1
|
Teng L, Li L, Cui D, An R, Jin J. Polymyalgia rheumatica and giant cell arteritis: A bidirectional Mendelian randomization study. Medicine (Baltimore) 2024; 103:e39723. [PMID: 39312384 PMCID: PMC11419444 DOI: 10.1097/md.0000000000039723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Polymyalgia rheumatica (PMR) and giant cell arteritis (GCA) as 2 types of autoimmune diseases are frequently concomitant, and Mendelian randomization (MR) was applied in this study to assess the causal relationship between them. In this study, single-nucleotide polymorphism (SNP) was used as the instrumental variable for Mendelian analysis, and the SNP data of GCA and PMR were obtained from the FinnGen Biobank databases. SNPs are significantly correlated with GCA and PMR and were screened based on preset thresholds. Inverse variance weighted analysis was used as the main analysis, supplemented with MR-Egger and weighted median. The evidence of the impact of GCA on PMR risk was found in inverse variance weighted results (odds ratio, 1.22 [95% confidence interval, 1.11-1.34]; P < .01), and the evidence of the impact of PMR on GCA risk has also been found (odds ratio, 1.58 [95% confidence interval, 1.28-1.96]; P < .01). Finally, the stability and reliability of the results were tested using the retention method, heterogeneity test, and horizontal gene pleiotropy test. MR analysis indicates that GCA increases the risk of PMR and PMR is an important risk factor for GCA, with a causal relationship. The potential value of reasonable management of PMR in patients with GCA has received high attention. In addition, novel GCA therapeutics may be indicated for PMR, and it is a potential for further investigation.
Collapse
Affiliation(s)
- Lin Teng
- Yanbian University Hospital, Yanji, China
| | - Lei Li
- Yanbian University Hospital, Yanji, China
| | - Dinglu Cui
- Yanbian University Hospital, Yanji, China
| | | | | |
Collapse
|
2
|
Karpouzas GA, Ormseth SR, van Riel PLCM, Gonzalez-Gay MA, Corrales A, Rantapää-Dahlqvist S, Sfikakis PP, Dessein P, Tsang L, Hitchon C, El-Gabalawy H, Pascual-Ramos V, Contreras-Yáñez I, Colunga-Pedraza IJ, Galarza-Delgado DA, Azpiri-Lopez JR, Semb AG, Misra DP, Hauge EM, Kitas G. Biological use influences the impact of inflammation on risk of major adverse cardiovascular events in rheumatoid arthritis. RMD Open 2024; 10:e004546. [PMID: 39043615 PMCID: PMC11268070 DOI: 10.1136/rmdopen-2024-004546] [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: 05/16/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
OBJECTIVES Chronic inflammation promotes cardiovascular risk in rheumatoid arthritis (RA). Biological disease-modifying antirheumatic drugs (bDMARDs) improve disease activity and cardiovascular disease outcomes. We explored whether bDMARDs influence the impact of disease activity and inflammatory markers on long-term cardiovascular risk in RA. METHODS We studied 4370 participants without cardiovascular disease in a 10-country observational cohort of patients with RA. Endpoints were (1) major adverse cardiovascular events (MACE) encompassing myocardial infarction, stroke and cardiovascular death; and (2) any ischaemic cardiovascular events (iCVE) including MACE plus revascularisation, angina, transient ischaemic attack and peripheral arterial disease. RESULTS Over 26 534 patient-years, 239 MACE and 362 iCVE occurred. The interaction between 28-joint Disease Activity Score with C-reactive protein (DAS28-CRP) and bDMARD use was significant for MACE (p=0.017), suggesting the effect of DAS28-CRP on MACE risk differed among bDMARD users (n=515) and non-users (n=3855). DAS28-CRP (per unit increase) is associated with MACE risk in bDMARD non-users (HR 1.21 (95% CI 1.07 to 1.37)) but not users (HR 0.69 (95% CI 0.40 to 1.20)). The interaction between CRP (per log unit increase) and bDMARD use was also significant for MACE (p=0.011). CRP associated with MACE risk in bDMARD non-users (HR 1.16 (95% CI 1.04 to 1.30)), but not users (HR 0.65 (95% CI 0.36 to 1.17)). No interaction was observed between bDMARD use and DAS28-CRP (p=0.167) or CRP (p=0.237) for iCVE risk. CONCLUSIONS RA activity and inflammatory markers associated with risk of MACE in bDMARD non-users but not users suggesting the possibility of biological-specific benefits locally on arterial wall independently of effects on systemic inflammation.
Collapse
Affiliation(s)
- George Athanasios Karpouzas
- Internal Medicine - Rheumatology, The Lundquist Institute, Torrance, California, USA
- Rheumatology, Harbor-UCLA Medical Center, Torrance, California, USA
| | | | | | - Miguel A Gonzalez-Gay
- Rheumatology, Hospital Universitario Marques de Valdecilla, Santander, Cantabria, Spain
- IIS-Fundacion Jimenez Diaz, Madrid, Spain
| | - Alfonso Corrales
- Hospital Universitario Marques de Valdecilla, Santander, Cantabria, Spain
| | | | - Petros P Sfikakis
- First Dept. of Propedeutic Medicine, University of Athens, Athens, Attica, Greece
| | - Patrick Dessein
- School of Physiology, University of the Witwatersrand Johannesburg, Johannesburg, South Africa
| | - Linda Tsang
- Vrije Universiteit Brussel, Brussel, Belgium
| | - Carol Hitchon
- Rheumatology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hani El-Gabalawy
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Virginia Pascual-Ramos
- Immunology and Rheumatology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico City, Mexico
| | - Irazú Contreras-Yáñez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Ciudad de Mexico, Mexico
| | - Iris J Colunga-Pedraza
- Rheumatology, Hospital Universitario Dr José Eleuterio González, Monterrey, Nuevo León, Mexico
| | | | | | - Anne Grete Semb
- Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Durga Prasanna Misra
- Clinical Immunology and Rheumatology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Ellen-Margrethe Hauge
- Department of Joint and Connective Tissue Diseases, Aarhus Universitetshospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus Universitet, Aarhus, Midtjylland, Denmark
| | - George Kitas
- Department of Rheumatology, The Dudley Group NHS Foundation Trust, Dudley, West Midlands, UK
| |
Collapse
|
3
|
van der Geest KSM, Sandovici M, Bley TA, Stone JR, Slart RHJA, Brouwer E. Large vessel giant cell arteritis. THE LANCET. RHEUMATOLOGY 2024; 6:e397-e408. [PMID: 38574745 DOI: 10.1016/s2665-9913(23)00300-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 04/06/2024]
Abstract
Giant cell arteritis is the principal form of systemic vasculitis affecting people over 50. Large-vessel involvement, termed large vessel giant cell arteritis, mainly affects the aorta and its branches, often occurring alongside cranial giant cell arteritis, but large vessel giant cell arteritis without cranial giant cell arteritis can also occur. Patients mostly present with constitutional symptoms, with localising large vessel giant cell arteritis symptoms present in a minority of patients only. Large vessel giant cell arteritis is usually overlooked until clinicians seek to exclude it with imaging by ultrasonography, magnetic resonance angiography (MRA), computed tomography angiography (CTA), or [18F]fluorodeoxyglucose-PET-CT. Although the role of imaging in treatment monitoring remains uncertain, imaging by MRA or CTA is crucial for identifying aortic aneurysm formation during patient follow up. In this Series paper, we define the large vessel subset of giant cell arteritis and summarise its clinical challenges. Furthermore, we identify areas for future research regarding the management of large vessel giant cell arteritis.
Collapse
Affiliation(s)
- Kornelis S M van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Thorsten A Bley
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - James R Stone
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Riemer H J A Slart
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Netherlands; Department of Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, Enschede, Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
4
|
Nageswaran P, Ahmed S, Tahir H. Review of phase 2/3 trials in polymyalgia rheumatica and giant cell arteritis. Expert Opin Emerg Drugs 2024; 29:5-17. [PMID: 38180809 DOI: 10.1080/14728214.2024.2303093] [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: 10/20/2023] [Accepted: 01/04/2024] [Indexed: 01/07/2024]
Abstract
INTRODUCTION GCA (giant cell arteritis) and PMR (polymyalgia rheumatica) are two overlapping inflammatory rheumatic conditions that are seen exclusively in older adults, sharing some common features. GCA is a clinical syndrome characterized by inflammation of the medium and large arteries, with both cranial and extracranial symptoms. PMR is a clinical syndrome characterized by stiffness in the neck, shoulder, and pelvic girdle muscles. Both are associated with constitutional symptoms. AREAS COVERED In this review, we assess the established and upcoming treatments for GCA and PMR. We review the current treatment landscape, completed trials, and upcoming trials in these conditions, to identify new and promising therapies. EXPERT OPINION Early use of glucocorticoids (GC) remains integral to the immediate management of PMR and GCA but being aware of patient co-morbidities that may influence treatment toxicity is paramount. As such GC sparing agents are required in the treatment of PMR. Currently there are limited treatment options available for PMR and GCA, and significant unmet needs remain. Newer mechanisms of action, and hence therapeutic options being studied include CD4 T cell co-stimulation blockade, IL-17 inhibition, IL-12/23 inhibition, GM-CSF inhibition, IL-1β inhibition, TNF-α antagonist and Jak inhibition, among others, which will be discussed in this review.
Collapse
Affiliation(s)
| | - Saad Ahmed
- Department of Rheumatology, East Suffolk and North Essex Foundation Trust, Colchester, UK
| | - Hasan Tahir
- Department of Rheumatology, Royal Free London NHS Trust, London, UK
- Department of Medicine, University College London, London, UK
| |
Collapse
|
5
|
Palamidas DA, Chatzis L, Papadaki M, Gissis I, Kambas K, Andreakos E, Goules AV, Tzioufas AG. Current Insights into Tissue Injury of Giant Cell Arteritis: From Acute Inflammatory Responses towards Inappropriate Tissue Remodeling. Cells 2024; 13:430. [PMID: 38474394 DOI: 10.3390/cells13050430] [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: 01/31/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Giant cell arteritis (GCA) is an autoimmune disease affecting large vessels in patients over 50 years old. It is an exemplary model of a classic inflammatory disorder with IL-6 playing the leading role. The main comorbidities that may appear acutely or chronically are vascular occlusion leading to blindness and thoracic aorta aneurysm formation, respectively. The tissue inflammatory bulk is expressed as acute or chronic delayed-type hypersensitivity reactions, the latter being apparent by giant cell formation. The activated monocytes/macrophages are associated with pronounced Th1 and Th17 responses. B-cells and neutrophils also participate in the inflammatory lesion. However, the exact order of appearance and mechanistic interactions between cells are hindered by the lack of cellular and molecular information from early disease stages and accurate experimental models. Recently, senescent cells and neutrophil extracellular traps have been described in tissue lesions. These structures can remain in tissues for a prolonged period, potentially favoring inflammatory responses and tissue remodeling. In this review, current advances in GCA pathogenesis are discussed in different inflammatory phases. Through the description of these-often overlapping-phases, cells, molecules, and small lipid mediators with pathogenetic potential are described.
Collapse
Affiliation(s)
- Dimitris Anastasios Palamidas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Loukas Chatzis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Maria Papadaki
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Ilias Gissis
- Department of Thoracic and Cardiovascular Surgery, Evangelismos General Hospital, 11473 Athens, Greece
| | - Konstantinos Kambas
- Laboratory of Molecular Genetics, Department of Immunology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Andreas V Goules
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Athanasios G Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
- Research Institute for Systemic Autoimmune Diseases, 11527 Athens, Greece
| |
Collapse
|
6
|
Britsch S, Langer H, Duerschmied D, Becher T. The Evolving Role of Dendritic Cells in Atherosclerosis. Int J Mol Sci 2024; 25:2450. [PMID: 38397127 PMCID: PMC10888834 DOI: 10.3390/ijms25042450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis, a major contributor to cardiovascular morbidity and mortality, is characterized by chronic inflammation of the arterial wall. This inflammatory process is initiated and maintained by both innate and adaptive immunity. Dendritic cells (DCs), which are antigen-presenting cells, play a crucial role in the development of atherosclerosis and consist of various subtypes with distinct functional abilities. Following the recognition and binding of antigens, DCs become potent activators of cellular responses, bridging the innate and adaptive immune systems. The modulation of specific DC subpopulations can have either pro-atherogenic or atheroprotective effects, highlighting the dual pro-inflammatory or tolerogenic roles of DCs. In this work, we provide a comprehensive overview of the evolving roles of DCs and their subtypes in the promotion or limitation of atherosclerosis development. Additionally, we explore antigen pulsing and pharmacological approaches to modulate the function of DCs in the context of atherosclerosis.
Collapse
Affiliation(s)
- Simone Britsch
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 13092 Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Harald Langer
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 13092 Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Daniel Duerschmied
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 13092 Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Tobias Becher
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
| |
Collapse
|
7
|
Cho MJ, Lee MR, Park JG. Aortic aneurysms: current pathogenesis and therapeutic targets. Exp Mol Med 2023; 55:2519-2530. [PMID: 38036736 PMCID: PMC10766996 DOI: 10.1038/s12276-023-01130-w] [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: 06/29/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 12/02/2023] Open
Abstract
Aortic aneurysm is a chronic disease characterized by localized expansion of the aorta, including the ascending aorta, arch, descending aorta, and abdominal aorta. Although aortic aneurysms are generally asymptomatic, they can threaten human health by sudden death due to aortic rupture. Aortic aneurysms are estimated to lead to 150,000 ~ 200,000 deaths per year worldwide. Currently, there are no effective drugs to prevent the growth or rupture of aortic aneurysms; surgical repair or endovascular repair is the only option for treating this condition. The pathogenic mechanisms and therapeutic targets for aortic aneurysms have been examined over the past decade; however, there are unknown pathogenic mechanisms involved in cellular heterogeneity and plasticity, the complexity of the transforming growth factor-β signaling pathway, inflammation, cell death, intramural neovascularization, and intercellular communication. This review summarizes the latest research findings and current pathogenic mechanisms of aortic aneurysms, which may enhance our understanding of aortic aneurysms.
Collapse
Affiliation(s)
- Min Ji Cho
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Mi-Ran Lee
- Department of Biomedical Laboratory Science, Jungwon University, 85 Munmu-ro, Goesan-eup, Goesan-gun, Chungbuk, 28024, Republic of Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
- Department of Bioscience, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
8
|
Aronova A, Tosato F, Naser N, Asare Y. Innate Immune Pathways in Atherosclerosis-From Signaling to Long-Term Epigenetic Reprogramming. Cells 2023; 12:2359. [PMID: 37830572 PMCID: PMC10571887 DOI: 10.3390/cells12192359] [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: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Innate immune pathways play a crucial role in the development of atherosclerosis, from sensing initial danger signals to the long-term reprogramming of immune cells. Despite the success of lipid-lowering therapy, anti-hypertensive medications, and other measures in reducing complications associated with atherosclerosis, cardiovascular disease (CVD) remains the leading cause of death worldwide. Consequently, there is an urgent need to devise novel preventive and therapeutic strategies to alleviate the global burden of CVD. Extensive experimental research and epidemiological studies have demonstrated the dominant role of innate immune mechanisms in the progression of atherosclerosis. Recently, landmark trials including CANTOS, COLCOT, and LoDoCo2 have provided solid evidence demonstrating that targeting innate immune pathways can effectively reduce the risk of CVD. These groundbreaking trials mark a significant paradigm shift in the field and open new avenues for atheroprotective treatments. It is therefore crucial to comprehend the intricate interplay between innate immune pathways and atherosclerosis for the development of targeted therapeutic interventions. Additionally, unraveling the mechanisms underlying long-term reprogramming may offer novel strategies to reverse the pro-inflammatory phenotype of immune cells and restore immune homeostasis in atherosclerosis. In this review, we present an overview of the innate immune pathways implicated in atherosclerosis, with a specific focus on the signaling pathways driving chronic inflammation in atherosclerosis and the long-term reprogramming of immune cells within atherosclerotic plaque. Elucidating the molecular mechanisms governing these processes presents exciting opportunities for the development of a new class of immunotherapeutic approaches aimed at reducing inflammation and promoting plaque stability. By addressing these aspects, we can potentially revolutionize the management of atherosclerosis and its associated cardiovascular complications.
Collapse
Affiliation(s)
| | | | | | - Yaw Asare
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), 80539 Munich, Germany
| |
Collapse
|
9
|
Parreau S, Molina E, Dumonteil S, Goulabchand R, Naves T, Bois MC, Akil H, Terro F, Fauchais AL, Liozon E, Jauberteau MO, Weyand CM, Ly KH. Use of high-plex data provides novel insights into the temporal artery processes of giant cell arteritis. Front Immunol 2023; 14:1237986. [PMID: 37744332 PMCID: PMC10512077 DOI: 10.3389/fimmu.2023.1237986] [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: 06/10/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Objective To identify the key coding genes underlying the biomarkers and pathways associated with giant cell arteritis (GCA), we performed an in situ spatial profiling of molecules involved in the temporal arteries of GCA patients and controls. Furthermore, we performed pharmacogenomic network analysis to identify potential treatment targets. Methods Using human formalin-fixed paraffin-embedded temporal artery biopsy samples (GCA, n = 9; controls, n = 7), we performed a whole transcriptome analysis using the NanoString GeoMx Digital Spatial Profiler. In total, 59 regions of interest were selected in the intima, media, adventitia, and perivascular adipose tissue (PVAT). Differentially expressed genes (DEGs) (fold-change > 2 or < -2, p-adjusted < 0.01) were compared across each layer to build a spatial and pharmacogenomic network and to explore the pathophysiological mechanisms of GCA. Results Most of the transcriptome (12,076 genes) was upregulated in GCA arteries, compared to control arteries. Among the screened genes, 282, 227, 40, and 5 DEGs were identified in the intima, media, adventitia, and PVAT, respectively. Genes involved in the immune process and vascular remodeling were upregulated within GCA temporal arteries but differed across the arterial layers. The immune-related functions and vascular remodeling were limited to the intima and media. Conclusion This study is the first to perform an in situ spatial profiling characterization of the molecules involved in GCA. The pharmacogenomic network analysis identified potential target genes for approved and novel immunotherapies.
Collapse
Affiliation(s)
- Simon Parreau
- Division of Rheumatology, Mayo Clinic, Rochester, MN, United States
- Division of Internal Medicine, Dupuytren University Hospital, Limoges, France
- INSERM U1308, Faculty of Medicine, University of Limoges, Limoges, France
| | - Elsa Molina
- Stem Cell Genomics Core, Stem Cell Program, University of California, San Diego, La Jolla, CA, United States
- Next Generation Sequencing Core, Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Stéphanie Dumonteil
- Division of Internal Medicine, Dupuytren University Hospital, Limoges, France
| | - Radjiv Goulabchand
- Division of Internal Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Thomas Naves
- INSERM U1308, Faculty of Medicine, University of Limoges, Limoges, France
| | - Melanie C. Bois
- Division of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Hussein Akil
- INSERM U1308, Faculty of Medicine, University of Limoges, Limoges, France
| | - Faraj Terro
- Cell Biology, Dupuytren University Hospital, Limoges, France
| | - Anne-Laure Fauchais
- Division of Internal Medicine, Dupuytren University Hospital, Limoges, France
- INSERM U1308, Faculty of Medicine, University of Limoges, Limoges, France
| | - Eric Liozon
- Division of Internal Medicine, Dupuytren University Hospital, Limoges, France
| | | | | | - Kim-Heang Ly
- Division of Internal Medicine, Dupuytren University Hospital, Limoges, France
- INSERM U1308, Faculty of Medicine, University of Limoges, Limoges, France
| |
Collapse
|
10
|
Li H, Xu S, Xu B, Zhang Y, Yin J, Yang Y. Unraveling the Links between Chronic Inflammation, Autoimmunity, and Spontaneous Cervicocranial Arterial Dissection. J Clin Med 2023; 12:5132. [PMID: 37568534 PMCID: PMC10419694 DOI: 10.3390/jcm12155132] [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: 04/12/2023] [Revised: 06/12/2023] [Accepted: 07/18/2023] [Indexed: 08/13/2023] Open
Abstract
Advances in imaging techniques have led to a rise in the diagnosis of spontaneous cervicocranial arterial dissection (SCCAD), which is now considered a common cause of stroke in young adults. However, our understanding of the pathophysiological mechanisms underlying SCCAD remains limited. Prior studies have proposed various factors contributing to arterial wall weakness or stress as potential causes for SCCAD. A combination of biopsies, case reports, and case-control studies suggests that inflammatory changes and autoimmunity may play roles in the cascade of events leading to SCCAD. In this review, we examine the close relationship between SCCAD, chronic inflammation, and autoimmune diseases, aiming to explore potential underlying pathophysiological mechanisms connecting these conditions. While some relevant hypotheses and studies exist, direct evidence on this topic is still relatively scarce. Further investigation of the underlying mechanisms in larger clinical cohorts is needed, and the exploration of animal models may provide novel insights.
Collapse
Affiliation(s)
- Hao Li
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (H.L.); (B.X.)
| | - Shiyao Xu
- The Institute of Neuroscience, Soochow University, Suzhou 215006, China; (S.X.); (Y.Z.)
| | - Beibei Xu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (H.L.); (B.X.)
| | - Yutong Zhang
- The Institute of Neuroscience, Soochow University, Suzhou 215006, China; (S.X.); (Y.Z.)
| | - Jun Yin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yi Yang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (H.L.); (B.X.)
| |
Collapse
|
11
|
Vats V, Patel K, Sharma DD, Almansouri NE, Makkapati NSR, Nimal S, Ramteke P, Mohammed Arifuddin B, Jagarlamudi NS, Narain A, Raut YD. Exploring Cardiovascular Manifestations in Vasculitides: An In-Depth Review. Cureus 2023; 15:e44417. [PMID: 37791229 PMCID: PMC10543473 DOI: 10.7759/cureus.44417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2023] [Indexed: 10/05/2023] Open
Abstract
Systemic vasculitides encompass a cluster of autoimmune diseases that affect blood vessels, and are characterized by immune-mediated injury to either small- or large-sized blood vessels. Individuals afflicted with systemic vasculitides experience notable morbidity and mortality attributable to cardiovascular manifestations. Noteworthy among these are ischemic heart disease, venous thromboembolism, aortic involvement, valvular irregularities, myocarditis, and pericarditis. This narrative review investigated and evaluated the prevalent cardiovascular disturbances commonly associated with different types of vasculitides. This review also discusses the mechanisms that underlie these manifestations. It also provides a thorough explanation of the many diagnostic techniques essential for detecting the disease at its occult stage. It is essential for healthcare professionals to have knowledge of the cardiovascular complications caused by vasculitides, as this enables them to promptly recognize these symptoms and employ suitable diagnostic techniques early on. By doing so, timely detection can be ensured, which will subsequently aid in initiating appropriate treatment strategies that are vital for decreasing morbidity and mortality in patients with systemic vasculitides.
Collapse
Affiliation(s)
- Vaibhav Vats
- Internal Medicine, Smt. Kashibai Navale Medical College and General Hospital, Mumbai, IND
| | - Kriyesha Patel
- Internal Medicine, MP Shah Medical College, Jamnagar, IND
| | | | | | | | - Simran Nimal
- Internal Medicine, Byramjee Jeejeebhoy (BJ) Government Medical College, Pune, IND
| | - Palash Ramteke
- Medical School, NKP Salve Institute of Medical Sciences, Nagpur, IND
| | | | | | - Archit Narain
- Internal Medicine, Lala Lajpat Rai Memorial Medical College, Meerut, IND
| | - Yogesh D Raut
- Miscellaneous, NKP Salve Institute of Medical Sciences, Nagpur, IND
| |
Collapse
|
12
|
Bhandari S, Butt SRR, Ishfaq A, Attaallah MH, Ekhator C, Halappa Nagaraj R, Mulmi A, Kamran M, Karski A, Vargas KI, Lazarevic S, Zaman MU, Lakshmipriya Vetrivendan G, Shahzed SMI, Das A, Yadav V, Bellegarde SB, Ullah A. Pathophysiology, Diagnosis, and Management of Takayasu Arteritis: A Review of Current Advances. Cureus 2023; 15:e42667. [PMID: 37525862 PMCID: PMC10386905 DOI: 10.7759/cureus.42667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2023] [Indexed: 08/02/2023] Open
Abstract
Takayasu arteritis (TA) is a rare, chronic, inflammatory vasculitis that primarily affects large arteries, causing significant morbidity and mortality. This review provides an overview of the pathophysiology, diagnosis, and management of TA based on current advances in the field. TA is characterized by autoimmune-mediated inflammation, vascular remodeling, and endothelial dysfunction. The disease progresses through three stages (active, chronic, and healing phase) each presenting distinct clinical features. Diagnosis of TA can be challenging due to non-specific clinical manifestations and the lack of specific diagnostic tests. Various imaging modalities, such as angiography, ultrasound, and Doppler techniques, play a crucial role in the diagnosis of TA by visualizing arterial involvement and assessing disease extent. Management of TA involves a multidisciplinary approach, with disease-modifying anti-rheumatic drugs (DMARDs) as the cornerstone of medical therapy. Synthetic and biologic DMARDs are used to induce remission, control inflammation, and prevent complications. Non-pharmacologic interventions, such as resistance exercises and curcumin supplementation, show potential benefits. Invasive interventions, including endovascular therapy and open surgery, are used for managing vascular lesions. However, challenges remain in disease understanding and management, including the heterogeneity of disease presentation and the lack of standardized treatment guidelines. The future of TA management lies in precision medicine, utilizing biomarkers and molecular profiling to personalize treatment approaches and improve patient outcomes. Further research is needed to unravel the underlying mechanisms of TA and develop targeted therapies.
Collapse
Affiliation(s)
| | - Samia Rauf R Butt
- General Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | | | - Mohamed H Attaallah
- Medicine, Cairo University, Cairo, EGY
- Medicine, University of Louisville, Louisville, USA
| | - Chukwuyem Ekhator
- Neuro-Oncology, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, USA
| | | | | | | | - Amanda Karski
- Emergency Medicine, American University of Antigua, Miami, USA
| | - Karla I Vargas
- Medicine, Universidad Juárez del Estado de Durango, Durango, MEX
| | | | | | | | | | - Archana Das
- Internal Medicine, North East Medical College and Hospital, Sylhet, BGD
| | - Vikas Yadav
- Internal Medicine, Pt. Bhagwat Dayal (BD) Sharma Postgraduate Institute of Medical Sciences, Rohtak, IND
| | - Sophia B Bellegarde
- Pathology and Laboratory Medicine, American University of Antigua, St. John's, ATG
| | | |
Collapse
|
13
|
Tomelleri A, van der Geest KSM, Khurshid MA, Sebastian A, Coath F, Robbins D, Pierscionek B, Dejaco C, Matteson E, van Sleen Y, Dasgupta B. Disease stratification in GCA and PMR: state of the art and future perspectives. Nat Rev Rheumatol 2023:10.1038/s41584-023-00976-8. [PMID: 37308659 DOI: 10.1038/s41584-023-00976-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 06/14/2023]
Abstract
Giant cell arteritis (GCA) and polymyalgia rheumatica (PMR) are closely related conditions characterized by systemic inflammation, a predominant IL-6 signature, an excellent response to glucocorticoids, a tendency to a chronic and relapsing course, and older age of the affected population. This Review highlights the emerging view that these diseases should be approached as linked conditions, unified under the term GCA-PMR spectrum disease (GPSD). In addition, GCA and PMR should be seen as non-monolithic conditions, with different risks of developing acute ischaemic complications and chronic vascular and tissue damage, different responses to available therapies and disparate relapse rates. A comprehensive stratification strategy for GPSD, guided by clinical findings, imaging and laboratory data, facilitates appropriate therapy and cost-effective use of health-economic resources. Patients presenting with predominant cranial symptoms and vascular involvement, who usually have a borderline elevation of inflammatory markers, are at an increased risk of sight loss in early disease but have fewer relapses in the long term, whereas the opposite is observed in patients with predominant large-vessel vasculitis. How the involvement of peripheral joint structures affects disease outcomes remains uncertain and understudied. In the future, all cases of new-onset GPSD should undergo early disease stratification, with their management adapted accordingly.
Collapse
Affiliation(s)
- Alessandro Tomelleri
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Hospital, Milan, Italy
| | - Kornelis S M van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Alwin Sebastian
- Department of Rheumatology, University Hospital Limerick, Limerick, Ireland
| | - Fiona Coath
- Rheumatology Department, Mid and South Essex University Hospitals NHS Foundation Trust, Southend University Hospital, Westcliff-on-sea, UK
| | - Daniel Robbins
- Medical Technology Research Centre, School of Allied Health, Anglia Ruskin University, Chelmsford, UK
| | - Barbara Pierscionek
- Faculty of Health Education Medicine and Social Care, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford Campus, Chelmsford, UK
| | - Christian Dejaco
- Department of Rheumatology, Hospital of Bruneck (ASAA-SABES), Teaching Hospital of the Paracelsus Medical University, Bruneck, Italy
- Department of Rheumatology and Immunology, Medical University of Graz, Graz, Austria
| | - Eric Matteson
- Division of Rheumatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Yannick van Sleen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Bhaskar Dasgupta
- Rheumatology Department, Mid and South Essex University Hospitals NHS Foundation Trust, Southend University Hospital, Westcliff-on-sea, UK.
| |
Collapse
|
14
|
Gollmann-Tepeköylü C, Graber M, Hirsch J, Mair S, Naschberger A, Pölzl L, Nägele F, Kirchmair E, Degenhart G, Demetz E, Hilbe R, Chen HY, Engert JC, Böhm A, Franz N, Lobenwein D, Lener D, Fuchs C, Weihs A, Töchterle S, Vogel GF, Schweiger V, Eder J, Pietschmann P, Seifert M, Kronenberg F, Coassin S, Blumer M, Hackl H, Meyer D, Feuchtner G, Kirchmair R, Troppmair J, Krane M, Weiss G, Tsimikas S, Thanassoulis G, Grimm M, Rupp B, Huber LA, Zhang SY, Casanova JL, Tancevski I, Holfeld J. Toll-Like Receptor 3 Mediates Aortic Stenosis Through a Conserved Mechanism of Calcification. Circulation 2023; 147:1518-1533. [PMID: 37013819 PMCID: PMC10192061 DOI: 10.1161/circulationaha.122.063481] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/08/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is characterized by a phenotypic switch of valvular interstitial cells to bone-forming cells. Toll-like receptors (TLRs) are evolutionarily conserved pattern recognition receptors at the interface between innate immunity and tissue repair. Type I interferons (IFNs) are not only crucial for an adequate antiviral response but also implicated in bone formation. We hypothesized that the accumulation of endogenous TLR3 ligands in the valvular leaflets may promote the generation of osteoblast-like cells through enhanced type I IFN signaling. METHODS Human valvular interstitial cells isolated from aortic valves were challenged with mechanical strain or synthetic TLR3 agonists and analyzed for bone formation, gene expression profiles, and IFN signaling pathways. Different inhibitors were used to delineate the engaged signaling pathways. Moreover, we screened a variety of potential lipids and proteoglycans known to accumulate in CAVD lesions as potential TLR3 ligands. Ligand-receptor interactions were characterized by in silico modeling and verified through immunoprecipitation experiments. Biglycan (Bgn), Tlr3, and IFN-α/β receptor alpha chain (Ifnar1)-deficient mice and a specific zebrafish model were used to study the implication of the biglycan (BGN)-TLR3-IFN axis in both CAVD and bone formation in vivo. Two large-scale cohorts (GERA [Genetic Epidemiology Research on Adult Health and Aging], n=55 192 with 3469 aortic stenosis cases; UK Biobank, n=257 231 with 2213 aortic stenosis cases) were examined for genetic variation at genes implicated in BGN-TLR3-IFN signaling associating with CAVD in humans. RESULTS Here, we identify TLR3 as a central molecular regulator of calcification in valvular interstitial cells and unravel BGN as a new endogenous agonist of TLR3. Posttranslational BGN maturation by xylosyltransferase 1 (XYLT1) is required for TLR3 activation. Moreover, BGN induces the transdifferentiation of valvular interstitial cells into bone-forming osteoblasts through the TLR3-dependent induction of type I IFNs. It is intriguing that Bgn-/-, Tlr3-/-, and Ifnar1-/- mice are protected against CAVD and display impaired bone formation. Meta-analysis of 2 large-scale cohorts with >300 000 individuals reveals that genetic variation at loci relevant to the XYLT1-BGN-TLR3-interferon-α/β receptor alpha chain (IFNAR) 1 pathway is associated with CAVD in humans. CONCLUSIONS This study identifies the BGN-TLR3-IFNAR1 axis as an evolutionarily conserved pathway governing calcification of the aortic valve and reveals a potential therapeutic target to prevent CAVD.
Collapse
Affiliation(s)
| | - Michael Graber
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob Hirsch
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Sophia Mair
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Naschberger
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955, Saudi Arabia
| | - Leo Pölzl
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Nägele
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Elke Kirchmair
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerald Degenhart
- Department of Radiology, Core Facility for Micro-CT, Medical University of Innsbruck, Innsbruck, Austria
| | - Egon Demetz
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Hao-Yu Chen
- Preventive and Genomic Cardiology, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - James C. Engert
- Preventive and Genomic Cardiology, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Anna Böhm
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Nadja Franz
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Lobenwein
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Lener
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Christiane Fuchs
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
| | - Anna Weihs
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
| | - Sonja Töchterle
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Georg F. Vogel
- Department of Pediatrics/Institute of Cell biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Victor Schweiger
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Jonas Eder
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Pietschmann
- Division of Cellular and Molecular Pathophysiology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Markus Seifert
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Blumer
- Institute of Clinical and Functional Anatomy, Innsbruck Medical University, Innsbruck, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Dirk Meyer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Gudrun Feuchtner
- Department of Radiology, Core Facility for Micro-CT, Medical University of Innsbruck, Innsbruck, Austria
| | - Rudolf Kirchmair
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob Troppmair
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, University of Innsbruck, Innsbruck, Innsbruck, Austria
| | - Markus Krane
- Department of Cardiovascular Surgery, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Günther Weiss
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Sotirios Tsimikas
- Division of Cardiovascular Diseases, University of California, San Diego, La Jolla, USA
| | - George Thanassoulis
- Preventive and Genomic Cardiology, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Michael Grimm
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Bernhard Rupp
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas A. Huber
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
- Austrian Drug Screening Institute, ADSI, Innsbruck, Austria
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
| | - Ivan Tancevski
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Holfeld
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Chu CQ. Animal models for large vessel vasculitis - The unmet need. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2023; 4:4-10. [PMID: 37138652 PMCID: PMC10150876 DOI: 10.2478/rir-2023-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 02/19/2023] [Indexed: 05/05/2023]
Abstract
Our understanding of the pathogenesis of large vessel vasculitis (LVV) are mainly achieved by studying the arteries taken from temporal artery biopsy in giant cell arteries (GCA) or surgical or autopsy specimens in Takayasu arteritis (TAK). These artery specimens provide invaluable information about pathological changes in these conditions that GCA and TAK are similar but are distinctly different in immune cell infiltrate and distribution of inflammatory cells in anatomical locations. However, these specimens of established arteritis do not provide information of the arteritis initiation and early events which are impossible to obtain in human artery specimens. Animal models for LVV are needed but not available. Here, several approaches are proposed for experimentation to generate animal models to aid in delineating the interaction of immune reaction with arterial wall components.
Collapse
Affiliation(s)
- Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, Oregon97239, USA
- Innovent Biologics (USA), Rockville, Maryland20850, USA
| |
Collapse
|
16
|
Monocyte subsets and monocyte-related chemokines in Takayasu arteritis. Sci Rep 2023; 13:2092. [PMID: 36746990 PMCID: PMC9902560 DOI: 10.1038/s41598-023-29369-3] [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: 10/17/2022] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of Takayasu arteritis (TAK) is poorly understood and no previous studies have analyzed monocytes in TAK. This study evaluated monocyte subsets and monocyte-related chemokines in the peripheral blood of TAK patients and healthy controls (HC). Monocyte subsets were identified as classical (CD14+CD16-), intermediate (CD14+CD16dim), and non-classical (CD14dimCD16high) in the peripheral blood. The chemokines CCL (C-C chemokine ligand)2, CCL3, CCL4, CCL5, CCL7, CXCL (C-X-C motif ligand)10, and CX3CL (C-X3-C motif ligand)1 were measured in the sera. Thirty-two TAK patients and 30 HC were evaluated. Intermediate monocytes were higher in TAK than HC [25.0 cells ×106/L (16.7-52.0) vs. 17.2 cells ×106/L (9.2-25.3); p = 0.014]. Active disease was associated with monocytosis (p = 0.004), increased classical (p = 0.003), and intermediate (p < 0.001) subsets than HC. Prednisone reduced the percentage of non-classical monocytes (p = 0.011). TAK patients had lower CCL3 (p = 0.033) and CCL4 (p = 0.023) levels than HC, whereas CCL22 levels were higher in active TAK compared to the remission state (p = 0.008). Glucocorticoids were associated with lower CXCL10 levels (p = 0.012). In TAK, CCL4 correlated with total (Rho = 0.489; p = 0.005), classical and intermediate monocytes (Rho = 0.448; p = 0.010 and Rho = 0.412; p = 0.019). In conclusion, TAK is associated with altered counts of monocyte subsets in the peripheral blood compared to HC and CCL22 is the chemokine with the strongest association with active disease in TAK.
Collapse
|
17
|
Yang M, Xu J, Zhang F, Luo P, Xu K, Feng R, Xu P. Large-Scale Genetic Correlation Analysis between Spondyloarthritis and Human Blood Metabolites. J Clin Med 2023; 12:jcm12031201. [PMID: 36769847 PMCID: PMC9917834 DOI: 10.3390/jcm12031201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/17/2023] [Accepted: 01/22/2023] [Indexed: 02/05/2023] Open
Abstract
The aim was to study the genetic correlation and causal relationship between spondyloarthritis (SpA) and blood metabolites based on the large-scale genome-wide association study (GWAS) summary data. The GWAS summary data (3966 SpA and 448,298 control cases) of SpA were from the UK Biobank, and the GWAS summary data (486 blood metabolites) of human blood metabolites were from a published study. First, the genetic correlation between SpA and blood metabolites was analyzed by linkage disequilibrium score (LDSC) regression. Next, we used Mendelian randomization (MR) analysis to perform access causal relationship between SpA and blood metabolites. Random effects inverse variance weighted (IVW) was the main analysis method, and the MR Egger, weighted median, simple mode, and weighted mode were supplementary methods. The MR analysis results were dominated by the random effects IVW. The Cochran's Q statistic (MR-IVW) and Rucker's Q statistic (MR Egger) were used to check heterogeneity. MR Egger and MR pleiotropy residual sum and outlier (MR-PRESSO) were used to check horizontal pleiotropy. The MR-PRESSO was also used to check outliers. The "leave-one-out" analysis was used to assess whether the MR analysis results were affected by a single SNP and thus test the robustness of the MR results. Finally, we identified seven blood metabolites that are genetically related to SpA: X-10395 (correlation coefficient = -0.546, p = 0.025), pantothenate (correlation coefficient = -0.565, p = 0.038), caprylate (correlation coefficient = -0.333, p = 0.037), pelargonate (correlation coefficient = -0.339, p = 0.047), X-11317 (correlation coefficient = -0.350, p = 0.038), X-12510 (correlation coefficient = -0.399, p = 0.034), and X-13859 (Correlation coefficient = -0.458, p = 0.015). Among them, X-10395 had a positive genetic causal relationship with SpA (p = 0.014, OR = 1.011). The blood metabolites that have genetic correlation and causal relationship with SpA found in this study provide a new idea for the study of the pathogenesis of SpA and the determination of diagnostic indicators.
Collapse
Affiliation(s)
- Mingyi Yang
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Jiawen Xu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Pan Luo
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Ruoyang Feng
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Correspondence:
| |
Collapse
|
18
|
Abstract
Giant cell arteritis is an autoimmune disease of medium and large arteries, characterized by granulomatous inflammation of the three-layered vessel wall that results in vaso-occlusion, wall dissection, and aneurysm formation. The immunopathogenesis of giant cell arteritis is an accumulative process in which a prolonged asymptomatic period is followed by uncontrolled innate immunity, a breakdown in self-tolerance, the transition of autoimmunity from the periphery into the vessel wall and, eventually, the progressive evolution of vessel wall inflammation. Each of the steps in pathogenesis corresponds to specific immuno-phenotypes that provide mechanistic insights into how the immune system attacks and damages blood vessels. Clinically evident disease begins with inappropriate activation of myeloid cells triggering the release of hepatic acute phase proteins and inducing extravascular manifestations, such as muscle pains and stiffness diagnosed as polymyalgia rheumatica. Loss of self-tolerance in the adaptive immune system is linked to aberrant signaling in the NOTCH pathway, leading to expansion of NOTCH1+CD4+ T cells and the functional decline of NOTCH4+ T regulatory cells (Checkpoint 1). A defect in the endothelial cell barrier of adventitial vasa vasorum networks marks Checkpoint 2; the invasion of monocytes, macrophages and T cells into the arterial wall. Due to the failure of the immuno-inhibitory PD-1 (programmed cell death protein 1)/PD-L1 (programmed cell death ligand 1) pathway, wall-infiltrating immune cells arrive in a permissive tissues microenvironment, where multiple T cell effector lineages thrive, shift toward high glycolytic activity, and support the development of tissue-damaging macrophages, including multinucleated giant cells (Checkpoint 3). Eventually, the vascular lesions are occupied by self-renewing T cells that provide autonomy to the disease process and limit the therapeutic effectiveness of currently used immunosuppressants. The multi-step process deviating protective to pathogenic immunity offers an array of interception points that provide opportunities for the prevention and therapeutic management of this devastating autoimmune disease.
Collapse
Affiliation(s)
- Cornelia M. Weyand
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
- Department of Cardiovascular Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, USA
- Department of Immunology, Mayo Clinic College of Medicine and Science
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306
| | - Jörg J. Goronzy
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic College of Medicine and Science
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306
| |
Collapse
|
19
|
Weyand CM. Large Vessel Vasculitides. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00060-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
20
|
Aymonnier K, Amsler J, Lamprecht P, Salama A, Witko‐Sarsat V. The neutrophil: A key resourceful agent in immune‐mediated vasculitis. Immunol Rev 2022; 314:326-356. [PMID: 36408947 DOI: 10.1111/imr.13170] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The term "vasculitis" refers to a group of rare immune-mediated diseases characterized by the dysregulated immune system attacking blood vessels located in any organ of the body, including the skin, lungs, and kidneys. Vasculitides are classified according to the size of the vessel that is affected. Although this observation is not specific to small-, medium-, or large-vessel vasculitides, patients show a high circulating neutrophil-to-lymphocyte ratio, suggesting the direct or indirect involvement of neutrophils in these diseases. As first responders to infection or inflammation, neutrophils release cytotoxic mediators, including reactive oxygen species, proteases, and neutrophil extracellular traps. If not controlled, this dangerous arsenal can injure the vascular system, which acts as the main transport route for neutrophils, thereby amplifying the initial inflammatory stimulus and the recruitment of immune cells. This review highlights the ability of neutrophils to "set the tone" for immune cells and other cells in the vessel wall. Considering both their long-established and newly described roles, we extend their functions far beyond their direct host-damaging potential. We also review the roles of neutrophils in various types of primary vasculitis, including immune complex vasculitis, anti-neutrophil cytoplasmic antibody-associated vasculitis, polyarteritis nodosa, Kawasaki disease, giant cell arteritis, Takayasu arteritis, and Behçet's disease.
Collapse
Affiliation(s)
- Karen Aymonnier
- INSERM U1016, Institut Cochin, Université Paris Cité, CNRS 8104 Paris France
| | - Jennifer Amsler
- INSERM U1016, Institut Cochin, Université Paris Cité, CNRS 8104 Paris France
| | - Peter Lamprecht
- Department of Rheumatology and Clinical Immunology University of Lübeck Lübeck Germany
| | - Alan Salama
- Department of Renal Medicine, Royal Free Hospital University College London London UK
| | | |
Collapse
|
21
|
Liegise A, Joseph G, Varghese M, Thomson V. Spontaneous delayed expansion of self-expanding stents in stenotic arteries in Takayasu arteritis. Diagn Interv Radiol 2022; 28:586-592. [PMID: 36550759 PMCID: PMC9885721 DOI: 10.5152/dir.2022.20940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Despite frequent use of self-expanding stents (SES) in treating obstructive arterial lesions in Takayasu arteritis (TA), spontaneous delayed stent expansion (SDSE) in TA remains unstudied. This study aimed primarily to document and quantify SDSE and secondarily to determine factors that might be associated with this process. METHODS Consecutive TA patients with obstructive arterial lesions undergoing routine percutaneous intervention involving SES use (sized 1:1 with normal vessel diameter but dilated only to 4mm/5mm) were recruited prospectively. Final stent diameters obtained were measured at 1cm intervals along the length of the stent using fluoroscopic images and an indwelling marker catheter. At angiographic follow-up, stent diameters were measured again in identical fashion. Interval change in stent diameter at each point was averaged for each stent. In a small sub-study intravascular ultrasound was used at follow-up to obtain potential mechanistic insights. RESULTS Seventeen TA patients (age 33 ± 13 years, 15 female) had 22 arterial obstructive lesions (16 occlusions, 18 subclavian) treated with one SES each. Follow-up obtained in all patients after 8.7 ± 3.8 months (range 3-18 months) showed interval increase in mean stent diameter of 1.6 ± 0.5 mm, range 0.7-2.8 mm (P < 0.001); 36% of stents achieved 100% of the nominal diameter at follow-up, while 90% of stents achieved ≥90%. The degree of SDSE did not correlate with the segment of artery stented or with TA disease activity at baseline. Intravascular ultrasound in four lesions showed that SDSE was associated with positive medial-adventitial remodelling and that neointimal hyperplasia occurs concurrently, causing in-stent luminal narrowing. CONCLUSION SDSE, to diameters equal or close to nominal, occurs in all stenotic TA lesions treated with SES. The degree of SDSE does not correlate with the segment of artery stented or with TA disease activity at baseline. Preliminary results suggest that the mechanism by which SDSE is accommodated by the arterial wall is by positive medial-adventitial remodelling.
Collapse
Affiliation(s)
- Avitso Liegise
- Department of Cardiology, Christian Medical College, Vellore, India
| | - George Joseph
- Department of Cardiology, Christian Medical College, Vellore, India
| | - Mithun Varghese
- Department of Cardiology, Christian Medical College, Vellore, India
| | - Viji Thomson
- Department of Cardiology, Christian Medical College, Vellore, India
| |
Collapse
|
22
|
Liu L, Chen J, Li J, Yang Y, Zeng X, Tian X. Whole Exome Sequencing Revealed Variants That Predict Pulmonary Artery Involvement in Patients with Takayasu Arteritis. J Inflamm Res 2022; 15:4817-4831. [PMID: 36046661 PMCID: PMC9420927 DOI: 10.2147/jir.s377402] [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: 06/13/2022] [Accepted: 08/06/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To conduct the first whole exome sequencing (WES) on Takayasu arteritis (TAK) to identify common and rare variants responsible for disease susceptibility. Patients and Methods A total of 200 patients and 1675 healthy controls from China were recruited for this study. Site-based association analysis for common variants and gene-based burden analysis for rare variants were conducted. A weighted genetic risk score (wGRS) was calculated for each patient with TAK based on the independent risk alleles identified in the association analyses. The ability of the patient wGRS to discriminate between different phenotypes was evaluated. Results In the site-based analysis, the top association signal was CCHCR1 (rs1265067, p = 8.27 × 10-12, OR = 2.41), a proxy for HLA-B*52:01. HLA-DQB1 (rs9273902), HLA-DQB2 (rs34109750), and a haplotype block in the human leukocyte antigen (HLA) class III region (represented by rs3130618) also exhibited significant associations independently. In addition, four novel non-HLA susceptibility loci were identified: PRRT4, TLL2, LRP1B, and DLGAP2. Twelve independently associated single nucleotide polymorphisms were used to calculate the wGRS. TAK patients with a higher wGRS were found to have an increased risk of pulmonary artery involvement compared with those with a lower wGRS (p = 5.76 × 10-7, OR = 13.92). The wGRS algorithm showed good predictive capability for pulmonary artery involvement in TAK (sensitivity, 92.1%; specificity, 59.9%). In the gene-based analysis, risk genes that reached exome-wide significance were not identified. Conclusion This WES study on TAK supports a previously reported association within the HLA region. Moreover, novel susceptibility loci were identified outside the HLA region. These risk alleles showed potential associations with pulmonary artery involvement in TAK. However, additional studies are warranted to verify our findings.
Collapse
Affiliation(s)
- Lingyu Liu
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, People's Republic of China
| | - Jing Chen
- Department of Rheumatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, People's Republic of China
| | - Yunjiao Yang
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, People's Republic of China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, People's Republic of China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, People's Republic of China
| |
Collapse
|
23
|
Cen X, Wang B, Liang Y, Chen Y, Xiao Y, Du S, Nandakumar KS, Yin H, Liu S, Cheng K. Small molecule SMU-CX24 targeting toll-like receptor 3 counteracts inflammation: A novel approach to atherosclerosis therapy. Acta Pharm Sin B 2022; 12:3667-3681. [PMID: 36176917 PMCID: PMC9513496 DOI: 10.1016/j.apsb.2022.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/15/2022] [Accepted: 05/26/2022] [Indexed: 12/03/2022] Open
Abstract
Toll-like receptor 3 (TLR3), as an important pattern recognition receptor (PRR), dominates the innate and adaptive immunity regulating many acute and chronic inflammatory diseases. Atherosclerosis is proved as an inflammatory disease, and inflammatory events involved in the entire process of initiation and deterioration. However, the contribution of TLR3 to atherosclerosis remains unclear. Herein, we identified the clinical relevance of TLR3 upregulation and disease processes in human atherosclerosis. Besides, activation of TLR3 also directly led to significant expression of atherogenic chemokines and adhesion molecules. Conversely, silencing TLR3 inhibited the uptake of oxLDL by macrophages and significantly reduced foam cell formation. Given the aberrance in TLR3 functions on atherosclerosis progression, we hypothesized that TLR3 could serve as novel target for clinical atherosclerosis therapy. Therefore, we developed the novel ellipticine derivative SMU-CX24, which specifically inhibited TLR3 (IC50 = 18.87 ± 2.21 nmol/L). In vivo, atherosclerotic burden was alleviated in Western diet fed ApoE-/- mice in response to SMU-CX24 treatment, accompanying notable reductions in TLR3 expression and inflammation infiltration within atherosclerotic lesion. Thus, for the first time, we revealed that pharmacological downregulation of TLR3 with specific inhibitor regenerated inflammatory environment to counteract atherosclerosis progression, thereby proposing a new strategy and probe for atherosclerosis therapy.
Collapse
Affiliation(s)
- Xiaohong Cen
- Guangdong Provincial Key Laboratory of New Drug Screening and State Key Laboratory of Organ Failure Research, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Baoqu Wang
- Guangdong Provincial Key Laboratory of New Drug Screening and State Key Laboratory of Organ Failure Research, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuqing Liang
- Guangdong Provincial Key Laboratory of New Drug Screening and State Key Laboratory of Organ Failure Research, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanlin Chen
- Guangdong Provincial Key Laboratory of New Drug Screening and State Key Laboratory of Organ Failure Research, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yu Xiao
- Guangdong Provincial Key Laboratory of New Drug Screening and State Key Laboratory of Organ Failure Research, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shaohua Du
- Department of Musculoskeletal Oncology, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510642, China
| | - Kutty Selva Nandakumar
- Guangdong Provincial Key Laboratory of New Drug Screening and State Key Laboratory of Organ Failure Research, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hang Yin
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening and State Key Laboratory of Organ Failure Research, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Kui Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening and State Key Laboratory of Organ Failure Research, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Musculoskeletal Oncology, the Third Affiliated Hospital of Southern Medical University, Guangzhou 510642, China
| |
Collapse
|
24
|
Danda D, Manikuppam P, Tian X, Harigai M. Advances in Takayasu arteritis: An Asia Pacific perspective. Front Med (Lausanne) 2022; 9:952972. [PMID: 36045929 PMCID: PMC9423100 DOI: 10.3389/fmed.2022.952972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Takayasu Arteritis (TA) is a rare form of chronic granulomatous large vessel vasculitis that is more common in Asia compared to other parts of the world. There have been several developments in the field of Takayasu arteritis in relation to genetics, classification, clinical features, imaging, disease activity assessment and management and much of these works have been done in the Asia Pacific region. We will be discussing selected few in the current review.
Collapse
Affiliation(s)
- Debashish Danda
- Department of Clinical Immunology and Rheumatology, Christian Medical College and Hospital, Vellore, India
| | - Prathyusha Manikuppam
- Department of Clinical Immunology and Rheumatology, Christian Medical College and Hospital, Vellore, India
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Peking Union Medical College Hospital (PUMCH), Beijing, China
| | - Masayoshi Harigai
- Division of Epidemiology and Pharmacoepidemiology of Rheumatic Diseases, Institute of Rheumatology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
25
|
Rizzo C, La Barbera L, Miceli G, Tuttolomondo A, Guggino G. The innate face of Giant Cell Arteritis: Insight into cellular and molecular innate immunity pathways to unravel new possible biomarkers of disease. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:933161. [PMID: 39086970 PMCID: PMC11285707 DOI: 10.3389/fmmed.2022.933161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/11/2022] [Indexed: 08/02/2024]
Abstract
Giant cell arteritis (GCA) is an inflammatory chronic disease mainly occurring in elderly individuals. The pathogenesis of GCA is still far from being completely elucidated. However, in susceptible arteries, an aberrant immune system activation drives the occurrence of vascular remodeling which is mainly characterized by intimal hyperplasia and luminal obstruction. Vascular damage leads to ischemic manifestations involving extra-cranial branches of carotid arteries, mostly temporal arteries, and aorta. Classically, GCA was considered a pathological process resulting from the interaction between an unknown environmental trigger, such as an infectious agent, with local dendritic cells (DCs), activated CD4 T cells and effector macrophages. In the last years, the complexity of GCA has been underlined by robust evidence suggesting that several cell subsets belonging to the innate immunity can contribute to disease development and progression. Specifically, a role in driving tissue damage and adaptive immunity activation was described for dendritic cells (DCs), monocytes and macrophages, mast cells, neutrophils and wall components, such as endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). In this regard, molecular pathways related to cytokines, chemokines, growth factors, vasoactive molecules and reactive oxygen species may contribute to the inflammatory process underlying GCA. Altogether, innate cellular and molecular pathways may clarify many pathogenetic aspects of the disease, paving the way for the identification of new biomarkers and for the development of new treatment targets for GCA. This review aims to deeply dissect past and new evidence on the innate immunological disruption behind GCA providing a comprehensive description of disease development from the innate perspective.
Collapse
Affiliation(s)
- Chiara Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Palermo, Italy
| | - Lidia La Barbera
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Palermo, Italy
| | - Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Unit of Internal Medicine and Stroke Care, University of Palermo, Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Unit of Internal Medicine and Stroke Care, University of Palermo, Palermo, Italy
| | - Giuliana Guggino
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Palermo, Italy
| |
Collapse
|
26
|
Zhou H, Wang L, Liu S, Wang W. The role of phosphoinositide 3-kinases in immune-inflammatory responses: potential therapeutic targets for abdominal aortic aneurysm. Cell Cycle 2022; 21:2339-2364. [PMID: 35792922 DOI: 10.1080/15384101.2022.2094577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm (AAA) includes inflammatory responses, matrix metalloproteinases (MMPs) degradation, VSMC apoptosis, oxidative stress, and angiogenesis, among which the inflammatory response plays a key role. At present, surgery is the only curing treatment, and no effective drug can delay AAA progression in clinical practice. Therefore, searching for a signaling pathway related to the immune-inflammatory response is an essential direction for developing drugs targeting AAA. Recent studies have confirmed that the PI3K family plays an important role in many inflammatory diseases and is involved in regulating various cellular functions, especially in the immune-inflammatory response. This review focuses on the role of each isoform of PI3K in each stage of AAA immune-inflammatory response, making available explorations for a deeper understanding of the mechanism of inflammation and immune response during the formation and development of AAA.
Collapse
Affiliation(s)
- Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
27
|
The Yin and Yang of toll-like receptors in endothelial dysfunction. Int Immunopharmacol 2022; 108:108768. [DOI: 10.1016/j.intimp.2022.108768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
|
28
|
Zhuang C, Chen R, Zheng Z, Lu J, Hong C. Toll-Like Receptor 3 in Cardiovascular Diseases. Heart Lung Circ 2022; 31:e93-e109. [PMID: 35367134 DOI: 10.1016/j.hlc.2022.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023]
Abstract
Toll-like receptor 3 (TLR3) is an important member of the innate immune response receptor toll-like receptors (TLRs) family, which plays a vital role in regulating immune response, promoting the maturation and differentiation of immune cells, and participating in the response of pro-inflammatory factors. TLR3 is activated by pathogen-associated molecular patterns and damage-associated molecular patterns, which support the pathophysiology of many diseases related to inflammation. An increasing number of studies have confirmed that TLR3, as a crucial medium of innate immunity, participates in the occurrence and development of cardiovascular diseases (CVDs) by regulating the transcription and translation of various cytokines, thus affecting the structure and physiological function of resident cells in the cardiovascular system, including vascular endothelial cells, vascular smooth muscle cells, cardiomyocytes, fibroblasts and macrophages. The dysfunction and structural damage of vascular endothelial cells and proliferation of vascular smooth muscle cells are the key factors in the occurrence of vascular diseases such as pulmonary arterial hypertension, atherosclerosis, myocardial hypertrophy, myocardial infarction, ischaemia/reperfusion injury, and heart failure. Meanwhile, cardiomyocytes, fibroblasts, and macrophages are involved in the development of CVDs. Therefore, the purpose of this review was to explore the latest research published on TLR3 in CVDs and discuss current understanding of potential mechanisms by which TLR3 contributes to CVDs. Even though TLR3 is a developing area, it has strong treatment potential as an immunomodulator and deserves further study for clinical translation.
Collapse
Affiliation(s)
- Chunying Zhuang
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; First Clinical School, Guangzhou Medical University, Guangzhou, China
| | - Riken Chen
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenzhen Zheng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Guangzhou, China
| | - Jianmin Lu
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cheng Hong
- China State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
29
|
Watanabe R, Hashimoto M. Vasculitogenic T Cells in Large Vessel Vasculitis. Front Immunol 2022; 13:923582. [PMID: 35784327 PMCID: PMC9240193 DOI: 10.3389/fimmu.2022.923582] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Vasculitis is an autoimmune disease of unknown etiology that causes inflammation of the blood vessels. Large vessel vasculitis is classified as either giant cell arteritis (GCA), which occurs exclusively in the elderly, or Takayasu arteritis (TAK), which mainly affects young women. Various cell types are involved in the pathogenesis of large vessel vasculitis. Among these, dendritic cells located between the adventitia and the media initiate the inflammatory cascade as antigen-presenting cells, followed by activation of macrophages and T cells contributing to vessel wall destruction. In both diseases, naive CD4+ T cells are polarized to differentiate into Th1 or Th17 cells, whereas differentiation into regulatory T cells, which suppress vascular inflammation, is inhibited. Skewed T cell differentiation is the result of aberrant intracellular signaling, such as the mechanistic target of rapamycin (mTOR) or the Janus kinase signal transducer and activator of transcription (JAK-STAT) pathways. It has also become clear that tissue niches in the vasculature fuel activated T cells and maintain tissue-resident memory T cells. In this review, we outline the most recent understanding of the pathophysiology of large vessel vasculitis. Then, we provide a summary of skewed T cell differentiation in the vasculature and peripheral blood. Finally, new therapeutic strategies for correcting skewed T cell differentiation as well as aberrant intracellular signaling are discussed.
Collapse
|
30
|
New Insights into the Pathogenesis of Giant Cell Arteritis: Mechanisms Involved in Maintaining Vascular Inflammation. J Clin Med 2022; 11:jcm11102905. [PMID: 35629030 PMCID: PMC9143803 DOI: 10.3390/jcm11102905] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
The giant cell arteritis (GCA) pathophysiology is complex and multifactorial, involving a predisposing genetic background, the role of immune aging and the activation of vascular dendritic cells by an unknown trigger. Once activated, dendritic cells recruit CD4 T cells and induce their activation, proliferation and polarization into Th1 and Th17, which produce interferon-gamma (IFN-γ) and interleukin-17 (IL-17), respectively. IFN-γ triggers the production of chemokines by vascular smooth muscle cells, which leads to the recruitment of additional CD4 and CD8 T cells and also monocytes that differentiate into macrophages. Recent data have shown that IL-17, IFN-γ and GM-CSF induce the differentiation of macrophage subpopulations, which play a role in the destruction of the arterial wall, in neoangiogenesis or intimal hyperplasia. Under the influence of different mediators, mainly endothelin-1 and PDGF, vascular smooth muscle cells migrate to the intima, proliferate and change their phenotype to become myofibroblasts that further proliferate and produce extracellular matrix proteins, increasing the vascular stenosis. In addition, several defects in the immune regulatory mechanisms probably contribute to chronic vascular inflammation in GCA: a defect in the PD-1/PD-L1 pathway, a quantitative and qualitative Treg deficiency, the implication of resident cells, the role of GM-CSF and IL-6, the implication of the NOTCH pathway and the role of mucosal‑associated invariant T cells and tissue‑resident memory T cells.
Collapse
|
31
|
Li Y, Ou K, Wang Y, Luo L, Chen Z, Wu J. TLR9 agonist suppresses choroidal neovascularization by restricting endothelial cell motility via ERK/c-Jun pathway. Microvasc Res 2022; 141:104338. [PMID: 35150733 DOI: 10.1016/j.mvr.2022.104338] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/08/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Choroidal neovascularization (CNV) is the feature of neovascular age-related macular degeneration (AMD). It has been demonstrated that inflammation plays a key role in the development of CNV. Here we aim to investigate how TLR9 agonist (CpG-ODN), one of the key regulators of inflammatory responses, suppresses CNV in vivo. MATERIALS AND METHODS The cell viability was assessed by MTT and EdU test after CpG-ODN treatment. Endothelial cells gap assay, tube formation assay and transwell assay were practiced to observe how CpG-ODN affected the endothelial cells functions. The choroidal explants and laser-induced CNV model were built to investigate how CpG-ODN suppressed angiogenesis. The ERK and c-Jun expression were evaluated to assess if CpG-ODN affected cell proliferation. Flow cytometry and qPCR was practiced to observe how CpG-ODN regulated cell proliferation. RESULTS Our data showed that CpG-ODN not only reduced CNV area in vivo, but also decreased the RPE damage. CpG-ODN inhibited endothelial cells from migration and forming tubes, while the effect was not toxic. EdU test and MTT test suggested that CpG-ODN inhibited endothelial cells proliferation. CpG-ODN significantly increased protein expression of phosphorylated c-Jun but reduced phosphorylated ERK in HUVECs, which was confirmed in ERK transfected 293T cells. JNK inhibitor abolished the suppression of endothelial cells migration and tube formation by CpG-ODN. The findings were also in agreement with the observation in CpG-ODN treated CNV eyes in vivo. The flow cytometry and qPCR data revealed that the suppression of cell motility by CpG-ODN was achieved by arresting endothelial cells cell cycle at G0/G1 phase. CONCLUSIONS Our study demonstrated that CpG-ODN suppressed endothelial cell motility by restricting the cell cycle progression at G0/G1 phase, the effect of which was achieved by interacting with ERK/c-Jun pathways.
Collapse
Affiliation(s)
- Youjian Li
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Kepeng Ou
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Yuwei Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Liying Luo
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Zhongzhu Chen
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, IATTI, Chongqing University of Arts and Sciences, Chongqing, China
| | - Jiahui Wu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.
| |
Collapse
|
32
|
Pugh D, Karabayas M, Basu N, Cid MC, Goel R, Goodyear CS, Grayson PC, McAdoo SP, Mason JC, Owen C, Weyand CM, Youngstein T, Dhaun N. Large-vessel vasculitis. Nat Rev Dis Primers 2022; 7:93. [PMID: 34992251 PMCID: PMC9115766 DOI: 10.1038/s41572-021-00327-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 02/08/2023]
Abstract
Large-vessel vasculitis (LVV) manifests as inflammation of the aorta and its major branches and is the most common primary vasculitis in adults. LVV comprises two distinct conditions, giant cell arteritis and Takayasu arteritis, although the phenotypic spectrum of primary LVV is complex. Non-specific symptoms often predominate and so patients with LVV present to a range of health-care providers and settings. Rapid diagnosis, specialist referral and early treatment are key to good patient outcomes. Unfortunately, disease relapse remains common and chronic vascular complications are a source of considerable morbidity. Although accurate monitoring of disease activity is challenging, progress in vascular imaging techniques and the measurement of laboratory biomarkers may facilitate better matching of treatment intensity with disease activity. Further, advances in our understanding of disease pathophysiology have paved the way for novel biologic treatments that target important mediators of disease in both giant cell arteritis and Takayasu arteritis. This work has highlighted the substantial heterogeneity present within LVV and the importance of an individualized therapeutic approach. Future work will focus on understanding the mechanisms of persisting vascular inflammation, which will inform the development of increasingly sophisticated imaging technologies. Together, these will enable better disease prognostication, limit treatment-associated adverse effects, and facilitate targeted development and use of novel therapies.
Collapse
Affiliation(s)
- Dan Pugh
- British Hearth Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Maira Karabayas
- Centre for Arthritis & Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Neil Basu
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Maria C Cid
- Department of Autoimmune Diseases, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Ruchika Goel
- Department of Clinical Immunology & Rheumatology, Christian Medical College, Vellore, India
| | - Carl S Goodyear
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Peter C Grayson
- National Institute of Arthritis & Musculoskeletal & Skin Diseases, National Institutes of Health, Bethesda, MA, USA
| | - Stephen P McAdoo
- Department of Immunology & Inflammation, Imperial College London, London, UK
| | - Justin C Mason
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | - Cornelia M Weyand
- Centre for Translational Medicine, Stanford University, Stanford, California, USA
| | - Taryn Youngstein
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Neeraj Dhaun
- British Hearth Foundation/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
33
|
The Immunogenetics of Vasculitis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:299-334. [DOI: 10.1007/978-3-030-92616-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
34
|
Abstract
Two vasculitides, giant cell arteritis (GCA) and Takayasu arteritis (TAK), are recognized as autoimmune and autoinflammatory diseases that manifest exclusively within the aorta and its large branches. In both entities, the age of the affected host is a critical risk factor. TAK manifests during the 2nd-4th decade of life, occurring while the immune system is at its height of performance. GCA is a disease of older individuals, with infrequent cases during the 6th decade and peak incidence during the 8th decade of life. In both vasculitides, macrophages and T cells infiltrate into the adventitia and media of affected vessels, induce granulomatous inflammation, cause vessel wall destruction, and reprogram vascular cells to drive adventitial and neointimal expansion. In GCA, abnormal immunity originates in an aged immune system and evolves within the aged vascular microenvironment. One hallmark of the aging immune system is the preferential loss of CD8+ T cell function. Accordingly, in GCA but not in TAK, CD8+ effector T cells play a negligible role and anti-inflammatory CD8+ T regulatory cells are selectively impaired. Here, we review current evidence of how the process of immunosenescence impacts the risk for GCA and how fundamental differences in the age of the immune system translate into differences in the granulomatous immunopathology of TAK versus GCA.
Collapse
|
35
|
Moll M, Reichel K, Nurjadi D, Förmer S, Krall LJ, Heeg K, Hildebrand D. Notch Ligand Delta-Like 1 Is Associated With Loss of Vascular Endothelial Barrier Function. Front Physiol 2021; 12:766713. [PMID: 34955884 PMCID: PMC8703021 DOI: 10.3389/fphys.2021.766713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
Vascular leakage associated with vascular endothelial cell (vEC) dysfunction is a hallmark of sepsis. Causative for the decreased integrity of the vascular endothelium (vE) is a complex concurrence of pathogen components, inflammation-associated host factors, and the interaction of vECs and activated circulating immune cells. One signaling pathway that regulates the integrity of the vE is the Notch cascade, which is activated through the binding of a Notch ligand to its respective Notch receptor. Recently, we showed that the soluble form of the Notch ligand Delta-like1 (sDLL1) is highly abundant in the blood of patients with sepsis. However, a direct connection between DLL1-activated Notch signaling and loss of vEC barrier function has not been addressed so far. To study the impact of infection-associated sDLL1, we used human umbilical vein cells (HUVEC) grown in a transwell system and cocultured with blood. Stimulation with sDLL1 induced activation as well as loss of endothelial tight structure and barrier function. Moreover, LPS-stimulated HUVEC activation and increase in endothelial cell permeability could be significantly decreased by blocking DLL1-receptor binding and Notch signaling, confirming the involvement of the cascade in LPS-mediated endothelial dysfunction. In conclusion, our results suggest that during bacterial infection and LPS recognition, DLL1-activated Notch signaling is associated with vascular permeability. This finding might be of clinical relevance in terms of preventing vascular leakage and the severity of sepsis.
Collapse
Affiliation(s)
- Maximilian Moll
- Medical Microbiology and Hygiene, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Konrad Reichel
- Medical Microbiology and Hygiene, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Dennis Nurjadi
- Medical Microbiology and Hygiene, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Sandra Förmer
- Medical Microbiology and Hygiene, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Lars Johannes Krall
- Medical Microbiology and Hygiene, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus Heeg
- Medical Microbiology and Hygiene, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Dagmar Hildebrand
- Medical Microbiology and Hygiene, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
36
|
Ciccia F, Macaluso F, Mauro D, Nicoletti GF, Croci S, Salvarani C. New insights into the pathogenesis of giant cell arteritis: are they relevant for precision medicine? THE LANCET. RHEUMATOLOGY 2021; 3:e874-e885. [PMID: 38287633 DOI: 10.1016/s2665-9913(21)00253-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/15/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
Giant cell arteritis is a primary granulomatous vasculitis characterised by a strict tissue tropism for large and medium-size vessels, occurring in people older than 50 years. Although considerable progress in understanding some of the pathophysiological mechanisms involved in the pathogenesis of giant cell arteritis has been made in the past 10 years, specific triggers of disease and mechanisms of chronic damage have not yet been identified. The definition of a specific pro-inflammatory hierarchy between the multiple cell types and the different cytokines or chemokines involved in the inflammatory process are still unexplored areas of study. The overall goal of precision medicine is to identify the best possible therapeutic approach for an individual or group of individuals with a given disease. The fundamental prerequisite of this approach is the identification, at baseline, of clinical and imaging findings and of molecular biomarkers that allow a precise stratification of patients and an adequate prediction of the therapeutic response. In this regard, the possibility of obtaining temporal artery biopsies for diagnostic purposes offers incredible exploratory possibilities to define different disease pathotypes potentially susceptible to different therapeutic interventions. In this Series paper, we will describe the most recent evidence relating to the pathogenesis of giant cell arteritis, trying to define, if possible, a new pathogenetic-centred approach to patients with giant cell arteritis.
Collapse
Affiliation(s)
- Francesco Ciccia
- Department of Precision Medicine, Section of Rheumatology, Università della Campania L Vanvitelli, Naples, Italy.
| | - Federica Macaluso
- Department of Precision Medicine, Section of Rheumatology, Università della Campania L Vanvitelli, Naples, Italy; Dipartimento Specialità Mediche, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Daniele Mauro
- Department of Precision Medicine, Section of Rheumatology, Università della Campania L Vanvitelli, Naples, Italy
| | - Giovanni Francesco Nicoletti
- Dipartimento Multidisciplinare di Specialità Medico-Chirurgiche e Odontoiatriche, Università della Campania L Vanvitelli, Naples, Italy
| | - Stefania Croci
- Autoimmunità, Allergologia e Biotecnologie Innovative, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carlo Salvarani
- Dipartimento Specialità Mediche, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy; Dipartimento Chirurgico, Medico, Odontoiatrico e di Scienze Morfologiche con interesse Trapiantologico, Oncologico e di Medicina Rigenerativa, Università di Modena e Reggio Emilia, Modena, Italy.
| |
Collapse
|
37
|
Tomas L, Prica F, Schulz C. Trafficking of Mononuclear Phagocytes in Healthy Arteries and Atherosclerosis. Front Immunol 2021; 12:718432. [PMID: 34759917 PMCID: PMC8573388 DOI: 10.3389/fimmu.2021.718432] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Monocytes and macrophages play essential roles in all stages of atherosclerosis – from early precursor lesions to advanced stages of the disease. Intima-resident macrophages are among the first cells to be confronted with the influx and retention of apolipoprotein B-containing lipoproteins at the onset of hypercholesterolemia and atherosclerosis development. In this review, we outline the trafficking of monocytes and macrophages in and out of the healthy aorta, as well as the adaptation of their migratory behaviour during hypercholesterolemia. Furthermore, we discuss the functional and ontogenetic composition of the aortic pool of mononuclear phagocytes and its link to the atherosclerotic disease process. The development of mouse models of atherosclerosis regression in recent years, has enabled scientists to investigate the behaviour of monocytes and macrophages during the resolution of atherosclerosis. Herein, we describe the dynamics of these mononuclear phagocytes upon cessation of hypercholesterolemia and how they contribute to the restoration of tissue homeostasis. The aim of this review is to provide an insight into the trafficking, fate and disease-relevant dynamics of monocytes and macrophages during atherosclerosis, and to highlight remaining questions. We focus on the results of rodent studies, as analysis of cellular fates requires experimental manipulations that cannot be performed in humans but point out findings that could be replicated in human tissues. Understanding of the biology of macrophages in atherosclerosis provides an important basis for the development of therapeutic strategies to limit lesion formation and promote plaque regression.
Collapse
Affiliation(s)
- Lukas Tomas
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Filip Prica
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Schulz
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
38
|
Functionally Heterogenous Macrophage Subsets in the Pathogenesis of Giant Cell Arteritis: Novel Targets for Disease Monitoring and Treatment. J Clin Med 2021; 10:jcm10214958. [PMID: 34768479 PMCID: PMC8585092 DOI: 10.3390/jcm10214958] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/20/2021] [Accepted: 10/23/2021] [Indexed: 12/19/2022] Open
Abstract
Giant cell arteritis (GCA) is a granulomatous large-vessel vasculitis that affects adults above 50 years of age. In GCA, circulating monocytes are recruited to the inflamed arteries. With cues from the vascular microenvironment, they differentiate into macrophages and play important roles in the pathogenesis of GCA via pro-inflammatory cytokine production and vascular remodeling. However, a deeper understanding of macrophage heterogeneity in GCA pathogenesis is needed to assist the development of novel diagnostic tools and targeted therapies. Here, we review the current knowledge on macrophage heterogeneity and diverse functions of macrophage subsets in the pathogenesis of GCA. We next discuss the possibility to exploit their heterogeneity as a source of novel biomarkers and as targets for nuclear imaging. Finally, we discuss novel macrophage-targeted therapies and future directions for targeting these cells in GCA.
Collapse
|
39
|
Wagner AD, Wittkop U, Thalmann J, Willmen T, Gödecke V, Hodam J, Ronicke S, Zenke M. Glucocorticoid Effects on Tissue Residing Immune Cells in Giant Cell Arteritis: Importance of GM-CSF. Front Med (Lausanne) 2021; 8:709404. [PMID: 34557501 PMCID: PMC8452956 DOI: 10.3389/fmed.2021.709404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/17/2021] [Indexed: 11/13/2022] Open
Abstract
Giant cell arteritis (GCA) is a systemic granulomatous vasculitis clinically characterized by a prompt response to glucocorticoid therapy. Dendritic cells (DCs) play a central role in the pathogenesis of the disease and are increased in temporal arteries from GCA patients. The aim of this study was to determine the effects of glucocorticoid therapy on granulomatous infiltrates and on peripheral DCs of GCA patients. Immunohistochemical staining of temporal artery specimens from 41 GCA patients revealed a rapid reduction of the number of DCs after initiation of glucocorticoid treatment. TUNEL staining was performed to quantify apoptotic S100+ DC, CD3+ T cells, and CD68+ macrophages in the granulomatous infiltrates. An increase of apoptotic cells up to 9 ± 2% after 4–5 days of glucocorticoid therapy and up to 27 ± 5% (p < 0.001, compared to earlier timepoints) after 6–10 days was detected. A decrease of CCL19 and CCL21 expression was observed after starting glucocorticoid therapy. Granulocyte-macrophage colony-stimulating factor (GM-CSF) expression also significantly decreased under glucocorticoid therapy. No GM-CSF expression was detected in the control specimens. Glucocorticoid therapy leads to a rapid, time-dependent reduction of DCs in temporal arteries from GCA patients and reduction of mediators for cell migration. Our data suggest GM-CSF as a novel therapeutic target of GCA.
Collapse
Affiliation(s)
- Annette D Wagner
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Ulrike Wittkop
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Jessica Thalmann
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Tina Willmen
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Vega Gödecke
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Justyna Hodam
- Deutsches Rotes Kreuz (DRK) Clinic for Child and Adolescent Psychiatry, Bad Neuenahr, Germany
| | - Simon Ronicke
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Martin Zenke
- Department of Cell Biology, Institute for Biomedical Engineering, Rheinisch-Westfälische Technische Hochschule Aachen (RWTH) Aachen University Medical School, Aachen, Germany.,Helmholtz Institute for Biomedical Engineering, Rheinisch-Westfälische Technische Hochschule Aachen (RWTH) Aachen University, Aachen, Germany
| |
Collapse
|
40
|
Carbon Monoxide-Releasing Molecule-2 Ameliorates Particulate Matter-Induced Aorta Inflammation via Toll-Like Receptor/NADPH Oxidase/ROS/NF- κB/IL-6 Inhibition. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2855042. [PMID: 34336088 PMCID: PMC8292097 DOI: 10.1155/2021/2855042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022]
Abstract
Particulate matter (PM), a major air pollutant, may be associated with adverse cardiovascular effects. Reactive oxygen species- (ROS-) dependent proinflammatory cytokine production, such as interleukin-6 (IL-6), is a possible underlying mechanism. Carbon monoxide- (CO-) releasing molecule-2 (CORM-2) which liberates exogenous CO can exert many beneficial effects, particularly anti-inflammation and antioxidant effects. The purpose of this study was to explore the protective effects and underpinning mechanisms of CORM-2 on PM-induced aorta inflammation. Here, human aortic vascular smooth muscle cells (HASMCs) were utilized as in vitro models for the assessment of signaling pathways behind CORM-2 activities against PM-induced inflammatory responses, including Toll-like receptors (TLRs), NADPH oxidase, ROS, nuclear factor-kappa B (NF-κB), and IL-6. The modulation of monocyte adherence and HASMC migration, that are two critical cellular events of inflammatory process, along with their regulators, including intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and matrix metalloproteinase-2 (MMP-2) and MMP-9, in response to PM by CORM-2, were further evaluated. Finally, mice experiments under different conditions were conducted for the in vivo evaluation of CORM-2 benefits on the expression of inflammatory molecules including IL-6, ICAM-1, VCAM-1, MMP-2, and MMP-9. Our results found that PM could induce aorta inflammation in vitro and in vivo, as evidenced by the increase of IL-6 expression that was regulated by the TLR2 and TLR4/NADPH oxidase/ROS/NF-κB signaling pathway, thereby promoting ICAM-1- and VCAM-1-dependent monocyte adhesion and MMP-2- and MMP-9-dependent HASMC migration. Importantly, our experimental models demonstrated that CORM-2-liberated CO effectively inhibited the whole identified PM-induced inflammatory cascade in HASMCs and tissues. In conclusion, CORM-2 treatment may elicit multiple beneficial effects on inflammatory responses of aorta due to PM exposure, thereby providing therapeutic value in the context of inflammatory diseases of the cardiovascular system.
Collapse
|
41
|
Matsumoto K, Suzuki K, Yoshimoto K, Ishigaki S, Yoshida H, Magi M, Matsumoto Y, Kaneko Y, Takeuchi T. Interleukin-1 pathway in active large vessel vasculitis patients with a poor prognosis: a longitudinal transcriptome analysis. Clin Transl Immunology 2021; 10:e1307. [PMID: 34249359 PMCID: PMC8251870 DOI: 10.1002/cti2.1307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/14/2021] [Accepted: 06/08/2021] [Indexed: 12/23/2022] Open
Abstract
Objectives Large vessel vasculitis (LVV) is characterised by a high relapse rate. Because accurate assessment of the LVV disease status can be difficult, an accurate prognostic marker for initial risk stratification is required. We conducted a comprehensive longitudinal investigation of next‐generation RNA‐sequencing data for patients with LVV to explore useful biomarkers associated with clinical characteristics. Methods Key molecular pathways relevant to LVV pathogenesis were identified by examining the whole blood RNA from patients with LVV and healthy controls (HCs). The data were examined by pathway analysis and weighted gene correlation network analysis (WGCNA) to identify functional gene sets that were differentially expressed between LVV patients and HCs, and associated with clinical features. We then compared the expression of the selected genes during week 0, week 6, remission and relapse. Results The whole‐transcriptome gene expression data for 108 samples obtained from LVV patients (n = 27) and HCs (n = 12) were compared. The pathway analysis and WGCNA revealed that molecular pathway related to interleukin (IL)‐1 was significantly upregulated in LVV patients compared with HCs, which correlated with the positron emission tomography vascular activity score, a disease‐extent score based on the distribution of affected arteries. Further analysis revealed that the expression levels of genes in the IL‐1 signalling pathway remained high after conventional treatment and were associated with disease relapse. Conclusion Upregulation of the IL‐1 signalling pathway was a characteristic of LVV patients and was associated with the extent of disease and a poor prognosis.
Collapse
Affiliation(s)
- Kotaro Matsumoto
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Katsuya Suzuki
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Keiko Yoshimoto
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Sho Ishigaki
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | | | - Mayu Magi
- Chugai Pharmaceutical Co. Ltd Kanagawa Japan
| | | | - Yuko Kaneko
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| |
Collapse
|
42
|
Karpouzas GA, Bui VL, Ronda N, Hollan I, Ormseth SR. Biologics and atherosclerotic cardiovascular risk in rheumatoid arthritis: a review of evidence and mechanistic insights. Expert Rev Clin Immunol 2021; 17:355-374. [PMID: 33673792 DOI: 10.1080/1744666x.2021.1899809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Cardiovascular disease is a leading comorbidity in rheumatoid arthritis. Timely introduction of biologic therapies in a treat-to-target approach has optimized disease-related outcomes and attenuated accrual of comorbidities, including cardiovascular risk.Areas covered: A literature search in MEDLINE (via PubMed) was performed between January 2009 and November 2020. This manuscript explores recent developments in atherosclerotic cardiovascular risk in RA compared with non-RA individuals; it synopsizes differences in vascular function and inflammation, prevalence, burden, vulnerability, and progression of atherosclerotic plaque and their underlying cellular and molecular mechanisms. Finally, it reviews the recent literature on cardioprotective benefits of biologics and draws mechanistic links with inhibition of new plaque formation, stabilization of high-risk lesions and improvement in endothelial function, arterial stiffness, lipid metabolism, and traditional cardiac risk factors.Expert opinion: Increasing evidence points to a solid cardioprotective influence of earlier, longer, and ongoing use of biologic treatments in RA. Nevertheless, the precise mechanistic effects of plaque progression and remodeling, vascular stiffness, endothelial dysfunction, lipid metabolism, and traditional cardiac risk factors are less rigorously characterized.
Collapse
Affiliation(s)
- George A Karpouzas
- Division of Rheumatology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Viet L Bui
- Division of Rheumatology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Ivana Hollan
- The Norwegian University of Science and Technology, Gjøvik, Norway.,Beitostølen Sport and Health Centre, Beitostølen, Norway
| | - Sarah R Ormseth
- Division of Rheumatology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| |
Collapse
|
43
|
Desbois AC, Ciocan D, Saadoun D, Perlemuter G, Cacoub P. Specific microbiome profile in Takayasu's arteritis and giant cell arteritis. Sci Rep 2021; 11:5926. [PMID: 33723291 PMCID: PMC7961033 DOI: 10.1038/s41598-021-84725-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
Recent studies have provided evidence of a close link between specific microbiota and inflammatory disorders. While the vessel wall microbiota has been recently described in large vessel vasculitis (LVV) and controls, the blood microbiome in these diseases has not been previously reported (LVV). We aimed to analyse the blood microbiome profile of LVV patients (Takayasu’s arteritis [TAK], giant cell arteritis [GCA]) and healthy blood donors (HD). We studied the blood samples of 13 patients with TAK (20 samples), 9 patients with GCA (11 samples) and 15 HD patients. We assessed the blood microbiome profile by sequencing the 16S rDNA blood bacterial DNA. We used linear discriminant analysis (LDA) coupled with linear discriminant effect size measurement (LEfSe) to investigate the differences in the blood microbiome profile between TAK and GCA patients. An increase in the levels of Clostridia, Cytophagia and Deltaproteobacteria and a decrease in Bacilli at the class level were found in TAK patients compared with HD patients (LDA > 2, p < 0.05). Active TAK patients had significantly lower levels of Staphylococcus compared with inactive TAK patients. Samples of GCA patients had an increased abundance of Rhodococcus and an unidentified member of the Cytophagaceae family. Microbiota of TAK compared with GCA patients was found to show higher levels of Candidatus Aquiluna and Cloacibacterium (LDA > 2; p < 0.05). Differences highlighted in the blood microbiome were also associated with a shift of bacterial predicted metabolic functions in TAK in comparison with HD. Similar results were also found in patients with active versus inactive TAK. In conclusion, patients with TAK were found to present a specific blood microbiome profile in comparison with healthy donors and GCA subjects. Significant changes in the blood microbiome profiles of TAK patients were associated with specific metabolic functions.
Collapse
Affiliation(s)
- Anne Claire Desbois
- INSERM, UMR_S 959, Inflammation-Immunopathology-Biotherapy Department, Sorbonne Université, UPMC University of Paris, Paris, France. .,Department of Internal Medicine and Clinical Immunology, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France. .,Department of Internal Medicine and Laboratory I3 "Immunology, Immunopathology, Immunotherapy" UMR 7211 (CNRS/UPMC) INSERM U959, Hôpital Pitié-Salpêtrière, 47-83 boulevard de l'Hôpital, 75013, Paris, France.
| | - Dragos Ciocan
- INSERM U996, Inflammation Chemokines and Immunopathology, DHU Hépatinov, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France.,APHP-Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
| | - David Saadoun
- INSERM, UMR_S 959, Inflammation-Immunopathology-Biotherapy Department, Sorbonne Université, UPMC University of Paris, Paris, France.,Department of Internal Medicine and Clinical Immunology, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Gabriel Perlemuter
- INSERM U996, Inflammation Chemokines and Immunopathology, DHU Hépatinov, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France.,APHP-Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
| | - Patrice Cacoub
- INSERM, UMR_S 959, Inflammation-Immunopathology-Biotherapy Department, Sorbonne Université, UPMC University of Paris, Paris, France.,Department of Internal Medicine and Clinical Immunology, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| |
Collapse
|
44
|
Bi X, Du C, Wang X, Wang X, Han W, Wang Y, Qiao Y, Zhu Y, Ran L, Liu Y, Xiong J, Huang Y, Liu M, Liu C, Zeng C, Wang J, Yang K, Zhao J. Mitochondrial Damage-Induced Innate Immune Activation in Vascular Smooth Muscle Cells Promotes Chronic Kidney Disease-Associated Plaque Vulnerability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002738. [PMID: 33717842 PMCID: PMC7927614 DOI: 10.1002/advs.202002738] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/06/2020] [Indexed: 05/02/2023]
Abstract
Chronic kidney disease (CKD) is associated with accelerated atherosclerosis progression and high incidence of cardiovascular events, hinting that atherosclerotic plaques in CKD may be vulnerable. However, its cause and mechanism remain obscure. Here, it is shown that apolipoprotein E-deficient (ApoE-/-) mouse with CKD (CKD/ApoE-/- mouse) is a useful model for investigating the pathogenesis of plaque vulnerability, and premature senescence and phenotypic switching of vascular smooth muscle cells (VSMCs) contributes to CKD-associated plaque vulnerability. Subsequently, VSMC phenotypes in patients with CKD and CKD/ApoE-/- mice are comprehensively investigated. Using multi-omics analysis and targeted and VSMC-specific gene knockout mice, VSMCs are identified as both type-I-interferon (IFN-I)-responsive and IFN-I-productive cells. Mechanistically, mitochondrial damage resulting from CKD-induced oxidative stress primes the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway to trigger IFN-I response in VSMCs. Enhanced IFN-I response then induces VSMC premature senescence and phenotypic switching in an autocrine/paracrine manner, resulting in the loss of fibrous cap VSMCs and fibrous cap thinning. Conversely, blocking IFN-I response remarkably attenuates CKD-associated plaque vulnerability. These findings reveal that IFN-I response in VSMCs through immune sensing of mitochondrial damage is essential for the pathogenesis of CKD-associated plaque vulnerability. Mitigating IFN-I response may hold promise for the treatment of CKD-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Xianjin Bi
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Changhong Du
- State Key Laboratory of TraumaBurns and Combined InjuryInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xinmiao Wang
- State Key Laboratory of TraumaBurns and Combined InjuryInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xue‐Yue Wang
- Laboratory of Stem Cell & Developmental BiologyDepartment of Histology and EmbryologyCollege of Basic Medical SciencesArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Wenhao Han
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yue Wang
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yu Qiao
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yingguo Zhu
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Li Ran
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yong Liu
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jiachuan Xiong
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yinghui Huang
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Mingying Liu
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Chi Liu
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Chunyu Zeng
- Department of CardiologyDaping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Junping Wang
- State Key Laboratory of TraumaBurns and Combined InjuryInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Ke Yang
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jinghong Zhao
- Department of Nephrologythe Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of ChongqingKidney Center of PLAXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| |
Collapse
|
45
|
Akiyama M, Ohtsuki S, Berry GJ, Liang DH, Goronzy JJ, Weyand CM. Innate and Adaptive Immunity in Giant Cell Arteritis. Front Immunol 2021; 11:621098. [PMID: 33717054 PMCID: PMC7947610 DOI: 10.3389/fimmu.2020.621098] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022] Open
Abstract
Autoimmune diseases can afflict every organ system, including blood vessels that are critically important for host survival. The most frequent autoimmune vasculitis is giant cell arteritis (GCA), which causes aggressive wall inflammation in medium and large arteries and results in vaso-occlusive wall remodeling. GCA shares with other autoimmune diseases that it occurs in genetically predisposed individuals, that females are at higher risk, and that environmental triggers are suspected to beget the loss of immunological tolerance. GCA has features that distinguish it from other autoimmune diseases and predict the need for tailored diagnostic and therapeutic approaches. At the core of GCA pathology are CD4+ T cells that gain access to the protected tissue niche of the vessel wall, differentiate into cytokine producers, attain tissue residency, and enforce macrophages differentiation into tissue-destructive effector cells. Several signaling pathways have been implicated in initiating and sustaining pathogenic CD4+ T cell function, including the NOTCH1-Jagged1 pathway, the CD28 co-stimulatory pathway, the PD-1/PD-L1 co-inhibitory pathway, and the JAK/STAT signaling pathway. Inadequacy of mechanisms that normally dampen immune responses, such as defective expression of the PD-L1 ligand and malfunction of immunosuppressive CD8+ T regulatory cells are a common theme in GCA immunopathology. Recent studies are providing a string of novel mechanisms that will permit more precise pathogenic modeling and therapeutic targeting in GCA and will fundamentally inform how abnormal immune responses in blood vessels lead to disease.
Collapse
Affiliation(s)
- Mitsuhiro Akiyama
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Shozo Ohtsuki
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - David H Liang
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Jörg J Goronzy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Cornelia M Weyand
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
46
|
Hofeld BC, Puppala VK, Tyagi S, Ahn KW, Anger A, Jia S, Salzman NH, Hessner MJ, Widlansky ME. Lactobacillus plantarum 299v probiotic supplementation in men with stable coronary artery disease suppresses systemic inflammation. Sci Rep 2021; 11:3972. [PMID: 33597583 PMCID: PMC7889883 DOI: 10.1038/s41598-021-83252-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
Recent trials demonstrate that systemic anti-inflammatory therapy reduces cardiovascular events in coronary artery disease (CAD) patients. We recently demonstrated Lactobacillus plantarum 299v (Lp299v) supplementation improved vascular endothelial function in men with stable CAD. Whether this favorable effect is in part due to anti-inflammatory action remains unknown. Testing this hypothesis, we exposed plasma obtained before and after Lp299v supplementation from these subjects to a healthy donor's PBMCs and measured differences in the PBMC transciptome, performed gene ontological analyses, and compared Lp299v-induced transcriptome changes with changes in vascular function. Daily alcohol users (DAUs) (n = 4) had a significantly different response to Lp299v and were separated from the main analyses. Non-DAUs- (n = 15) showed improved brachial flow-mediated dilation (FMD) and reduced circulating IL-8, IL-12, and leptin. 997 genes were significantly changed. I.I.com decreased (1.01 ± 0.74 vs. 0.22 ± 0.51; P < 0.0001), indicating strong anti-inflammatory effects. Pathway analyses revealed downregulation of IL-1β, interferon-stimulated pathways, and toll-like receptor signaling, and an increase in regulator T-cell (Treg) activity. Reductions in GBP1, JAK2, and TRAIL expression correlated with improved FMD. In non-DAU men with stable CAD, post-Lp299v supplementation plasma induced anti-inflammatory transcriptome changes in human PBMCs that could benefit CAD patients. Future studies should delineate changes in circulating metabolites responsible for these effects.
Collapse
Affiliation(s)
- Benjamin C Hofeld
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Venkata K Puppala
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sudhi Tyagi
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kwang Woo Ahn
- Department of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amberly Anger
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shuang Jia
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nita H Salzman
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Martin J Hessner
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael E Widlansky
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Division of Cardiovascular Medicine, Professor of Medicine and Pharmacology, Medical College of Wisconsin, Hub for Collaborative Medicine, 5th Floor A5743, 8701 W. Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
47
|
Kabeerdoss J, Goel R, Mohan H, Danda D. High expression of pro-inflammatory cytokine genes IL-1β and IL-1R2 upon TLR4 activation in Takayasu arteritis. Rheumatol Int 2021; 42:535-543. [PMID: 33528653 DOI: 10.1007/s00296-020-04785-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/31/2020] [Indexed: 10/22/2022]
Abstract
Toll-like receptors (TLR) 4 and its endogenous ligands are highly expressed in aorta. In the present study, we have explored the effect of TLR-4 activation by pro-inflammatory and angiogenic factors in PBMCs of patients with Takayasu Arteritis (TA). In the screening cohort, PBMCs of TA (n = 6) and healthy controls (n = 6) were stimulated with LPS and cultured. mRNA expression of 84 genes were quantitated by RT2 Profiler™ PCR Array kit in PBMCs. Validation set of additional PBMCs from TA (n = 7) and healthy controls [HC) (n = 7) were then stimulated with LPS to study expression of selected genes with delta Ct > 0.1 in the screening cohort. Significant gene expressions were correlated with Indian Takayasu arteritis activity scores (ITAS 2010). Increased expression of CCL2 was observed only in unstimulated PBMCs of patients with TA [median relative difference (RD) of 2.37] as compared to HC (RD 1.37, p < 0.03) in validation cohort, while stimulation with TLR4 ligand led to increased mRNA expression of IL-1β (RD 7.9, p < 0.028) and IL-1R2 (RD 0.08 p < 0.013) genes as compared to that of HC [RD of 5.32 for IL-1β and 0.01 for IL-1R2, respectively] in validation cohort. TLR4 activation also led to significantly higher expression of HPSE, TIMP1 and low expression of VEGFB, S1PR1, SERPINF1, ANGPLT4, ANGPT2, TIE1 and NOS3 genes in the screening cohort. But expression of VEGFB was not significant in validation cohort. The significant gene expressions, however, did not correlate with ITAS [ITAS2010 and ITAS-A (CRP)]. TLR4 activation leads to increased expression of IL-1β and IL-1R2 genes in PBMCs of patients with TA.
Collapse
Affiliation(s)
- Jayakanthan Kabeerdoss
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Ruchika Goel
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Hindumathi Mohan
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Debashish Danda
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Tamil Nadu, Vellore, 632004, India.
| |
Collapse
|
48
|
McCoy MG, Nascimento DW, Veleeparambil M, Murtazina R, Gao D, Tkachenko S, Podrez E, Byzova TV. Endothelial TLR2 promotes proangiogenic immune cell recruitment and tumor angiogenesis. Sci Signal 2021; 14. [PMID: 33986920 DOI: 10.1126/scisignal.abc5371] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Toll-like receptor 2 (TLR2) is implicated in various pathologies, mainly in terms of its function within innate immune cells. However, TLR2 is also present in endothelial cells. Here, we explored the physiological and pathophysiological roles of endothelial TLR2 signaling. We found that TLR2 was highly abundant in the endothelium within various tissues using TLR2-IRES-EGFP reporter mice and was required for proinflammatory endothelial cell function. Endothelial cells lacking TLR2 exhibited reduced proinflammatory potential at the protein, cell, and tissue levels. Loss of endothelial TLR2 blunted the inflammatory response to both exogenous and endogenous danger signals in endothelial cells in culture and in vivo. Endothelial TLR2 promoted tumor growth, angiogenesis, and protumorigenic immune cell recruitment in a mouse model of prostate cancer. Furthermore, the cell surface localization of P-selectin and the subsequent production of other critical cell adhesion molecules (such as E-selectin, ICAM-1 and VCAM-1) that recruit immune cells required endothelial TLR2. Our findings demonstrate that endothelial cells actively contribute to innate immune pathways and propose that endothelial TLR2 has a pathological role in proinflammatory conditions.
Collapse
Affiliation(s)
- Michael G McCoy
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, OH, USA 44195
| | - Daniel W Nascimento
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, OH, USA 44195
| | - Manoj Veleeparambil
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, OH, USA 44195
| | - Rakhylia Murtazina
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, OH, USA 44195
| | - Detao Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, OH, USA 44195
| | - Svyatoslav Tkachenko
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, OH, USA 44195
| | - Eugene Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, OH, USA 44195
| | - Tatiana V Byzova
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, OH, USA 44195
| |
Collapse
|
49
|
Simon S, Ninan J, Hissaria P. Diagnosis and management of giant cell arteritis: Major review. Clin Exp Ophthalmol 2021; 49:169-185. [PMID: 33426764 DOI: 10.1111/ceo.13897] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/06/2020] [Accepted: 12/13/2020] [Indexed: 12/13/2022]
Abstract
Giant cell arteritis is a medical emergency because of the high risk of irreversible blindness and cerebrovascular accidents. While elevated inflammatory markers, temporal artery biopsy and modern imaging modalities are useful diagnostic aids, thorough history taking and clinical acumen still remain key elements in establishing a timely diagnosis. Glucocorticoids are the cornerstone of treatment but are associated with high relapse rates and side effects. Targeted biologic agents may open up new treatment approaches in the future.
Collapse
Affiliation(s)
- Sumu Simon
- Department of Ophthalmology and South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Jem Ninan
- Department of Rheumatology, Modbury Public Hospital, Modbury, South Australia, Australia
| | - Pravin Hissaria
- Department of Immunology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
50
|
Pereira-da-Silva T, Napoleão P, Pinheiro T, Selas M, Silva F, Ferreira RC, Carmo MM. The Proinflammatory Soluble CD40 Ligand Is Associated with the Systemic Extent of Stable Atherosclerosis. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:39. [PMID: 33406736 PMCID: PMC7824733 DOI: 10.3390/medicina57010039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/26/2020] [Accepted: 12/29/2020] [Indexed: 01/05/2023]
Abstract
Background and objectives: Polyvascular atherosclerosis is frequent and associated with a high cardiovascular risk, although the mechanisms regulating the atherosclerosis extent to single or multiple arterial territories are still poorly understood. Inflammation regulates atherogenesis and soluble CD40 ligand (sCD40L) is an inflammatory mediator associated with the presence of single-territorial atherosclerosis. We assessed whether the sCD40L expression is associated with the atherosclerosis extent to single or multiple arterial territories and with the atherosclerosis severity in different territories. Materials and Methods: We prospectively enrolled 94 participants with no atherosclerosis (controls, n = 26); isolated coronary atherosclerosis (group 1, n = 20); coronary and lower extremity (LE) atherosclerosis (group 2, n = 18); coronary and carotid atherosclerosis (group 3, n = 12); and coronary, LE, and carotid atherosclerosis (group 4, n = 18). Serum sCD40L levels were quantified. Results: The sCD40L levels (ng/mL, mean (standard deviation)) were 4.0 (1.5), 5.6 (2.6), 7.2 (4.2), 5.9 (3.7), and 5.1 (2.4) in controls and groups 1 to 4, respectively (ANOVA p = 0.012). In nonrevascularized patients, the sCD40L levels were significantly higher in group 2 than in group 1 and were correlated with the number of LE diseased segments. Prior LE bypass surgery was associated with lower sCD40L levels. Coexistence of coronary and LE atherosclerosis was independently associated with the sCD40L levels. Conclusions: The sCD40L levels were increased in stable atherosclerosis, particularly in polyvascular coronary and LE atherosclerosis. The number of LE diseased segments and prior LE revascularization were associated with sCD40L expression. To our knowledge, these are novel data, which provide insights into the mechanisms underlying multi-territorial atherosclerosis expression. sCD40L may be a promising noninvasive tool for refining the stratification of the systemic atherosclerotic burden.
Collapse
Affiliation(s)
- Tiago Pereira-da-Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| | - Patrícia Napoleão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Teresa Pinheiro
- Instituto de Bioengenharia e Biociências, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Lisbon, Portugal;
| | - Mafalda Selas
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Filipa Silva
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Rui Cruz Ferreira
- Department of Cardiology, Hospital de Santa Marta, Centro Hospitalar Universitário de Lisboa Central, 1169-024 Lisbon, Portugal; (M.S.); (F.S.); (R.C.F.)
| | - Miguel Mota Carmo
- Chronic Diseases Research Center (CEDOC), NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal;
| |
Collapse
|