1
|
Drosos AA, Venetsanopoulou AA, Pelechas E, Voulgari PV. Exploring Cardiovascular Risk Factors and Atherosclerosis in Rheumatoid Arthritis. Eur J Intern Med 2024; 128:1-9. [PMID: 39048336 DOI: 10.1016/j.ejim.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease mainly affecting the peripheral diarthrodial joints symmetrically and also presenting many extra-articular manifestations. Morbidity and mortality in RA patients are higher compared to the general population. Cardiovascular (CV) disease is one of the most common causes of death in these patients. Classical or traditional risk factors for atherosclerosis development occur more frequently in RA patients compared to those without this condition. Studies have showed that RA patients often present comorbidities such as hypertension, dyslipidemia, diabetes mellitus and obesity. However, the high incidence of CV events occurring in RA patients is not explained by the presence of traditional risk factors. Systemic inflammation, as it is expressed with the presence of proinflammatory cytokines and increased acute phase reactants, may contribute to the development of premature atherosclerosis in these patients. In this review, we explore the risk factors for CV disease, the generation of dyslipidemia, the lipid paradox and the role of systemic inflammation in the atherosclerotic process in RA. We discuss also the role of early therapeutic intervention that suppresses inflammation which may have beneficial effects on CV disease in RA patients.
Collapse
Affiliation(s)
- Alexandros A Drosos
- Department of Rheumatology, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece.
| | - Aliki A Venetsanopoulou
- Department of Rheumatology, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Eleftherios Pelechas
- Department of Rheumatology, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Paraskevi V Voulgari
- Department of Rheumatology, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
2
|
Zhou H, Zhang R, Li M, Wang F, Gao Y, Fang K, Zong J, Chang X. Methazolamide Can Treat Atherosclerosis by Increasing Immunosuppressive Cells and Decreasing Expressions of Genes Related to Proinflammation, Calcification, and Tissue Remodeling. J Immunol Res 2024; 2024:5009637. [PMID: 39081633 PMCID: PMC11288698 DOI: 10.1155/2024/5009637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/01/2024] [Accepted: 06/29/2024] [Indexed: 08/02/2024] Open
Abstract
It has been reported that carbonic anhydrase I (CA1) is a target for the diagnosis and therapy of atherosclerosis (AS) since CA1 can promote AS aortic calcification. We also found that methazolamide (MTZ), a drug for glaucoma treatment and an inhibitor of carbonic anhydrases, can treat AS by inhibiting calcification in aortic tissues. This study focused on the therapeutic mechanism of MTZ and the pathogenic mechanism of AS. In this study, a routine AS animal model was established in ApoE-/- mice, which were treated with MTZ. The aortic tissues were analyzed using single-cell sequencing. MTZ significantly increased the proportions of B-1/MZB B cells with high expressions of Nr4A1 and Ccr7, CD8+CD122+ Treg-like cells with high Nr4A1 expression, and smooth muscle cells with high Tpm2 expression. These cells or their marker genes were reported to exert immunosuppressive, anti-proinflammatory, and atheroprotective effects. MTZ also decreased the proportions of endothelial cells with high expressions of Retn, Apoc1, Lcn2, Mt1, Serpina3, Lpl, and Lgals3; nonclassical CD14+CD16++ monocytes with high expressions of Mt1, Tyrobp, Lgals3, and Cxcl2; and Spp1+ macrophages with high expressions of Mmp-12, Trem2, Mt1, Lgals3, Cxcl2, and Lpl. These cells or their marker genes have been reported to promote inflammation, calcification, tissue remodeling, and atherogenesis. A significant decrease in the proportion of CD8+CD183 (CXCR3)+ T cells, the counterpart of murine CD8+CD122+ T cells, was detected in the peripheral blood of newly diagnosed AS patients rather than in that of patients receiving anti-AS treatments. These results suggest that MTZ can treat AS by increasing immunosuppressive cells and decreasing expressions of genes related to inflammation, calcification, and tissue remodeling.
Collapse
Affiliation(s)
- Hongji Zhou
- Medical Research CenterThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao 266000, China
- Department of CardiologyThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao 266000, China
| | - Rui Zhang
- Department of CardiologyThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao 266000, China
| | - Min Li
- Clinical Laboratory and Central LaboratoryQingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Road Renmin 4, Qingdao 266033, Shandong Province, China
| | - Fuyan Wang
- Clinical Laboratory and Central LaboratoryQingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Road Renmin 4, Qingdao 266033, Shandong Province, China
| | - Yuxia Gao
- Shandong Engineering Research Center of Bacterial Anti-tumor Drugs and Cell Therapy, Jingshi Road 7000, Jinan 250000, Shandong Province, China
| | - Kehua Fang
- Clinical LaboratoryThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao, Shandong 266000, China
| | - Jinbao Zong
- Clinical Laboratory and Central LaboratoryQingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Road Renmin 4, Qingdao 266033, Shandong Province, China
| | - Xiaotian Chang
- Medical Research CenterThe Affiliated Hospital of Qingdao University, Wutaishan Road 1677, Qingdao 266000, China
| |
Collapse
|
3
|
He J, Gao Y, Yang C, Guo Y, Liu L, Lu S, He H. Navigating the landscape: Prospects and hurdles in targeting vascular smooth muscle cells for atherosclerosis diagnosis and therapy. J Control Release 2024; 366:261-281. [PMID: 38161032 DOI: 10.1016/j.jconrel.2023.12.047] [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: 09/21/2023] [Revised: 12/02/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Vascular smooth muscle cells (VSMCs) have emerged as pivotal contributors throughout all phases of atherosclerotic plaque development, effectively dispelling prior underestimations of their prevalence and significance. Recent lineage tracing studies have unveiled the clonal nature and remarkable adaptability inherent to VSMCs, thereby illuminating their intricate and multifaceted roles in the context of atherosclerosis. This comprehensive review provides an in-depth exploration of the intricate mechanisms and distinctive characteristics that define VSMCs across various physiological processes, firmly underscoring their paramount importance in shaping the course of atherosclerosis. Furthermore, this review offers a thorough examination of the significant strides made over the past two decades in advancing imaging techniques and therapeutic strategies with a precise focus on targeting VSMCs within atherosclerotic plaques, notably spotlighting meticulously engineered nanoparticles as a promising avenue. We envision the potential of VSMC-targeted nanoparticles, thoughtfully loaded with medications or combination therapies, to effectively mitigate pro-atherogenic VSMC processes. These advancements are poised to contribute significantly to the pivotal objective of modulating VSMC phenotypes and enhancing plaque stability. Moreover, our paper also delves into recent breakthroughs in VSMC-targeted imaging technologies, showcasing their remarkable precision in locating microcalcifications, dynamically monitoring plaque fibrous cap integrity, and assessing the therapeutic efficacy of medical interventions. Lastly, we conscientiously explore the opportunities and challenges inherent in this innovative approach, providing a holistic perspective on the potential of VSMC-targeted strategies in the evolving landscape of atherosclerosis research and treatment.
Collapse
Affiliation(s)
- Jianhua He
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Yu Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Can Yang
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yujie Guo
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Lisha Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Shan Lu
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Hongliang He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
4
|
Falkenberg C, Bartholdy C, Koch J, Toft M, Skov S, Hansen CHF, Hansen A. Induction of CD8 + immune memory and enhanced inflammation in a skin inflammation model through pre-immunization with inactivated pathogens. Clin Transl Sci 2024; 17:e13697. [PMID: 38082552 PMCID: PMC10766028 DOI: 10.1111/cts.13697] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024] Open
Abstract
Laboratory mice live in specific pathogen-free (SPF) conditions, resulting in an immature immune system comparable to that of newborns rather than adult humans or mice from pet shops. This condition may compromise their translational value. Reintroducing pathogens would lead to the uncontrolled spread of infections and associated diseases, so research facilities should seek safer alternatives. We immunized laboratory mice with a cocktail of pathogens, which were inactivated by ultraviolet irradiation and mixed with the adjuvant AddaVax. This immunization resulted in a higher percentage of CD8+ effector memory T cells compared to untreated mice, although the response was not as robust as in pet shop mice. In a model of skin inflammation, pre-immunization led to an increased skin inflammatory response compared to non-immunized mice. All immunized mice seroconverted to the pathogens in the mixture, while none of the non-immunized mice housed together seroconverted to the pathogens applied to the pre-immunized mice. In conclusion, pre-immunization of mice impacts the immune system, which includes increasing the levels of CD8+ effector memory T cells.
Collapse
Affiliation(s)
- Caroline Falkenberg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenFrederiksberg CDenmark
| | - Christina Bartholdy
- Translational Sciences, Research & Early Development, LEO Pharma A/SBallerupDenmark
| | - Janne Koch
- Translational Sciences, Research & Early Development, LEO Pharma A/SBallerupDenmark
| | | | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenFrederiksberg CDenmark
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenFrederiksberg CDenmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenFrederiksberg CDenmark
| |
Collapse
|
5
|
Pickett JR, Wu Y, Zacchi LF, Ta HT. Targeting endothelial vascular cell adhesion molecule-1 in atherosclerosis: drug discovery and development of vascular cell adhesion molecule-1-directed novel therapeutics. Cardiovasc Res 2023; 119:2278-2293. [PMID: 37595265 PMCID: PMC10597632 DOI: 10.1093/cvr/cvad130] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/14/2023] [Accepted: 07/04/2023] [Indexed: 08/20/2023] Open
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) has been well established as a critical contributor to atherosclerosis and consequently as an attractive therapeutic target for anti-atherosclerotic drug candidates. Many publications have demonstrated that disrupting the VCAM-1 function blocks monocyte infiltration into the sub-endothelial space, which effectively prevents macrophage maturation and foam cell transformation necessary for atherosclerotic lesion formation. Currently, most VCAM-1-inhibiting drug candidates in pre-clinical and clinical testing do not directly target VCAM-1 itself but rather down-regulate its expression by inhibiting upstream cytokines and transcriptional regulators. However, the pleiotropic nature of these regulators within innate immunity means that optimizing dosage to a level that suppresses pathological activity while preserving normal physiological function is extremely challenging and oftentimes infeasible. In recent years, highly specific pharmacological strategies that selectively inhibit VCAM-1 function have emerged, particularly peptide- and antibody-based novel therapeutics. Studies in such VCAM-1-directed therapies so far remain scarce and are limited by the constraints of current experimental atherosclerosis models in accurately representing the complex pathophysiology of the disease. This has prompted the need for a comprehensive review that recounts the evolution of VCAM-1-directed pharmaceuticals and addresses the current challenges in novel anti-atherosclerotic drug development.
Collapse
Affiliation(s)
- Jessica R Pickett
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, West Creek Road, Nathan, QLD 4111, Australia
- School of Environment and Science, Griffith University, Kessels Road, Nathan, QLD 4111, Australia
| | - Yuao Wu
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, West Creek Road, Nathan, QLD 4111, Australia
| | - Lucia F Zacchi
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, St. Lucia, QLD 4072, Australia
- School of Chemistry and Molecular Biosciences, the University of Queensland, St. Lucia, QLD 4072, Australia
| | - Hang T Ta
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, West Creek Road, Nathan, QLD 4111, Australia
- School of Environment and Science, Griffith University, Kessels Road, Nathan, QLD 4111, Australia
| |
Collapse
|
6
|
Han S, Lu H, Yu Y, Liu X, Jing F, Wang L, Zhao Y, Hou M. Hyperlipidemia in immune thrombocytopenia: a retrospective study. Thromb J 2023; 21:102. [PMID: 37784127 PMCID: PMC10544441 DOI: 10.1186/s12959-023-00545-9] [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: 04/22/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Immune thrombocytopenia (ITP) is an autoimmune hemorrhagic disease characterized by low platelet count and bleeding manifestations. However, some patients also suffered from atherosclerosis or even infarction. Apart from activated platelets, lipid metabolism takes a large part in the formation of atherosclerosis and metabolic syndrome. The lipid metabolic state in ITP patients is still unknown. METHODS We retrospectively reviewed 302 hospitalized ITP patients in our cohort, comparing their blood lipids, bleeding symptoms, metabolic diseases and treatment responses. RESULTS We found a high proportion of ITP patients suffered from hyperlipidemia, and other metabolic diseases including cardiovascular or cerebral atherosclerosis or infarction, hypertension, and type 2 diabetes. Hyperlipidemia was associated with severe bleeding and treatment refractoriness in ITP. Statins could alleviate thrombocytopenia and bleeding severity, and facilitate ITP treatment, while improving hyperlipidemia in ITP patients. CONCLUSIONS Our present study demonstrated that lipid metabolism might play an indispensable role in ITP pathogenesis and development.
Collapse
Affiliation(s)
- Shouqing Han
- Department of Hematology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong, China
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, China
| | - Hui Lu
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yafei Yu
- Department of Hematology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong, China
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinguang Liu
- Department of Hematology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong, China
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, China
| | - Fangmiao Jing
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Liang Wang
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China.
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, China.
| | - Yajing Zhao
- Department of Hematology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong, China.
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, China.
| | - Ming Hou
- Department of Hematology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong, China.
- Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
7
|
Xiao Y, Powell DW, Liu X, Li Q. Cardiovascular manifestations of inflammatory bowel diseases and the underlying pathogenic mechanisms. Am J Physiol Regul Integr Comp Physiol 2023; 325:R193-R211. [PMID: 37335014 PMCID: PMC10979804 DOI: 10.1152/ajpregu.00300.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/21/2023]
Abstract
Inflammatory bowel disease (IBD), consisting of ulcerative colitis and Crohn's disease, mainly affects the gastrointestinal tract but is also known to have extraintestinal manifestations because of long-standing systemic inflammation. Several national cohort studies have found that IBD is an independent risk factor for the development of cardiovascular disorders. However, the molecular mechanisms by which IBD impairs the cardiovascular system are not fully understood. Although the gut-heart axis is attracting more attention in recent years, our knowledge of the organ-to-organ communication between the gut and the heart remains limited. In patients with IBD, upregulated inflammatory factors, altered microRNAs and lipid profiles, as well as dysbiotic gut microbiota, may induce adverse cardiac remodeling. In addition, patients with IBD have a three- to four times higher risk of developing thrombosis than people without IBD, and it is believed that the increased risk of thrombosis is largely due to increased procoagulant factors, platelet count/activity, and fibrinogen concentration, in addition to decreased anticoagulant factors. The predisposing factors for atherosclerosis are present in IBD and the possible mechanisms may involve oxidative stress system, overexpression of matrix metalloproteinases, and changes in vascular smooth muscle phenotype. This review focuses mainly on 1) the prevalence of cardiovascular diseases associated with IBD, 2) the potential pathogenic mechanisms of cardiovascular diseases in patients with IBD, and 3) adverse effects of IBD drugs on the cardiovascular system. Also, we introduce here a new paradigm for the gut-heart axis that includes exosomal microRNA and the gut microbiota as a cause for cardiac remodeling and fibrosis.
Collapse
Affiliation(s)
- Ying Xiao
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
- Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas, United States
| | - Don W Powell
- Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas, United States
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
| | - Qingjie Li
- Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, Texas, United States
| |
Collapse
|
8
|
Gusev E, Sarapultsev A. Atherosclerosis and Inflammation: Insights from the Theory of General Pathological Processes. Int J Mol Sci 2023; 24:ijms24097910. [PMID: 37175617 PMCID: PMC10178362 DOI: 10.3390/ijms24097910] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Recent advances have greatly improved our understanding of the molecular mechanisms behind atherosclerosis pathogenesis. However, there is still a need to systematize this data from a general pathology perspective, particularly with regard to atherogenesis patterns in the context of both canonical and non-classical inflammation types. In this review, we analyze various typical phenomena and outcomes of cellular pro-inflammatory stress in atherosclerosis, as well as the role of endothelial dysfunction in local and systemic manifestations of low-grade inflammation. We also present the features of immune mechanisms in the development of productive inflammation in stable and unstable plaques, along with their similarities and differences compared to canonical inflammation. There are numerous factors that act as inducers of the inflammatory process in atherosclerosis, including vascular endothelium aging, metabolic dysfunctions, autoimmune, and in some cases, infectious damage factors. Life-critical complications of atherosclerosis, such as cardiogenic shock and severe strokes, are associated with the development of acute systemic hyperinflammation. Additionally, critical atherosclerotic ischemia of the lower extremities induces paracoagulation and the development of chronic systemic inflammation. Conversely, sepsis, other critical conditions, and severe systemic chronic diseases contribute to atherogenesis. In summary, atherosclerosis can be characterized as an independent form of inflammation, sharing similarities but also having fundamental differences from low-grade inflammation and various variants of canonical inflammation (classic vasculitis).
Collapse
Affiliation(s)
- Evgenii Gusev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080 Chelyabinsk, Russia
| |
Collapse
|
9
|
Karakasis P, Lefkou E, Pamporis K, Nevras V, Bougioukas KI, Haidich AB, Fragakis N. Risk of Subclinical Atherosclerosis in Patients with Antiphospholipid Syndrome and Subjects With Antiphospholipid Antibody Positivity: A Systematic Review and Meta-analysis. Curr Probl Cardiol 2023; 48:101672. [PMID: 36841314 DOI: 10.1016/j.cpcardiol.2023.101672] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/26/2023]
Abstract
Currently, the guidelines for the prevention of atherosclerosis in patients with antiphospholipid syndrome (APS) do not differ substantially from those in the general population. We aimed to assess the risk of subclinical atherosclerosis in patients with APS and subjects with antiphospholipid antibody (aPL) positivity. Systematic literature search was conducted through Medline and Scopus until January 2023. Random effects meta-analyses were performed to examine the differences in markers of subclinical atherosclerosis between APS patients, subjects positive for aPLs and healthy controls. Patients with APS had significantly higher values of common carotid artery (CCA) intima-media thickness (IMT) (MD = 0.07 mm; P < 0.0001), internal carotid artery IMT (MD = 0.06 mm; P < 0.01), carotid bifurcation IMT (MD = 0.14 mm; P < 0.01) and were more frequently diagnosed with atherosclerotic plaques compared to controls (OR = 3.73; P < 0.01). Similarly, APS patients showed a decreased flow and nitrate-mediated dilation (MD = -4.52 %; <0.01, MD = -1.25 %; P < 0.05, respectively). Interestingly, comparable were the results for subjects with aPL positivity, who had higher CCA-IMT (MD = 0.06 mm; P < 0.01) and higher prevalence of atherosclerotic plaques (OR = 2.59; P = 0.08) compared to controls. Sensitivity analysis conducted on primary APS patients revealed that the risk of atherosclerosis is associated with APS per se and is not exclusively driven by other underlying conditions. Patients with APS and subjects with aPLs have an increased risk of subclinical atherosclerosis and require early and disease-specific prevention of atherosclerosis.
Collapse
Affiliation(s)
- Paschalis Karakasis
- Department of Hygiene, Social-Preventive Medicine & Medical Statistics, Medical School, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece.
| | - Elmina Lefkou
- Perigenesis, Institute of Obstetric Haematology, Thessaloniki, Greece
| | - Konstantinos Pamporis
- Department of Hygiene, Social-Preventive Medicine & Medical Statistics, Medical School, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Vasileios Nevras
- Cardiology Department, G. Gennimatas General Hospital, Thessaloniki, Greece
| | - Konstantinos I Bougioukas
- Department of Hygiene, Social-Preventive Medicine & Medical Statistics, Medical School, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Anna-Bettina Haidich
- Department of Hygiene, Social-Preventive Medicine & Medical Statistics, Medical School, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Nikolaos Fragakis
- Second Cardiology Department, Hippokrateion General Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
10
|
The Clinical Significance of Salusins in Systemic Sclerosis-A Cross-Sectional Study. Diagnostics (Basel) 2023; 13:diagnostics13050848. [PMID: 36899991 PMCID: PMC10001236 DOI: 10.3390/diagnostics13050848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/14/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Background: Systemic sclerosis (SSc) is a connective tissue disease manifesting with progressive fibrosis of the skin and internal organs. Its pathogenesis is strictly associated with vascular disfunction and damage. Salusin-α and salusin-β, endogenous peptides regulating secretion of pro-inflammatory cytokines and vascular smooth muscle proliferation, may potentially play a role in SSc pathogenesis. Objectives: The aim of this study was to assess the concentration of salusins in sera of patients with SSc and healthy controls and to evaluate correlations between the salusins levels and selected clinical parameters within the study group. Materials and methods: 48 patients with SSc (44 women; mean age, 56.4, standard deviation, 11.4) and 25 adult healthy volunteers (25 women; mean age, 55.2, standard deviation, 11.2) were enrolled. All patients with SSc were treated with vasodilators and twenty-seven of them (56%) also received immunosuppressive therapy. Results: Circulating salusin-α was significantly elevated in patients with SSc in comparison to healthy controls (U = 350.5, p = 0.004). Patients with SSc receiving immunosuppression had higher serum salusin-α concentrations compared with those without immunosuppressive therapy (U = 176.0, p = 0.026). No correlation was observed between salusins concentrations and skin or internal organ involvement parameters. Conclusions: Salusin-α, a bioactive peptide mitigating the endothelial disfunction, was elevated in patients with systemic sclerosis receiving vasodilators and immunosuppressants. Increased salusin-α concertation may be associated with the initiation of atheroprotective processes in patients with SSc managed pharmacologically, which requires verification in future studies.
Collapse
|
11
|
Titanium dioxide nanoparticle-based hydroxyl and superoxide radical production for oxidative stress biological simulations. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2022.114290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
12
|
Chen J, Liu Y, Liu Y, Peng J. Resveratrol protects against ox-LDL-induced endothelial dysfunction in atherosclerosis via depending on circ_0091822/miR-106b-5p-mediated upregulation of TLR4. Immunopharmacol Immunotoxicol 2022; 44:915-924. [PMID: 35736860 DOI: 10.1080/08923973.2022.2093740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Atherosclerosis (AS) is the most common inducer of cardiovascular diseases, and resveratrol (RSV) has played a protective function in the endothelial injury of AS. This study was to explore the molecular mechanism of RSV in oxidized low-density lipoprotein (ox-LDL)-mediated endothelial dysfunction. METHODS Circ_0091822, microRNA-106b-5p (miR-106b-5p) or toll-like receptor (TLR4) levels were examined using reverse transcription-quantitative polymerase chain reaction assay. Cell viability was detected via Cell Counting Kit-8 assay and angiogenesis was assessed by tube formation assay. Cell apoptosis was determined through flow cytometry. The protein analysis was conducted via western blot. Inflammatory cytokines were measured by enzyme-linked immunosorbent assay. The oxidative injury was evaluated using the commercial kits. The binding detection was performed via dual-luciferase reporter assay and RNA pull-down assay. RESULTS Circ_0091822 was downregulated by RSV in ox-LDL-treated endothelial cells. RSV promoted cell viability and angiogenesis while inhibiting apoptosis, inflammation, and oxidative stress after exposure to ox-LDL. The circ_0091822 knockdown relieved the ox-LDL-induced cell damages. RSV suppressed the ox-LDL-caused endothelial dysfunction via inducing the downregulation of circ_0091822. Circ_0091822 could target miR-106b-5p, and the reversal of circ_0091822 for RSV function was achieved by sponging miR-106b-5p. Circ_0091822 absorbed miR-106b-5p to elevate the level of TLR4. RSV impeded ox-LDL-induced damages by regulating miR-106b-5p/TLR4 axis. CONCLUSION All these findings suggested that RSV acted as an inhibitory factor in ox-LDL-induced endothelial injury via downregulating circ_0091822 to upregulate miR-106b-5p-related TLR4.
Collapse
Affiliation(s)
- Jinsong Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hengyang Medcial School, University of South China, Hengyang City, China
| | - Yang Liu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hengyang Medcial School, University of South China, Hengyang City, China
| | - Yunyang Liu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hengyang Medcial School, University of South China, Hengyang City, China
| | - Jianye Peng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hengyang Medcial School, University of South China, Hengyang City, China
| |
Collapse
|
13
|
CHENG X, ZHAO C, JIN Z, HU J, ZHANG Z, ZHANG C. Natural products: potential therapeutic agents for atherosclerosis. Chin J Nat Med 2022; 20:830-845. [DOI: 10.1016/s1875-5364(22)60219-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Indexed: 11/24/2022]
|
14
|
Wei N, Xu Y, Li Y, Shi J, Zhang X, You Y, Sun Q, Zhai H, Hu Y. A bibliometric analysis of T cell and atherosclerosis. Front Immunol 2022; 13:948314. [PMID: 36311729 PMCID: PMC9606647 DOI: 10.3389/fimmu.2022.948314] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/30/2022] [Indexed: 08/29/2023] Open
Abstract
Atherosclerosis (AS) is widespread and develops into circulatory system problems. T cells play an essential regulatory role in AS occurrence and development. So far, there is no bibliometric research on T cells and AS. To learn more about T cell and AS development, 4,381 records were retrieved from Web of Science™ Core Collection. Then, these records were scientometrically analyzed using CiteSpace and VOSviewer in terms of spatiotemporal distribution, author distribution, subject categories, topic distribution, references, and keywords. Our analysis provides basic information on research in the field, demonstrates that the field has stabilized over the past decade, and identifies potential partners for interested researchers. Current research hotspots in this field mainly include the inflammatory mechanism, immune mechanism, related diseases, and related cytokines of AS. B cell, mortality, inhibition, and monocyte represent the frontiers of research in this field, undergoing an explosive phase. We hope that this work will provide new ideas for advancing the scientific research and clinical application of T cell and AS.
Collapse
Affiliation(s)
- Namin Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Xu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ya’nan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingjing Shi
- Department of Cardiovascular Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuesong Zhang
- Department of Cardiovascular Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yaping You
- Department of Cardiovascular Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qianqian Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Huaqiang Zhai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanhui Hu
- Department of Cardiovascular Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Qi W, Zhao J, Huang C, Jiang N, Li J, Wu C, Zhang S, Hu C, Xu D, Wang Q, Li M, Tian X, Zhao Y, Zeng X. Clinical characteristics and prognosis of patients with antiphospholipid antibodies based on cluster analysis: an 8-year cohort study. Arthritis Res Ther 2022; 24:140. [PMID: 35690831 PMCID: PMC9188169 DOI: 10.1186/s13075-022-02814-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/12/2022] [Indexed: 12/26/2022] Open
Abstract
Background Antiphospholipid syndrome (APS) is an autoimmune disease characterized by persistent antiphospholipid antibodies (aPLs) positivity with a wide manifestation spectrum. A risk stratification is needed for management guidance and prognosis assessment. We aimed to identify phenotypes among aPL-positive patients and assess the prognosis of each phenotype. Methods This was a single-center, prospective cohort study of aPL-positive patients presented to Peking Union Medical College Hospital from 2012 to 2020. Demographic characteristics, aPL-related manifestations, cardiovascular risk factors, and antibodies profiles were recorded. The primary endpoint was defined as a combination of newly onset thrombosis, major bleeding events, non-criteria manifestations, and all-cause death. Hierarchical cluster analysis and Kaplan-Meier survival analysis were performed. Results Four clusters among 383 patients (70.2% female; mean age 37.7 years) were identified. Cluster 1 (n = 138): patients with systemic lupus erythematosus (SLE) and non-criteria manifestations; cluster 2 (n = 112): patients with multiple cardiovascular risk factors; cluster 3 (n = 83): female patients with obstetric morbidity; cluster 4 (n = 50): patients with isolated lupus anticoagulant (LA) positivity. Non-criteria manifestations were found aggregated with SLE from cluster analysis of variables. Cluster 3 showed the best outcome, while cluster 2 suffered highest frenquency of newly onset arterial thrombosis. Conclusions We identified 4 clinical phenotypes of aPL-positive patients. Non-criteria manifestations may indicate underlying SLE, for which immunosuppressive therapy besides anticoagulation may be necessary. Patients with isolated LA positivity suffered similar risks with secondary APS and patients with multiple cardiovascular risk factors. Attention should be paid to male patients, and the screening of cardiovascular risk factors should never be ignored. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02814-w.
Collapse
Affiliation(s)
- Wanting Qi
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China.,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Can Huang
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Nan Jiang
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Chanyuan Wu
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Shangzhu Zhang
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Chaojun Hu
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Dong Xu
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China.,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China.,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China.,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China.,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China.,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. .,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China. .,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, China. .,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.
| |
Collapse
|
16
|
Tektonidou MG. Cardiovascular disease risk in antiphospholipid syndrome: Thrombo-inflammation and atherothrombosis. J Autoimmun 2022; 128:102813. [PMID: 35247655 DOI: 10.1016/j.jaut.2022.102813] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/23/2022] [Accepted: 02/27/2022] [Indexed: 12/11/2022]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disorder characterized by the presence of antiphospholipid antibodies (aPL) (lupus anticoagulant, anticardiolipin antibodies and anti-beta2glycoprotein I (anti-β2GPI) antibodies) and a plethora of macro- and micro-vascular manifestations, affecting predominantly young adults. Cardiovascular events are the leading causes of morbidity and mortality in APS. APL-mediated thrombo-inflammation and atherothrombosis are emerging pathogenetic mechanisms of cardiovascular disease (CVD) in APS, involving endothelial cell and monocyte activation, cytokines and adhesion molecules expression, complement and neutrophils activation, neutrophil extracellular traps formation, platelet cell activation and aggregation, and subsequent thrombin generation, in parallel with an oxidized low-density lipoprotein (oxLDL)-β2GPI complex induced macrophage differentiation to foam cells. High risk aPL profile, especially the presence of lupus anticoagulant and triple aPL positivity (all three aPL subtypes), co-existence with Systemic Lupus Erythematosus (SLE), as well as traditional risk factors such as smoking, hypertension, hyperlipemia and obesity are associated with both subclinical atherosclerosis and cardiovascular events in APS. Increased awareness of CVD risk by the physicians and patients, regular assessment and strict control of traditional risk factors, and lifestyle modifications are recommended. Use of low-dose aspirin should be considered for cardiovascular prevention in asymptomatic aPL carriers or SLE patients with high-risk aPL profile. The role of older agents such as hydroxychloroquine and statins or new potential targeted treatments against immuno- and athero-thrombosis has been demonstrated by experimental and some clinical studies and needs to be further evaluated by randomized controlled studies. This review summarizes the available evidence about the pathogenetic mechanisms and prevalence of cardiovascular events and subclinical atherosclerosis, the interrelationship between traditional and disease-related CVD risk factors, and the cardiovascular risk assessment and management in APS.
Collapse
Affiliation(s)
- Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| |
Collapse
|
17
|
Akam EY, Nuako AA, Daniel AK, Stanford FC. Racial Disparities and Cardiometabolic Risk: New Horizons of Intervention and Prevention. Curr Diab Rep 2022; 22:129-136. [PMID: 35175453 PMCID: PMC9908372 DOI: 10.1007/s11892-022-01451-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/03/2022] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW Cardiometabolic diseases are a leading cause of morbidity and mortality in the USA and disproportionately impact racial and ethnic minorities. Multiple factors contribute to this disparity including genetic and socioeconomic factors, the latter of which contributes to disparities both through systemic barriers such as healthcare access and by directly impacting metabolism through epigenetics and environment-related alterations in the gut microbiome. This review will discuss advances in medicine that can be used to identify, prognosticate, and treat cardiometabolic diseases, and how these may be used to address existing disparities. RECENT FINDINGS There is growing research aimed at identifying novel cardiometabolic disease targets and expanding the use of existing pharmacotherapies based on comorbidities. Advances in metabolomics and genomics can give insight into an individual's unique biochemical profile, providing the means for earlier identification of disease and specific treatment targets. Moreover, developments in telehealth and related medical device technologies can expand access to underserved minority populations and improve control of chronic conditions such as diabetes and hypertension. Precision medicine may be integral to bridging the racial gap in cardiometabolic disease outcomes. Developments in genomics, metabolomics, wearable medical devices, and telehealth can result in personalized treatments for patients that account for the socioeconomic and genetic factors that contribute to poor health outcomes in minorities. As research in this field rapidly progresses, special efforts must be made to ensure inclusion of racial and ethnic minority populations in clinical research and equal access to all treatment modalities.
Collapse
Affiliation(s)
- Eftitan Y Akam
- Massachusetts General Hospital Department of Medicine, 55 Fruit Street, Boston, MA, 02114, USA.
- Massachusetts General Hospital Department of Pediatrics, 55 Fruit Street, Boston, MA, 02114, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| | - Akua A Nuako
- Massachusetts General Hospital Department of Medicine, 55 Fruit Street, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Afkera K Daniel
- Massachusetts General Hospital Department of Medicine, 55 Fruit Street, Boston, MA, 02114, USA
- Massachusetts General Hospital Department of Pediatrics, 55 Fruit Street, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Fatima Cody Stanford
- Harvard Medical School, Boston, MA, 02115, USA
- Massachusetts General Hospital, MGH Weight Center, Department of Medicine-Division of Endocrinology-Neuroendocrine, Department of Pediatrics-Division of Endocrinology, Nutrition Obesity Research Center at Harvard (NORCH), 50 Staniford Street, Boston, MA, 02114, USA
| |
Collapse
|
18
|
Vilela EM, Fontes-Carvalho R. Inflammation and ischemic heart disease: The next therapeutic target? Rev Port Cardiol 2021; 40:785-796. [PMID: 34857118 DOI: 10.1016/j.repce.2021.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammation plays an important role in several stages of the cardiovascular continuum. In recent decades a plethora of studies have provided new data highlighting the role of inflammation in atherogenesis and atherothrombosis in two-way interactions with various cardiovascular risk factors and further influencing these dynamic processes. The concept of targeting residual inflammatory risk among individuals with ischemic heart disease (IHD) is therefore gaining increasing attention. Recently, several landmark randomized controlled trials have assessed different pharmacological approaches that may mitigate this residual risk. The results of some of these studies, such as CANTOS with canakinumab and COLCOT and LoDoCo2 with colchicine, are promising and have provided data to support this concept. Moreover, though several aspects remain to be clarified, these trials have shown the potential of modulating inflammation as a new target to reduce the risk of cardiovascular events in secondary prevention patients. In the present review, we aim to present a pragmatic overview of the complex interplay between inflammation and IHD, and to critically appraise the current evidence on this issue while presenting future perspectives on this topic of pivotal contemporary interest.
Collapse
Affiliation(s)
- Eduardo M Vilela
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.
| | - Ricardo Fontes-Carvalho
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal; Cardiovascular Research Center (UniC), Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
19
|
Vilela EM, Fontes-Carvalho R. Inflammation and ischemic heart disease: The next therapeutic target? Rev Port Cardiol 2021; 40:S0870-2551(21)00321-8. [PMID: 34456098 DOI: 10.1016/j.repc.2021.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/07/2021] [Accepted: 02/16/2021] [Indexed: 10/20/2022] Open
Abstract
Inflammation plays an important role in several stages of the cardiovascular continuum. In recent decades a plethora of studies have provided new data highlighting the role of inflammation in atherogenesis and atherothrombosis in two-way interactions with various cardiovascular risk factors and further influencing these dynamic processes. The concept of targeting residual inflammatory risk among individuals with ischemic heart disease (IHD) is therefore gaining increasing attention. Recently, several landmark randomized controlled trials have assessed different pharmacological approaches that may mitigate this residual risk. The results of some of these studies, such as CANTOS with canakinumab and COLCOT and LoDoCo2 with colchicine, are promising and have provided data to support this concept. Moreover, though several aspects remain to be clarified, these trials have shown the potential of modulating inflammation as a new target to reduce the risk of cardiovascular events in secondary prevention patients. In the present review, we aim to present a pragmatic overview of the complex interplay between inflammation and IHD, and to critically appraise the current evidence on this issue while presenting future perspectives on this topic of pivotal contemporary interest.
Collapse
Affiliation(s)
- Eduardo M Vilela
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.
| | - Ricardo Fontes-Carvalho
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal; Cardiovascular Research Center (UniC), Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
20
|
Law CC, Puranik R, Fan J, Fei J, Hambly BD, Bao S. Clinical Implications of IL-32, IL-34 and IL-37 in Atherosclerosis: Speculative Role in Cardiovascular Manifestations of COVID-19. Front Cardiovasc Med 2021; 8:630767. [PMID: 34422917 PMCID: PMC8377289 DOI: 10.3389/fcvm.2021.630767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 07/13/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis, which is a primary cause of cardiovascular disease (CVD) deaths around the world, is a chronic inflammatory disease that is characterised by the accumulation of lipid plaques in the arterial wall, triggering inflammation that is regulated by cytokines/chemokines that mediate innate and adaptive immunity. This review focuses on IL-32, -34 and -37 in the stable vs. unstable plaques from atherosclerotic patients. Dysregulation of the novel cytokines IL-32, -34 and -37 has been discovered in atherosclerotic plaques. IL-32 and -34 are pro-atherogenic and associated with an unstable plaque phenotype; whereas IL-37 is anti-atherogenic and maintains plaque stability. It is speculated that these cytokines may contribute to the explanation for the increased occurrence of atherosclerotic plaque rupture seen in patients with COVID-19 infection. Understanding the roles of these cytokines in atherogenesis may provide future therapeutic perspectives, both in the management of unstable plaque and acute coronary syndrome, and may contribute to our understanding of the COVID-19 cytokine storm.
Collapse
Affiliation(s)
- Ching Chee Law
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Rajesh Puranik
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jingchun Fan
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jian Fei
- Shanghai Engineering Research Centre for Model Organisms, SMOC, Shanghai, China
| | - Brett D Hambly
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Shisan Bao
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
21
|
Tabares-Guevara JH, Villa-Pulgarin JA, Hernandez JC. Atherosclerosis: immunopathogenesis and strategies for immunotherapy. Immunotherapy 2021; 13:1231-1244. [PMID: 34382409 DOI: 10.2217/imt-2021-0009] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis, a chronic inflammatory condition in which atheroma accumulates within the intima of the arterial wall, is a life-threatening manifestation of cardiovascular disease, due to atheroma rupture, chronic luminal narrowing and thrombosis. Current knowledge of the role of a protective immune response in atherosclerotic lesions has provided promising opportunities to develop new immunotherapeutic strategies. In particular, Tregs exert an atheroprotective role by releasing anti-inflammatory cytokines (IL-10/TGF-β) and suppressing autoreactive T lymphocytes. In vivo animal experiments have shown that this can be achieved by developing vaccines that stimulate immunological tolerance to atheroma antigens. Here, we present an overview of the current knowledge of the proatherogenic immune response, and we discuss the strategies currently used as immunoregulatory therapy.
Collapse
Affiliation(s)
| | - Janny A Villa-Pulgarin
- Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| | - Juan C Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
| |
Collapse
|
22
|
Recognition of Oxidized Lipids by Macrophages and Its Role in Atherosclerosis Development. Biomedicines 2021; 9:biomedicines9080915. [PMID: 34440119 PMCID: PMC8389651 DOI: 10.3390/biomedicines9080915] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a multifactorial chronic disease that has a prominent inflammatory component. Currently, atherosclerosis is regarded as an active autoimmune process that involves both innate and adaptive immune pathways. One of the drivers of this process is the presence of modified low-density lipoprotein (LDL). For instance, lipoprotein oxidation leads to the formation of oxidation-specific epitopes (OSE) that can be recognized by the immune cells. Macrophage response to OSEs is recognized as a key trigger for initiation and a stimulator of progression of the inflammatory process in the arteries. At the same time, the role of oxidized LDL components is not limited to pro-inflammatory stimulation, but includes immunoregulatory effects that can have protective functions. It is, therefore, important to better understand the complexity of oxidized LDL effects in atherosclerosis in order to develop new therapeutic approaches to correct the inflammatory and metabolic imbalance associated with this disorder. In this review, we discuss the process of oxidized LDL formation, mechanisms of OSE recognition by macrophages and the role of these processes in atherosclerosis.
Collapse
|
23
|
Effect of GLP-1/GLP-1R on the Polarization of Macrophages in the Occurrence and Development of Atherosclerosis. Mediators Inflamm 2021; 2021:5568159. [PMID: 33854404 PMCID: PMC8019627 DOI: 10.1155/2021/5568159] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/22/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Aims To investigate the effect of GLP-1/GLP-1R on the polarization of macrophages in the occurrence and development of atherosclerosis. Methods Totally, 49 patients with coronary heart disease (CHD) and 52 cases of health control (HC) were recruited, all subjects accept coronary angiography gold standard inspection. One or more major coronary arteries (LM, LAD, LCx, and RCA) stenosis degree in 50% of patients as CHD group; the rest of the stenosis less than 50% or not seen obvious stenosis are assigned to the HC group. Flow cytometry were used to detect the percentage of (CD14+) M macrophages, (CD14+CD80+) M1 macrophages, (CD14+CD206+) M2 macrophages, and their surface GLP-1R expression differences in the two groups, using BD cytokine kit to detect the levels of IL-8, IL-1β, IL-6, IL-10, TNF, and IL-12p70. Results GLP-1R expression on the surface of total macrophages and M2 macrophages was different between the CHD group and the HC group (P < 0.05). There was no difference in the percentage of total, M1 or M2 macrophages (P > 0.05). Concentration of IL-8 in the HC group was higher than that in the CHD group (P < 0.05). There is no significant difference in the cytokine IL-1β, IL-6, IL-10, TNF, and IL-12p70 in the two groups (P > 0.05). After controlling for potential confounders including age, gender, smoking status (S.S.), drinking status (D.S.), HR, SBP, DBP, PP, TC, TG, HDL-C, LDL-C, GHbA1c, M, M1, M2, GLP-1R_M, GLP-1R_M1, GLP-1R_M2, IL-8, IL-1β, IL-6, IL-10, TNF, and IL-12p70 by multiple linear regression, decreasing Gensini Score was significantly associated with increased percentage of M1 macrophage. Conclusion GLP-1R agonist is independent of the hypoglycemic effect of T2DM and has protective effect on cardiovascular system. GLP-1R may regulate the polarization of macrophages toward M2, thus playing a protective role in the progression of coronary atherosclerosis.
Collapse
|
24
|
Sangha GS, Goergen CJ, Prior SJ, Ranadive SM, Clyne AM. Preclinical techniques to investigate exercise training in vascular pathophysiology. Am J Physiol Heart Circ Physiol 2021; 320:H1566-H1600. [PMID: 33385323 PMCID: PMC8260379 DOI: 10.1152/ajpheart.00719.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Atherosclerosis is a dynamic process starting with endothelial dysfunction and inflammation and eventually leading to life-threatening arterial plaques. Exercise generally improves endothelial function in a dose-dependent manner by altering hemodynamics, specifically by increased arterial pressure, pulsatility, and shear stress. However, athletes who regularly participate in high-intensity training can develop arterial plaques, suggesting alternative mechanisms through which excessive exercise promotes vascular disease. Understanding the mechanisms that drive atherosclerosis in sedentary versus exercise states may lead to novel rehabilitative methods aimed at improving exercise compliance and physical activity. Preclinical tools, including in vitro cell assays, in vivo animal models, and in silico computational methods, broaden our capabilities to study the mechanisms through which exercise impacts atherogenesis, from molecular maladaptation to vascular remodeling. Here, we describe how preclinical research tools have and can be used to study exercise effects on atherosclerosis. We then propose how advanced bioengineering techniques can be used to address gaps in our current understanding of vascular pathophysiology, including integrating in vitro, in vivo, and in silico studies across multiple tissue systems and size scales. Improving our understanding of the antiatherogenic exercise effects will enable engaging, targeted, and individualized exercise recommendations to promote cardiovascular health rather than treating cardiovascular disease that results from a sedentary lifestyle.
Collapse
Affiliation(s)
- Gurneet S Sangha
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Steven J Prior
- Department of Kinesiology, University of Maryland School of Public Health, College Park, Maryland.,Baltimore Veterans Affairs Geriatric Research, Education, and Clinical Center, Baltimore, Maryland
| | - Sushant M Ranadive
- Department of Kinesiology, University of Maryland School of Public Health, College Park, Maryland
| | - Alisa M Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
25
|
Liu H, Ma X, Wang X, Ma X, Mao Z, Zhu J, Chen F. Hsa_circ_0000345 regulates the cellular development of ASMCs in response to oxygenized low-density lipoprotein. J Cell Mol Med 2020; 24:11849-11857. [PMID: 32865338 PMCID: PMC7578870 DOI: 10.1111/jcmm.15801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/19/2020] [Accepted: 08/08/2020] [Indexed: 12/31/2022] Open
Abstract
The interaction between circRNAs and atherosclerosis has been extensively studied. However, more novel circRNAs need to be explored to help establish a perfect regulatory network. In the present research, hsa_circ_0000345 was demonstrated to regulate cellular development of oxygenized low‐density lipoprotein (ox‐LDL)‐treated aortic smooth muscle cells (ASMCs), which was closely related to the occurrence and progress of atherosclerosis. Ox‐LDL exposure remarkably decreased hsa_circ_0000345 expression in ASMCs. Transfection‐induced hsa_circ_0000345 overexpression activated cell viability (detected by an MTT assay) and restrained cellular apoptosis (analysed by flow cytometry) in the atherosclerosis cellular model. While down‐regulation of hsa_circ_0000345 reduced cell viability and promoted cell apoptosis. In addition, the data of the cell cycle distribution analysis and trans‐well assay indicated that cell cycle progression was arrested at the G1 phase while cell invasion was enhanced in ASMCs following treatment of ox‐LDL in the context of hsa_circ_0000345 OE plasmids. In addition, up‐regulation of hsa_circ_0000345 supported HIF‐1α at both the mRNA and protein level, and down‐regulation of hsa_circ_0000345 reduced HIF‐1α expression. Overall, the above findings revealed that hsa_circ_0000345 was a dramatic regulator of ASMCs proliferation, apoptosis and invasion in response to ox‐LDL treatment. Hsa_circ_0000345 was identified as a protector of cell viability during ox‐LDL induced cell development.
Collapse
Affiliation(s)
- Huifang Liu
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowen Ma
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Wang
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xirui Ma
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziming Mao
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhu
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengling Chen
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
26
|
Chu CS, Law SH, Lenzen D, Tan YH, Weng SF, Ito E, Wu JC, Chen CH, Chan HC, Ke LY. Clinical Significance of Electronegative Low-Density Lipoprotein Cholesterol in Atherothrombosis. Biomedicines 2020; 8:biomedicines8080254. [PMID: 32751498 PMCID: PMC7460408 DOI: 10.3390/biomedicines8080254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the numerous risk factors for atherosclerotic cardiovascular diseases (ASCVD), cumulative evidence shows that electronegative low-density lipoprotein (L5 LDL) cholesterol is a promising biomarker. Its toxicity may contribute to atherothrombotic events. Notably, plasma L5 LDL levels positively correlate with the increasing severity of cardiovascular diseases. In contrast, traditional markers such as LDL-cholesterol and triglyceride are the therapeutic goals in secondary prevention for ASCVD, but that is controversial in primary prevention for patients with low risk. In this review, we point out the clinical significance and pathophysiological mechanisms of L5 LDL, and the clinical applications of L5 LDL levels in ASCVD can be confidently addressed. Based on the previously defined cut-off value by receiver operating characteristic curve, the acceptable physiological range of L5 concentration is proposed to be below 1.7 mg/dL. When L5 LDL level surpass this threshold, clinically relevant ASCVD might be present, and further exams such as carotid intima-media thickness, pulse wave velocity, exercise stress test, or multidetector computed tomography are required. Notably, the ultimate goal of L5 LDL concentration is lower than 1.7 mg/dL. Instead, with L5 LDL greater than 1.7 mg/dL, lipid-lowering treatment may be required, including statin, ezetimibe or PCSK9 inhibitor, regardless of the low-density lipoprotein cholesterol (LDL-C) level. Since L5 LDL could be a promising biomarker, we propose that a high throughput, clinically feasible methodology is urgently required not only for conducting a prospective, large population study but for developing therapeutics strategies to decrease L5 LDL in the blood.
Collapse
Affiliation(s)
- Chih-Sheng Chu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan;
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Shi Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - David Lenzen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - Yong-Hong Tan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - Shih-Feng Weng
- Department of Healthcare Administration and Medical Informatics, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
| | - Etsuro Ito
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
- Department of Biology, Waseda University, Tokyo 162-8480, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 162-8480, Japan
| | - Jung-Chou Wu
- Division of Cardiology, Department of Internal Medicine, Pingtung Christian Hospital, Pingtung 90059, Taiwan;
| | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA;
| | - Hua-Chen Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan;
- Correspondence: (H.-C.C.); (L.-Y.K.); Tel.: +886-73121101 (ext. 2296); Fax: +886-73111996 (L.-Y.K.)
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
- Graduate Institute of Medicine, College of Medicine, & Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Correspondence: (H.-C.C.); (L.-Y.K.); Tel.: +886-73121101 (ext. 2296); Fax: +886-73111996 (L.-Y.K.)
| |
Collapse
|
27
|
Berretta M, Bignucolo A, Di Francia R, Comello F, Facchini G, Ceccarelli M, Iaffaioli RV, Quagliariello V, Maurea N. Resveratrol in Cancer Patients: From Bench to Bedside. Int J Mol Sci 2020; 21:E2945. [PMID: 32331450 PMCID: PMC7216168 DOI: 10.3390/ijms21082945] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Resveratrol (3,5,4'-trihydroxystilbene) is a natural phytoalexin that accumulates in several vegetables and fruits like nuts, grapes, apples, red fruits, black olives, capers, red rice as well as red wines. Being both an extremely reactive molecule and capable to interact with cytoplasmic and nuclear proteins in human cells, resveratrol has been studied over the years as complementary and alternative medicine (CAM) for the therapy of cancer, metabolic and cardiovascular diseases like myocardial ischemia, myocarditis, cardiac hypertrophy and heart failure. This review will describe the main biological targets, cardiovascular outcomes, physico-chemical and pharmacokinetic properties of resveratrol in preclinical and clinical models implementing its potential use in cancer patients.
Collapse
Affiliation(s)
- Massimiliano Berretta
- Department of Medical Oncology-Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alessia Bignucolo
- Experimental and Clinical Pharmacology-Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (A.B.); (F.C.)
| | - Raffaele Di Francia
- Gruppo Oncologico Ricercatori Italiani, GORI-ONLUS, 33170 Pordenone (PN), Italy;
| | - Francesco Comello
- Experimental and Clinical Pharmacology-Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (A.B.); (F.C.)
| | - Gaetano Facchini
- UOC Oncologia, ASL Napoli 2 Nord, P.O. “S.M. delle Grazie”, Pozzuoli-Ischia, 80078 Napoli, Italy;
| | - Manuela Ceccarelli
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Catania, 95122 Catania, Italy;
| | - Rosario Vincenzo Iaffaioli
- Association for Multidisciplinary Studies in Oncology and Mediterranean Diet, Piazza Nicola Amore, 80138 Naples, Italy;
| | - Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (N.M.)
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (N.M.)
| |
Collapse
|