1
|
Bedathuru D, Rengaswamy M, Channavazzala M, Ray T, Packrisamy P, Kumar R. Multiscale, mechanistic model of Rheumatoid Arthritis to enable decision making in late stage drug development. NPJ Syst Biol Appl 2024; 10:126. [PMID: 39496637 PMCID: PMC11535547 DOI: 10.1038/s41540-024-00454-1] [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: 10/03/2023] [Accepted: 10/13/2024] [Indexed: 11/06/2024] Open
Abstract
Rheumatoid Arthritis (RA) is a chronic autoimmune inflammatory disease that affects about 0.1% to 2% of the population worldwide. Despite the development of several novel therapies, there is only limited benefit for many patients. Thus, there is room for new approaches to improve response to therapy, including designing better trials e.g., by identifying subpopulations that can benefit from specific classes of therapy and enabling reverse translation by analyzing completed clinical trials. We have developed an open-source, mechanistic multi-scale model of RA, which captures the interactions of key immune cells and mediators in an inflamed joint. The model consists of a treatment-naive Virtual Population (Vpop) that responds appropriately (i.e. as reported in clinical trials) to standard-of-care treatment options-Methotrexate (MTX) and Adalimumab (ADA, anti-TNF-α) and an MTX inadequate responder sub-population that responds appropriately to Tocilizumab (TCZ, anti-IL-6R) therapy. The clinical read-outs of interest are the American College of Rheumatology score (ACR score) and Disease Activity Score (DAS28-CRP), which is modeled to be dependent on the physiological variables in the model. Further, we have validated the Vpop by predicting the therapy response of TCZ on ADA Non-responders. This paper aims to share our approach, equations, and code to enable community evaluation and greater adoption of mechanistic models in drug development for autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | - Tamara Ray
- Vantage Research Inc, Lewes, Lewes, DE, USA
| | | | | |
Collapse
|
2
|
Al-Nasser MM, Al-Saeedi MJ, Alhowaiti SA, Shinwari Z, Alhamlan FS, Alothaid H, Alkahtani S, Al-Qahtani AA. Combination of Methotrexate and Resveratrol Reduces Pro-Inflammatory Chemokines in Human THP-1 Cells. J Inflamm Res 2024; 17:8085-8098. [PMID: 39507267 PMCID: PMC11539838 DOI: 10.2147/jir.s482503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Methotrexate (MTX) is a widely used anti-metabolite drug in cancer therapy, but its efficacy is often hindered by reactive oxygen species (ROS)-induced cellular toxicity. Resveratrol, a natural polyphenol, possesses antioxidant and anticancer properties. Therefore, this in vitro study aimed to investigate the synergistic anti-proliferative and anti-inflammatory effects of MTX and resveratrol in human THP-1 cells. Methods THP-1 cells were differentiated into macrophage-like cells using phorbol 12-myristate 13-acetate. In vitro experiments assessed the impact of various concentrations of MTX and resveratrol on cell viability and proliferation using the MTT assay. Concentration-effect curves were generated to elucidate their relationship. Gene expression was analyzed by RT-qPCR, while chemokine expression was measured via ELISA. Phagocytic and migratory activities were also evaluated. Results Differentiated THP-1 cells were treated with MTX and resveratrol and stimulated with dimethyl sulfoxide (DMSO) and lipopolysaccharide (LPS) as inflammatory stimuli. The combination of MTX and resveratrol exhibited an anti-proliferative effect in THP-1 cells (p = 0.03). The concentration-effect curve revealed IC50 values of 49.15 µg at 24 hours (R = 0.8236) and 2.029e-005 µg at 48 hours (R = 0.97) for MTX, and 20.17 µg at 48 hours (R = 1.000) and 55.38 µg at 96 hours (R = 0.9666) for resveratrol. Co-treatment with MTX and resveratrol significantly reduced mRNA and chemokine expression of CCL2, CCL3, CCL4, CCL5, and CXCL10 (p < 0.05). Moreover, decreased phagocytic and migratory activities were confirmed by phagocytosis and migration assays (p < 0.05). Conclusion The combination of MTX and resveratrol effectively attenuated pro-inflammatory activity in THP-1 cells, as evidenced by the downregulation of mRNA and chemokine expression. These findings suggest that the synergistic effects of MTX and resveratrol hold promise for enhancing cancer therapeutics.
Collapse
Affiliation(s)
- Moonerah M Al-Nasser
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mashael J Al-Saeedi
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Saltana A Alhowaiti
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Zakia Shinwari
- Therapeutics & Biomarker Discovery for Clinical Applications, Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Fatimah S Alhamlan
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A Al-Qahtani
- Department of Infection and Immunity, Research Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Yang M, Liu X, Jiang M, Hu J, Xiao Z. TAX1BP1/A20 inhibited TLR2-NF-κB activation to induce tolerant expression of IL-6 in endothelial cells. Int Immunopharmacol 2024; 139:112789. [PMID: 39079200 DOI: 10.1016/j.intimp.2024.112789] [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: 03/21/2024] [Revised: 06/06/2024] [Accepted: 07/23/2024] [Indexed: 09/25/2024]
Abstract
The inflammatory cascadedriven by interleukin-6 (IL-6) plays a crucial role in the initiation and progression of chronic inflammatory conditions such as atherosclerosis. Research has demonstrated that prolonged exposure to inflammatory stimuli leads to the development of "immune tolerance" in specialized immune cells such as monocytes and macrophages, serving as a mechanism to prevent tissue damage and curb the inflammatory cascade. However, our recent investigation revealed that immune tolerance did not effectively regulate the production of IL-6 in human umbilical vein endothelial cells (HUVECs) when stimulated by a Toll-like receptor 2 (TLR2) ligand Pam3CSK4, which is a potent activator of the pro-inflammatory transcription factor NF-κB. Furthermore, the negative regulator of NF-κB signaling, A20, was ineffective in suppressing TLR2-induced IL-6 synthesis in this context. Notably, all A20 auxiliary molecules, with the exception of TAX1BP1, were found to be significantly expressed in HUVECs. DNA methylation in TAX1BP1 was confirmed in GEO database. According to the information provided, it is hypothesized that altered DNA methylation in HUVECs could potentially lead to decreased expression of TAX1BP1, thereby impeding A20's capacity to modulate continuous activation of the TLR2-NF-κB pathway. This may consequently lead to unregulated production of IL-6, evading immune tolerance mechanisms. Subsequent investigations suggested that demethylating TAX1BP1 could enhance its expression, potentially reducing the endogenous IL-6 levels induced by repeated TLR2 stimulation and restoring A20's inhibitory role in NF-κB signaling. Additionally, over-expression of TAX1BP1 coulddecrease the production of atherosclerosis-associated cytokines like IL-6, MCP-1, ICAM-1, and VCAM-1, while increasing NO release following repeated Pam3cks4 stimulation, along with enhanced co-localization of TAX1BP1 and A20. These findings indicate that inducing immune tolerance in endothelial cells may effectively suppress endogenous IL-6 production and halt the IL-6-mediated inflammatory cascade, with TAX1BP1/A20 identified as crucial components in this process.These insights provide novel perspectives and potential targets for therapeutic strategies in inflammatoryimmunological disorders involving the overproduction of IL-6.
Collapse
Affiliation(s)
- Mei Yang
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Zhilin Xiao
- Department of Geriatric Cardiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
4
|
Ding Y, Cao Q, Yang W, Xu J, Xiao P. Macrophage: Hidden Criminal in Therapy Resistance. J Innate Immun 2024; 16:188-202. [PMID: 38442696 PMCID: PMC10990480 DOI: 10.1159/000538212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/29/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Although substantial efforts have been made by researchers to develop drugs, a disappointing reality is that the emergence of drug resistance is an unavoidable reality for the majority of patients. In recent years, emerging evidence suggests a connection between drug resistance and immune dysregulation. SUMMARY As a ubiquitously distributed, versatile innate immune cell, macrophages play essential roles in maintaining tissue homeostasis in a steady state. Nevertheless, it is becoming aware that macrophages undermine the action of therapeutic drugs across various disease types. Reprogramming macrophage function has been proven to be effective in restoring patient responsiveness to treatment. Herein, we comprehensively reviewed how macrophages respond to drugs and the mechanisms by which they contribute to treatment unresponsiveness in cancer, inflammatory diseases, and metabolic diseases. In addition, future prospects in macrophage-based combination therapy were discussed. KEY MESSAGES Targeting macrophages is a promising strategy for overcoming drug resistance in immune disorders.
Collapse
Affiliation(s)
- Yimin Ding
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjuan Yang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Junjie Xu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
5
|
Tang J, Yu Z, Xia J, Jiang R, Chen S, Ye D, Sheng H, Lin J. METTL14-Mediated m6A Modification of TNFAIP3 Involved in Inflammation in Patients With Active Rheumatoid Arthritis. Arthritis Rheumatol 2023; 75:2116-2129. [PMID: 37327357 DOI: 10.1002/art.42629] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 05/03/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVE The aim of the study was to investigate the role of N6 -methyladenosine (m6A) modification in the progression of rheumatoid arthritis (RA). METHODS Peripheral blood mononuclear cells (PBMCs) from patients with RA and healthy controls were collected. The expression of m6A modification-related proteins and m6A levels were detected using polymerase chain reaction (PCR), western blot, and m6A enzyme-linked immunosorbent assay (ELISA). The roles of methyltransferase-like 14 (METTL14) in the regulation of inflammation in RA was explored using methylated RNA immunoprecipitation (MeRIP) sequencing and RNA immunoprecipitation assays. Collagen antibody-induced arthritis (CAIA) mice were used as an in vivo model to study the role of METTL14 in the inflammation progression of RA. RESULTS We found that m6A writer METTL14 and m6A levels were decreased in PBMCs of patients with active RA and correlated negatively with the disease activity score using 28 joint counts (DAS28). Knockdown of METTL14 downregulated m6A and promoted the secretion of inflammatory cytokines interleukin 6 (IL-6) and IL-17 in PBMCs of patients with RA. Consistently, METTL14 knockdown promoted joint inflammation accompanied by upregulation of IL-6 and IL-17 in CAIA mice. MeRIP sequencing and functional studies confirmed that tumor necrosis factor α induced protein 3 (TNFAIP3), a key suppressor of the nuclear factor-κB inflammatory pathway, was involved in m6A-regulated PBMCs. Mechanistic investigations revealed that m6A affected TNFAIP3 expression by regulation of messenger RNA stability and translocation in TNFAIP3 protein coding sequence. CONCLUSIONS Our study highlights the critical roles of m6A on regulation of inflammation in RA progression. Treatment strategies targeting m6A modification may represent a new option for management of RA.
Collapse
Affiliation(s)
- Jifeng Tang
- Department of Laboratory Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China and Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ziqing Yu
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China and Department of Pathology, Fujian Cancer Hospital, Fuzhou, China
| | - Jinfang Xia
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Renquan Jiang
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shuhui Chen
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Detai Ye
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Huiming Sheng
- Department of Laboratory Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinpiao Lin
- Department of Laboratory Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China and Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
6
|
Jeljeli MM, Adamopoulos IE. Innate immune memory in inflammatory arthritis. Nat Rev Rheumatol 2023; 19:627-639. [PMID: 37674048 PMCID: PMC10721491 DOI: 10.1038/s41584-023-01009-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 09/08/2023]
Abstract
The concept of immunological memory was demonstrated in antiquity when protection against re-exposure to pathogens was observed during the plague of Athens. Immunological memory has been linked with the adaptive features of T and B cells; however, in the past decade, evidence has demonstrated that innate immune cells can exhibit memory, a phenomenon called 'innate immune memory' or 'trained immunity'. Innate immune memory is currently being defined and is transforming our understanding of chronic inflammation and autoimmunity. In this Review, we provide an up-to-date overview of the memory-like features of innate immune cells in inflammatory arthritis and the crosstalk between chronic inflammatory milieu and cell reprogramming. Aberrant pro-inflammatory signalling, including cytokines, regulates the metabolic and epigenetic reprogramming of haematopoietic progenitors, leading to exacerbated inflammatory responses and osteoclast differentiation, in turn leading to bone destruction. Moreover, imprinted memory on mature cells including terminally differentiated osteoclasts alters responsiveness to therapies and modifies disease outcomes, commonly manifested by persistent inflammatory flares and relapse following medication withdrawal.
Collapse
Affiliation(s)
- Maxime M Jeljeli
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Iannis E Adamopoulos
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Wroński J, Ciechomska M, Kuca-Warnawin E. Impact of methotrexate treatment on vaccines immunogenicity in adult rheumatological patients - Lessons learned from the COVID-19 pandemic. Biomed Pharmacother 2023; 165:115254. [PMID: 37542854 DOI: 10.1016/j.biopha.2023.115254] [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: 06/16/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/07/2023] Open
Abstract
Despite the development of new biological and synthetic targeted therapies, methotrexate remains one of the most commonly used immunomodulatory drugs in rheumatology. However, its effect on the immunogenicity of vaccines has been studied only to a limited extent until recently, resulting in the lack of clear guidelines on the use of methotrexate during vaccination. Significant progress was made during the COVID-19 pandemic due to the dynamic development of research on vaccines, including patients with autoimmune inflammatory rheumatic diseases. In the following literature review, we present a summary of what we know so far on the impact of methotrexate on post-vaccination response in adult rheumatology patients, taking into account the lessons learned from the COVID-19 pandemic. Studies on the effect of methotrexate on the immunogenicity of influenza, pneumococcal, herpes zoster, tetanus/diphtheria/pertussis, hepatitis A, yellow fever, and COVID-19 vaccines are described in detail, including the effect of methotrexate on the humoral and cellular response of individual vaccines. The available evidence for recommendations for withholding methotrexate in the post-vaccination period is presented. Lastly, an overview of potential immunological mechanisms through which MTX modulates the immunogenicity of vaccinations is also provided.
Collapse
Affiliation(s)
- Jakub Wroński
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland.
| | - Marzena Ciechomska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland
| | - Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland
| |
Collapse
|
8
|
Lang MB, Leung KY, Greene ND, Malone KM, Saginc G, Randi AM, Kiprianos A, Maughan RT, Pericleous C, Mason JC. The actions of methotrexate on endothelial cells are dependent on the shear stress-induced regulation of one carbon metabolism. Front Immunol 2023; 14:1209490. [PMID: 37457690 PMCID: PMC10349526 DOI: 10.3389/fimmu.2023.1209490] [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: 04/20/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Objectives The disease-modifying anti-rheumatic drug methotrexate (MTX) is recognized to reduce cardiovascular risk in patients with systemic inflammatory diseases. However, the molecular basis for these cardioprotective effects remains incompletely understood. This study evaluated the actions of low-dose MTX on the vascular endothelium. Methods Human endothelial cells (EC) were studied under in vitro conditions relevant to inflammatory arthritis. These included culture in a pro-inflammatory microenvironment and exposure to fluid shear stress (FSS) using a parallel plate model. Respectively treated cells were analyzed by RNA sequencing and quantitative real-time PCR for gene expression, by immunoblotting for protein expression, by phosphokinase activity arrays, by flow cytometry for cell cycle analyses and by mass spectrometry to assess folate metabolite levels. Results In static conditions, MTX was efficiently taken up by EC and caused cell cycle arrest concurrent with modulation of cell signaling pathways. These responses were reversed by folinic acid (FA), suggesting that OCM is a predominant target of MTX. Under FSS, MTX did not affect cell proliferation or pro-inflammatory gene expression. Exposure to FSS downregulated endothelial one carbon metabolism (OCM) as evidenced by decreased expression of key OCM genes and metabolites. Conclusion We found that FSS significantly downregulated OCM and thereby rendered EC less susceptible to the effects of MTX treatment. The impact of shear stress on OCM suggested that MTX does not directly modulate endothelial function. The cardioprotective actions of MTX likely reflect direct actions on inflammatory cells and indirect benefit on the vascular endothelium.
Collapse
Affiliation(s)
- Marie B. Lang
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kit-Yi Leung
- Developmental Biology & Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Nicholas D.E. Greene
- Developmental Biology & Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Kerri M. Malone
- European Bioinformatics Institute, Cambridge, United Kingdom
| | - Gaye Saginc
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Anna M. Randi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Allan Kiprianos
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Robert T. Maughan
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Charis Pericleous
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Justin C. Mason
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
9
|
Jia Y, Yin C, Ke W, Liu J, Guo B, Wang X, Zhao P, Hu S, Zhang C, Li X, Liu R, Zheng X, Wang Y, Wang G, Pan H, Hu W, Song Z. Alpha-ketoglutarate alleviates cadmium-induced inflammation by inhibiting the HIF1A-TNFAIP3 pathway in hepatocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 878:163069. [PMID: 36996991 DOI: 10.1016/j.scitotenv.2023.163069] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 05/13/2023]
Abstract
The threat to public health posed by rapidly increasing levels of cadmium (Cd) in the environment is receiving worldwide attention. Although, Cd is known to be absorbed into the body and causes non-negligible damage to the liver, the detailed mechanisms underlying its hepatoxicity are incompletely understood. In the present study, investigated the effect of TNFAIP3 and α-ketoglutarate (AKG) on Cd-induced liver inflammation and hepatocyte death. Male C57BL/6 mice were exposed to cadmium chloride (1.0 mg/kg) while being fed a diet with 2 % AKG for two weeks. We found that Cd induced hepatocyte injury and inflammatory infiltration. In addition, TNFAIP3 expression was inhibited in the liver tissues and cells of CdCl2-treated mice. Mouse hepatocyte-specific TNFAIP3 overexpression by tail vein injection of an adeno-associated virus (AAV) vector effectively alleviated Cd-induced hepatic necrosis and inflammation, which was mediated by the NF-κB signaling pathway. Notably, this inhibitory effect of TNFAIP3 on Cd-induced liver injury was dependent on AKG. Exogenous addition of AKG prevented Cd exposure-induced increases in serum ALT, AST and LDH levels, production of pro-inflammatory cytokines, activation of the NF-κB signaling pathway, and even significantly reduced Cd-induced oxidative stress and hepatocyte death. Mechanistically, AKG exerted its anti-inflammatory effect by promoting the hydroxylation and degradation of HIF1A to reduce its Cd-induced overexpression in vivo and in vitro, avoiding the inhibition of the TNFAIP3 promoter by HIF1A. Moreover, the protective effect of AKG was significantly weaker in Cd-treated primary hepatocytes transfected with HIF1A pcDNA. Overall, our results reveal a novel mechanism of Cd-induced hepatotoxicity.
Collapse
Affiliation(s)
- Yinzhao Jia
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Wenbo Ke
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jing Liu
- Key Laboratory of Coal Science and Technology of Ministry of Education, College of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan 030000, China
| | - Bing Guo
- Insitute for Genome Sciences, University of Maryland School of Medical, Baltimore, MD 21201, United States
| | - Xiaofei Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Peng Zhao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shaobo Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xuan Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ran Liu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xichuan Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yaofeng Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Gengqiao Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Wenjun Hu
- School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| |
Collapse
|
10
|
Muller IB, Lin M, de Jonge R, Will N, López-Navarro B, van der Laken C, Struys EA, Oudejans CBM, Assaraf YG, Cloos J, Puig-Kröger A, Jansen G. Methotrexate Provokes Disparate Folate Metabolism Gene Expression and Alternative Splicing in Ex Vivo Monocytes and GM-CSF- and M-CSF-Polarized Macrophages. Int J Mol Sci 2023; 24:9641. [PMID: 37298590 PMCID: PMC10253671 DOI: 10.3390/ijms24119641] [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: 04/14/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Macrophages constitute important immune cell targets of the antifolate methotrexate (MTX) in autoimmune diseases, including rheumatoid arthritis. Regulation of folate/MTX metabolism remains poorly understood upon pro-inflammatory (M1-type/GM-CSF-polarized) and anti-inflammatory (M2-type/M-CSF-polarized) macrophages. MTX activity strictly relies on the folylpolyglutamate synthetase (FPGS) dependent intracellular conversion and hence retention to MTX-polyglutamate (MTX-PG) forms. Here, we determined FPGS pre-mRNA splicing, FPGS enzyme activity and MTX-polyglutamylation in human monocyte-derived M1- and M2-macrophages exposed to 50 nmol/L MTX ex vivo. Moreover, RNA-sequencing analysis was used to investigate global splicing profiles and differential gene expression in monocytic and MTX-exposed macrophages. Monocytes displayed six-eight-fold higher ratios of alternatively-spliced/wild type FPGS transcripts than M1- and M2-macrophages. These ratios were inversely associated with a six-ten-fold increase in FPGS activity in M1- and M2-macrophages versus monocytes. Total MTX-PG accumulation was four-fold higher in M1- versus M2-macrophages. Differential splicing after MTX-exposure was particularly apparent in M2-macrophages for histone methylation/modification genes. MTX predominantly induced differential gene expression in M1-macrophages, involving folate metabolic pathway genes, signaling pathways, chemokines/cytokines and energy metabolism. Collectively, macrophage polarization-related differences in folate/MTX metabolism and downstream pathways at the level of pre-mRNA splicing and gene expression may account for variable accumulation of MTX-PGs, hence possibly impacting MTX treatment efficacy.
Collapse
Affiliation(s)
- Ittai B. Muller
- Department of Laboratory Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (I.B.M.); (M.L.); (R.d.J.); (E.A.S.); (C.B.M.O.)
| | - Marry Lin
- Department of Laboratory Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (I.B.M.); (M.L.); (R.d.J.); (E.A.S.); (C.B.M.O.)
| | - Robert de Jonge
- Department of Laboratory Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (I.B.M.); (M.L.); (R.d.J.); (E.A.S.); (C.B.M.O.)
| | - Nico Will
- Facility for Environment and Natural Science, Brandenburg Technical University Cottbus-Senftenberg, 01968 Senftenberg, Germany;
| | - Baltasar López-Navarro
- Laboratorio de Inmuno-Metabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital Gregorio Marañón, 28007 Madrid, Spain; (B.L.-N.); (A.P.-K.)
| | - Conny van der Laken
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center–location VUmc, 1081 HV Amsterdam, The Netherlands;
| | - Eduard A. Struys
- Department of Laboratory Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (I.B.M.); (M.L.); (R.d.J.); (E.A.S.); (C.B.M.O.)
| | - Cees B. M. Oudejans
- Department of Laboratory Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands; (I.B.M.); (M.L.); (R.d.J.); (E.A.S.); (C.B.M.O.)
| | - Yehuda G. Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel;
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam University Medical Center–location VUmc, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Amaya Puig-Kröger
- Laboratorio de Inmuno-Metabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital Gregorio Marañón, 28007 Madrid, Spain; (B.L.-N.); (A.P.-K.)
| | - Gerrit Jansen
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center–location VUmc, 1081 HV Amsterdam, The Netherlands;
| |
Collapse
|
11
|
Zhang Q, Lin X, Wang W, Zhang X, Lü M, Shao Z, Shi D, Zhang R, Shi H, Zhang Y, Pan J, Song G, Cheng K, Ge L, Wang L, Han J. Evaluation of 18F-FAPI-04 Imaging in Assessing the Therapeutic Response of Rheumatoid Arthritis. Mol Imaging Biol 2023:10.1007/s11307-023-01817-6. [PMID: 37020126 DOI: 10.1007/s11307-023-01817-6] [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: 11/15/2022] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 04/07/2023]
Abstract
PURPOSE Fibroblast activating protein (FAP) is highly expressed in the synovial tissues of rheumatoid arthritis (RA) patients. The aim of this study was to determine the feasibility of PET imaging with an Al[18F] F-NOTA-labeled FAP inhibitor 04(18F-FAPI-04) for the evaluation of arthritic progression and therapeutic response in experimental arthritis. METHODS Fibroblast-like synoviocytes (FLSs) were obtained from patients with RA or osteoarthritis (OA), and the relationship between 18F-FAPI-04 uptake and the inflammatory activity of RA FLSs was investigated. Collagen-induce arthritis (CIA) mice models were established and treated with methotrexate (MTX) or etanercept (ETC). Then, PET imaging was performed 24 h following 18F-FAPI-04 injection. The imaging results were compared by assessing macroscopic arthritis scores and histological staining. RESULTS 18F-FAPI-04 uptake was obvious in RA FLSs that characterizing FAP activation. The higher the uptake of 18F-FAPI-04, the more severity of the inflammatory phenotype in RA FLS. Furthermore, the uptake of 18F-FAPI-04 in inflamed joints could be found even before the deformity of the parental joints could be observed by histological examination. Both MTX and ETC were effective in inhibiting the progression of arthritis in CIA mice was confirmed by macroscopic, histological, and radiographic pathology scores. Importantly, 18F-FAPI-04 uptake declined accordingly in CIA models following MTX and ETC treatment. CONCLUSIONS These findings suggest that PET imaging of 18F-FAPI-04 can be used to monitor treatment response in RA, and is more sensitive in disease speculation than macroscopic arthritis scoring.
Collapse
Affiliation(s)
- Qingyun Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China
| | - Xuehong Lin
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China
| | - Weiqi Wang
- College of Preventive Medical Sciences (Institute of Radiation Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, China
| | - Xiaofan Zhang
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Mengxue Lü
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China
| | - Zhurui Shao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China
| | - Dandan Shi
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China
| | - Ruojia Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China
| | - Haojun Shi
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yuang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China
| | - Jihong Pan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China
| | - Guanhua Song
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Kai Cheng
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan 250117, Shandong, China
| | - Luna Ge
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China.
| | - Lin Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China.
| | - Jinxiang Han
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, #6699, Qingdao Road, Jinan, 250017, China.
| |
Collapse
|
12
|
Gu Y, Hsu ACY, Zuo X, Guo X, Zhou Z, Jiang S, Ouyang Z, Wang F. Chronic exposure to low-level lipopolysaccharide dampens influenza-mediated inflammatory response via A20 and PPAR network. Front Immunol 2023; 14:1119473. [PMID: 36726689 PMCID: PMC9886269 DOI: 10.3389/fimmu.2023.1119473] [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: 12/08/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
Influenza A virus (IAV) infection leads to severe inflammation, and while epithelial-driven inflammatory responses occur via activation of NF-κB, the factors that modulate inflammation, particularly the negative regulators are less well-defined. In this study we show that A20 is a crucial molecular switch that dampens IAV-induced inflammatory responses. Chronic exposure to low-dose LPS environment can restrict this excessive inflammation. The mechanisms that this environment provides to suppress inflammation remain elusive. Here, our evidences show that chronic exposure to low-dose LPS suppressed IAV infection or LPS stimulation-induced inflammation in vitro and in vivo. Chronic low-dose LPS environment increases A20 expression, which in turn positively regulates PPAR-α and -γ, thus dampens the NF-κB signaling pathway and NLRP3 inflammasome activation. Knockout of A20 abolished the inhibitory effect on inflammation. Thus, A20 and its induced PPAR-α and -γ play a key role in suppressing excessive inflammatory responses in the chronic low-dose LPS environment.
Collapse
Affiliation(s)
- Yinuo Gu
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Alan Chen-Yu Hsu
- Signature Research Program in Emerging Infectious Diseases, Duke - National University of Singapore (NUS) Graduate Medical School, Singapore, Singapore,School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia,Viruses, Infections/Immunity, Vaccines and Asthma, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Xu Zuo
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaoping Guo
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zhengjie Zhou
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Shengyu Jiang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zhuoer Ouyang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Fang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China,*Correspondence: Fang Wang,
| |
Collapse
|
13
|
Vitale A, Alivernini S, Caporali R, Cassone G, Bruno D, Cantarini L, Lopalco G, Rossini M, Atzeni F, Favalli EG, Conti F, Gremese E, Iannone F, Ferraccioli GF, Lapadula G, Sebastiani M. From Bench to Bedside in Rheumatoid Arthritis from the "2022 GISEA International Symposium". J Clin Med 2023; 12:jcm12020527. [PMID: 36675455 PMCID: PMC9863451 DOI: 10.3390/jcm12020527] [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: 11/14/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
While precision medicine is still a challenge in rheumatic disease, in recent years many advances have been made regarding pathogenesis, the treatment of inflammatory arthropathies, and their interaction. New insight into the role of inflammasome and synovial tissue macrophage subsets as predictors of drug response give hope for future tailored therapeutic strategies and a personalized medicine approach in inflammatory arthropathies. Here, we discuss the main pathogenetic mechanisms and therapeutic approaches towards precision medicine in rheumatoid arthritis from the 2022 International GISEA/OEG Symposium.
Collapse
Affiliation(s)
- Antonio Vitale
- Department of Medical Sciences, Surgery and Neurosciences, Research Center of Systemic Autoinflammatory Diseases and Behçet’s Disease Clinic, University of Siena, 53100 Siena, SI, Italy
| | - Stefano Alivernini
- Immunology Research Core Facility, Gemelli Science and Technology Park, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, RM, Italy
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, RM, Italy
| | - Roberto Caporali
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, 20122 Milano, MI, Italy
- Department of Clinical Sciences and Community Health, Research Center for Pediatric and Adult Rheumatic Diseases (RECAP.RD), University of Milan, 20122 Milano, MI, Italy
| | - Giulia Cassone
- Rheumatology Unit, Azienda Ospedaliera Policlinico di Modena, University of Modena and Reggio Emilia, 41121 Modena, MO, Italy
| | - Dario Bruno
- Immunology Research Core Facility, Gemelli Science and Technology Park, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, RM, Italy
| | - Luca Cantarini
- Department of Medical Sciences, Surgery and Neurosciences, Research Center of Systemic Autoinflammatory Diseases and Behçet’s Disease Clinic, University of Siena, 53100 Siena, SI, Italy
| | - Giuseppe Lopalco
- Rheumatology Unit, Department of Emergency Surgery and Organ Transplantations, University of Bari, 70121 Bari, BA, Italy
| | - Maurizio Rossini
- Rheumatology Unit, University of Verona, Policlinico G.B. Rossi, Piazzale A. Scuro, 37134 Verona, VR, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, 98122 Messina, ME, Italy
| | - Ennio Giulio Favalli
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, 20122 Milano, MI, Italy
- Department of Clinical Sciences and Community Health, Research Center for Pediatric and Adult Rheumatic Diseases (RECAP.RD), University of Milan, 20122 Milano, MI, Italy
| | - Fabrizio Conti
- Lupus Clinic, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, 00185 Roma, RM, Italy
| | - Elisa Gremese
- Immunology Research Core Facility, Gemelli Science and Technology Park, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, RM, Italy
- Division of Clinical Immunology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 20123 Milano, MI, Italy
| | - Florenzo Iannone
- Rheumatology Unit, Department of Emergency Surgery and Organ Transplantations, University of Bari, 70121 Bari, BA, Italy
| | | | - Giovanni Lapadula
- Rheumatology Unit, Department of Emergency Surgery and Organ Transplantations, University of Bari, 70121 Bari, BA, Italy
| | - Marco Sebastiani
- Rheumatology Unit, Azienda Ospedaliera Policlinico di Modena, University of Modena and Reggio Emilia, 41121 Modena, MO, Italy
- Correspondence:
| |
Collapse
|
14
|
Mangoni AA, Sotgia S, Zinellu A, Carru C, Pintus G, Damiani G, Erre GL, Tommasi S. Methotrexate and cardiovascular prevention: an appraisal of the current evidence. Ther Adv Cardiovasc Dis 2023; 17:17539447231215213. [PMID: 38115784 PMCID: PMC10732001 DOI: 10.1177/17539447231215213] [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: 09/05/2023] [Accepted: 11/02/2023] [Indexed: 12/21/2023] Open
Abstract
New evidence continues to accumulate regarding a significant association between excessive inflammation and dysregulated immunity (local and systemic) and the risk of cardiovascular events in different patient cohorts. Whilst research has sought to identify novel atheroprotective therapies targeting inflammation and immunity, several marketed drugs for rheumatological conditions may serve a similar purpose. One such drug, methotrexate, has been used since 1948 for treating cancer and, more recently, for a wide range of dysimmune conditions. Over the last 30 years, epidemiological and experimental studies have shown that methotrexate is independently associated with a reduced risk of cardiovascular disease, particularly in rheumatological patients, and exerts several beneficial effects on vascular homeostasis and blood pressure control. This review article discusses the current challenges with managing cardiovascular risk and the new frontiers offered by drug discovery and drug repurposing targeting inflammation and immunity with a focus on methotrexate. Specifically, the article critically appraises the results of observational, cross-sectional and intervention studies investigating the effects of methotrexate on overall cardiovascular risk and individual risk factors. It also discusses the putative molecular mechanisms underpinning the atheroprotective effects of methotrexate and the practical advantages of using methotrexate in cardiovascular prevention, and highlights future research directions in this area.
Collapse
Affiliation(s)
- Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA 5042, Australia
| | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Giovanni Damiani
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Italian Centre of Precision Medicine and Chronic Inflammation, Milan, Italy
| | - Gian Luca Erre
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital (AOUSS) and University of Sassari, Sassari, Italy
| | - Sara Tommasi
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, SA, Australia
| |
Collapse
|
15
|
Chen YT, Chang YH, Pathak N, Tzou SC, Luo YC, Hsu YC, Li TN, Lee JY, Chen YC, Huang YW, Yang HJ, Hsu NY, Tsai HP, Chang TY, Hsu SC, Liu PC, Chin YF, Lin WC, Yang CM, Wu HL, Lee CY, Hsu HL, Liu YC, Chu JW, Wang LHC, Wang JY, Huang CH, Lin CH, Hsieh PS, Wu Lee YH, Hung YJ, Yang JM. Methotrexate inhibition of SARS-CoV-2 entry, infection and inflammation revealed by bioinformatics approach and a hamster model. Front Immunol 2022; 13:1080897. [PMID: 36618412 PMCID: PMC9811668 DOI: 10.3389/fimmu.2022.1080897] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Background Drug repurposing is a fast and effective way to develop drugs for an emerging disease such as COVID-19. The main challenges of effective drug repurposing are the discoveries of the right therapeutic targets and the right drugs for combating the disease. Methods Here, we present a systematic repurposing approach, combining Homopharma and hierarchal systems biology networks (HiSBiN), to predict 327 therapeutic targets and 21,233 drug-target interactions of 1,592 FDA drugs for COVID-19. Among these multi-target drugs, eight candidates (along with pimozide and valsartan) were tested and methotrexate was identified to affect 14 therapeutic targets suppressing SARS-CoV-2 entry, viral replication, and COVID-19 pathologies. Through the use of in vitro (EC50 = 0.4 μM) and in vivo models, we show that methotrexate is able to inhibit COVID-19 via multiple mechanisms. Results Our in vitro studies illustrate that methotrexate can suppress SARS-CoV-2 entry and replication by targeting furin and DHFR of the host, respectively. Additionally, methotrexate inhibits all four SARS-CoV-2 variants of concern. In a Syrian hamster model for COVID-19, methotrexate reduced virus replication, inflammation in the infected lungs. By analysis of transcriptomic analysis of collected samples from hamster lung, we uncovered that neutrophil infiltration and the pathways of innate immune response, adaptive immune response and thrombosis are modulated in the treated animals. Conclusions We demonstrate that this systematic repurposing approach is potentially useful to identify pharmaceutical targets, multi-target drugs and regulated pathways for a complex disease. Our findings indicate that methotrexate is established as a promising drug against SARS-CoV-2 variants and can be used to treat lung damage and inflammation in COVID-19, warranting future evaluation in clinical trials.
Collapse
Affiliation(s)
- Yun-Ti Chen
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yu-Hsiu Chang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan,Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Nikhil Pathak
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Shey-Cherng Tzou
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan,Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan,Center for Intelligent Drug Systems and Smart Bio-Devices, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yong-Chun Luo
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yen-Chao Hsu
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Tian-Neng Li
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Jung-Yu Lee
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yi-Cyun Chen
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yu-Wei Huang
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Hsin-Ju Yang
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Nung-Yu Hsu
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Hui-Ping Tsai
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Tein-Yao Chang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan,Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Chen Hsu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Ping-Cheng Liu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Fan Chin
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Chin Lin
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chuen-Mi Yang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Hsueh-Ling Wu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Ying Lee
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Hui-Ling Hsu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan,Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Chun Liu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Jhih-Wei Chu
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan,Center for Intelligent Drug Systems and Smart Bio-Devices, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Lily Hui-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Jann-Yuan Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Heng Huang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan,Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Hung Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan,Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan,Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Po-Shiuan Hsieh
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yan-Hwa Wu Lee
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan,Center for Intelligent Drug Systems and Smart Bio-Devices, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yi-Jen Hung
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan,Division of Endocrine and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan,*Correspondence: Yi-Jen Hung, ; Jinn-Moon Yang,
| | - Jinn-Moon Yang
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan,Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan,Center for Intelligent Drug Systems and Smart Bio-Devices, National Yang Ming Chiao Tung University, Hsinchu, Taiwan,*Correspondence: Yi-Jen Hung, ; Jinn-Moon Yang,
| |
Collapse
|
16
|
Ríos I, López-Navarro B, Torres-Torresano M, Soler Palacios B, Simón-Fuentes M, Domínguez-Soto Á, Muller IB, Jansen G, Corbí ÁL, Puig-Kröger A. GSK3β Inhibition Prevents Macrophage Reprogramming by High-Dose Methotrexate. J Innate Immun 2022; 15:283-296. [PMID: 36380627 PMCID: PMC10643894 DOI: 10.1159/000526622] [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: 05/16/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2023] Open
Abstract
Methotrexate (MTX) is an antifolate drug used as a chemotherapeutic agent for acute lymphoblastic leukemia, where MTX improves patients' prognosis. Macrophage reprogramming is being increasingly assessed as an antitumor therapeutic strategy. However, and although MTX limits the pathogenic action of macrophages in chronic inflammatory diseases, its effects on tumor-promoting macrophages have not been previously explored. We now report that MTX shapes the transcriptional and functional profile of M-CSF-dependent human macrophages, whose transcriptome is highly enriched in the gene signature that defines pathogenic tumor-associated macrophages ("large TAM"). Specifically, MTX prompted the acquisition of the gene signature of antitumoral "small TAM" and skewed macrophages toward an IL-6high IFNβ1high IL-10low phenotype upon subsequent stimulation. Mechanistically, the MTX-induced macrophage reprogramming effect correlated with a reduction of the M-CSF receptor CSF1R expression and function, as well as a diminished expression of MAF and MAFB transcription factors, primary determinants of pro-tumoral macrophages whose transcriptional activity is dependent on GSK3β. Indeed, the ability of MTX to transcriptionally reprogram macrophages toward an antitumoral phenotype was abrogated by inhibition of GSK3β. Globally, our results establish MTX as a macrophage reprogramming drug and indicate that its ability to modulate macrophage polarization may also underlie its therapeutic benefits. Since GSK3β inhibition abrogates the reprogramming action of MTX, our results suggest that the GSK3β-MAFB/MAF axis constitutes a target for the macrophage-centered antitumor strategies.
Collapse
Affiliation(s)
- Israel Ríos
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Baltasar López-Navarro
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Mónica Torres-Torresano
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Blanca Soler Palacios
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | | | | - Ittai B. Muller
- Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Gerrit Jansen
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Ángel L. Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, Madrid, Spain
| | - Amaya Puig-Kröger
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| |
Collapse
|
17
|
Wang W, Bale S, Wei J, Yalavarthi B, Bhattacharyya D, Yan JJ, Abdala-Valencia H, Xu D, Sun H, Marangoni RG, Herzog E, Berdnikovs S, Miller SD, Sawalha AH, Tsou PS, Awaji K, Yamashita T, Sato S, Asano Y, Tiruppathi C, Yeldandi A, Schock BC, Bhattacharyya S, Varga J. Fibroblast A20 governs fibrosis susceptibility and its repression by DREAM promotes fibrosis in multiple organs. Nat Commun 2022; 13:6358. [PMID: 36289219 PMCID: PMC9606375 DOI: 10.1038/s41467-022-33767-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 09/29/2022] [Indexed: 02/04/2023] Open
Abstract
In addition to autoimmune and inflammatory diseases, variants of the TNFAIP3 gene encoding the ubiquitin-editing enzyme A20 are also associated with fibrosis in systemic sclerosis (SSc). However, it remains unclear how genetic factors contribute to SSc pathogenesis, and which cell types drive the disease due to SSc-specific genetic alterations. We therefore characterize the expression, function, and role of A20, and its negative transcriptional regulator DREAM, in patients with SSc and disease models. Levels of A20 are significantly reduced in SSc skin and lungs, while DREAM is elevated. In isolated fibroblasts, A20 mitigates ex vivo profibrotic responses. Mice haploinsufficient for A20, or harboring fibroblasts-specific A20 deletion, recapitulate major pathological features of SSc, whereas DREAM-null mice with elevated A20 expression are protected. In DREAM-null fibroblasts, TGF-β induces the expression of A20, compared to wild-type fibroblasts. An anti-fibrotic small molecule targeting cellular adiponectin receptors stimulates A20 expression in vitro in wild-type but not A20-deficient fibroblasts and in bleomycin-treated mice. Thus, A20 has a novel cell-intrinsic function in restraining fibroblast activation, and together with DREAM, constitutes a critical regulatory network governing the fibrotic process in SSc. A20 and DREAM represent novel druggable targets for fibrosis therapy.
Collapse
Affiliation(s)
- Wenxia Wang
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
| | - Swarna Bale
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jun Wei
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
| | - Bharath Yalavarthi
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dibyendu Bhattacharyya
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jing Jing Yan
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Dan Xu
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Hanshi Sun
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Roberta G Marangoni
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
| | - Erica Herzog
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Sergejs Berdnikovs
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Amr H Sawalha
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Pei-Suen Tsou
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kentaro Awaji
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takashi Yamashita
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Chinnaswamy Tiruppathi
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Anjana Yeldandi
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Bettina C Schock
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, UK
| | - Swati Bhattacharyya
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA.
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - John Varga
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA.
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
18
|
Triguero-Martínez A, Roy-Vallejo E, Montes N, de la Fuente H, Ortiz AM, Castañeda S, González-Álvaro I, Lamana A. Genetic LGALS1 Variants Are Associated with Heterogeneity in Galectin-1 Serum Levels in Patients with Early Arthritis. Int J Mol Sci 2022; 23:7181. [PMID: 35806182 PMCID: PMC9266574 DOI: 10.3390/ijms23137181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 11/17/2022] Open
Abstract
Galectin 1 (Gal1) exerts immunomodulatory effects leading to therapeutic effects in autoimmune animal models. Patients with rheumatoid arthritis have been reported to show higher Gal1 serum levels than the healthy population. Our study aimed to find genetic variants on the Gal1 gene (LGALS1) modulating its expression and/or clinical features in patients with early arthritis (EA). LGALS1 was sequenced in 53 EA patients to characterize all genetic variants. Then, we genotyped rs9622682, rs929039, and rs4820293, which covered the main genetic variation in LGALS1, in 532 EA patients. Gal1 and IL-6 serum levels were measured by ELISA and Gal1 also by western blot (WB) in lymphocytes from patients with specific genotypes. Once disease activity improved with treatment, patients with at least one copy of the minor allele in rs9622682 and rs929039 or those with GG genotype in rs4820293 showed significantly higher Gal1 serum levels (p < 0.05). These genotypic combinations were also associated with higher Gal1 expression in lymphocytes by WB and lower IL-6 serum levels in EA patients. In summary, our study suggests that genetic variants studied in LGALS1 can explain heterogeneity in Gal1 serum levels showing that patients with higher Gal1 levels have lower serum IL-6 levels.
Collapse
Affiliation(s)
- Ana Triguero-Martínez
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain; (A.T.-M.); (N.M.); (A.M.O.); (S.C.)
| | - Emilia Roy-Vallejo
- Internal Medicine Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain;
| | - Nuria Montes
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain; (A.T.-M.); (N.M.); (A.M.O.); (S.C.)
| | - Hortensia de la Fuente
- Immunology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain;
| | - Ana María Ortiz
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain; (A.T.-M.); (N.M.); (A.M.O.); (S.C.)
| | - Santos Castañeda
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain; (A.T.-M.); (N.M.); (A.M.O.); (S.C.)
| | - Isidoro González-Álvaro
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain; (A.T.-M.); (N.M.); (A.M.O.); (S.C.)
| | - Amalia Lamana
- Cell Biology Department, Facultad de Biología, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
19
|
Yu N, Yang F, Zhao X, Guo Y, Xu Y, Pang G, Gong Y, Wang S, Liu Y, Fang Y, Yu K, Yao L, Wang H, Zhang K, Liu B, Wang Z, Guo Y, Xu Z. Manual acupuncture at ST36 attenuates rheumatoid arthritis by inhibiting M1 macrophage polarization and enhancing Treg cell populations in adjuvant-induced arthritic rats. Acupunct Med 2022; 41:96-109. [PMID: 35585798 DOI: 10.1177/09645284221085278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Acupuncture has been found to be effective at relieving many inflammatory pain conditions, including rheumatoid arthritis (RA). We aimed to assess the anti-inflammatory potential of manual acupuncture (MA) treatment of RA using adjuvant-induced arthritic (AIA) rats and to explore the underlying mechanisms. METHODS The anti-inflammatory and analgesic actions of MA at ST36 (Zusanli) in AIA rats were assessed using paw withdrawal latency and swelling, histological examination and cytokine detection by enzyme-linked immunoassay (ELISA). The cell-cell communication (CCC) network was analyzed with a multiplex immunoassay of 24 immune factors expressed in the inflamed joints, and the macrophage and Treg populations and associated cytokines regulated by MA were investigated using reverse-transcription quantitative polymerase chain reaction (RT-qPCR), ELISA and flow cytometry. RESULTS MA markedly decreased heat hyperalgesia and paw swelling in AIA rats. MA-treated rats also exhibited decreased levels of pro-inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-1β) coupled with increased anti-inflammatory cytokines (IL-10, transforming growth factor (TGF)-β1) in the ankle joints at protein and mRNA levels. CCC network analysis confirmed that macrophages are of critical importance and are potential therapeutic targets in RA. Repeated treatment with MA triggered a macrophage phenotypic switch in the paws, with fewer M1 macrophages. Prominent increases in the Treg cell population and TGF-β1 in the popliteal lymph nodes demonstrated the immunomodulatory effects of MA. Furthermore, a selective TGF-β1-receptor inhibitor, SB431542, attenuated the anti-inflammatory effects of MA and MA-induced suppression of the levels of M1-released cytokines. CONCLUSION These findings provide novel evidence that the anti-inflammatory and analgesic effects of MA on RA act through phenotypic modulation involving the inhibition of M1 macrophage polarization and an increase in the Treg cell population, highlighting the potential therapeutic advantages of acupuncture in controlling pain and ameliorating inflammatory conditions.
Collapse
Affiliation(s)
- Nannan Yu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,Department of Traditional Chinese Medicine, the First Affiliated Hospital, the Air Force Medical University, Xi'an, P.R. China
| | - Fuming Yang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Xue Zhao
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yongming Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yuan Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Guangchang Pang
- Tianjin Key Laboratory of Food Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, P.R. China
| | - Yinan Gong
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Shenjun Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yangyang Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Yuxin Fang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Kun Yu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Lin Yao
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Hui Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Kuo Zhang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Baohu Liu
- Department of Rehabilitation, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, P.R. China
| | - Zhenguo Wang
- Xi'an Encephalopathy Hospital of Traditional Chinese Medicine, Xi'an, P.R. China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, P.R. China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| |
Collapse
|
20
|
González de la Aleja A, Herrero C, Torres-Torresano M, de la Rosa JV, Alonso B, Capa-Sardón E, Muller IB, Jansen G, Puig-Kröger A, Vega MA, Castrillo A, Corbí ÁL. Activation of LXR Nuclear Receptors Impairs the Anti-Inflammatory Gene and Functional Profile of M-CSF-Dependent Human Monocyte-Derived Macrophages. Front Immunol 2022; 13:835478. [PMID: 35280993 PMCID: PMC8907538 DOI: 10.3389/fimmu.2022.835478] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/07/2022] [Indexed: 12/30/2022] Open
Abstract
Liver X Receptors (LXR) control cholesterol metabolism and exert anti-inflammatory actions but their contribution to human macrophage polarization remains unclear. The LXR pathway is enriched in pro-inflammatory macrophages from rheumatoid arthritis as well as in tumors-associated macrophages from human tumors. We now report that LXR activation inhibits the anti-inflammatory gene and functional profile of M-CSF-dependent human macrophages, and prompts the acquisition of a pro-inflammatory gene signature, with both effects being blocked by an LXR inverse agonist. Mechanistically, the LXR-stimulated macrophage polarization shift correlates with diminished expression of MAFB and MAF, which govern the macrophage anti-inflammatory profile, and with enhanced release of activin A. Indeed, LXR activation impaired macrophage polarization in response to tumor-derived ascitic fluids, as well as the expression of MAF- and MAFB-dependent genes. Our results demonstrate that LXR activation limits the anti-inflammatory human macrophage polarization and prompts the acquisition of an inflammatory transcriptional and functional profile.
Collapse
Affiliation(s)
- Arturo González de la Aleja
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Cristina Herrero
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mónica Torres-Torresano
- Unidad de Inmuno-Metabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Juan Vladimir de la Rosa
- Unidad de Biomedicina (Unidad Asociada al Consejo Superior de Investigaciones Científicas (CSIC)), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Grupo de Investigación Medio Ambiente y Salud, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Bárbara Alonso
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Enrique Capa-Sardón
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Ittai B. Muller
- Department of Clinical Chemistry, Amsterdam University Medical Center, Location VUmc, Amsterdam, Netherlands
| | - Gerrit Jansen
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Location VUmc, Amsterdam, Netherlands
| | - Amaya Puig-Kröger
- Unidad de Inmuno-Metabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Miguel A. Vega
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Antonio Castrillo
- Unidad de Biomedicina (Unidad Asociada al Consejo Superior de Investigaciones Científicas (CSIC)), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Grupo de Investigación Medio Ambiente y Salud, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Instituto Investigaciones Biomédicas “Alberto Sols” (IIBM), and Centro Mixto Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid (Consejo Superior de Investigaciones Científicas (ICSIC)-UAM), Madrid, Spain
| | - Ángel L. Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
21
|
Degboé Y, Poupot R, Poupot M. Repolarization of Unbalanced Macrophages: Unmet Medical Need in Chronic Inflammation and Cancer. Int J Mol Sci 2022; 23:1496. [PMID: 35163420 PMCID: PMC8835955 DOI: 10.3390/ijms23031496] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Monocytes and their tissue counterpart macrophages (MP) constitute the front line of the immune system. Indeed, they are able to rapidly and efficiently detect both external and internal danger signals, thereby activating the immune system to eradicate the disturbing biological, chemical, or physical agents. They are also in charge of the control of the immune response and account for the repair of the damaged tissues, eventually restoring tissue homeostasis. The balance between these dual activities must be thoroughly controlled in space and time. Any sustained unbalanced response of MP leads to pathological disorders, such as chronic inflammation, or favors cancer development and progression. In this review, we take advantage of our expertise in chronic inflammation, especially in rheumatoid arthritis, and in cancer, to highlight the pivotal role of MP in the physiopathology of these disorders and to emphasize the repolarization of unbalanced MP as a promising therapeutic strategy to control these diseases.
Collapse
Affiliation(s)
- Yannick Degboé
- Infinity, Université Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France;
- Département de Rhumatologie, CHU Toulouse, 31029 Toulouse, France
| | - Rémy Poupot
- Infinity, Université Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France;
| | - Mary Poupot
- Centre de Recherche en Cancérologie de Toulouse, Université Toulouse, INSERM, UPS, 31037 Toulouse, France;
| |
Collapse
|
22
|
Fuentelsaz-Romero S, Barrio-Alonso C, García Campos R, Torres Torresano M, Muller IB, Triguero-Martínez A, Nuño L, Villalba A, García-Vicuña R, Jansen G, Miranda-Carús ME, González-Álvaro I, Puig-Kröger A. The Macrophage Reprogramming Ability of Antifolates Reveals Soluble CD14 as a Potential Biomarker for Methotrexate Response in Rheumatoid Arthritis. Front Immunol 2021; 12:776879. [PMID: 34804067 PMCID: PMC8602851 DOI: 10.3389/fimmu.2021.776879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/14/2021] [Indexed: 12/24/2022] Open
Abstract
The identification of “trained immunity/tolerance” in myeloid cells has changed our perception of the performance of monocytes and macrophages during inflammatory and immune responses. Pemetrexed (PMX) and methotrexate (MTX) are blockers of the one-carbon metabolism (OCM) and commonly used therapeutic agents in cancer and rheumatoid arthritis (RA). We have previously showed that MTX promotes trained immunity in human macrophages. In the present manuscript, we have assessed the anti-inflammatory effects of PMX and MTX and found that OCM blockers alter the functional and gene expression profile of human macrophages and that OCM blockade reprograms macrophages towards a state of lipopolysaccharide (LPS) tolerance at the signaling and functional levels. Moreover, OCM blockade reduced macrophage LPS responsiveness by impairing the expression of membrane-bound and soluble CD14 (sCD14). The therapeutic relevance of these results was later confirmed in early RA patients, as MTX-responder RA patients exhibit lower sCD14 serum levels, with baseline sCD14 levels predicting MTX response. As a whole, our results demonstrate that OCM is a metabolic circuit that critically mediates the acquisition of innate immune tolerance and positions sCD14 as a valuable tool to predict MTX response in RA patients.
Collapse
Affiliation(s)
- Sara Fuentelsaz-Romero
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Celia Barrio-Alonso
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Raquel García Campos
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Mónica Torres Torresano
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Ittai B Muller
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, Netherlands
| | - Ana Triguero-Martínez
- Servicio de Reumatología, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Hospital Universitario La Princesa, Madrid, Spain
| | - Laura Nuño
- Department of Rheumatology, Hospital Universitario La Paz-IdiPaz, Madrid, Spain
| | - Alejandro Villalba
- Department of Rheumatology, Hospital Universitario La Paz-IdiPaz, Madrid, Spain
| | - Rosario García-Vicuña
- Servicio de Reumatología, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Hospital Universitario La Princesa, Madrid, Spain
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, Netherlands
| | | | - Isidoro González-Álvaro
- Servicio de Reumatología, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Hospital Universitario La Princesa, Madrid, Spain
| | - Amaya Puig-Kröger
- Unidad de Inmunometabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| |
Collapse
|
23
|
Ibrahim SSA, Kandil LS, Ragab GM, El-Sayyad SM. Micro RNAs 26b, 20a inversely correlate with GSK-3 β/NF-κB/NLRP-3 pathway to highlight the additive promising effects of atorvastatin and quercetin in experimental induced arthritis. Int Immunopharmacol 2021; 99:108042. [PMID: 34426107 DOI: 10.1016/j.intimp.2021.108042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 12/23/2022]
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease with challenging therapeutic potential due to the implication of cross-talking intracellular pathways in the pathogenesis of the disease. This study aimed to evaluate the effects of the combination therapy of atorvastatin and quercetin on glycogen synthase kinase-3 beta/ nuclear factor kappa-B/ nucleotide-binding oligomerization domain-like receptor family pyrin domain containing-3 or inflammasome (GSK-3β/NF-KB/NLRP-3) pathway as well as on microRNAs 26b and 20a (miR-26b, miR-20a) and to investigate the possible beneficial outcomes of the combination to offer a better treatment option than methotrexate (MTX) in adjuvant-induced arthritis (AIA). Assessment of arthritis progression, serum inflammatory, and oxidative parameters were done. The tibiotarsal tissue expression of the inflammatory parameters was evaluated. Western blot analysis was done to assess the expression level of the important members in the GSK-3β/NF-κB/NLRP-3 pathway. Furthermore, the expression level of both microRNAs and serum level of transaminases were determined. All treatments, especially the combination regimen, abated arthritis progression, the elevated serum level of inflammatory and oxidative stress parameters in arthritic rats. Moreover, They down-regulated the gene expression of the important members of the aforementioned signaling pathway, amended the tissue levels of inflammatory parameters and elevated the expression level of miR-26b and miR-20a. Finally, we concluded that the combination therapy modulated miR-26b and miR-20a as well as GSK-3β/NF-κB/NLRP-3 pathway, provided additive anti-inflammatory and anti-oxidant effects and offered an additional hepatoprotective effect as compared to untreated arthritic rats and MTX-treated groups, suggesting its promising role to be used as replacement therapy to MTX in RA.
Collapse
Affiliation(s)
| | - Lamia Said Kandil
- Department of Pharmacology & Therapeutics, Pharos University in Alexandria, Alexandria, Egypt; Department of Biochemistry, School of Biological Sciences, University of East Anglia, England
| | - Ghada M Ragab
- Department of Pharmacology and Toxicology, Misr University for Science and Technology, 6(th) of October City, Egypt
| | - Shorouk M El-Sayyad
- Department of Pharmacology & Toxicology, October 6 University, 12585, Giza, Egypt
| |
Collapse
|
24
|
Targeting Toll-like Receptor (TLR) Pathways in Inflammatory Arthritis: Two Better Than One? Biomolecules 2021; 11:biom11091291. [PMID: 34572504 PMCID: PMC8464963 DOI: 10.3390/biom11091291] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory arthritis is a cluster of diseases caused by unregulated activity of the immune system. The lost homeostasis is followed by the immune attack of one’s self, what damages healthy cells and tissues and leads to chronic inflammation of various tissues and organs (e.g., joints, lungs, heart, eyes). Different medications to control the excessive immune response are in use, however, drug resistances, flare-reactions and adverse effects to the current therapies are common in the affected patients. Thus, it is essential to broaden the spectrum of alternative treatments and to develop disease-modifying drugs. In the last 20 years, the involvement of the innate immune receptors TLRs in inflammatory arthritis has been widely investigated and targeting either the receptor itself or the proteins in the downstream signalling cascades has emerged as a promising therapeutic strategy. Yet, concerns about the use of pharmacological agents that inhibit TLR activity and may leave the host unprotected against invading pathogens and toxicity issues amid inhibition of downstream kinases crucial in various cellular functions have arisen. This review summarizes the existing knowledge on the role of TLRs in inflammatory arthritis; in addition, the likely druggable related targets and the developed inhibitors, and discusses the pros and cons of their potential clinical use.
Collapse
|
25
|
Decarriere G, Barnetche T, Combe B, Gaujoux-Viala C, Lukas C, Morel J, Daien C. Most Appropriate Conventional Disease-Modifying Antirheumatic Drug to Combine With Different Advanced Therapies in Rheumatoid Arthritis: A Systematic Literature Review With Meta-Analysis. Arthritis Care Res (Hoboken) 2021; 73:873-884. [PMID: 32216091 DOI: 10.1002/acr.24195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 03/17/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE In rheumatoid arthritis, the association between advanced therapies (including biologic disease-modifying antirheumatic drugs [DMARDs] and targeted synthetic DMARDs) and methotrexate (MTX) is recommended by international societies. When MTX cannot be used, other conventional synthetic DMARDs (csDMARDs) may be proposed. We aimed to compare the safety and efficacy of MTX and non-MTX csDMARDs in combination with advanced therapies. METHODS We systematically searched the literature for studies comparing the effectiveness, retention rate, and safety of MTX versus non-MTX csDMARDs (leflunomide or others) in combination with tumor necrosis factor inhibitors (TNFi), abatacept, rituximab, tocilizumab, and JAK inhibitors. Meta-analysis was performed with RevMan, using an inverse variance approach with fixed or random-effects models. Risk ratios (RRs) and 95% confidence intervals (95% CIs) were estimated. RESULTS The literature search revealed 3,842 articles; 41 studies were included for the systematic literature review and 21 for the meta-analysis: 13 with TNFi, 3 with abatacept, and 5 with rituximab. For TNFi, the European Alliance of Associations for Rheumatology (EULAR) response at 6 months was lower for patients receiving non-MTX csDMARDs than for those using MTX (RR 0.93 [95% CI 0.87, 1.0], P = 0.04; n = 3,843; I2 = 28%), with a lower retention rate at 12 months. For abatacept, effectiveness and safety were similar between the 2 groups. For rituximab, a good EULAR response was higher with leflunomide than MTX (RR 1.38 [95% CI 1.13, 1.68], P = 0.001; n = 2,078; I2 = 0%), with similar adverse event rates. Meta-analysis for tocilizumab or JAK inhibitors could not be performed. CONCLUSION The different csDMARDs seem safe and efficient to combine with advanced therapies in RA patients. Although MTX seems slightly superior to other csDMARDs in combination with TNFi, leflunomide might be superior to MTX in combination with rituximab.
Collapse
Affiliation(s)
| | | | - Bernard Combe
- CHU Montpellier and Montpellier University, Montpellier, France
| | | | - Cédric Lukas
- CHU Montpellier and Montpellier University, Montpellier, France
| | - Jacques Morel
- CHU Montpellier and Montpellier University, Montpellier, France
| | - Claire Daien
- CHU Montpellier and Montpellier University, Montpellier, France
| |
Collapse
|
26
|
Wu CY, Yang HY, Huang JL, Lai JH. Signals and Mechanisms Regulating Monocyte and Macrophage Activation in the Pathogenesis of Juvenile Idiopathic Arthritis. Int J Mol Sci 2021; 22:ijms22157960. [PMID: 34360720 PMCID: PMC8347893 DOI: 10.3390/ijms22157960] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Monocytes (Mos) and macrophages (Mφs) are key players in the innate immune system and are critical in coordinating the initiation, expansion, and regression of many autoimmune diseases. In addition, they display immunoregulatory effects that impact inflammation and are essential in tissue repair and regeneration. Juvenile idiopathic arthritis (JIA) is an umbrella term describing inflammatory joint diseases in children. Accumulated evidence suggests a link between Mo and Mφ activation and JIA pathogenesis. Accordingly, topics regarding the signals and mechanisms regulating Mo and Mφ activation leading to pathologies in patients with JIA are of great interest. In this review, we critically summarize recent advances in the understanding of how Mo and Mφ activation is involved in JIA pathogenesis and focus on the signaling pathways and mechanisms participating in the related cell activation processes.
Collapse
Affiliation(s)
- Chao-Yi Wu
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-Y.W.); (J.-L.H.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Huang-Yu Yang
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Jing-Long Huang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-Y.W.); (J.-L.H.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei City 236, Taiwan
| | - Jenn-Haung Lai
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan
- National Defense Medical Center, Graduate Institute of Medical Science, Taipei 114, Taiwan
- Correspondence: ; Tel./Fax: +886-2-8791-8382
| |
Collapse
|
27
|
Boutet MA, Courties G, Nerviani A, Le Goff B, Apparailly F, Pitzalis C, Blanchard F. Novel insights into macrophage diversity in rheumatoid arthritis synovium. Autoimmun Rev 2021; 20:102758. [PMID: 33476818 DOI: 10.1016/j.autrev.2021.102758] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 11/24/2020] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease affecting joints and causing progressive damage and disability. Macrophages are of critical importance in the initiation and perpetuation of synovitis in RA, they can function as antigen presenting cells leading to T-cell dependent B-cell activation, assume a variety of inflammatory cell states with the production of destructive cytokines, but also contribute to tissue homeostasis/repair. The recent development of high-throughput technologies, including bulk and single cells RNA-sequencing, has broadened our understanding of synovial cell diversity, and opened novel perspectives to the discovery of new potential therapeutic targets in RA. In this review, we will focus on the relationship between the synovial macrophage infiltration and clinical disease severity and response to treatment. We will then provide a state-of-the-art picture of the biological roles of synovial macrophages and distinct macrophage subsets described in RA. Finally, we will review the effects of approved conventional and biologic drugs on the synovial macrophage component and highlight the therapeutic potential of future strategies to re-program macrophage phenotypes in RA.
Collapse
Affiliation(s)
- Marie-Astrid Boutet
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Gabriel Courties
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.
| | - Alessandra Nerviani
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Benoit Le Goff
- INSERM UMR1238, Bone Sarcoma and Remodelling of Calcified Tissues, Nantes University, Nantes, France; Rheumatology Department, Nantes University Hospital, Nantes, France.
| | | | - Costantino Pitzalis
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Frédéric Blanchard
- INSERM UMR1238, Bone Sarcoma and Remodelling of Calcified Tissues, Nantes University, Nantes, France.
| |
Collapse
|
28
|
Rajaiah R, Abhilasha KV, Shekar MA, Vogel SN, Vishwanath BS. Evaluation of mechanisms of action of re-purposed drugs for treatment of COVID-19. Cell Immunol 2020; 358:104240. [PMID: 33137649 PMCID: PMC7558230 DOI: 10.1016/j.cellimm.2020.104240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/16/2020] [Accepted: 10/09/2020] [Indexed: 12/23/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a global health emergency caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). The rapid worldwide spread of SARS-CoV-2 infection has necessitated a global effort to identify effective therapeutic strategies in the absence of vaccine. Among the re-purposed drugs being tested currently, hydroxychloroquine (HCQ), without or with zinc ion (Zn++) and the antibiotic azithromycin (AZM), has been administered to prevent or treat patients with COVID-19. The outcome of multiple clinical studies on HCQ has been mixed. Zn++ interferes with viral replication by inhibiting replicative enzymes and its entry into cells may be facilitated by HCQ. Another immunomodulatory drug, methotrexate (MTX), is well known for its ability to mitigate overactive immune system by upregulating the anti-inflammatory protein, A20. However, its beneficial effect in treating COVID-19 has not drawn much attention. This review provides an overview of the virology of SARS-CoV-2 and an analysis of the mechanisms by which these anti-inflammatory agents may act in the treatment of COVID-19 patients. We propose a rationale for the combinatorial use of these re-purposed drugs that may help to combat this ongoing pandemic health emergency.
Collapse
Affiliation(s)
- Rajesh Rajaiah
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysuru, Karnataka, India.
| | - Kandahalli V Abhilasha
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| | - Mysore A Shekar
- Chowdaiah Medical Center & Apoorva Diabetes Foundation, Mysuru, Karnataka, India
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine (UMSOM), Baltimore, MD, USA
| | - Bannikuppe S Vishwanath
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysuru, Karnataka, India.
| |
Collapse
|
29
|
Triguero-Martínez A, de la Fuente H, Montes N, Ortiz AM, Roy-Vallejo E, Castañeda S, González-Alvaro I, Lamana A. Validation of galectin-1 as potential diagnostic biomarker of early rheumatoid arthritis. Sci Rep 2020; 10:17799. [PMID: 33082382 PMCID: PMC7576119 DOI: 10.1038/s41598-020-74185-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/21/2020] [Indexed: 01/01/2023] Open
Abstract
Galectin 1 (Gal1) is a lectin with a wide cellular expression that functions as a negative regulator of the immune system in several animal models of autoimmune diseases. Identification of patients with rheumatoid arthritis (RA) has improved during the last decade, although there is still a need for biomarkers allowing an early diagnosis. In this regard, it has been recently proposed that Gal1 serum levels are increased in patients with RA compared to the general population. However, this topic is controversial in the literature. In this work, we provide additional information about the potential usefulness of Gal1 serum levels as a biomarker for RA diagnosis. We studied Gal1 serum and synovial fluid levels and clinical parameters in samples from 62 patients with early arthritis belonging to the PEARL study. In addition, 24 healthy donors were studied. We found that both patients fulfilling RA criteria and patients with undifferentiated arthritis showed higher Gal1 levels than healthy donors. Similar findings were observed in synovial fluid, which showed even higher levels than serum. However, we did not find correlation between Gal1 levels and disease activity or disability. Therefore, our results suggest that Gal1 could be a diagnostic but not a severity biomarker.
Collapse
Affiliation(s)
- Ana Triguero-Martínez
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Hortensia de la Fuente
- Immunology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Nuria Montes
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Ana María Ortiz
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Emilia Roy-Vallejo
- Internal Medicine Service, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Santos Castañeda
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain
| | - Isidoro González-Alvaro
- Rheumatology Department, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-IP), Madrid, Spain.
| | - Amalia Lamana
- Cell Biology Department, Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
30
|
Fattahi N, Bahari A, Ramazani A, Koolivand D. In vitro immunobiological assays of methotrexate-stearic acid conjugate in human PBMCs. Immunobiology 2020; 225:151984. [DOI: 10.1016/j.imbio.2020.151984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/04/2020] [Indexed: 10/23/2022]
|
31
|
Jara González FE, Jimenez Alulima GDF, Sananay Auquilla EL, Murillo Sanclemente JC, Molina Vasquez PA, Vélez Páez JL. Hipercoagulabilidad, trombosis intravascular y trombocitosis asociada al COVID-19. Reporte de un caso. BIONATURA 2020. [DOI: 10.21931/rb/2020.05.02.11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Una nueva pandemia en el siglo XXI, generada por un agente viral del tipo coronavirus denominado SARS CoV-2, que produce la enfermedad COVID-19, la misma que debutó a finales del 2019 en China, con una mortalidad importante, que afecta el tracto respiratorio inferior y se manifiesta como neumonía en humanos.
A medida de su expansión mundial, el virus ha presentado mutaciones y por ende variantes fenitípicas que han generado cuadros clínicos que distan mucho de los síntomas descritos inicialmente; más bien, las alteraciones de la coagulación y la intensa inflamación (tormenta de citoquinas) han generado síntomas atípicos y enfoques terapéuticos diferentes; situando a la ferritina y el dímero D como biomarcadores cardinales para decidir anticoagulación y/o terapia anti inflamatoria como corticoterapia, el monoclonal anti IL6 Tocilizumab o el metotrexate, entre otros.
A continuación, presentamos el caso de un paciente diagnosticado de COVID-19 con RT-PCR, que presenta a más de sus manifestaciones respiratorias, un estado protrombótico que desencadenó trombosis intravasculares y posiblemente embolismo pulmonar, con valores elevados de ferritina y dimero D, que luego de anticoagulación y administración de corticoides superó su estado de gravedad extrema y egreso de Terapia Intensiva.
Collapse
|
32
|
Kania G, Rudnik M, Distler O. Involvement of the myeloid cell compartment in fibrogenesis and systemic sclerosis. Nat Rev Rheumatol 2020; 15:288-302. [PMID: 30953037 DOI: 10.1038/s41584-019-0212-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Systemic sclerosis (SSc) is an autoimmune fibrotic disease of unknown aetiology that is characterized by vascular changes in the skin and visceral organs. Autologous haematopoietic stem cell transplantation can improve skin and organ fibrosis in patients with progressive disease and a high risk of organ failure, indicating that cells originating in the bone marrow are important contributors to the pathogenesis of SSc. Animal studies also indicate a pivotal function of myeloid cells in the development of fibrosis leading to changes in the tissue architecture and dysfunction in multiple organs such as the heart, lungs, liver and kidney. In this Review, we summarize current knowledge about the function of myeloid cells in fibrogenesis that occurs in patients with SSc. Targeted therapies currently in clinical studies for SSc might affect myeloid cell-related pathways. Therefore, myeloid cells might be used as cellular biomarkers of disease through the application of high-dimensional techniques such as mass cytometry and single-cell RNA sequencing.
Collapse
Affiliation(s)
- Gabriela Kania
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Michal Rudnik
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
33
|
Methotrexate and its mechanisms of action in inflammatory arthritis. Nat Rev Rheumatol 2020; 16:145-154. [PMID: 32066940 DOI: 10.1038/s41584-020-0373-9] [Citation(s) in RCA: 311] [Impact Index Per Article: 77.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2020] [Indexed: 11/08/2022]
Abstract
Despite the introduction of numerous biologic agents for the treatment of rheumatoid arthritis (RA) and other forms of inflammatory arthritis, low-dose methotrexate therapy remains the gold standard in RA therapy. Methotrexate is generally the first-line drug for the treatment of RA, psoriatic arthritis and other forms of inflammatory arthritis, and it enhances the effect of most biologic agents in RA. Understanding the mechanism of action of methotrexate could be instructive in the appropriate use of the drug and in the design of new regimens for the treatment of RA. Although methotrexate is one of the first examples of intelligent drug design, multiple mechanisms potentially contribute to the anti-inflammatory actions of methotrexate, including the inhibition of purine and pyrimidine synthesis, transmethylation reactions, translocation of nuclear factor-κB (NF-κB) to the nucleus, signalling via the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway and nitric oxide production, as well as the promotion of adenosine release and expression of certain long non-coding RNAs.
Collapse
|
34
|
Riffo-Vasquez Y, Kanabar V, Keir SD, E-Lacerda RR, Man F, Jackson DJ, Corrigall V, Coates ARM, Page CP. Modulation of allergic inflammation in the lung by a peptide derived from Mycobacteria tuberculosis chaperonin 60.1. Clin Exp Allergy 2020; 50:508-519. [PMID: 31845415 DOI: 10.1111/cea.13550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/25/2019] [Accepted: 12/01/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND We have previously demonstrated that Mycobacteria tuberculosis chaperonin 60.1 inhibits leucocyte diapedesis and bronchial hyperresponsiveness in a murine model of allergic lung inflammation. METHODS In the present study, we have investigated the effect of a shorter peptide sequence derived from Cpn 60.1, named IRL201104, on allergic lung inflammation induced by ovalbumin (OVA) in mice and by house dust mite (HDM) in guinea pigs, as well as investigating the action of IRL201104 on human cells in vitro. RESULTS Pre-treatment of mice or guinea pigs with IRL201104 inhibits the infiltration of eosinophils to the lung, cytokine release, and in guinea pig skin, inhibits allergen-induced vascular permeability. The protective effect of intranasal IRL201104 against OVA-induced eosinophilia persisted for up to 20 days post-treatment. Moreover, OVA-sensitized mice treated intranasally with 20 ng/kg of IRL201104 show a significant increase in the expression of the anti-inflammatory molecule ubiquitin A20 and significant inhibition of the activation of NF-κB in lung tissue. Our results also show that A20 expression was significantly reduced in blood leucocytes and ASM obtained from patients with asthma compared to cells obtained from healthy subjects which were restored after incubation with IRL201104 in vitro, when added alone, or in combination with LPS or TNF-α in ASM. CONCLUSIONS Our results suggest that a peptide derived from mycobacterial Cpn60.1 has a long-lasting anti-inflammatory and immunomodulatory activity which may help explain some of the protective effects of TB against allergic diseases.
Collapse
Affiliation(s)
- Yanira Riffo-Vasquez
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Sciences, King's College London, London, UK
| | - Varsha Kanabar
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Sciences, King's College London, London, UK
| | - Sandra D Keir
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Sciences, King's College London, London, UK
| | - Rodrigo R E-Lacerda
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Sciences, King's College London, London, UK
| | - Francis Man
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Sciences, King's College London, London, UK
| | - David J Jackson
- Asthma Care Guy's & St Thomas' NHS Trust, London, UK.,Faculty of Life Sciences and Medicine, MRC & Asthma UK Centre, Guy's Hospital, King's College London, London, UK
| | - Valerie Corrigall
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Sciences, King's College London, London, UK
| | - Anthony R M Coates
- Medical Microbiology, Institute of Infection and Immunity, St George's, University of London, London, UK
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Sciences, King's College London, London, UK
| |
Collapse
|
35
|
Jekic B, Maksimovic N, Damnjanovic T. Methotrexate pharmacogenetics in the treatment of rheumatoid arthritis. Pharmacogenomics 2019; 20:1235-1245. [DOI: 10.2217/pgs-2019-0121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
For many decades, methotrexate (MXT) has remained the drug of choice in the treatment of rheumatoid arthritis (RA). Unfortunately, a considerable number of patients do not achieve an appropriate therapeutic response. Pharmacogenetics studies do not give usable results regarding differences in MTX response among RA patients. The mechanism of MTX action in RA is not completely understood. We present and discuss data regarding the molecular basis of folate and adenosine pathways, the most obvious MTX targets, to explain possible causes of therapy failure. The molecular basis of the disease could also have an impact on therapy outcomes and in this review we explore this. Finally, we make a short review of available pharmacogenetics study results.
Collapse
Affiliation(s)
- Biljana Jekic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade,11000 Belgrade, Serbia
| | - Nela Maksimovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade,11000 Belgrade, Serbia
| | - Tatjana Damnjanovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade,11000 Belgrade, Serbia
| |
Collapse
|
36
|
Frigerio B, Bizzoni C, Jansen G, Leamon CP, Peters GJ, Low PS, Matherly LH, Figini M. Folate receptors and transporters: biological role and diagnostic/therapeutic targets in cancer and other diseases. J Exp Clin Cancer Res 2019; 38:125. [PMID: 30867007 PMCID: PMC6417013 DOI: 10.1186/s13046-019-1123-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/28/2019] [Indexed: 01/28/2023] Open
Abstract
Folate receptors and transporters and one-carbon metabolism continue to be important areas of study given their essential roles in an assortment of diseases and as targets for treatment of cancer and inflammation. Reflecting this, every 2 years, the Folate Receptor Society organizes an international meeting, alternating between North America and Europe, where basic and translational scientists, clinical oncologists and rheumatologists from both academia and industry convene in an informal setting. The 7th International Symposium on Folate Receptors and Transporters was held in Sant'Alessio Siculo (ME), Taormina, Italy from 1st to 5th of October 2018, organized by Dr. Mariangela Figini from Fondazione IRCCS Istituto Nazionale dei Tumori, Milan. Following the format of previous meetings, more than 50 scientists from 9 different countries attended the 2018 meeting to share ongoing developments, discuss current research challenges and identify new avenues in basic and translational research. An important feature of this meeting was the participation of young investigators and trainees in this area, two (A. Dekhne and N. Verweij) of whom were awarded fellowships to attend this meeting as a recognition of the high scientific quality of their work. This report provides a synopsis of the highlights presented in the following sessions: Barton Kamen Lecture; Targeting one-carbon metabolism in cytosol and mitochondria; Structure and biology of the one-carbon solute transporters; Physiology and pathophysiology of folate receptors and transporters; Folate receptors for targeting tumors and inflammatory diseases; Conventional and new anti-folate drugs for treating inflammatory diseases and cancer; Imaging; Ongoing clinical trials; and Chimeric Antigen Receptor cell therapies of cancer.
Collapse
Affiliation(s)
- Barbara Frigerio
- Dipartimento di Ricerca Applicata e Sviluppo Tecnologico, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Bizzoni
- Dipartimento di Ricerca Applicata e Sviluppo Tecnologico, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Present address: ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Center, location Vrije Universiteit, Amsterdam, The Netherlands
| | | | - Godefridus J. Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Philip S. Low
- Purdue University Institute for Drug Discovery, West Lafayette, Indiana, USA
| | - Larry H. Matherly
- Barbara Ann Karmanos Cancer Institute and Wayne State University School of Medicine, Detroit, MI USA
| | - Mariangela Figini
- Dipartimento di Ricerca Applicata e Sviluppo Tecnologico, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
37
|
Tabata R, Tabata C, Uesugi H, Takei Y. Highly aggressive plasmablastic neoplasms in patients with rheumatoid arthritis treated with methotrexate. Int Immunopharmacol 2019; 68:213-217. [DOI: 10.1016/j.intimp.2019.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 10/27/2022]
|
38
|
Mangoni AA, Tommasi S, Zinellu A, Sotgia S, Carru C, Piga M, Erre GL. Repurposing existing drugs for cardiovascular risk management: a focus on methotrexate. Drugs Context 2018; 7:212557. [PMID: 30459819 PMCID: PMC6239018 DOI: 10.7573/dic.212557] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
About 20% of patients with a history of atherosclerotic cardiovascular disease will experience further cardiovascular events despite maximal pharmacological treatment with cardioprotective drugs. This highlights the presence of residual cardiovascular risk in a significant proportion of patients and the need for novel, more effective therapies. These therapies should ideally target different pathophysiological pathways involved in the onset and the progression of atherosclerosis, particularly the inflammatory and immune pathways. Methotrexate is a first-line disease-modifying antirheumatic drug that is widely used for the management of autoimmune and chronic inflammatory disorders. There is some in vitro and in vivo evidence that methotrexate might exert a unique combination of anti-inflammatory, blood pressure lowering, and vasculoprotective effects. Pending the results of large prospective studies investigating surrogate end-points as well as morbidity and mortality, repurposing methotrexate for cardiovascular risk management might represent a cost-effective strategy with immediate public health benefits. This review discusses the current challenges in the management of cardiovascular disease; the available evidence on the effects of methotrexate on inflammation, blood pressure, and surrogate markers of arterial function; suggestions for future research directions; and practical considerations with the use of methotrexate in this context.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Sara Tommasi
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Matteo Piga
- Rheumatology Unit, University Clinic and AOU of Cagliari, Italy
| | - Gian Luca Erre
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital (AOUSS) and University of Sassari, Sassari, Italy
| |
Collapse
|
39
|
Tabata R, Tabata C, Okamura M, Takei Y, Ohshima K. Successful treatment of monomorphic epitheliotropic intestinal T cell lymphoma with pralatrexate. Ann Hematol 2018; 98:1301-1303. [PMID: 30173289 DOI: 10.1007/s00277-018-3491-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Rie Tabata
- Department of Hematology and Rheumatology, Saiseikai-Noe Hospital, 1-3-25 Furuichi, Joto-ku, Osaka, 536-0001, Japan.
| | - Chiharu Tabata
- Cancer Center, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | | | - Yusuke Takei
- Department of Pathology, Saiseikai Noe Hospital, Osaka, Japan
| | - Koichi Ohshima
- Department of Pathology, School of Medicine, Kurume University, Kurume, Japan
| |
Collapse
|