/s13073-024-01335-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) and Parkinson's disease (PD) are chronic disorders that have been suggested to share common pathophysiological processes. LRRK2 has been implicated as playing a role in both diseases. Exploring the genetic basis of the IBD-PD comorbidity through studying high-impact rare genetic variants can facilitate the identification of the novel shared genetic factors underlying this comorbidity. METHODS We analyzed whole exomes from the BioMe BioBank and UK Biobank, and whole genomes from a cohort of 67 European patients diagnosed with both IBD and PD to examine the effects of LRRK2 missense variants on IBD, PD and their co-occurrence (IBD-PD). We performed optimized sequence kernel association test (SKAT-O) and network-based heterogeneity clustering (NHC) analyses using high-impact rare variants in the IBD-PD cohort to identify novel candidate genes, which we further prioritized by biological relatedness approaches. We conducted phenome-wide association studies (PheWAS) employing BioMe BioBank and UK Biobank whole exomes to estimate the genetic relevance of the 14 prioritized genes to IBD-PD. RESULTS The analysis of LRRK2 missense variants revealed significant associations of the G2019S and N2081D variants with IBD-PD in addition to several other variants as potential contributors to increased or decreased IBD-PD risk. SKAT-O identified two significant genes, LRRK2 and IL10RA, and NHC identified 6 significant gene clusters that are biologically relevant to IBD-PD. We observed prominent overlaps between the enriched pathways in the known IBD, PD, and candidate IBD-PD gene sets. Additionally, we detected significantly enriched pathways unique to the IBD-PD, including MAPK signaling, LPS/IL-1 mediated inhibition of RXR function, and NAD signaling. Fourteen final candidate IBD-PD genes were prioritized by biological relatedness methods. The biological importance scores estimated by protein-protein interaction networks and pathway and ontology enrichment analyses indicated the involvement of genes related to immunity, inflammation, and autophagy in IBD-PD. Additionally, PheWAS provided support for the associations of candidate genes with IBD and PD. CONCLUSIONS Our study confirms and uncovers new LRRK2 associations in IBD-PD. The identification of novel inflammation and autophagy-related genes supports and expands previous findings related to IBD-PD pathogenesis, and underscores the significance of therapeutic interventions for reducing systemic inflammation.
Collapse
Affiliation(s)
- Meltem Ece Kars
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yiming Wu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- College of Life Science, China West Normal University, Nan Chong, Si Chuan, 637009, China
| | - Peter D Stenson
- Institute of Medical Genetics, Cardiff University, Cardiff, CF14 4XN, UK
| | - David N Cooper
- Institute of Medical Genetics, Cardiff University, Cardiff, CF14 4XN, UK
| | - Johan Burisch
- Gastrounit, Medical Division, Copenhagen University Hospital - Amager and Hvidovre, Kettegård Alle 30, Hvidovre, Copenhagen, 2650, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents and Adults, Copenhagen University Hospital - Amager and Hvidovre, Kettegård Alle 30, Hvidovre, Copenhagen, 2650, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen, 2200, Denmark
| | - Inga Peter
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Yuval Itan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
26
|
Kameda H, Tasaka S, Takahashi T, Suzuki K, Soeda N, Tanaka Y. Safety and effectiveness of sarilumab in Japanese patients with rheumatoid arthritis refractory to previous treatments: An interim analysis of a post-marketing surveillance. Mod Rheumatol 2024; 34:444-452. [PMID: 37300807 DOI: 10.1093/mr/road055] [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: 02/01/2023] [Revised: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023]
Abstract
OBJECTIVES An interim analysis of post-marketing surveillance data to assess the safety and effectiveness of sarilumab in Japanese patients with rheumatoid arthritis refractory to previous treatment. METHODS The interim analysis included patients who initiated sarilumab therapy between June 2018 and January 2021. The primary objective of this surveillance was safety. RESULTS In total, 1036 patients were enrolled and registered by 12 January 2021 (interim cut-off date). Of these, 678 were included in the safety analysis [75.4% female; mean age (± standard deviation) 65.8 ± 13.0 years]. Adverse drug reactions, defined as adverse events classified as possibly or probably related to sarilumab, were reported in 170 patients (incidence: 25.1%), with white blood cell count decreased (4.4%) and neutrophil count decreased (1.6%) most frequently reported. Serious haematologic disorders (3.4%) and serious infections (including tuberculosis) (2.5%) were the most frequently reported priority surveillance items. No malignant tumour was reported. An absolute neutrophil count (ANC) below the minimum standard value did not increase the incidence of serious infections. CONCLUSIONS Sarilumab was well tolerated, and no new safety signals were noted in this analysis. There was no difference in the frequency of serious infections between patients with an ANC below or above normal.
Collapse
Affiliation(s)
- Hideto Kameda
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Toho University, Tokyo, Japan
| | - Sadatomo Tasaka
- Department of Respiratory Medicine, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | | | | | - Naoki Soeda
- Medical Affairs Department, Asahi Kasei Pharma Corporation, Tokyo, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
27
|
Dao TNP, Onikanni SA, Fadaka AO, Sibuyi NRS, Le MH, Chang HH. Phytotherapeutic potential of compounds identified from fractionated extracts of Morus alba L., as an inhibitor of interleukin-6 in the treatment of rheumatoid arthritis: computational approach. J Biomol Struct Dyn 2024:1-14. [PMID: 38525928 DOI: 10.1080/07391102.2024.2330713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 03/06/2024] [Indexed: 03/26/2024]
Abstract
The presence of HLA-DRB1 alleles that encode critical points associated with environmental interactions is associated with increased risk of rheumatoid arthritis caused by anti-citrullinated protein antibodies. Therefore, interleukin-6 (IL-6), a multifunctional cytokine that controls both local and systemic acute inflammatory responses through its ability to induce a phase response, plays a serious role. Its overexpression leads to pathological challenges such as rheumatoid arthritis and menopausal osteoporosis. However, targeting the IL-6 receptor and its region could be the major step in controlling the overexpression of this cytokine for therapeutic importance. Therefore, our research explored the computational insight needed to investigate the anti-RFA potential of phytochemicals from fractionated extracts of Morus alba L. against receptors, which have been implicated as druggable targets for the treatment of rheumatoid arthritis. In this study, fifty-nine (59) previously isolated and characterized phytochemicals from M. alba L. were identified from the literature and retrieved from the PubChem database. In silico screening was used to assess the mode of action of these phytochemicals from M. alba L. against receptors that may serve as therapeutic targets for rheumatoid arthritis. Molecular docking studies, toxicity prediction, drug visualization and molecular dynamics simulation (MD) of the ligands together with the receptor-identified target were carried out using the Schrodinger Molecular Drug Discovery Suite. The findings indicated that a selected group of ligands displayed significant binding strength to specific amino acid residues, revealing an important link between the building blocks of proteins (amino acids) and ligands at the inhibitor binding site through traditional chemical interactions, such as interactions between hydrophobic and hydrogen bonds. The binding affinities of the receptors were carefully checked via comparison with those of the approved ligands, and the results suggested structural and functional changes in the lead compounds. Therefore, the bioactive component from M. alba L. could be a lead foot interleukin-6 (IL-6) inhibitor and could be a promising lead compound for the treatment of rheumatoid arthritis and related challenges.
Collapse
Affiliation(s)
- Tran Nhat Phong Dao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan (ROC)
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Sunday Amos Onikanni
- College of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taiwan (ROC)
- Department of Chemical Sciences, Biochemistry Unit, Afe-Babalola University, Ado-Ekiti, Nigeria
| | | | - Nicole Remaliah Samantha Sibuyi
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, University of the Western Cape, Bellville, South Africa
| | - Minh Hoang Le
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Hen-Hong Chang
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan (ROC)
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan (ROC)
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan (ROC)
| |
Collapse
|
28
|
Dao TNP, Onikanni SA, Fadaka AO, Klein A, Tran VD, Le MH, Wang CH, Chang HH. In silico identification of compounds from Piper sarmentosum Roxb leaf fractionated extract inhibit interleukin-6 to prevent rheumatoid arthritis. Front Pharmacol 2024; 15:1358037. [PMID: 38576490 PMCID: PMC10991700 DOI: 10.3389/fphar.2024.1358037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Objective: Medicinal herbs with a phytonutrient background has been applied globally as major alternatives to ameliorate the continuous increase in rheumatoid arthritis cases worldwide. We herein aimed to critically examine the bioactive components of the medicinal herb Piper sarmentosum Roxb leaf fractionated extract for its potential to inhibit the influx of interleukin-6 (IL-6) in rheumatoid arthritis. Methods: The Schrödinger platform was employed as the main computational acumen for the screening of bioactive compounds identified and reference compounds subjected to molecular simulation (MDS) for analyzing the stability of docked complexes to assess fluctuations and conformational changes during protein-ligand interactions. Results: The values of the simulatory properties and principal component analysis (PCA) revealed the good stability of these phytochemicals in the active pocket of interleukin-6 (IL-6). Discussion: Our findings reveal new strategies in which these phytochemicals are potential inhibitory agents that can be modified and further evaluated to develop more effective agents for the management of rheumatoid arthritis, thereby providing a better understanding and useful model for the reproduction and/or discovery of new drugs for the management of rheumatoid arthritis and its complications.
Collapse
Affiliation(s)
- Tran Nhat Phong Dao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Sunday Amos Onikanni
- College of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Chemical Sciences, Biochemistry Unit, Afe-Babalola University, Ado-Ekiti, Nigeria
| | | | - Ashwil Klein
- Department of Biotechnology, University of the Western Cape, Bellville, South Africa
| | - Van De Tran
- Department of Health Organization and Management, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Minh Hoang Le
- Faculty of Traditional Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Chih-Hao Wang
- College of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Cell Biology, China Medical University, Taichung, Taiwan
| | - Hen-Hong Chang
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
29
|
Royaei M, Tahoori MT, Bardania H, Shams A, Dehghan A. Amelioration of inflammation through reduction of oxidative stress in rheumatoid arthritis by treating fibroblast-like synoviocytes (FLS) with DMF-loaded PLGA nanoparticles. Int Immunopharmacol 2024; 129:111617. [PMID: 38309093 DOI: 10.1016/j.intimp.2024.111617] [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: 11/15/2023] [Revised: 01/19/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory condition, and Dimethyl fumarate (DMF) is known for inducing antioxidant enzymes and reducing reactive oxygen species (ROS). Fibroblast-like synoviocytes (FLS) contribute to joint damage by releasing interleukins (IL-1β, IL-6, and IL-8) in response to ROS. Given ROS's impact on FLS acquiring an invasive phenotype, our study explored the effects of poly lactic-co-glycolic acid (PLGA) nanoparticles containing DMF on the expression of the HO-1 enzyme and the inflammatory cytokines IL-1β, IL-6, and IL-8 in FLS cells. METHODS In this study, we evaluated and compared the impact of Free-DMF and PLGA-DMF, on the gene expression of the HO-1 and inflammatory cytokines (IL-1β, IL-6, and IL-8) in FLS cells derived from 13 patients with rheumatoid arthritis. qRT-PCR method was used to quantify the gene expression levels. RESULTS PLGA-DMF nanoparticles demonstrated a significant increase in HO-1 expression and a significant decrease in IL-1β gene expression. Also, a significant decrease in IL-6 gene expression was seen under the effect of Free-DMF. These results indicate the potential effectiveness of PLGA-DMF nanoparticles in reducing inflammation and improving rheumatoid arthritis symptoms. DISCUSSION According to the findings, PLGA-DMF nanoparticles are expected to be effective in reducing inflammation and improving the symptoms of rheumatoid arthritis. Also, further studies on other factors affected by oxidative stress such as cell invasion factors and survival factors after the effect of PLGA-DMF nanoparticle are recommended.
Collapse
Affiliation(s)
- Mohammadreza Royaei
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Taher Tahoori
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran; Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran; Clinical Research Development Unit, Imam Sajad Hospital, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Ali Shams
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Ali Dehghan
- Department of Internal Medicine, Shahid Sadoughi Hospital, Yazd University of Medical Sciences, Yazd, Iran
| |
Collapse
|
30
|
Mohammed RS, Ibrahim FM, El-Akad RH, Al-Mokaddem AK, Ahmed KA, Ashour WES, Attia HN. Antiarthritic activity of Physalis peruviana fruit extract via inhibition of inflammatory mediators: Integrated in vitro, in vivo and in silico study. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117502. [PMID: 38030020 DOI: 10.1016/j.jep.2023.117502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE P. peruviana fruit, native to Andean region, is cultivated worldwide for its adaptability to various soil natures and climatic conditions. It is increasingly consumed for its high nutritional profile and history of ethnomedical uses including treatment of arthritis. Little pharmacological evidences support this folk use except for previous in vitro study that reported significant inhibition of protein denaturation. AIM OF THE STUDY The study aims at providing new in vivo evidence on antiarthritic activity of P. peruviana fruits in vivo that justifies its traditional use through mechanism-based experiment. MATERIAL AND METHODS Inhibition of inflammatory mediators is considered one of the key treatments to alleviate painful symptoms of rheumatoid arthritis (RA). Anti-inflammatory activity was assessed against COX-1 and COX-2 activity in vitro. Serum TNFα, IL-1β and IL-6 were traced using in vivo model of adjuvant-induced arthritis. Gross/inflammatory changes in rat paw, relative mass indices of spleen and liver were further investigated together with joint tissue histoarchitecture. Seven metabolites from different phytochemical classes, that were previously reported in P. peruviana fruit, were evaluated in silico against TNF-α target protein (PDB ID: 2AZ5) to assess their inhibitory effect. This was followed by assessment of their drug-likeness based on Lipinski's rule according to their physicochemical and pharmacokinetic properties. RESULTS High dose of extract (E-1000 mg) improved adjuvant-induced cachexia and attenuated immune-inflammatory responses in paw and serum parameters, with equipotent effect to MTX, in addition to minimal side effect profile on spleen and liver. Histopathological study of knee joint tissues confirmed dose-dependent improvement in arthritic groups treated with P. peruviana fruit extracts. The insilico study recommended steroidal lactones withaperuvin E/C and hydroxywithanolide E as promising lead compounds for inhibiting TNF enzyme as evidenced by docking scores of 6.301, 5.488 and 5.763 kcal/mol, respectively, fitting as well the Lipinski's rule of drug likeness. CONCLUSION The study provided novel approach that rationalize folk use of P. peruviana fruit in treatment of arthritis.
Collapse
Affiliation(s)
- Reda S Mohammed
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Giza, Dokki, P.O. Box 12622, Egypt.
| | - Faten M Ibrahim
- Medicinal and Aromatic Plants Research Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Giza, Dokki, P.O. Box 12622, Egypt.
| | - Radwa H El-Akad
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Giza, Dokki, P.O. Box 12622, Egypt.
| | - Asmaa K Al-Mokaddem
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, 12211, Giza, Egypt.
| | - Kawkab A Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, 12211, Giza, Egypt.
| | - Wedian E-S Ashour
- Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Giza, Dokki, P.O. Box 12622, Egypt.
| | - Hanan N Attia
- Medicinal and Pharmaceutical Chemistry Department (Pharmacology group), Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki-Giza- P.O. Box 12622, Egypt.
| |
Collapse
|
31
|
Zubrzycka-Sienkiewicz A, Klama K, Ullmann M, Petit-Frere C, Baker P, Monnet J, Illes A. Comparison of the efficacy and safety of a proposed biosimilar MSB11456 with tocilizumab reference product in subjects with moderate-to-severe rheumatoid arthritis: results of a randomised double-blind study. RMD Open 2024; 10:e003596. [PMID: 38316489 PMCID: PMC11148666 DOI: 10.1136/rmdopen-2023-003596] [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: 08/09/2023] [Accepted: 01/20/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVE To evaluate the efficacy, immunogenicity and safety of the proposed biosimilar MSB11456 versus European Union (EU)-approved tocilizumab reference product in patients with rheumatoid arthritis (RA) in a multicentre, randomised, double-blind, multinational, parallel-group study (NCT04512001). METHODS Adult patients with moderate-to-severe active RA and inadequate clinical response to ≥1 disease-modifying antirheumatic drug (synthetic or biologic) receiving methotrexate were randomised to receive 24 weekly subcutaneous 162 mg injections of either MSB11456 or EU-approved tocilizumab. Equivalence between treatments was considered if the 95% CI (European Medicines Agency)/90% CI (US Food and Drug Administration) for the difference in mean change from baseline to week 24 in Disease Activity Score-28 Joint Count with erythrocyte sedimentation rate (DAS28-ESR) between treatments was entirely within prespecified equivalence intervals (-0.6 to 0.6 and -0.6 to 0.5, respectively). At week 24, patients were rerandomised to continued treatment or MSB11456. Secondary efficacy endpoints to week 52, and safety and immunogenicity to week 55 were also evaluated. RESULTS At week 24, the least squares mean difference in the change from baseline in DAS28-ESR between treatments was 0.01 (95% CI -0.19 to 0.22) in the 604 randomised patients. Similarity between treatments was shown for all other efficacy, safety and immunogenicity endpoints, including in patients who switched from EU-approved tocilizumab to MSB114466. CONCLUSIONS Therapeutic equivalence was demonstrated for efficacy endpoints, and safety and immunogenicity analyses support the similarity of the two treatments. The results of this study strengthen the evidence that the proposed biosimilar MSB11456 and EU-approved tocilizumab exert similar clinical effects.
Collapse
Affiliation(s)
| | | | | | | | - Peter Baker
- Fresenius Kabi SwissBioSim GmbH, Eysins, Switzerland
| | - Joëlle Monnet
- Fresenius Kabi SwissBioSim GmbH, Eysins, Switzerland
| | - Andras Illes
- Fresenius Kabi SwissBioSim GmbH, Eysins, Switzerland
| |
Collapse
|
32
|
Lee YH, Song GG. Comparison of the efficacy and safety of olokizumab at different dosages in patients with active rheumatoid arthritis: a network meta-analysis of randomized controlled trials. Z Rheumatol 2024; 83:107-114. [PMID: 37266677 DOI: 10.1007/s00393-023-01367-w] [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] [Accepted: 04/07/2023] [Indexed: 06/03/2023]
Abstract
OBJECTIVE This study aimed to assess the relative efficacy and safety of olokizumab at different dosages in patients with active rheumatoid arthritis (RA). METHODS We performed a Bayesian network meta-analysis to combine direct and indirect evidence from randomized controlled trials (RCTs) to examine the efficacy and safety of olokizumab administered intravenously to RA patients at 64 mg/kg every 2 or 4 weeks (Q2 or Q4W). RESULTS Five RCTs comprising 2609 patients met the inclusion criteria. Both olokizumab Q2 and Q4W treatments achieved a significant American College of Rheumatology 20% response (ACR20) compared with the placebo (odds ratio [OR] 3.21, 95% credible interval [CrI] 2.53-4.09; OR 3.05, 95% CrI 2.43-3.86). However, olokizumab Q2W was associated with the most favorable surface using the cumulative ranking curve (SUCRA) for the ACR20 response rate. The ranking probability based on the SUCRA indicated that olokizumab Q2W had the highest probability of being considered the best treatment option for achieving the ACR20 response rate, followed by olokizumab Q4W, adalimumab, and placebo. The ACR50 and 70 response rates showed a similar distribution pattern to the ACR20 response rate, except that olokizumab Q4W had a higher-ranking probability than olokizumab Q2W for ACR50. The SUCRA rating likelihood of adverse events (AEs) and withdrawal due to AEs showed that a placebo was likely to be the best intervention. CONCLUSION Both olokizumab Q2 and Q4W were efficacious and well-tolerated treatments for active RA.
Collapse
Affiliation(s)
- Young Ho Lee
- Department of Rheumatology, Korea University Anam Hospital, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, 02841, Seoul, Korea (Republic of).
| | - Gwan Gyu Song
- Department of Rheumatology, Korea University Anam Hospital, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, 02841, Seoul, Korea (Republic of)
| |
Collapse
|
33
|
Ho Lee Y, Gyu Song G. Comparison of the efficacy and safety of tocilizumab, sarilumab, and olokizumab in patients with active rheumatoid arthritis: a network meta-analysis of randomized controlled trials. Z Rheumatol 2024; 83:97-106. [PMID: 36607422 DOI: 10.1007/s00393-022-01315-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVE This study compared the relative efficacy and safety of olokizumab, tocilizumab, and sarilumab in rheumatoid arthritis (RA) patients who were intolerant or responding inadequately to methotrexate (MTX). METHODS We performed a Bayesian network meta-analysis to combine direct and indirect evidence from randomized controlled trials (RCTs) to examine the efficacy and safety of olokizumab, tocilizumab, and sarilumab in RA patients who were intolerant or responding inadequately to MTX. RESULTS Six RCTs comprising 4439 patients met the inclusion criteria. Tocilizumab, sarilumab, olokizumab, and adalimumab treatments achieved a significant American College of Rheumatology 20% (ACR20) response rate compared with placebo. However, tocilizumab was associated with the most favorable surface area using the cumulative ranking curve (SUCRA) for the ACR20 response rate. The ranking probability based on the SUCRA indicated that tocilizumab treatment had the highest probability of providing the best ACR20 response rate, followed by sarilumab, olokizumab every 2 weeks (Q2W), olokizumab Q4W, adalimumab 40 mg, and placebo. The ACR50 and 70 response rates showed a distribution pattern similar to that of the ACR20 response rate. However, olokizumab Q4W had a higher ranking probability than olokizumab Q2W. The SUCRA rating showed that the placebo was the best intervention with the least adverse events (AEs) and withdrawal due to AEs, followed by interleukin‑6 inhibitors. CONCLUSION Tocilizumab, sarilumab, and olokizumab are more effective than adalimumab and have similar efficacy and safety in RA patients with inadequate responses to MTX.
Collapse
Affiliation(s)
- Young Ho Lee
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea (Republic of).
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, 73, Goryeodae-ro, 02841, Seongbuk-gu, Seoul, Korea (Republic of).
| | - Gwan Gyu Song
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea (Republic of)
| |
Collapse
|
34
|
Lee YE, Lee SH, Kim WU. Cytokines, Vascular Endothelial Growth Factors, and PlGF in Autoimmunity: Insights From Rheumatoid Arthritis to Multiple Sclerosis. Immune Netw 2024; 24:e10. [PMID: 38455464 PMCID: PMC10917575 DOI: 10.4110/in.2024.24.e10] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
In this review, we will explore the intricate roles of cytokines and vascular endothelial growth factors in autoimmune diseases (ADs), with a particular focus on rheumatoid arthritis (RA) and multiple sclerosis (MS). AD is characterized by self-destructive immune responses due to auto-reactive T lymphocytes and Abs. Among various types of ADs, RA and MS possess inflammation as a central role but in different sites of the patients. Other common aspects among these two ADs are their chronicity and relapsing-remitting symptoms requiring continuous management. First factor inducing these ADs are cytokines, such as IL-6, TNF-α, and IL-17, which play significant roles in the pathogenesis by contributing to inflammation, immune cell activation, and tissue damage. Secondly, vascular endothelial growth factors, including VEGF and angiopoietins, are crucial in promoting angiogenesis and inflammation in these two ADs. Finally, placental growth factor (PlGF), an emerging factor with bi-directional roles in angiogenesis and T cell differentiation, as we introduce as an "angio-lymphokine" is another key factor in ADs. Thus, while angiogenesis recruits more inflammatory cells into the peripheral sites, cytokines secreted by effector cells play critical roles in the pathogenesis of ADs. Various therapeutic interventions targeting these soluble molecules have shown promise in managing autoimmune pathogenic conditions. However, delicate interplay between cytokines, angiogenic factors, and PlGF has more to be studied when considering their complementary role in actual pathogenic conditions. Understanding the complex interactions among these factors provides valuable insights for the development of innovative therapies for RA and MS, offering hope for improved patient outcomes.
Collapse
Affiliation(s)
- Young eun Lee
- Graduate School of Medical Science and Engineering (GSMSE), Biomedical Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seung-Hyo Lee
- Graduate School of Medical Science and Engineering (GSMSE), Biomedical Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
35
|
Prabha J, Kumar M, Kumar D, Chopra S, Bhatia A. Nano-platform Strategies of Herbal Components for the Management of Rheumatoid Arthritis: A Review on the Battle for Next-Generation Formulations. Curr Drug Deliv 2024; 21:1082-1105. [PMID: 37622715 DOI: 10.2174/1567201821666230825102748] [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: 03/10/2023] [Revised: 06/01/2023] [Accepted: 07/06/2023] [Indexed: 08/26/2023]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that initially affects small joints and then spreads to the bigger joints. It also affects other organs of the body such as lungs, eyes, kidneys, heart, and skin. In RA, there is destruction of cartilage and joints, and ligaments and tendons become brittle. Damage to the joints leads to abnormalities and bone degradation, which may be quite painful for the patient. METHOD The nano-carriers such as liposomes, phytosomes, nanoparticles, microcapsules, and niosomes are developed to deliver the encapsulated phytoconstituents to targeted sites for the better management of RA. RESULTS The phytoconstituents loaded nano-carriers have been used in order to increase bioavailability, stability and reduce the dose of an active compound. In one study, the curcumin-loaded phytosomes increase the bioavailability of curcumin and also provides relief from RA symptoms. The drug-loaded nano-carriers are the better option for the management of RA. CONCLUSION In conclusion, there are many anti-arthritic herbal and synthetic medicine available in the market that are currently used in the treatment of RA. However, chronic use of these medications may result in a variety of side effects. Because therapy for RA is frequently necessary for the rest of ones life. The use of natural products may be a better option for RA management. These phytoconstituents, however, have several disadvantages, including limited bioavailability, low stability, and the need for a greater dosage. These problems can be rectified by using nano-technology.
Collapse
Affiliation(s)
- Jyoti Prabha
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Mohit Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Devesh Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Shruti Chopra
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh - 201313, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| |
Collapse
|
36
|
Pan S, Wu S, Wei Y, Liu J, Zhou C, Chen T, Zhu J, Tan W, Huang C, Feng S, Zhang B, Wei W, Zhan X, Liu C. Exploring the causal relationship between inflammatory cytokines and inflammatory arthritis: A Mendelian randomization study. Cytokine 2024; 173:156446. [PMID: 37979213 DOI: 10.1016/j.cyto.2023.156446] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 11/14/2023] [Indexed: 11/20/2023]
Abstract
OBJECTIVES Previous studies have reported an association between inflammatory cytokines and inflammatory arthritis, including Ankylosing spondylitis (AS), rheumatoid arthritis (RA), and psoriatic arthritis (PsA). This study aims to explore the causal relationship between inflammatory cytokines and AS, RA, and PsA using Mendelian randomization (MR). METHODS We conducted a bidirectional two-sample MR analysis using genetic summary data from a publicly available genome-wide association study (GWAS) that included 41 genetic variations of inflammatory cytokines, as well as genetic variant data for AS, RA, and PsA from the FinnGen consortium. The main analysis method used was Inverse variance weighted (IVW) to investigate the causal relationship between exposure and outcome. Additionally, other methods such as MR Egger, weighted median (WM), simple mode, and weighted mode were employed to strengthen the final results. Sensitivity analysis was also performed to ensure the reliability of the findings. RESULTS The results showed that macrophage colony-stimulating factor (MCSF) was associated with an increased risk of AS (OR = 1.163, 95 % CI = 1.016-1.33, p = 0.028). Conversely, high levels of TRAIL and beta nerve growth factor (β-NGF) were associated with a decreased risk of AS (OR = 0.892, 95 % CI = 0.81-0.982, p = 0.002; OR = 0.829, 95 % CI = 0.696-0.988, p = 0.036). Four inflammatory cytokines were found to be associated with an increased risk of PsA: vascular endothelial growth factor (VEGF) (OR = 1.161, 95 % CI = 1.057-1.275, p = 0.002); Interleukin 12p70 (IL12p70) (OR = 1.189, 95 % CI = 1.049-1.346, p = 0.007); IL10 (OR = 1.216, 95 % CI = 1.024-1.444, p = 0.026); IL13 (OR = 1.159, 95 % CI = 1.05-1.28, p = 0.004). Interleukin 1 receptor antagonist (IL-1rα) was associated with an increased risk of seropositive RA (OR = 1.181, 95 % CI = 1.044-1.336, p = 0.008). Similarly, genetic susceptibility to inflammatory arthritis was found to be causally associated with multiple inflammatory cytokines. Lastly, the sensitivity analysis supported the robustness of these findings. CONCLUSIONS This study provides additional insights into the relationship between inflammatory cytokines and inflammatory arthritis, and may offer new clues for the etiology, diagnosis, and treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Shixin Pan
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shaofeng Wu
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yating Wei
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingjing Liu
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province (First People's Hospital of Honghe State), Yunnan, China
| | - Chenxing Zhou
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tianyou Chen
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jichong Zhu
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weiming Tan
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chengqian Huang
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Sitan Feng
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bin Zhang
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wendi Wei
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xinli Zhan
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Chong Liu
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
37
|
Makris A, Barkas F, Sfikakis PP, Liberopoulos E, Filippatos TD, Ray KK, Agouridis AP. Lipoprotein(a), Interleukin-6 inhibitors, and atherosclerotic cardiovascular disease: Is there an association? ATHEROSCLEROSIS PLUS 2023; 54:1-6. [PMID: 37720252 PMCID: PMC10500445 DOI: 10.1016/j.athplu.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/09/2023] [Accepted: 09/06/2023] [Indexed: 09/19/2023]
Abstract
Background and aims Lipoprotein(a) [Lp(a)] and interleuking-6 (IL-6), an inflammation biomarker, have been established as distinct targets of the residual atherosclerotic cardiovascular disease (ASCVD) risk. We aimed to investigate the association between them, and the potential clinical implications in ASCVD prevention. Methods A literature search was conducted in PubMed until December 31st, 2022, using relevant keywords. Results Elevated lipoprotein(a) [Lp(a)] levels constitute the most common inherited lipid disorder associated with ASCVD. Although Lp(a) levels are mostly determined genetically by the LPA gene locus, they may be altered by acute conditions of stress and chronic inflammatory diseases. Considering its resemblance with low-density lipoproteins, Lp(a) is involved in atherosclerosis, but it also exerts oxidative, thrombotic, antifibrinolytic and inflammatory properties. The cardiovascular efficacy of therapies lowering Lp(a) by >90% is currently investigated. On the other hand, interleukin (IL)-1b/IL-6 pathway also plays a pivotal role in atherosclerosis and residual ASCVD risk. IL-6 receptor inhibitors [IL-6(R)i] lower Lp(a) by 16-41%, whereas ongoing trials are investigating their potential anti-atherosclerotic effect. The Lp(a)-lowering effect of IL-6(R)i might be attributed to the inhibition of the IL-6 response elements in the promoter region of the LPA gene. Conclusions Although the effect of IL-6(R)i on Lp(a) levels is inferior to that of available Lp(a)-lowering therapies, the dual effect of the former on both inflammation and apolipoprotein (a) synthesis may prove of equal or even greater significance when it comes ASCVD outcomes. More trials are required to establish IL-6(R)i in ASCVD prevention and elucidate their interplay with Lp(a) as well as its clinical significance.
Collapse
Affiliation(s)
- Anastasios Makris
- School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Fotios Barkas
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Petros P. Sfikakis
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Evangelos Liberopoulos
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | | | - Kausik K. Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Aris P. Agouridis
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Department of Internal Medicine, German Oncology Center, Limassol, Cyprus
| |
Collapse
|
38
|
Hoffman JM, Robinson R, Greenway G, Glass J, Budkin S, Sharma S. Blockade of interleukin-6 trans-signaling prevents mitochondrial dysfunction and cellular senescence in retinal endothelial cells. Exp Eye Res 2023; 237:109721. [PMID: 37956941 PMCID: PMC10759313 DOI: 10.1016/j.exer.2023.109721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
Interleukin-6 (IL-6) is a multifaceted cytokine implicated in the pathogenesis of diabetic retinopathy (DR). Its activity extends through cis- and trans-signaling (TS) pathways, with cis-signaling limited to specific cell types possessing the membrane-bound IL-6 receptor, while trans-signaling broadly activates various cells without the membrane bound IL-6 receptor, including retinal endothelial cells. In this study, we determined the effects of interleukin-6 trans-signaling on mitochondrial dysfunction and cellular senescence in human retinal endothelial cells (HRECs). HRECs were cultured and treated with IL-6 + soluble IL-6R or Hyper IL-6 to activate trans-signaling, along with sgp130Fc for inhibition. RT-PCR was used to analyze gene expression changes associated with inflammation and senescence. Cellular senescence was assessed using SA β-gal staining. Mitochondrial function was evaluated using Seahorse XFe24 Bioanalyzer. IL-6 trans-signaling induced inflammatory gene expression as indicated by the upregulation of ICAM1, MCP1, and SERPINA3 levels. Additionally, it reduced mitochondrial respiration and oxidative phosphorylation, and these effects were counteracted by sgp130Fc. Moreover, IL-6 trans-signaling led to altered expression of apoptosis-associated genes, including downregulation of FIS1, BCL2, and MCL1, while promoting cellular senescence, a phenomenon mitigated by sgp130Fc. These results not only deepen our understanding of IL-6 in DR but also carry broader implications for age-related diseases and the aging process itself. This study underscores the potential therapeutic value of targeting IL-6 trans-signaling with sgp130Fc as a promising anti-inflammatory approach for DR and potentially other inflammatory conditions. Further in-vivo investigations are warranted to elucidate the function of IL-6 trans-signaling in aging-related pathologies and overall organismal health.
Collapse
Affiliation(s)
- Jessica M Hoffman
- Department of Biological Sciences, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| | - Rebekah Robinson
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Grace Greenway
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Joshua Glass
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Stepan Budkin
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Shruti Sharma
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA; Department of Ophthalmology, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
39
|
Lai R, Deng X, Lv X, Liu Q, Zhou K, Peng D. Causal relationship between rheumatoid arthritis and hypothyroidism or hyperthyroidism: a bidirectional two-sample univariable and multivariable Mendelian randomization study. Front Endocrinol (Lausanne) 2023; 14:1256208. [PMID: 38093966 PMCID: PMC10716525 DOI: 10.3389/fendo.2023.1256208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Objective The causal relationship between Rheumatoid arthritis (RA) and hypothyroidism/hyperthyroidism remains controversial due to the limitations of conventional observational research, such as confounding variables and reverse causality. We aimed to examine the potential causal relationship between RA and hypothyroidism/hyperthyroidism using Mendelian randomization (MR). Method We conducted a bidirectional two-sample univariable analysis to investigate the potential causal relationship between hypothyroidism/hyperthyroidism and RA. Furthermore, we performed a multivariate analysis to account for the impact of body mass index (BMI), smoking quantity, and alcohol intake frequency. Results The univariable analysis indicated that RA has a causative influence on hypothyroidism (odds ratio [OR]=1.07, 95% confidence interval [CI]=1.01-1.14, P=0.02) and hyperthyroidism (OR=1.32, 95% CI=1.15-1.52, P<0.001). When hypothyroidism/hyperthyroidism was considered as an exposure variable, we only observed a causal relationship between hypothyroidism (OR=1.21, 95% CI=1.05-1.40, P=0.01) and RA, whereas no such connection was found between hyperthyroidism (OR=0.91, 95% CI=0.83-1.01, P=0.07) and RA. In the multivariate MR analyses, after separately and jointly adjusting for the effects of daily smoking quantity, alcohol intake frequency, and BMI, the causal impact of RA on hypothyroidism/hyperthyroidism and hypothyroidism on RA remained robust. However, there is no evidence to suggest a causal effect of hyperthyroidism on the risk of RA (P >0.05). Conclusion Univariate and multivariate MR analyses have validated the causal association between RA and hypothyroidism/hyperthyroidism. Hypothyroidism confirmed a causal relationship with RA when employed as an exposure variable, whereas no such relationship was found between hyperthyroidism and RA.
Collapse
Affiliation(s)
- Rui Lai
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinmin Deng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofeng Lv
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Liu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kun Zhou
- The Third Clinical School of Zhejiang Chinese Medicine University, Hangzhou, China
| | - Dezhong Peng
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
40
|
Palmisano B, Riminucci M, Karsenty G. Interleukin-6 signaling in osteoblasts regulates bone remodeling during exercise. Bone 2023; 176:116870. [PMID: 37586472 DOI: 10.1016/j.bone.2023.116870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/29/2023] [Accepted: 08/07/2023] [Indexed: 08/18/2023]
Abstract
Aerobic exercise has many beneficial effects on human health. One of them, is to influence positively bone remodeling through, however, incompletely understood mechanisms. Given its recently demonstrated role as a mediator of the bone to muscle to bone crosstalk during exercise, we hypothesized that interleukin-6 (IL-6) signaling in bone may contribute to the beneficial effect that exercise has on bone homeostasis. In this study, we first show that aerobic exercise increases the expression of Il6r in bones of WT mice. Then, we analyzed a mutant mouse strain that lacks the IL-6 receptor alpha specifically in osteoblasts (Il6rosb-/-). As it has been reported in the case of Il6-/- mice, in sedentary conditions, bone mass and remodeling were normal in adult Il6rosb-/- mice when compared to controls. In contrast, Il6rosb-/- mice that were subjected to aerobic exercise did not show the increase in bone mass and remodeling parameters that control littermates demonstrated. Moreover, Il6rosb-/- mice undergoing aerobic exercise showed a severe impairment in bone formation, indicating that activation of bone-forming cells is defective when IL-6 signaling in osteoblasts is disrupted. In sum, this study provides evidence that a function of IL-6 signaling in osteoblasts is to promote high bone turnover during aerobic exercise.
Collapse
Affiliation(s)
- Biagio Palmisano
- Department of Genetics and Development, Columbia University Irving Medical Center, 701 W 168th street, New York, NY 10032, United States of America.
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University Irving Medical Center, 701 W 168th street, New York, NY 10032, United States of America.
| |
Collapse
|
41
|
Oberemok VV, Andreeva O, Laikova K, Alieva E, Temirova Z. Rheumatoid Arthritis Has Won the Battle but Not the War: How Many Joints Will We Save Tomorrow? MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1853. [PMID: 37893571 PMCID: PMC10608469 DOI: 10.3390/medicina59101853] [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: 09/11/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023]
Abstract
Rheumatoid arthritis refers to joint diseases of unclear etiology whose final stages can lead to unbearable pain and complete immobility of the affected joints. As one of the most widely known diseases of the joints, it serves as a study target for a large number of research groups and pharmaceutical companies. Modern treatment with anti-inflammatory drugs, including janus kinase (JAK) inhibitors, monoclonal antibodies, and botanicals (polyphenols, glycosides, alkaloids, etc.) has achieved some success and hope for improving the course of the disease. However, existing drugs against RA have a number of side effects which push researchers to elaborate on more selective and effective drug candidates. The avant-garde of research, which aims to develop treatment of rheumatoid arthritis using antisense oligonucleotides along with nonsteroidal drugs and corticosteroids against inflammation, increases the chances of success and expands the arsenal of drugs. The primary goal in the treatment of this disease is to find therapies that allow patients with rheumatoid arthritis to move their joints without pain. The main purpose of this review is to show the victories and challenges for the treatment of rheumatoid arthritis and the tortuous but promising path of research that aims to help patients experience the joy of freely moving joints without pain.
Collapse
Grants
- No. FZEG-2021-0009 Department of Molecular Genetics and Biotechnologies, Institute of Biochemical Technologies, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, Simferopol, Crimea
- No. FZEG-2021-0009 Department of Molecular Genetics and Biotechnologies, Institute of Biochemical Technologies, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, Simferopol, Crimea
Collapse
Affiliation(s)
- Volodymyr V. Oberemok
- Department of Molecular Genetics and Biotechnologies, Institute of Biochemical Technologies, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, Simferopol 295007, Crimea; (O.A.); (K.L.); (E.A.); (Z.T.)
| | | | | | | | | |
Collapse
|
42
|
Orsini F, Crotti C, Cincinelli G, Di Taranto R, Amati A, Ferrito M, Varenna M, Caporali R. Bone Involvement in Rheumatoid Arthritis and Spondyloartritis: An Updated Review. BIOLOGY 2023; 12:1320. [PMID: 37887030 PMCID: PMC10604370 DOI: 10.3390/biology12101320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023]
Abstract
Several rheumatologic diseases are primarily distinguished by their involvement of bone tissue, which not only serves as a mere target of the condition but often plays a pivotal role in its pathogenesis. This scenario is particularly prominent in chronic inflammatory arthritis such as rheumatoid arthritis (RA) and spondyloarthritis (SpA). Given the immunological and systemic nature of these diseases, in this review, we report an overview of the pathogenic mechanisms underlying specific bone involvement, focusing on the complex interactions that occur between bone tissue's own cells and the molecular and cellular actors of the immune system, a recent and fascinating field of interest defined as osteoimmunology. Specifically, we comprehensively elaborate on the distinct pathogenic mechanisms of bone erosion seen in both rheumatoid arthritis and spondyloarthritis, as well as the characteristic process of aberrant bone formation observed in spondyloarthritis. Lastly, chronic inflammatory arthritis leads to systemic bone involvement, resulting in systemic bone loss and consequent osteoporosis, along with increased skeletal fragility.
Collapse
Affiliation(s)
- Francesco Orsini
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Chiara Crotti
- Bone Diseases Unit, Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Gilberto Cincinelli
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Raffaele Di Taranto
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Andrea Amati
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Matteo Ferrito
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Massimo Varenna
- Bone Diseases Unit, Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| | - Roberto Caporali
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy (A.A.)
- Department of Rheumatology and Medical Sciences, ASST G.Pini-CTO, 20122 Milan, Italy
| |
Collapse
|
43
|
Lee YH, Song GG. Relative effectiveness and safety of interleukin-6 and Janus kinase inhibitors versus adalimumab in patients with rheumatoid arthritis: a network meta-analysis. Z Rheumatol 2023; 82:696-705. [PMID: 36427070 DOI: 10.1007/s00393-022-01290-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The relative efficacy and tolerability of interleukin‑6 (IL-6) and Janus kinase (JAK) inhibitor therapies were compared with those of adalimumab in patients with rheumatoid arthritis (RA) and inadequate responses to methotrexate (MTX). METHODS We performed a Bayesian network meta-analysis to combine direct and indirect evidence from randomized controlled trials (RCTs) to examine the efficacy and safety of IL‑6 inhibitors, JAK inhibitors, and adalimumab in patients with RA and inadequate responses to MTX. RESULTS Seven RCTs, which included 4428 patients (1066 for IL‑6 inhibitors and 3362 for JAK inhibitors), met the inclusion criteria. IL‑6 inhibitors were placed at the top left of the league table diagonal (Odds ratio, OR 1.43; 95% CrI 1.12-1.82) as these were correlated with the most beneficial ACR20 response rate. Conversely, the placebo was placed at the bottom right of the league table diagonal as it was correlated with the least desirable effects. IL‑6 and JAK inhibitors produced a substantial ACR20 response relative to adalimumab. The surface under the cumulative ranking curve (SUCRA) revealed that treatment with IL‑6 inhibitors had the greatest ability to reach the ACR20 response rate (SUCRA = 0.826), followed by treatment with JAK inhibitors (SUCRA = 0.672) and adalimumab (SUCRA = 0.001). The ACR50 and ACR70 rates displayed patterns similar to the ACR20 response rate. With regard to serious adverse events (SAEs), the SUCRA rating likelihood showed that adalimumab was likely to be the best intervention, followed by JAK and IL‑6 inhibitors. CONCLUSION Both IL‑6 and JAK inhibitors are more effective than adalimumab and have similar effects in patients with RA and an inadequate response to MTX. Adalimumab is likely to be safer than JAK and IL‑6 inhibitors.
Collapse
Affiliation(s)
- Young Ho Lee
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea (Republic of).
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, 73, Goryeodae-ro, 02841, Seongbuk-gu, Seoul, Korea (Republic of).
| | - Gwan Gyu Song
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea (Republic of)
| |
Collapse
|
44
|
Monteleone G, Moscardelli A, Colella A, Marafini I, Salvatori S. Immune-mediated inflammatory diseases: Common and different pathogenic and clinical features. Autoimmun Rev 2023; 22:103410. [PMID: 37597601 DOI: 10.1016/j.autrev.2023.103410] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
The term "immune-mediated inflammatory diseases (IMIDs)" refers to several inflammatory pathologies of multifactorial etiology and involving either simultaneously or sequentially more organs. IMIDs share some common pathogenic mechanisms, which account for some similarities in the clinical course and the impact that these diseases may have on other organs and systems of the body. However, there are some differences in the IMID-associated pathological process, including the synthesis and function of multiple inflammatory cytokines, which are supposed to perpetuate the tissue-damaging inflammation. This justifies the different indications and responsiveness to corticosteroids, immunosuppressors, small molecules, and biologics. Many individuals with IMIDs are, however, intolerant, or unresponsive to the current drugs, thus suggesting the necessity of novel therapeutic approaches, such as the combination of compounds that either inhibit more immuno-inflammatory networks selectively or simultaneously suppress inflammatory signals and activate counter-regulatory pathways. In this article, we highlight the most relevant features of IMIDs and discuss how clinicians can combat the detrimental immune response in such disorders.
Collapse
Affiliation(s)
- Giovanni Monteleone
- Gastroenterology Unit, Azienda Ospedaliera Policlinico Tor Vergata, Rome, Italy; Department of Systems Medicine, University of "Tor Vergata", Rome, Italy.
| | | | - Alice Colella
- Gastroenterology Unit, Azienda Ospedaliera Policlinico Tor Vergata, Rome, Italy
| | - Irene Marafini
- Gastroenterology Unit, Azienda Ospedaliera Policlinico Tor Vergata, Rome, Italy
| | - Silvia Salvatori
- Gastroenterology Unit, Azienda Ospedaliera Policlinico Tor Vergata, Rome, Italy; Department of Systems Medicine, University of "Tor Vergata", Rome, Italy
| |
Collapse
|
45
|
Hassan WM, Othman N, Daghestani M, Warsy A, Omair MA, Alqurtas E, Amin S, Ismail A, El-Ansary A, Bhat RS, Omair MA. The Fidelity of Rheumatoid Arthritis Multivariate Diagnostic Biomarkers Using Discriminant Analysis and Binary Logistic Regression. Biomolecules 2023; 13:1305. [PMID: 37759705 PMCID: PMC10526504 DOI: 10.3390/biom13091305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that causes multi-articular synovitis. The illness is characterized by worsening inflammatory synovitis, which causes joint swelling and pain. Synovitis erodes articular cartilage and marginal bone, resulting in joint deterioration. This bone injury is expected to be permanent. Cytokines play a prominent role in the etiology of RA and could be useful as early diagnostic biomarkers. This research was carried out at Riyadh's King Khalid University Hospital (KKUH). Patients were enrolled from the Rheumatology unit. Seventy-eight RA patients were recruited (67 (85.9%) females and 11 (14.1%) males). Patients were selected for participation by convenience sampling. Demographic data were collected, and disease activity measurements at 28 joints were recorded using the disease activity score (DAS-28). Age- and sex-matched controls from the general population were included in the study. A panel of 27 cytokines, chemokines, and growth factors was determined in patient and control sera. Binary logistic regression (BLR) and discriminant analysis (DA) were used to analyze the data. We show that multiple cytokine biomarker profiles successfully distinguished RA patients from healthy controls. IL-17, IL-4, and RANTES were among the most predictive variables and were the only biomarkers incorporated into both BLR and DA predictive models for pooled participants (men and women). In the women-only models, the significant cytokines incorporated in the model were IL-4, IL-17, MIP-1b, and RANTES for the BLR model and IL-4, IL-1Ra, GM-CSF, IL-17, and eotaxin for the DA model. The BLR and DA men-only models contained one cytokine each, eotaxin for BLR and platelet-derived growth factor-bb (PDGF-BB) for DA. We show that BLR has a higher fidelity in identifying RA patients than DA. We also found that the use of gender-specific models marginally improves detection fidelity, indicating a possible benefit in clinical diagnosis. More research is needed to determine whether this conclusion will hold true in various and larger patient populations.
Collapse
Affiliation(s)
- Wail M. Hassan
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA;
| | - Nashwa Othman
- Central Research Laboratory, Center for Science and Medical Studies for Girls, King Saud University, Riyadh 11495, Saudi Arabia; (N.O.); (A.W.)
| | - Maha Daghestani
- Department of Zoology, College of Science, Center for Science and Medical Studies for Girls, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Arjumand Warsy
- Central Research Laboratory, Center for Science and Medical Studies for Girls, King Saud University, Riyadh 11495, Saudi Arabia; (N.O.); (A.W.)
| | - Maha A. Omair
- Department of Statistics and Operations Research, College of Sciences, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Eman Alqurtas
- Rheumatology Unit, Department of Medicine, College of Medicine, King Saud University, Riyadh 11495, Saudi Arabia; (E.A.); (S.A.); (M.A.O.)
| | - Shireen Amin
- Rheumatology Unit, Department of Medicine, College of Medicine, King Saud University, Riyadh 11495, Saudi Arabia; (E.A.); (S.A.); (M.A.O.)
| | - Abdulaziz Ismail
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11495, Saudi Arabia;
| | - Afaf El-Ansary
- Central Research Laboratory, Center for Science and Medical Studies for Girls, King Saud University, Riyadh 11495, Saudi Arabia; (N.O.); (A.W.)
| | - Ramesa Shafi Bhat
- Biochemistry Department, College of Science, Center for Science and Medical Studies for Girls, King Saud University, Riyadh 11495, Saudi Arabia;
| | - Mohammed A. Omair
- Rheumatology Unit, Department of Medicine, College of Medicine, King Saud University, Riyadh 11495, Saudi Arabia; (E.A.); (S.A.); (M.A.O.)
| |
Collapse
|
46
|
Aripova N, Duryee MJ, England BR, Hunter CD, Mordeson JE, Ryan EM, Daubach EC, Romberger DJ, Thiele GM, Mikuls TR. Citrullinated and malondialdehyde-acetaldehyde modified fibrinogen activates macrophages and promotes an aggressive synovial fibroblast phenotype in patients with rheumatoid arthritis. Front Immunol 2023; 14:1203548. [PMID: 37654483 PMCID: PMC10467288 DOI: 10.3389/fimmu.2023.1203548] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/27/2023] [Indexed: 09/02/2023] Open
Abstract
Objective Post-translational protein modifications with malondialdehyde-acetaldehyde (MAA) and citrulline (CIT) are implicated in the pathogenesis of rheumatoid arthritis (RA). Although precise mechanisms have not been elucidated, macrophage-fibroblast interactions have been proposed to play a central role in the development and progression of RA. The purpose of our study was to evaluate the downstream effects of macrophage released soluble mediators, following stimulation with fibrinogen (FIB) modified antigens, on human fibroblast-like synoviocytes (HFLS). Methods PMA-treated U-937 monocytes (Mϕ) and macrophage-differentiated peripheral blood mononuclear cells (MP) were stimulated with FIB, FIB-MAA, FIB-CIT, or FIB-MAA-CIT. HFLS-RA cells were stimulated directly with FIB antigens or with supernatants (SN) from macrophages (Mϕ-SN or MP-SN) stimulated with FIB antigens. Genes associated with an aggressive HFLS phenotype, extracellular matrix proteins, and activated signaling pathways were evaluated. Results HFLS-RA cells treated with Mϕ-SNFIB-CIT and Mϕ-SNFIB-MAA-CIT demonstrated significant increases in mRNA expression of genes associated with an aggressive phenotype at 24-h as compared to direct stimulation with the same antigens. Similar results were obtained using MP-SN. Cellular morphology was altered and protein expression of vimentin (p<0.0001 vs. Mϕ-SNFIB) and type II collagen (p<0.0001) were significantly increased in HFLS-RA cells treated with any of the Mϕ-SN generated following stimulation with modified antigens. Phosphorylation of JNK, Erk1/2, and Akt were increased most substantially in HFLS-RA treated with Mϕ-SNFIB-MAA-CIT (p<0.05 vs Mϕ-SNFIB). These and other data suggested the presence of PDGF-BB in Mϕ-SN. Mϕ-SNFIB-MAA-CIT contained the highest concentration of PDGF-BB (p<0.0001 vs. Mϕ-SNFIB) followed by Mϕ-SNFIB-CIT then Mϕ-SNFIB-MAA. HFLS-RA cells treated with PDGF-BB showed similar cellular morphology to the Mϕ-SN generated following stimulation with modified FIB, as well as the increased expression of vimentin, type II collagen, and the phosphorylation of JNK, Erk1/2 and Akt signaling molecules. Conclusion Together, these findings support the hypothesis that in response to MAA-modified and/or citrullinated fibrinogen, macrophages release soluble factors including PDGF-BB that induce fibroblast activation and promote an aggressive fibroblast phenotype. These cellular responses were most robust following macrophage activation with dually modified fibrinogen, compared to single modification alone, providing novel insights into the combined role of multiple post-translational protein modifications in the development of RA.
Collapse
Affiliation(s)
- Nozima Aripova
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michael J. Duryee
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Bryant R. England
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Carlos D. Hunter
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Jack E. Mordeson
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Evan M. Ryan
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Eric C. Daubach
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Debra J. Romberger
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Omaha, NE, United States
| | - Geoffrey M. Thiele
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Ted R. Mikuls
- Department of Internal Medicine, Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Research Services 151, Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| |
Collapse
|
47
|
Weber BN, Giles JT, Liao KP. Shared inflammatory pathways of rheumatoid arthritis and atherosclerotic cardiovascular disease. Nat Rev Rheumatol 2023; 19:417-428. [PMID: 37231248 PMCID: PMC10330911 DOI: 10.1038/s41584-023-00969-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2023] [Indexed: 05/27/2023]
Abstract
The association between chronic inflammation and increased risk of cardiovascular disease in rheumatoid arthritis (RA) is well established. In the general population, inflammation is an established independent risk factor for cardiovascular disease, and much interest is placed on controlling inflammation to reduce cardiovascular events. As inflammation encompasses numerous pathways, the development of targeted therapies in RA provides an opportunity to understand the downstream effect of inhibiting specific pathways on cardiovascular risk. Data from these studies can inform cardiovascular risk management in patients with RA, and in the general population. This Review focuses on pro-inflammatory pathways targeted by existing therapies in RA and with mechanistic data from the general population on cardiovascular risk. Specifically, the discussions include the IL-1, IL-6 and TNF pathways, as well as the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signalling pathway, and the role of these pathways in RA pathogenesis in the joint alongside the development of atherosclerotic cardiovascular disease. Overall, some robust data support inhibition of IL-1 and IL-6 in decreasing the risk of cardiovascular disease, with growing data supporting IL-6 inhibition in both patients with RA and the general population to reduce the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Brittany N Weber
- Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jon T Giles
- Columbia University, Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Katherine P Liao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA.
- Rheumatology Section, VA Boston Medical Center, Boston, MA, USA.
| |
Collapse
|
48
|
Mashraqi MM, Alzamami A, Alturki NA, Alshamrani S, Alshahrani MM, Almasoudi HH, Basharat Z. Molecular Mimicry Mapping in Streptococcus pneumoniae: Cues for Autoimmune Disorders and Implications for Immune Defense Activation. Pathogens 2023; 12:857. [PMID: 37513704 PMCID: PMC10383125 DOI: 10.3390/pathogens12070857] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/08/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Streptococcus pneumoniae contributes to a range of infections, including meningitis, pneumonia, otitis media, and sepsis. Infections by this bacterium have been associated with the phenomenon of molecular mimicry, which, in turn, may contribute to the induction of autoimmunity. In this study, we utilized a bioinformatics approach to investigate the potential for S. pneumoniae to incite autoimmunity via molecular mimicry. We identified 13 S. pneumoniae proteins that have significant sequence similarity to human proteins, with 11 of them linked to autoimmune disorders such as psoriasis, rheumatoid arthritis, and diabetes. Using in silico tools, we predicted the sequence as well as the structural homology among these proteins. Database mining was conducted to establish links between these proteins and autoimmune disorders. The antigenic, non-allergenic, and immunogenic sequence mimics were employed to design and validate an immune response via vaccine construct design. Mimic-based vaccine construct can prove effective for immunization against the S. pneumoniae infections. Immune response simulation and binding affinity was assessed through the docking of construct C8 to human leukocyte antigen (HLA) molecules and TLR4 receptor, with promising results. Additionally, these mimics were mapped as conserved regions on their respective proteins, suggesting their functional importance in S. pneumoniae pathogenesis. This study highlights the potential for S. pneumoniae to trigger autoimmunity via molecular mimicry and the possibility of vaccine design using these mimics for triggering defense response.
Collapse
Affiliation(s)
- Mutaib M Mashraqi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Science, Shaqra University, AlQuwayiyah 11961, Saudi Arabia
| | - Norah A Alturki
- Clinical Laboratory Science Department, College of Applied Medical Science, King Saud University, Riyadh 11433, Saudi Arabia
| | - Saleh Alshamrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Mousa M Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | | |
Collapse
|
49
|
Makkar R, Sehgal A, Singh S, Sharma N, Rawat R, Rashid S, Vargas-De-La-Cruz C, Yadav S, Bungau SG, Behl T. Current trends in epigenetic, cellular and molecular pathways in management of rheumatoid arthritis. Inflammopharmacology 2023:10.1007/s10787-023-01262-5. [PMID: 37335368 DOI: 10.1007/s10787-023-01262-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/01/2023] [Indexed: 06/21/2023]
Abstract
Rheumatoid arthritis is a systemic chronic polyarticular autoimmune disorder of joints and joint membrane mainly affecting feet and hands. The pathological manifestation of the disease includes infiltration of immune cells, hyperplasia of the lining of synovium, formation of pannus and bone and cartilage destruction. If left untreated, the appearance of small focal necrosis, adhesion of granulation, and formation of fibrous tissue on the surface of articular cartilage is noted. The disease primarily affects nearly 1% of the population globally, women being more affected than men with a ratio 2:1 and can initiate regardless of any age. The synovial fibroblast in rheumatoid arthritis individuals exhibits an aggressive phenotype which upregulates the manifestation of protooncogenes, adhesive compounds, inflammatory cytokines and matrix-deteriorating enzymes. Apart from the inflammatory effects of cytokines, chemokines are also noted to induce swelling and pain in arthritic individuals by residing in synovial membrane and forming pannus. The current treatment of rheumatoid arthritis includes treatment with non-steroidal anti-inflammatory drugs, disease-modifying antirheumatic drugs, treatment with biologics such as inhibitors of TNF-α, interleukins, platelet activating factor, etc. which provides significant relief from symptoms and aids in management of the disease. The current review highlights the pathogenesis involved in the onset of rheumatoid arthritis and also covers epigenetic, cellular and molecular parameters associated with it to aid better and advanced therapeutic approaches for management of the debilitating disease.
Collapse
Affiliation(s)
- Rashita Makkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- GHG Khalsa College of Pharmacy, Gurusar Sadhar, Ludhiana, Punjab, India
| | - Sukhbir Singh
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Neelam Sharma
- Department of Pharmaceutics, MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Ravi Rawat
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Uttarakhand, India
| | - Summya Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Celia Vargas-De-La-Cruz
- Department of Pharmacology, Bromatology and Toxicology, Faculty of Pharmacy and Biochemistry, Universidad Nacional Mayor de San Marcos, Lima, 150001, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima, 15001, Peru
| | - Shivam Yadav
- School of Pharmacy, Babu Banarasi Das University, Lucknow, Uttar Pradesh, 226028, India
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028, Oradea, Romania.
- Doctoral School of Biomedical Sciences, University of Oradea, 410087, Oradea, Romania.
| | - Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Uttarakhand, India.
| |
Collapse
|
50
|
Torres-Sanchez A, Rivera-Robles M, Castillo-Pichardo L, Martínez-Ferrer M, Dorta-Estremera SM, Dharmawardhane S. Rac and Cdc42 inhibitors reduce macrophage function in breast cancer preclinical models. Front Oncol 2023; 13:1152458. [PMID: 37397366 PMCID: PMC10313121 DOI: 10.3389/fonc.2023.1152458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Background Metastatic disease lacks effective treatments and remains the primary cause of mortality from epithelial cancers, especially breast cancer. The metastatic cascade involves cancer cell migration and invasion and modulation of the tumor microenvironment (TME). A viable anti-metastasis strategy is to simultaneously target the migration of cancer cells and the tumor-infiltrating immunosuppressive inflammatory cells such as activated macrophages, neutrophils, and myeloid-derived suppressor cells (MDSC). The Rho GTPases Rac and Cdc42 are ideal molecular targets that regulate both cancer cell and immune cell migration, as well as their crosstalk signaling at the TME. Therefore, we tested the hypothesis that Rac and Cdc42 inhibitors target immunosuppressive immune cells, in addition to cancer cells. Our published data demonstrate that the Vav/Rac inhibitor EHop-016 and the Rac/Cdc42 guanine nucleotide association inhibitor MBQ-167 reduce mammary tumor growth and prevent breast cancer metastasis from pre-clinical mouse models without toxic effects. Methods The potential of Rac/Cdc42 inhibitors EHop-016 and MBQ-167 to target macrophages was tested in human and mouse macrophage cell lines via activity assays, MTT assays, wound healing, ELISA assays, and phagocytosis assays. Immunofluorescence, immunohistochemistry, and flow cytometry were used to identify myeloid cell subsets from tumors and spleens of mice following EHop-016 or MBQ-167 treatment. Results EHop-016 and MBQ-167 inhibited Rac and Cdc42 activation, actin cytoskeletal extensions, migration, and phagocytosis without affecting macrophage cell viability. Rac/Cdc42 inhibitors also reduced tumor- infiltrating macrophages and neutrophils in tumors of mice treated with EHop-016, and macrophages and MDSCs from spleens and tumors of mice with breast cancer, including activated macrophages and monocytes, following MBQ-167 treatment. Mice with breast tumors treated with EHop-016 significantly decreased the proinflammatory cytokine Interleukin-6 (IL-6) from plasma and the TME. This was confirmed from splenocytes treated with lipopolysaccharide (LPS) where EHop-016 or MBQ-167 reduced IL-6 secretion in response to LPS. Conclusion Rac/Cdc42 inhibition induces an antitumor environment via inhibition of both metastatic cancer cells and immunosuppressive myeloid cells in the TME.
Collapse
Affiliation(s)
- Anamaris Torres-Sanchez
- Department of Biology, University of Puerto Rico, San Juan, Puerto Rico
- Department of Biochemistry, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Michael Rivera-Robles
- Department of Biochemistry, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | | | - Magaly Martínez-Ferrer
- Department of Pharmaceutical Sciences, School of Pharmacy, San Juan, Puerto Rico
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
| | - Stephanie M. Dorta-Estremera
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
- Department of Microbiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Suranganie Dharmawardhane
- Department of Biochemistry, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
- Division of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
| |
Collapse
|