1
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [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: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
2
|
Markov M, Georgiev T, Angelov AK, Dimova M. Adhesion molecules and atherosclerosis in ankylosing spondylitis: implications for cardiovascular risk. Rheumatol Int 2024; 44:1837-1848. [PMID: 39180529 DOI: 10.1007/s00296-024-05693-3] [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: 05/27/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024]
Abstract
Ankylosing Spondylitis (AS) stands as a chronic inflammatory arthritis within the spondyloarthritis spectrum, notably increasing cardiovascular (CV) risk and mortality through accelerated atherosclerosis compared to the non-affected population. While evidence in some studies supports a higher cardiovascular morbidity in AS patients, results from other studies reveal no significant disparities in atherosclerotic markers between AS individuals and healthy controls. This discrepancy may arise from the complex interaction between traditional CV risk factors and AS inflammatory burden. Endothelial dysfunction, a recognized antecedent of atherosclerosis prevalent among most individuals with AS, demonstrates the synergistic impact of inflammation and conventional risk factors on endothelial injury, consequently hastening the progression of atherosclerosis. Remarkably, endothelial dysfunction can precede vascular pathology in AS, suggesting a unique relationship between inflammation, atherosclerosis, and vascular damage. The role of adhesion molecules in the development of atherosclerosis, facilitating leukocyte adherence and migration into vascular walls, underscores the predictive value of soluble intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) levels for cardiovascular events. Despite significant progress in comprehending the pathogenesis of AS and its associated cardiovascular implications, the interplay among inflammation, endothelial dysfunction, and atherosclerosis remains partially elucidated. Investigations into the efficacy of therapeutic approaches involving angiotensin receptor blockers and statins have demonstrated reduced cardiovascular risk in AS patients, underscoring the imperative for additional research in this domain.
Collapse
Affiliation(s)
- Miroslav Markov
- Department of Propedeutics of Internal Medicine, Faculty of Medicine, Medical University, Varna, 9002, Bulgaria
- Clinic of Internal Medicine, University Hospital St. Marina - Varna, Varna, 9010, Bulgaria
| | - Tsvetoslav Georgiev
- First Department of Internal Medicine, Faculty of Medicine, Medical University - Varna, Varna, 9002, Bulgaria.
- Clinic of Rheumatology, University Hospital St. Marina - Varna, Varna, 9010, Bulgaria.
| | | | - Maria Dimova
- Department of Propedeutics of Internal Medicine, Faculty of Medicine, Medical University, Varna, 9002, Bulgaria
- Clinic of Internal Medicine, University Hospital St. Marina - Varna, Varna, 9010, Bulgaria
| |
Collapse
|
3
|
Barzilay JI, Buzkova P, Bielinski SJ, Cotch MF, Kestenbaum B, Austin TR, Carbone L, Mukamal KJ, Budoff MJ. The association of microvascular disease and endothelial dysfunction with vertebral trabecular bone mineral density : The MESA study. Osteoporos Int 2024; 35:1595-1604. [PMID: 38913124 DOI: 10.1007/s00198-024-07152-y] [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: 02/26/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024]
Abstract
Retinopathy and albuminuria are associated with hip fracture risk. We investigated whether these disorders and endothelial dysfunction (which underlies microvascular diseases) were associated with low trabecular bone density. No significant associations were found, suggesting that microvascular diseases are not related to fracture risk through low trabecular bone density. PURPOSE Microvascular diseases of the eye, kidney, and brain are associated with endothelial dysfunction and increased hip fracture risk. To explore the basis for higher hip fracture risk, we comprehensively examined whether markers of microvascular disease and/or endothelial dysfunction are related to trabecular bone mineral density (BMD), a proximate risk factor for osteoporotic fractures. METHODS Among 6814 participants in the Multi-Ethnic Study of Atherosclerosis study (MESA), we derived thoracic vertebral trabecular BMD from computed tomography of the chest and measured urine albumin to creatinine ratios (UACR), retinal arteriolar and venular widths, flow mediated dilation (FMD) of the brachial artery after 5 min of ischemia; and levels of five soluble endothelial adhesion markers (ICAM-1, VCAM-1, L-selectin, P-selectin, and E-selectin). Linear regression models were used to examine the association of trabecular BMD with markers of microvascular disease and with markers of endothelial dysfunction. RESULTS We observed no significant associations of UACR, retinal arteriolar or venular widths, or FMD with BMD. We also observed no statistically significant association of spine trabecular BMD with levels of endothelial adhesion markers. Men and women had largely similar results. CONCLUSION We conclude that there is little evidence to connect thoracic spine trabecular BMD to microvascular disorders or to endothelial dysfunction among multi-ethnic middle-aged and older adults. Other factors beyond trabecular BMD (e.g., bone quality or predisposition to falling) may be responsible for the associations of microvascular disease with osteoporotic fractures.
Collapse
Affiliation(s)
- Joshua I Barzilay
- Division of Endocrinology, Kaiser Permanente of Georgia, 3650 Steve Reynolds Blvd, Duluth, GA, 30096, USA.
- Division of Endocrinology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Petra Buzkova
- Division of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Mary Frances Cotch
- Office of Vision Health and Population Sciences, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bryan Kestenbaum
- Division of Nephrology, University of Washington, Seattle, WA, USA
| | - Thomas R Austin
- Department of Epidemiology, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Laura Carbone
- Division of Rheumatology, Medical College of Georgia, Augusta, GA, USA
| | - Kenneth J Mukamal
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University, Brookline, MA, USA
| | - Matthew J Budoff
- Department of Medicine, Lundquist Institute at Harbor-UCLA, Torrance, CA, USA
| |
Collapse
|
4
|
Mangoni AA, Zinellu A. Circulating cell adhesion molecules in systemic sclerosis: a systematic review and meta-analysis. Front Immunol 2024; 15:1438302. [PMID: 39234240 PMCID: PMC11371573 DOI: 10.3389/fimmu.2024.1438302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Patients with systemic sclerosis (SSc) have an increased risk of endothelial dysfunction, atherosclerosis, and cardiovascular events compared to the general population. Therefore, the availability of robust circulating biomarkers of endothelial dysfunction and atherogenesis may facilitate early recognition and management of cardiovascular risk in SSc. We sought to address this issue by conducting a systematic review and meta-analysis of studies investigating various types of circulating cell adhesion molecules involved in endothelial dysfunction and atherogenesis (i.e., immunoglobulin-like vascular cell, VCAM-1, intercellular, ICAM-1, platelet endothelial cell, PECAM-1, neural cell, NCAM, Down syndrome cell, DSCAM, and endothelial cell-selective, ESAM, adhesion molecules, E-, L-, and P-selectin, integrins, and cadherins) in SSc patients and healthy controls. Methods We searched PubMed, Scopus, and Web of Science from inception to 1 May 2024. Risk of bias and certainty of evidence were assessed using validated tools. Results In 43 eligible studies, compared to controls, patients with SSc had significantly higher plasma or serum concentrations of ICAM-1 (standard mean difference, SMD=1.16, 95% CI 0.88 to 1.44, p<0.001; moderate certainty), VCAM-1 (SMD=1.09, 95% CI 0.72 to 1.46, p<0.001; moderate certainty), PECAM-1 (SMD=1.65, 95% CI 0.33 to 2.98, p=0.014; very low certainty), E-selectin (SMD=1.17, 95% CI 0.72 to 1.62, p<0.001; moderate certainty), and P-selectin (SMD=1.10, 95% CI 0.31 to 1.90, p=0.007; low certainty). There were no significant between-group differences in L-selectin concentrations (SMD=-0.35, 95% CI -1.03 to 0.32, p=0.31; very low certainty), whereas minimal/no evidence was available for cadherins, NCAM, DSCAM, ESAM, or integrins. Overall, no significant associations were observed between the effect size and various patient and study characteristics in meta-regression and subgroup analyses. Discussion The results of this systematic review and meta-analysis suggest that specific circulating cell adhesion molecules, i.e., ICAM-1, VCAM-1, PECAM-1, E-selectin, and P-selectin, can be helpful as biomarkers of endothelial dysfunction and atherogenesis in the assessment of cardiovascular risk in SSc patients. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024549710.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Adelaide, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, SA, Adelaide, Australia
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
5
|
Mangoni AA, Zinellu A. A systematic review and meta-analysis of circulating adhesion molecules in rheumatoid arthritis. Inflamm Res 2024; 73:305-327. [PMID: 38240792 PMCID: PMC10894129 DOI: 10.1007/s00011-023-01837-6] [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: 09/26/2023] [Revised: 11/18/2023] [Accepted: 12/12/2023] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The availability of robust biomarkers of endothelial activation might enhance the identification of subclinical atherosclerosis in rheumatoid arthritis (RA). We investigated this issue by conducting a systematic review and meta-analysis of cell adhesion molecules in RA patients. METHODS We searched electronic databases from inception to 31 July 2023 for case-control studies assessing the circulating concentrations of immunoglobulin-like adhesion molecules (vascular cell, VCAM-1, intercellular, ICAM-1, and platelet endothelial cell, PECAM-1, adhesion molecule-1) and selectins (E, L, and P selectin) in RA patients and healthy controls. Risk of bias and certainty of evidence were assessed using the JBI checklist and GRADE, respectively. RESULTS In 39 studies, compared to controls, RA patients had significantly higher concentrations of ICAM-1 (standard mean difference, SMD = 0.81, 95% CI 0.62-1.00, p < 0.001; I2 = 83.0%, p < 0.001), VCAM-1 (SMD = 1.17, 95% CI 0.73-1.61, p < 0.001; I2 = 95.8%, p < 0.001), PECAM-1 (SMD = 0.82, 95% CI 0.57-1.08, p < 0.001; I2 = 0.0%, p = 0.90), E-selectin (SMD = 0.64, 95% CI 0.42-0.86, p < 0.001; I2 = 75.0%, p < 0.001), and P-selectin (SMD = 1.06, 95% CI 0.50-1.60, p < 0.001; I2 = 84.8%, p < 0.001), but not L-selectin. In meta-regression and subgroup analysis, significant associations were observed between the effect size and use of glucocorticoids (ICAM-1), erythrocyte sedimentation rate (VCAM-1), study continent (VCAM-1, E-selectin, and P-selectin), and matrix assessed (P-selectin). CONCLUSIONS The results of our study support a significant role of cell adhesion molecules in mediating the interplay between RA and atherosclerosis. Further studies are warranted to determine whether the routine use of these biomarkers can facilitate the detection and management of early atherosclerosis in this patient group. PROSPERO Registration Number: CRD42023466662.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, SA, 5042, Australia.
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
6
|
Gerasimova EV, Shayakhmetova RU, Gerasimova DA, Popkova TV, Ananyeva LP. Systemic Sclerosis and Atherosclerosis: Potential Cellular Biomarkers and Mechanisms. Front Biosci (Schol Ed) 2023; 15:16. [PMID: 38163957 DOI: 10.31083/j.fbs1504016] [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: 10/22/2023] [Revised: 11/29/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Systemic sclerosis (SSc) is a rare systemic autoimmune disease of unknown etiology, which is characterized by endothelial dysfunction, pathologic vasculopathy, and increased tissue fibrosis. Traditionally, SSc has been regarded as a prototypical fibrotic disease in the family of systemic autoimmune diseases. Traditionally, emphasis has been placed on the three components of the pathogenesis of SSc: vascular, immune, and mesenchymal. Microvascular lesions, including endothelial dysfunction and smooth muscle cell migration into the intima of vessels in SSc, resemble the atherosclerotic process. Although microvascular disease is a hallmark of SSc, understanding the role of atherosclerotic vascular lesions in patients with SSc remains limited. It is still unknown whether the increased cardiovascular risk in SSc is related to specific cardiac complications (such as myocardial fibrosis) or the accelerated development of atherosclerosis. Different immune cell types appear to be involved in the immunopathogenesis of SSc via the activation of other immune cells, fibrosis, or vascular damage. Macrophages, B cells, T cells, dendritic cells, neutrophils, and endothelial cells have been reported to play the most important role in the pathogenesis of SSc and atherosclerosis. In our article, we reviewed the most significant and recent studies on the pathogenetic links between the development of SSc and the atherosclerotic process.
Collapse
Affiliation(s)
- Elena V Gerasimova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, 115522 Moscow, Russian Federation
| | - Rushana U Shayakhmetova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, 115522 Moscow, Russian Federation
| | - Daria A Gerasimova
- Department of Organization and Economics of Pharmacy, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russian Federation
| | - Tatiana V Popkova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, 115522 Moscow, Russian Federation
| | - Lidia P Ananyeva
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, 115522 Moscow, Russian Federation
| |
Collapse
|
7
|
Chetan IM, Vesa ȘC, Domokos Gergely B, Beyer RS, Tomoaia R, Cabau G, Vulturar DM, Pop D, Todea D. Increased Levels of VCAM-1 in Patients with High Cardiovascular Risk and Obstructive Sleep Apnea Syndrome. Biomedicines 2023; 12:48. [PMID: 38255155 PMCID: PMC10813101 DOI: 10.3390/biomedicines12010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
(1) Background: Although obstructive sleep apnea (OSA) is associated with increased cardiovascular morbidity, the link between OSA and cardiovascular disease (CVD) is not completely elucidated. Thus, we aim to assess cardiovascular risk (CVR) using SCORE 2 and SCORE 2 for older persons (SCORE 2OP), and to evaluate the association between the endothelial biomarkers VCAM-1, ICAM-1, epicardial fat, and sleep study parameters in order to improve current clinical practices and better understand the short-and long-term CVRs in OSA patients. (2) Methods: 80 OSA patients and 37 healthy volunteers were enrolled in the study. SCORE2 and SCORE 2 OP regional risk charts (validated algorithms to predict the 10-year risk of first-onset CVD) were used for the analysis of CVR. Two-dimensional echocardiography was performed on all patients and epicardial fat thickness was measured. VCAM-1 and ICAM-1 serum levels were assessed in all patients. (3) Results: OSA patients were classified as being at high CVR, regardless of the type of score achieved. Increased EFT was observed in the OSA group. VCAM-1 was associated with a high CVR in OSA patients, but no significant correlation was observed between adhesion molecules and epicardial fat thickness. (4) Conclusions: OSA patients have a high CVR according to the SCORE 2 and SCORE 2OP risk scores. VCAM-1 may be associated with a high CVR in OSA patients. Extending conventional risk stratification scores by adding other potential biomarkers improves the risk stratification and guide treatment eligibility for CVD prevention in the OSA population.
Collapse
Affiliation(s)
- Ioana-Maria Chetan
- Department of Pneumology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.-M.C.); (B.D.G.); (D.M.V.); (D.T.)
| | - Ștefan Cristian Vesa
- Department of Pharmacology, Toxicology and Clinical Pharmacology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Bianca Domokos Gergely
- Department of Pneumology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.-M.C.); (B.D.G.); (D.M.V.); (D.T.)
| | | | - Raluca Tomoaia
- Department of Cardiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (R.T.)
| | - Georgiana Cabau
- Department of Medical Genetics, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Damiana Maria Vulturar
- Department of Pneumology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.-M.C.); (B.D.G.); (D.M.V.); (D.T.)
| | - Dana Pop
- Department of Cardiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (R.T.)
| | - Doina Todea
- Department of Pneumology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.-M.C.); (B.D.G.); (D.M.V.); (D.T.)
| |
Collapse
|
8
|
Rojas GA, Saavedra N, Morales C, Saavedra K, Lanas F, Salazar LA. Modulation of the Cardiovascular Effects of Polycyclic Aromatic Hydrocarbons: Physical Exercise as a Protective Strategy. TOXICS 2023; 11:844. [PMID: 37888695 PMCID: PMC10610936 DOI: 10.3390/toxics11100844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/30/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023]
Abstract
Exposure to polycyclic aromatic hydrocarbons (PAHs) present in air pollution increases cardiovascular risk. On the contrary, physical exercise is a widely used therapeutic approach to mitigate cardiovascular risk, but its efficacy in an environment of air pollution, particularly with PAHs, remains unclear. This study investigates the effects of exercise on inflammation, endothelial dysfunction, and REDOX imbalance due to PAH exposure using a mouse model. Twenty male BALB/c mice were subjected to a mixture of PAHs (phenanthrene, fluoranthene, pyrene) in conjunction with aerobic exercise. The investigation evaluated serum levels of inflammatory cytokines, gene expression linked to inflammatory markers, endothelial dysfunction, and REDOX imbalance in aortic tissues. Furthermore, the study evaluated the expression of the ICAM-1 and VCAM-1 proteins. Exercise led to notable changes in serum inflammatory cytokines, as well as the modulation of genes associated with endothelial dysfunction and REDOX imbalance in aortic tissue. In turn, exercise produced a modulation in the protein expression of ICAM-1 and VCAM-1. The findings implicate the potential of exercise to counter PAH-induced damage, as demonstrated by changes in markers. In conclusion, exercise could mitigate the adverse effects related to exposure to PAHs present in air pollution, as evidenced by changes in inflammatory markers, endothelial dysfunction, and REDOX imbalance.
Collapse
Affiliation(s)
- Gabriel A. Rojas
- Center of Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile or (G.A.R.); (N.S.); (C.M.); (K.S.)
- PhD Program in Applied Cellular and Molecular Biology, Universidad de La Frontera, Temuco 4811230, Chile
- Escuela Kinesiología, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
| | - Nicolás Saavedra
- Center of Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile or (G.A.R.); (N.S.); (C.M.); (K.S.)
| | - Cristian Morales
- Center of Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile or (G.A.R.); (N.S.); (C.M.); (K.S.)
- PhD Program in Applied Cellular and Molecular Biology, Universidad de La Frontera, Temuco 4811230, Chile
- Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Temuco 4811230, Chile
| | - Kathleen Saavedra
- Center of Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile or (G.A.R.); (N.S.); (C.M.); (K.S.)
| | - Fernando Lanas
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Luis A. Salazar
- Center of Molecular Biology & Pharmacogenetics, Department of Basic Sciences, Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile or (G.A.R.); (N.S.); (C.M.); (K.S.)
| |
Collapse
|
9
|
Wu HD, Yang LW, Deng DY, Jiang RN, Song ZK, Zhou LT. The effects of brominated flame retardants (BFRs) on pro-atherosclerosis mechanisms. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115325. [PMID: 37544066 DOI: 10.1016/j.ecoenv.2023.115325] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/24/2023] [Accepted: 08/01/2023] [Indexed: 08/08/2023]
Abstract
Brominated flame-retardants (BFRs) are environmental endocrine disruptors, comprising several pollutants, which potentially affect the endocrine system and cause dysfunction and disease. Widespread BFR exposure may cause multisystem toxicity, including cardiovascular toxicity in some individuals. Studies have shown that BFRs not only increase heart rate, induce arrhythmia and cardiac hypertrophy, but also cause glycolipid metabolism disorders, vascular endothelial dysfunction, and inflammatory responses, all of which potentially induce pre-pathological changes in atherosclerosis. Experimental data indicated that BFRs disrupt gene expression or signaling pathways, which cause vascular endothelial dysfunction, lipid metabolism-related disease, inflammation, and possibly atherosclerosis. Considerable evidence now suggests that BFR exposure may be a pro-atherosclerotic risk factor. In this study, we reviewed putative BFR effects underpinning pro-atherosclerosis mechanisms, and focused on vascular endothelial cell dysfunction, abnormal lipid metabolism, pro-inflammatory cytokine production and foam cell formation. Consequently, we proposed a scientific basis for preventing atherosclerosis by BFRs and provided concepts for further research.
Collapse
Affiliation(s)
- Hai-Di Wu
- Department of Cardiology, The First Hospital of Jilin University, Changchun 130021, China
| | - Li-Wei Yang
- School of Public Health, Jilin University, Changchun 130021, China
| | - Da-Yong Deng
- Department of Radiology, Jilin Provincial Cancer Hospital, 1066 Jinhu Road, 130000 Changchun, China
| | - Rong-Na Jiang
- Department of Intensive Care Unit, Jilin Provincial Cancer Hospital, 1066 Jinhu Road, 130000 Changchun, China
| | - Zi-Kai Song
- Department of Cardiology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Li-Ting Zhou
- School of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
10
|
Zhang H, Zhang-Sun ZY, Xue CX, Li XY, Ren J, Jiang YT, Liu T, Yao HR, Zhang J, Gou TT, Tian Y, Lei WR, Yang Y. CTRP family in diseases associated with inflammation and metabolism: molecular mechanisms and clinical implication. Acta Pharmacol Sin 2023; 44:710-725. [PMID: 36207402 PMCID: PMC10042840 DOI: 10.1038/s41401-022-00991-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/27/2022] [Indexed: 11/08/2022]
Abstract
C1q/tumor necrosis factor (TNF) related proteins (CTRPs) is a newly discovered adipokine family with conservative structure and ubiquitous distribution and is secreted by adipose tissues. Recently, CTRPs have attracted increasing attention due to the its wide-ranging effects upon inflammation and metabolism. To-date, 15 members of CTRPs (CTRP1-15) with the characteristic C1q domain have been characterized. Earlier in-depth phenotypic analyses of mouse models of CTRPs deficiency have also unveiled ample function of CTRPs in inflammation and metabolism. This review focuses on the rise of CTRPs, with a special emphasis on the latest discoveries with regards to the effects of the CTRP family on inflammation and metabolism as well as related diseases. We first introduced the structure of characteristic domain and polymerization of CTRPs to reveal its pleiotropic biological functions. Next, intimate association of CTRP family with inflammation and metabolism, as well as the involvement of CTRPs as nodes in complex molecular networks, were elaborated. With expanding membership of CTRP family, the information presented here provides new perspectives for therapeutic strategies to improve inflammatory and metabolic abnormalities.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Zi-Yin Zhang-Sun
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Cheng-Xu Xue
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Xi-Yang Li
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Yu-Ting Jiang
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Tong Liu
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Hai-Rong Yao
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Juan Zhang
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Tian-Tian Gou
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Ye Tian
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Wang-Rui Lei
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Yang Yang
- Department of Cardiology, Xi'an No.3 Hospital/The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| |
Collapse
|
11
|
Hypotheses on Atherogenesis Triggering: Does the Infectious Nature of Atherosclerosis Development Have a Substruction? Cells 2023; 12:cells12050707. [PMID: 36899843 PMCID: PMC10001176 DOI: 10.3390/cells12050707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/03/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Since the end of the 20th century, it has been clear that atherosclerosis is an inflammatory disease. However, the main triggering mechanism of the inflammatory process in the vascular walls is still unclear. To date, many different hypotheses have been put forward to explain the causes of atherogenesis, and all of them are supported by strong evidence. Among the main causes of atherosclerosis, which underlies these hypotheses, the following can be mentioned: lipoprotein modification, oxidative transformation, shear stress, endothelial dysfunction, free radicals' action, homocysteinemia, diabetes mellitus, and decreased nitric oxide level. One of the latest hypotheses concerns the infectious nature of atherogenesis. The currently available data indicate that pathogen-associated molecular patterns from bacteria or viruses may be an etiological factor in atherosclerosis. This paper is devoted to the analysis of existing hypotheses for atherogenesis triggering, and special attention is paid to the contribution of bacterial and viral infections to the pathogenesis of atherosclerosis and cardiovascular disease.
Collapse
|
12
|
Zinellu A, Mangoni AA. A systematic review and meta-analysis of the effect of statin treatment on sVCAM-1 and sICAM-1. Expert Rev Clin Pharmacol 2022; 15:601-620. [PMID: 35485866 DOI: 10.1080/17512433.2022.2072294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND AND AIMS Statins might prevent cell adhesion to the endothelium, a key step in atherosclerosis. We conducted a systematic review and meta-analysis of the effect of statins on soluble vascular (sVCAM-1) and intercellular (sICAM-1) adhesion molecule 1. METHODS A systematic literature search was conducted in PubMed, Web of Science, and Scopus, from inception to July 2021. Risk of bias and certainty of evidence were assessed using the Joanna Briggs Institute Critical Appraisal Checklist for analytical studies and GRADE, respectively. RESULTS Statins significantly reduced both sVCAM-1 (standard mean difference, SMD=-0.28, 95% CI -0.44 to -0.12, p=0.001; 46 treatment arms; low certainty of evidence) and sICAM-1 (SMD=-0.75, 95% CI -1.00 to -0.50, p<0.001; 61 treatment arms; moderate certainty of evidence) concentrations. In sensitivity analysis, the SMD values were not modified when individual studies were sequentially removed. There were significant associations between SMD and publication year and, for sICAM-1, statin-induced changes in HDL-cholesterol. In subgroup analysis, the lowering effect was significant with liphophilic, but not hydrophilic, statins, and similar, for sICAM-1, in participants with or without clinically overt atherosclerosis. CONCLUSIONS Statins significantly lower sVCAM-1/sICAM-1. Prospective studies are required to determine whether this mediates their atheroprotective effects (PROSPERO registration number: CRD42021276825).
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.,Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
13
|
Li RL, Wang LY, Liu S, Duan HX, Zhang Q, Zhang T, Peng W, Huang Y, Wu C. Natural Flavonoids Derived From Fruits Are Potential Agents Against Atherosclerosis. Front Nutr 2022; 9:862277. [PMID: 35399657 PMCID: PMC8987282 DOI: 10.3389/fnut.2022.862277] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis, as a chronic inflammatory response, is one of the main causes of cardiovascular diseases. Atherosclerosis is induced by endothelial cell dysfunction, migration and proliferation of smooth muscle cells, accumulation of foam cells and inflammatory response, resulting in plaque accumulation, narrowing and hardening of the artery wall, and ultimately leading to myocardial infarction or sudden death and other serious consequences. Flavonoid is a kind of natural polyphenol compound widely existing in fruits with various structures, mainly including flavonols, flavones, flavanones, flavanols, anthocyanins, isoflavones, and chalcone, etc. Because of its potential health benefits, it is now used in supplements, cosmetics and medicines, and researchers are increasingly paying attention to its role in atherosclerosis. In this paper, we will focus on several important nodes in the development of atherosclerotic disease, including endothelial cell dysfunction, smooth muscle cell migration and proliferation, foam cell accumulation and inflammatory response. At the same time, through the classification of flavonoids from fruits, the role and potential mechanism of flavonoids in atherosclerosis were reviewed, providing a certain direction for the development of fruit flavonoids in the treatment of atherosclerosis drugs.
Collapse
Affiliation(s)
- Ruo-Lan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ling-Yu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuqin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hu-Xinyue Duan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Wei Peng,
| | - Yongliang Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Yongliang Huang,
| | - Chunjie Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chunjie Wu,
| |
Collapse
|
14
|
Chakravarty D, Ray AG, Chander V, Mabalirajan U, Mondal PC, Siddiqui KN, Sinha BP, Konar A, Bandyopadhyay A. Systemic deficiency of vitronectin is associated with aortic inflammation and plaque progression in ApoE-Knockout mice. FASEB Bioadv 2022; 4:121-137. [PMID: 35141476 PMCID: PMC8814562 DOI: 10.1096/fba.2021-00108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/30/2022] Open
Abstract
Optimal cell spreading and interplay of vascular smooth muscle cells (VSMC), inflammatory cells, and cell adhesion molecules (CAM) are critical for progressive atherosclerosis and cardiovascular complications. The role of vitronectin (VTN), a major cell attachment glycoprotein, in the pathogenesis of atherosclerosis remains elusive. In this study, we attempt to examine the pathological role of VTN in arterial plaque progression and inflammation. We found that, relative expression analysis of VTN from the liver of Apolipoprotein E (ApoE) Knockout mice revealed that atherosclerotic progression induced by feeding mice with high cholesterol diet (HCD) causes a significant downregulation of VTN mRNA as well as protein after 60 days. Promoter assay confirmed that cholesterol modulates the expression of VTN by influencing its promoter. Mimicking VTN reduction with siRNA in HCD fed ApoE Knockout mice, accelerated athero-inflammation with an increase in NF-kB, ICAM-1, and VCAM-1 at the site of the plaque along with upregulation of inflammatory proteins like MCP-1 and IL-1β in the plasma. Also, matrix metalloprotease (MMP)-9 and MMP-12 expression were increased and collagen content was decreased in the plaque, in VTN deficient condition. This might pose a challenge to plaque integrity. Human subjects with acute coronary syndrome or having risk factors of atherosclerosis have lower levels of VTN compared to healthy controls suggesting a clinical significance of plasma VTN in the pathophysiology of coronary artery disease. We establish that, VTN plays a pivotal role in cholesterol-driven atherosclerosis and aortic inflammation and might be a useful indicator for atherosclerotic plaque burden and stability.
Collapse
Affiliation(s)
- Devasmita Chakravarty
- Department of Cell Biology and PhysiologyCSIR‐Indian Institute of Chemical BiologyKolkataIndia
| | - Aleepta Guha Ray
- Department of Cell Biology and PhysiologyCSIR‐Indian Institute of Chemical BiologyKolkataIndia
| | - Vivek Chander
- Department of Cell Biology and PhysiologyCSIR‐Indian Institute of Chemical BiologyKolkataIndia
| | - Ulaganathan Mabalirajan
- Department of Cell Biology and PhysiologyCSIR‐Indian Institute of Chemical BiologyKolkataIndia
| | | | | | - Bishnu Prasad Sinha
- Department of Cancer Biology and Inflammatory DisorderCSIR‐Indian Institute of Chemical BiologyKolkataIndia
| | - Aditya Konar
- Department of Laboratory Animal FacilityCSIR‐Indian Institute of Chemical BiologyKolkataIndia
| | - Arun Bandyopadhyay
- Department of Cell Biology and PhysiologyCSIR‐Indian Institute of Chemical BiologyKolkataIndia
- Department of Cancer Biology and Inflammatory DisorderCSIR‐Indian Institute of Chemical BiologyKolkataIndia
- Department of Laboratory Animal FacilityCSIR‐Indian Institute of Chemical BiologyKolkataIndia
| |
Collapse
|
15
|
Yu L, Peng J, Mineo C. Lipoprotein sialylation in atherosclerosis: Lessons from mice. Front Endocrinol (Lausanne) 2022; 13:953165. [PMID: 36157440 PMCID: PMC9498574 DOI: 10.3389/fendo.2022.953165] [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] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
Sialylation is a dynamically regulated modification, which commonly occurs at the terminal of glycan chains in glycoproteins and glycolipids in eukaryotic cells. Sialylation plays a key role in a wide array of biological processes through the regulation of protein-protein interactions, intracellular localization, vesicular trafficking, and signal transduction. A majority of the proteins involved in lipoprotein metabolism and atherogenesis, such as apolipoproteins and lipoprotein receptors, are sialylated in their glycan structures. Earlier studies in humans and in preclinical models found a positive correlation between low sialylation of lipoproteins and atherosclerosis. More recent works using loss- and gain-of-function approaches in mice have revealed molecular and cellular mechanisms by which protein sialylation modulates causally the process of atherosclerosis. The purpose of this concise review is to summarize these findings in mouse models and to provide mechanistic insights into lipoprotein sialylation and atherosclerosis.
Collapse
Affiliation(s)
- Liming Yu
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jun Peng
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Chieko Mineo,
| |
Collapse
|
16
|
Zhou M, Wu J, Tan G. The relation of circulating cell division cycle 42 expression with Th1, Th2, and Th17 cells, adhesion molecules, and biochemical indexes in coronary heart disease patients. Ir J Med Sci 2021; 191:2085-2090. [PMID: 34811660 DOI: 10.1007/s11845-021-02836-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/21/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Cell division cycle 42 (CDC42) regulates macrophage polarization, vascular inflammation, atherosclerosis progression, and modifies differentiation of T helper (Th) cells, while its potential as a biomarker in coronary heart disease (CHD) patients is still lacking. This study aimed to evaluate CDC42 expression, its correlation with Th1, Th2, and Th17 cells, adhesion molecules, and biochemical indexes in CHD patients. METHODS One hundred two CHD patients and 50 controls were enrolled. CDC42 expression in peripheral blood mononuclear cells was assessed by reverse transcription quantitative polymerase chain reaction in all participants. In CHD patients, Th1, Th2, and Th17 cells were detected by flow cytometric analysis; meanwhile, serum levels of inflammatory cytokines and adhesion molecules were detected by enzyme-linked immunosorbent assay. RESULTS CDC42 was lower in CHD patients (median (interquartile range (IQR)) = 0.431 (0.304-0.722)) than in controls (median (IQR) = 0.985 (0.572-1.760)) (p < 0.001). CDC42 was positively associated with Th2 cells (p = 0.016) and interleukin (IL)-10 (p = 0.034), but negatively correlated with Th17 cells (p < 0.001) and IL-17A (p < 0.001) in CHD patients. However, no association was found in CDC42 with Th1 cells (p = 0.199) or interferon-γ (p = 0.367) in CHD patients. Besides, CDC42 was negatively correlated with vascular cell adhesion molecule-1 (p = 0.013) and intercellular cell adhesion molecule-1 (p = 0.001) in CHD patients. Additionally, CDC42 negatively associated with C-reactive protein (p < 0.001), Gensini score (p < 0.001), total cholesterol (p = 0.039), and low-density lipoprotein cholesterol (p = 0.014), but not with other biochemical indexes (p > 0.05) in CHD patients. CONCLUSION CDC42 correlates with Th2 cells, Th17 cells, and adhesion molecules, also reflects inflammation, coronary stenosis degree, and cholesterol level in CHD patients.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Jiang'an District, Hubei, 430030, China
| | - Jian Wu
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Jiang'an District, Hubei, 430030, China
| | - Gang Tan
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Jiang'an District, Hubei, 430030, China.
| |
Collapse
|
17
|
Zinellu A, Mangoni AA. Systematic Review and Meta-Analysis of the Effect of Statins on Circulating E-Selectin, L-Selectin, and P-Selectin. Biomedicines 2021; 9:biomedicines9111707. [PMID: 34829936 PMCID: PMC8615864 DOI: 10.3390/biomedicines9111707] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/30/2022] Open
Abstract
The pleiotropic effects of statins might involve preventing inflammatory cell adhesion to the endothelium, which is a critical step in the pathogenesis of atherosclerosis. We conducted a systematic review and meta-analysis of the effects of statins on the circulating cell adhesion molecules E-Selectin, L-Selectin, and P-Selectin. A literature search was conducted in PubMed, Web of Science, and Scopus, from inception to July 2021. Risk of bias and certainty of evidence were assessed using the Joanna Briggs Institute Critical Appraisal Checklist and GRADE, respectively. In 61 studies, statins significantly reduced P-selectin (standard mean difference, SMD = -0.39, 95% CI -0.55 to -0.22, p < 0.001; moderate certainty of evidence), L-selectin (SMD = -0.49, 95% CI -0.89 to -0.10, p = 0.014; very low certainty of evidence), and E-Selectin (SMD = -0.73, 95% CI -1.02 to -0.43, p < 0.001; moderate certainty of evidence), independently of baseline lipid profile and other study and patient characteristics. The corresponding pooled SMD values in sensitivity analysis were not substantially altered when individual studies were sequentially removed. Simvastatin had a significant lowering effect on both P-selectin and E-selectin. Therefore, statins significantly reduce circulating selectins. Further studies are required to investigate whether selectin lowering mediates cardiovascular risk reduction with these agents. (PROSPERO registration number: CRD42021282778).
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA 5042, Australia
- Correspondence:
| |
Collapse
|
18
|
Cimmino G, Conte S, Morello A, Pellegrino G, Marra L, Calì G, Golino P, Cirillo P. Colchicine inhibits the prothrombotic effects of oxLDL in human endothelial cells. Vascul Pharmacol 2021; 137:106822. [PMID: 33232770 DOI: 10.1016/j.vph.2020.106822] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/26/2020] [Accepted: 11/18/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Tissue Factor (TF) plays a pivotal role in coronary thrombosis. Oxidized low-density lipoproteins (oxLDL) are crucial in development of atherosclerosclerosis. Moreover, oxLDL are known to induce TF expression on several cell types including endothelial cells. The lectin-type oxidized LDL receptor 1 (LOX-1) represent the oxLDL receptor. Colchicine is an anti-mitotic drug recently proven to have beneficial effects in cardiovascular disease via unknown mechanisms. Thus, we aim at investigating colchicine effects on TF expression in oxLDL stimulated human vascular endothelial cells (HUVEC). Some molecular mechanism(s) potentially involved were investigated. METHODS HUVEC were pre-incubated with colchicine 10 μM for 1 h and then stimulated with oxLDL (50 μg/mL). TF gene (RT-PCR), protein (western blot), surface expression (FACS) and procoagulant activity (FXa generation assay) were measured. TF translocation to cell surface was investigated by immunofluorescence. NF-κB/IκB axis was examined by western blot analysis and translocation assay. Finally, LOX-1 expression was also investigated. RESULTS Colchicine significantly reduced TF gene and protein expression as well as its procoagulant activity in oxLDL-treated HUVEC. These effects seem to be related mainly to action of colchicine on microtubules that, in turn, modulate TF trafficking in the cytoplasm, NF-κB/IκB pathway and LOX-1 expression. CONCLUSIONS Data of the present study, although in vitro, indicate that one of the hypothetical mechanisms by which colchicine exert protective cardiovascular effects might be its ability to inhibit the pro-thrombotic activity of oxLDL.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Stefano Conte
- Department of Advanced Biomedical Sciences, Division of Cardiology, University of Naples, "Federico II", Naples, Italy
| | - Andrea Morello
- Department of Advanced Biomedical Sciences, Division of Cardiology, University of Naples, "Federico II", Naples, Italy; UOC Laboratorio Analisi, Azienda Sanitaria Regionale Molise, PO "Antonio Cardarelli", Campobasso, Italy
| | - Grazia Pellegrino
- Department of Advanced Biomedical Sciences, Division of Cardiology, University of Naples, "Federico II", Naples, Italy
| | - Laura Marra
- SC Cell Biology and Biotherapy, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Gaetano Calì
- Endocrinology and Experimental Oncology Institute, CNR, Naples, Italy
| | - Paolo Golino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, Division of Cardiology, University of Naples, "Federico II", Naples, Italy.
| |
Collapse
|
19
|
Kumboyono K, Nurwidyaningtyas W, Chomsy IN, Wihastuti TA. Early Detection of Negative Smoking Impacts: Vascular Adaptation Deviation Based on Quantification of Circulated Endothelial Activation Markers. Vasc Health Risk Manag 2021; 17:103-109. [PMID: 33790567 PMCID: PMC8001718 DOI: 10.2147/vhrm.s296293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/03/2021] [Indexed: 12/04/2022] Open
Abstract
Introduction Smoking can cause vascular damage in the form of an inflammatory reaction characterized by endothelial activation. Endothelial activation forms a pathological adaptation pattern so that it can induce the atherogenesis process. Several markers, such as E-selectin, platelet-derived micro particles (PMPs) and hematopoietic stem cell (HSC) can identify the activation of endothelial in circulating blood. Therefore, the deviation of vascular adaptation due to smoking can be detected early through the feedback mechanism between E-selectin, PMPs, and HSC. Purpose This study aims to analyze the initial picture of the negative impact of smoking on vascular adaptation by measuring E-selectin, PMPs, and HSC in the peripheral blood circulation. Participant criteria and methods: Peripheral blood samples (5 mL) were taken from each participant, both the smoking group (n = 30) and the non-smoker group (n = 31) to obtain peripheral blood mononuclear cells (PBMNC). PBMNC was isolated using ficoll-based gradient centrifugation. The flow cytometry assay method used to measure the E-selectin, PMPs and hematopoietic stem cells. Results The mean of circulating E-selectin in smokers was higher than that of non-smokers. On the other hand, the average number of PMPs and HSCs in smokers was lower than non-smokers. Conclusion Smoking increases the risk of accelerated vascular block formation, as indicated by an increase in the amount of circulating E-selectin. The increase in E-selectin in the blood vessels mediates the increased adhesion of PMPs in the vascular area so that the number of circulating PMPs in smokers decreases. The decrease in circulating PMPs decreases the signal of vascular repair, which is characterized by a decline in the number of HSCs.
Collapse
Affiliation(s)
- Kumboyono Kumboyono
- Nursing Department, Faculty of Medicine, University of Brawijaya, Malang, 65145, Indonesia
| | - Wiwit Nurwidyaningtyas
- Doctoral Program of Medical Science, Faculty of Medicine, University of Brawijaya, Malang, 65145, Indonesia
| | - Indah Nur Chomsy
- Doctoral Program of Medical Science, Faculty of Medicine, University of Brawijaya, Malang, 65145, Indonesia
| | - Titin Andri Wihastuti
- Department of Biomedicine, Faculty of Medicine, University of Brawijaya, Malang, 65145, Indonesia
| |
Collapse
|
20
|
Yuliana ND, Hunaefi D, Goto M, Ishikawa YT, Verpoorte R. Measuring the health effects of food by metabolomics. Crit Rev Food Sci Nutr 2021; 62:6359-6373. [PMID: 33749380 DOI: 10.1080/10408398.2021.1901256] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Metabolomics of human biological fluids or tissues is used to discover markers for diseases by comparing the metabolome of the patients against healthy individuals. Ultimately, these markers can be used in drug discovery to determine how medications normalize (at least in part) the human metabolome at specific disease stages to homeostatic. Likewise, the health effects of food can be studied. Even metabolomics of the food can be combined with metabolomics of the treated patients to correlate compounds from food with measurable health effects from clinical studies. Various chemometric analyses of these metabolomics data are used to identify markers for diseases and to obtain evidence for health effects. This review discusses recent researches (published from 2013 to 2021) on whether specific dietary intervention to humans suffering from metabolic disorders may improve their pathological status. The scope is limited to those associated with major lifestyle diseases such as diabetes, obesity, and cardiovascular diseases, for which food is thought may have detrimental as well as beneficial effects on human health. It includes metabolites characterization of different biological samples such as the human serum/plasma, urine, saliva, feces, or ileal fluid. Whether the study results supported the claimed health benefits and whether the research was conducted with appropriate study design, was criticized.
Collapse
Affiliation(s)
- Nancy Dewi Yuliana
- Department of Food Science and Technology, IPB University, IPB Darmaga Campus, Bogor, Indonesia.,Halal Science Center IPB University, IPB Baranangsiang Campus, Bogor, Indonesia
| | - Dase Hunaefi
- Department of Food Science and Technology, IPB University, IPB Darmaga Campus, Bogor, Indonesia.,Halal Science Center IPB University, IPB Baranangsiang Campus, Bogor, Indonesia
| | - Masao Goto
- Functionality Evaluation Unit, Food Function Division, Food Research Institute, NARO, Tsukuba-Ibaraki, Japan
| | - Yuko Takano Ishikawa
- Functionality Evaluation Unit, Food Function Division, Food Research Institute, NARO, Tsukuba-Ibaraki, Japan
| | - Robert Verpoorte
- Natural Products Laboratory, Institute of Biology, Leiden University, Leiden, RA, The Netherlands
| |
Collapse
|
21
|
Drake JD, Chambers AB, Ott BR, Daiello LA. Peripheral Markers of Vascular Endothelial Dysfunction Show Independent but Additive Relationships with Brain-Based Biomarkers in Association with Functional Impairment in Alzheimer's Disease. J Alzheimers Dis 2021; 80:1553-1565. [PMID: 33720880 PMCID: PMC8150492 DOI: 10.3233/jad-200759] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cerebrovascular dysfunction confers risk for functional decline in Alzheimer's disease (AD), yet the clinical interplay of these two pathogenic processes is not well understood. OBJECTIVE We utilized Alzheimer's Disease Neuroimaging Initiative (ADNI) data to examine associations between peripherally derived soluble cell adhesion molecules (CAMs) and clinical diagnostic indicators of AD. METHODS Using generalized linear regression models, we examined cross-sectional relationships of soluble plasma vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), and E-Selectin to baseline diagnosis and functional impairment (clinical dementia rating sum-of-boxes, CDR-SB) in the ADNI cohort (n = 112 AD, n = 396 mild cognitive impairment (MCI), n = 58 cognitively normal). We further analyzed associations of these biomarkers with brain-based AD biomarkers in a subset with available cerebrospinal fluid (CSF) data (n = 351). p-values derived from main effects and interaction terms from the linear regressions were used to assess the relationship between independent and dependent variables for significance (significance level was set at 0.05 a priori for all analysis). RESULTS Higher mean VCAM-1 (p = 0.0026) and ICAM-1 (p = 0.0189) levels were found in AD versus MCI groups; however, not in MCI versus cognitively normal groups. Only VCAM-1 was linked with CDR-SB scores (p = 0.0157), and APOE ɛ4 genotype modified this effect. We observed independent, additive associations when VCAM-1 and CSF amyloid-β (Aβ42), total tau, phosphorylated tau (P-tau), or P-tau/Aβ42 (all < p = 0.01) were combined in a CDR-SB model; ICAM-1 showed a similar pattern, but to a lesser extent. CONCLUSION Our findings indicate independent associations of plasma-based vascular biomarkers and CSF biomarkers with AD-related clinical impairment.
Collapse
Affiliation(s)
- Jonathan D Drake
- Alzheimer's Disease and Memory Disorders Center, Rhode Island Hospital, Providence, RI, USA.,Department of Neurology, Brown University Warren Alpert Medical School, Providence RI, USA
| | - Alison B Chambers
- Department of Medicine, Brown University Warren Alpert Medical School, Providence RI, USA
| | - Brian R Ott
- Alzheimer's Disease and Memory Disorders Center, Rhode Island Hospital, Providence, RI, USA.,Department of Neurology, Brown University Warren Alpert Medical School, Providence RI, USA
| | - Lori A Daiello
- Alzheimer's Disease and Memory Disorders Center, Rhode Island Hospital, Providence, RI, USA.,Department of Neurology, Brown University Warren Alpert Medical School, Providence RI, USA
| | | |
Collapse
|
22
|
Patel RB, Colangelo LA, Bielinski SJ, Larson NB, Ding J, Allen NB, Michos ED, Shah SJ, Lloyd-Jones DM. Circulating Vascular Cell Adhesion Molecule-1 and Incident Heart Failure: The Multi-Ethnic Study of Atherosclerosis (MESA). J Am Heart Assoc 2020; 9:e019390. [PMID: 33161805 PMCID: PMC7763727 DOI: 10.1161/jaha.120.019390] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Serum levels of vascular cell adhesion molecule‐1 (VCAM‐1) are reflective of endothelial activation. Although VCAM‐1 has been implicated in the pathogenesis of heart failure with preserved ejection fraction (HFpEF), the prospective association of VCAM‐1 with development of clinically overt heart failure (HF) across ejection fraction categories is unclear. Methods and Results In MESA (the Multi‐Ethnic Study of Atherosclerosis), we evaluated the association of VCAM‐1 at examination 2 (2002–2004) with incident HF (HFpEF and HF with reduced ejection fraction) after adjustment for cardiovascular risk factors. Incident HF was independently adjudicated as first hospitalization for symptomatic HF. Among 2297 participants (mean age, 63 years; women, 53%), those with higher VCAM‐1 were more likely to be White race, had higher blood pressure, and had lower kidney function. Over a median of 14.4 years, there were 102 HF events (HFpEF=65; HF with reduced ejection fraction=37). After covariate adjustment, each doubling of VCAM‐1 was associated with incident HF (hazard ratio [HR], 1.94; 95% CI, 1.17–3.23; P=0.01). This association appeared stronger among current/former smokers compared with never smokers. On evaluation of HF subtypes, VCAM‐1 was associated with incident HFpEF (HR, 1.97; 95% CI, 1.04–3.72; P=0.04) but not with incident HF with reduced ejection fraction, although risk estimates were consistent (HR, 1.82; 95% CI, 0.79–4.21; P=0.16). Conclusions In a multiethnic cohort, VCAM‐1 was significantly associated with incident HF over long‐term follow‐up. These findings suggest a potential role for endothelial activation in driving clinical HF, and specifically HFpEF. Therapies that decrease endothelial activation may prevent the progression from cardiovascular risk factors to clinical HF.
Collapse
Affiliation(s)
- Ravi B Patel
- Division of Cardiology Department of Medicine Northwestern University Feinberg School of Medicine Chicago IL.,Department of Preventive Medicine Northwestern University Feinberg School of Medicine Chicago IL
| | - Laura A Colangelo
- Department of Preventive Medicine Northwestern University Feinberg School of Medicine Chicago IL
| | | | | | - Jingzhong Ding
- Department of Internal Medicine Wake Forest School of Medicine Winston-Salem NC
| | - Norrina B Allen
- Department of Preventive Medicine Northwestern University Feinberg School of Medicine Chicago IL
| | - Erin D Michos
- Division of Cardiology Department of Medicine Johns Hopkins University School of Medicine Baltimore MD
| | - Sanjiv J Shah
- Division of Cardiology Department of Medicine Northwestern University Feinberg School of Medicine Chicago IL
| | - Donald M Lloyd-Jones
- Division of Cardiology Department of Medicine Northwestern University Feinberg School of Medicine Chicago IL.,Department of Preventive Medicine Northwestern University Feinberg School of Medicine Chicago IL
| |
Collapse
|
23
|
Cimmino G, Morello A, Conte S, Pellegrino G, Marra L, Golino P, Cirillo P. Vitamin D inhibits Tissue Factor and CAMs expression in oxidized low-density lipoproteins-treated human endothelial cells by modulating NF-κB pathway. Eur J Pharmacol 2020; 885:173422. [PMID: 32755551 DOI: 10.1016/j.ejphar.2020.173422] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022]
Abstract
Epidemiologic studies have clearly demonstrated the correlation existing between Vitamin D (Vit. D) deficiency and increased risk of developing cardiovascular disease, suggesting that it might have a protective role in this clinical setting. Although many experimental studies have investigated the molecular mechanisms by which Vit. D might exert these effects, its potential role in protecting against athero-thrombosis is still partially unknown. We have investigated whether Vit. D might exert anti athero-thombotic effects by preventing expression of adhesion molecules (CAMs) and Tissue Factor (TF), molecules involved in atherothrombotic pathophysiology, in oxLDL stimulated endothelial cells (HUVEC). Moreover, we have investigated whether Vit. D effects might be due to the NF-kB modulation. HUVEC cultivated in medium enriched with Vit. D (10 nM) were stimulated with oxLDL (50 μg/ml). TF gene (RT-PCR), protein (Western blot), surface expression (FACS) and procoagulant activity (FXa generation assay) were measured. Similarly, CAMs gene (RT-PCR), surface expression (FACS) and soluble values (ELISA) were measured. NF-kB translocation was also investigated. Vit. D significantly reduced TF gene as well protein expression and procoagulant activity in oxLDL-treated HUVEC. Similar effects were observed for CAMs. These effects were associated with Vit. D modulation of NF-κB pathway. This study, although in vitro, indicate that Vit. D has protective effect on endothelial cells by inhibiting expression of TF and CAMs, proteins involved in atherothrombotic pathophysiology. Further studies will be necessary to translate these findings to a clinical scenario to better define the potential therapeutical role of Vit. D supplementation in the management of cardiovascular disease in patients with Vit. D deficiency.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Andrea Morello
- Department of Advanced Biomedical Sciences, Section of Cardiology, University of Naples "Federico II", Naples, Italy
| | - Stefano Conte
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Grazia Pellegrino
- Department of Advanced Biomedical Sciences, Section of Cardiology, University of Naples "Federico II", Naples, Italy
| | - Laura Marra
- SC Cell Biology and Biotherapy, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Paolo Golino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, Section of Cardiology, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
24
|
Piqueras L, Sanz MJ. Angiotensin II and leukocyte trafficking: New insights for an old vascular mediator. Role of redox-signaling pathways. Free Radic Biol Med 2020; 157:38-54. [PMID: 32057992 DOI: 10.1016/j.freeradbiomed.2020.02.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/27/2020] [Accepted: 02/03/2020] [Indexed: 12/20/2022]
Abstract
Inflammation and activation of the immune system are key molecular and cellular events in the pathogenesis of cardiovascular diseases, including atherosclerosis, hypertension-induced target-organ damage, and abdominal aortic aneurysm. Angiotensin II (Ang-II) is the main effector peptide hormone of the renin-angiotensin system. Beyond its role as a potent vasoconstrictor and regulator of blood pressure and fluid homeostasis, Ang-II is intimately involved in the development of vascular lesions in cardiovascular diseases through the activation of different immune cells. The migration of leukocytes from circulation to the arterial subendothelial space is a crucial immune response in lesion development that is mediated through a sequential and coordinated cascade of leukocyte-endothelial cell adhesive interactions involving an array of cell adhesion molecules present on target leukocytes and endothelial cells and the generation and release of chemoattractants that activate and guide leukocytes to sites of emigration. In this review, we outline the key events of Ang-II participation in the leukocyte recruitment cascade, the underlying mechanisms implicated, and the corresponding redox-signaling pathways. We also address the use of inhibitor drugs targeting the effects of Ang-II in the context of leukocyte infiltration in these cardiovascular pathologies, and examine the clinical data supporting the relevance of blocking Ang-II-induced vascular inflammation.
Collapse
Affiliation(s)
- Laura Piqueras
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Spanish Ministry of Health, Madrid, Spain.
| | - Maria-Jesus Sanz
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Spanish Ministry of Health, Madrid, Spain.
| |
Collapse
|
25
|
Vascular Inflammation Is a Risk Factor Associated with Brain Atrophy and Disease Severity in Parkinson's Disease: A Case-Control Study. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2591248. [PMID: 32733633 PMCID: PMC7376437 DOI: 10.1155/2020/2591248] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/04/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022]
Abstract
Introduction Systemic inflammation with elevated oxidative stress causing neuroinflammation is considered a major factor in the pathogenesis of Parkinson's disease (PD). The interface between systemic circulation and the brain parenchyma is the blood-brain barrier (BBB), which also plays a role in maintaining neurovascular homeostasis. Vascular cell adhesion molecule-1 (VCAM-1) and microRNAs (miRNAs) regulate brain vessel endothelial function, neoangiogenesis, and, in turn, neuronal homeostasis regulation, such that their dysregulation can result in neurodegeneration, such as gray matter atrophy, in PD. Objective Our aim was to evaluate the associations among specific levels of gray matter atrophy, peripheral vascular adhesion molecules, miRNAs, and clinical disease severity in order to achieve a clearer understanding of PD pathogenesis. Methods Blood samples were collected from 33 patients with PD and 27 healthy volunteers, and the levels of VCAM-1 and several miRNAs in those samples were measured. Voxel-based morphometry (VBM) analysis was performed using 3 T magnetic resonance imaging (MRI) and SPM (Statistical Parametric Mapping software program). The associations among the vascular parameter, miRNAs, gray matter volume, and clinical disease severity measurements were evaluated by partial correlation analysis. Results The levels of VCAM-1, miRNA-22, and miRNA-29a expression were significantly elevated in the PD patients. The gray matter volume atrophy in the left parahippocampus, bilateral posterior cingulate gyrus, fusiform gyrus, left temporal gyrus, and cerebellum was significantly correlated with increased clinical disease severity, the upregulation of miRNA levels, and increased vascular inflammation. Conclusion Patients with PD seem to have abnormal levels of vascular inflammatory markers and miRNAs in the peripheral circulation, and these levels are correlated with specific brain volume changes. This study reinforces the associations among peripheral inflammation, the BBB interface, and gray matter atrophy in PD and further demonstrates that BBB dysfunction with neurovascular impairment may play an important role in PD progression.
Collapse
|
26
|
Patel RB, Colangelo LA, Reiner AP, Gross MD, Jacobs DR, Launer LJ, Lima JAC, Lloyd-Jones DM, Shah SJ. Cellular Adhesion Molecules in Young Adulthood and Cardiac Function in Later Life. J Am Coll Cardiol 2020; 75:2156-2165. [PMID: 32194198 DOI: 10.1016/j.jacc.2020.02.060] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND E-selectin and intercellular adhesion molecule (ICAM)-1 are biomarkers of endothelial activation, which has been implicated in the pathogenesis of heart failure (HF) with preserved ejection fraction (HFpEF). However, the temporal associations between E-selectin and ICAM-1 with subclinical cardiac dysfunction are unclear. OBJECTIVES This study sought to assess the longitudinal associations of E-selectin and ICAM-1 with subclinical alterations in cardiac function. METHODS In the Coronary Artery Disease Risk Development in Young Adults study, a cohort of black and white young adults, we evaluated the associations of E-selectin and ICAM-1, obtained at year (Y) 7 (Y7) and Y15 examinations, with cardiac function assessed at Y30 after adjustment for key covariates. RESULTS Higher E-selectin (n = 1,810) and ICAM-1 (n = 1,548) at Y7 were associated with black race, smoking, hypertension, and higher body mass index. After multivariable adjustment, higher E-selectin at Y7 (β coefficient per 1 SD higher: 0.22; SE: 0.06; p < 0.001) and Y15 (β coefficient per 1 SD higher: 0.19; SE: 0.06; p = 0.002) was associated with worse left ventricular (LV) global longitudinal strain (GLS). Additionally, higher Y15 ICAM-1 (β coefficient per 1 SD higher: 0.18; SE: 0.06; p = 0.004) and its increase from Y7 to Y15 (β coefficient per 1 SD higher: 0.16; SE: 0.07; p = 0.03) were also independently associated with worse LV GLS. E-selectin and ICAM-1 partially mediated the associations between higher body mass index and black race with worse GLS. Neither E-selectin nor ICAM-1 was associated with measures of LV diastolic function after multivariable adjustment. CONCLUSION Circulating levels of E-selectin and ICAM-1 and increases in ICAM-1 over the course of young adulthood are associated with worse indices of LV systolic function in midlife. These findings suggest associations of endothelial activation with subclinical HF with preserved ejection fraction.
Collapse
Affiliation(s)
- Ravi B Patel
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Laura A Colangelo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - Myron D Gross
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - David R Jacobs
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, Maryland
| | - Joao A C Lima
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Donald M Lloyd-Jones
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
27
|
Bindesbøll C, Aas A, Ogmundsdottir MH, Pankiv S, Reine T, Zoncu R, Simonsen A. NBEAL1 controls SREBP2 processing and cholesterol metabolism and is a susceptibility locus for coronary artery disease. Sci Rep 2020; 10:4528. [PMID: 32161285 PMCID: PMC7066131 DOI: 10.1038/s41598-020-61352-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/21/2020] [Indexed: 01/24/2023] Open
Abstract
Dysregulated cholesterol homeostasis promotes the pathology of atherosclerosis, myocardial infarction and strokes. Cellular cholesterol is mainly regulated at the transcriptional level by SREBP2, but also through uptake of extracellular cholesterol from low density lipoproteins (LDL) via expression of LDL receptors (LDLR) at the cell surface. Identification of the mechanisms involved in regulation of these processes are thus key to understand the pathology of coronary artery disease. Here, we identify the large and poorly characterized BEACH domain protein Neurobeachin-like (NBEAL) 1 as a Golgi- associated protein required for regulation of cholesterol metabolism. NBEAL1 is most abundantly expressed in arteries. Genetic variants in NBEAL1 are associated with decreased expression of NBEAL1 in arteries and increased risk of coronary artery disease in humans. We show that NBEAL1 regulates cholesterol metabolism by modulating LDLR expression in a mechanism involving interaction with SCAP and PAQR3 and subsequent SREBP2-processing. Thus, low expression of NBEAL1 may lead to increased risk of coronary artery disease by downregulation of LDLR levels.
Collapse
Affiliation(s)
- Christian Bindesbøll
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway.
| | - Aleksander Aas
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway
| | - Margret Helga Ogmundsdottir
- Department of Biochemistry and Molecular Biology, Biomedical Center, Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101, Reykjavik, Iceland
| | - Serhiy Pankiv
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway
| | - Trine Reine
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, 1112 Blindern, 0317, Oslo, Norway.,Section for Interphase genetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, 0424, Oslo, Norway
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway.
| |
Collapse
|
28
|
Bouwens E, van den Berg VJ, Akkerhuis KM, Baart SJ, Caliskan K, Brugts JJ, Mouthaan H, van Ramshorst J, Germans T, Umans VAWM, Boersma E, Kardys I. Circulating Biomarkers of Cell Adhesion Predict Clinical Outcome in Patients with Chronic Heart Failure. J Clin Med 2020; 9:E195. [PMID: 31936828 PMCID: PMC7020068 DOI: 10.3390/jcm9010195] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular inflammation and vascular endothelial dysfunction are involved in chronic heart failure (CHF), and cellular adhesion molecules are considered to play a key role in these mechanisms. We evaluated temporal patterns of 12 blood biomarkers of cell adhesion in patients with CHF. In 263 ambulant patients, serial, tri-monthly blood samples were collected during a median follow-up of 2.2 (1.4-2.5) years. The primary endpoint (PE) was a composite of cardiovascular mortality, HF hospitalization, heart transplantation and implantation of a left ventricular assist device and was reached in 70 patients. We selected the baseline blood samples in all patients, the two samples closest to a PE, or, for event-free patients, the last sample available. In these 567 samples, associations between biomarkers and PE were investigated by joint modelling. The median age was 68 (59-76) years, with 72% men and 74% New York Heart Association class I-II. Repeatedly measured levels of Complement component C1q receptor (C1qR), Cadherin 5 (CDH5), Chitinase-3-like protein 1 (CHI3L1), Ephrin type-B receptor 4 (EPHB4), Intercellular adhesion molecule-2 (ICAM-2) and Junctional adhesion molecule A (JAM-A) were independently associated with the PE. Their rates of change also predicted clinical outcome. Level of CHI3L1 was numerically the strongest predictor with a hazard ratio (HR) (95% confidence interval) of 2.27 (1.66-3.16) per SD difference in level, followed by JAM-A (2.10, 1.42-3.23) and C1qR (1.90, 1.36-2.72), adjusted for clinical characteristics. In conclusion, temporal patterns of C1qR, CDH5, CHI3L1, EPHB4, ICAM2 and JAM-A are strongly and independently associated with clinical outcome in CHF patients.
Collapse
|
29
|
Chen Q, Lv J, Yang W, Xu B, Wang Z, Yu Z, Wu J, Yang Y, Han Y. Targeted inhibition of STAT3 as a potential treatment strategy for atherosclerosis. Theranostics 2019; 9:6424-6442. [PMID: 31588227 PMCID: PMC6771242 DOI: 10.7150/thno.35528] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is the main pathological basis of ischemic cardiovascular and cerebrovascular diseases and has attracted more attention in recent years. Multiple studies have demonstrated that the signal transducer and activator of transcription 3 (STAT3) plays essential roles in the process of atherosclerosis. Moreover, aberrant STAT3 activation has been shown to contribute to the occurrence and development of atherosclerosis. Therefore, the study of STAT3 inhibitors has gradually become a focal research topic. In this review, we describe the crucial roles of STAT3 in endothelial cell dysfunction, macrophage polarization, inflammation, and immunity during atherosclerosis. STAT3 in mitochondria is mentioned as well. Then, we present a summary and classification of STAT3 inhibitors, which could offer potential treatment strategies for atherosclerosis. Furthermore, we enumerate some of the problems that have interfered with the development of mature therapies utilizing STAT3 inhibitors to treat atherosclerosis. Finally, we propose ideas that may help to solve these problems to some extent. Collectively, this review may be useful for developing future STAT3 inhibitor therapies for atherosclerosis.
Collapse
|
30
|
Randeria SN, Thomson GJA, Nell TA, Roberts T, Pretorius E. Inflammatory cytokines in type 2 diabetes mellitus as facilitators of hypercoagulation and abnormal clot formation. Cardiovasc Diabetol 2019; 18:72. [PMID: 31164120 PMCID: PMC6549308 DOI: 10.1186/s12933-019-0870-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/16/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The global burden of type 2 diabetes mellitus (T2DM), together with the presence of cardiovascular risk in this population, is reaching pandemic levels. A prominent feature of T2DM is chronic and systemic inflammation, with the accompanying presence of circulating and dysregulated inflammatory biomarkers; which in turn is associated with abnormal clot formation. METHODS Here, we investigate the correlation between abnormal blood clotting, using thromboelastography (TEG), clot ultrastructure using scanning electron microscopy (SEM) and the presence of a dysregulated inflammatory cytokine profile, by examining various circulating biomarkers. RESULTS Our results show that many biomarkers, across TEG, cytokine and lipid groups, were greatly dysregulated in the T2DM sample. Furthermore, our T2DM sample's coagulation profiles were significantly more hypercoagulable when compared to our heathy sample, and ultrastructural analysis confirmed a matted and denser clot structure in the T2DM sample. CONCLUSIONS We suggest that dysregulated circulating molecules may in part be responsible for a hypercoagulable state and vascular dysfunction in the T2DM sample. We propose further that a personalized approach could be of great value when planning treatment and tracking the patient health status after embarking on a treatment regimes, and that looking to novel inflammatory and vascular biomarkers might be crucial.
Collapse
Affiliation(s)
- Shehan N Randeria
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Greig J A Thomson
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Theo A Nell
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Timothy Roberts
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa.
- Department of Biochemistry, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa.
| |
Collapse
|
31
|
Fonkoue IT, Le N, Kankam ML, DaCosta D, Jones TN, Marvar PJ, Park J. Sympathoexcitation and impaired arterial baroreflex sensitivity are linked to vascular inflammation in individuals with elevated resting blood pressure. Physiol Rep 2019; 7:e14057. [PMID: 30968587 PMCID: PMC6456445 DOI: 10.14814/phy2.14057] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/15/2019] [Accepted: 03/21/2019] [Indexed: 12/31/2022] Open
Abstract
Elevated Resting Blood Pressure (ERBP) in the prehypertensive range is associated with increased risk of hypertension and cardiovascular disease, the mechanisms of which remain unclear. Prior studies have suggested that ERBP may be associated with overactivation and dysregulation of the sympathetic nervous system (SNS). We hypothesized that compared to normotensives (≤120/80 mmHg), ERBP (120/80-139/89 mmHg) has higher SNS activity, impaired arterial baroreflex sensitivity (BRS), and increased vascular inflammation. Twenty-nine participants were studied: 16 otherwise healthy individuals with ERBP (blood pressure (BP) 130 ± 2/85 ± 2 mmHg) and 13 matched normotensive controls (mean BP 114 ± 2/73 ± 2 mmHg). We measured muscle sympathetic nerve activity (MSNA), beat-to-beat BP, and continuous electrocardiogram at rest and during arterial BRS testing via the modified Oxford technique. Blood was analyzed for the following biomarkers of vascular inflammation: lipoprotein-associated phospholipase A2 (Lp-PLA2), E-selectin, and intercellular adhesion molecule 1 (ICAM-1). Resting MSNA burst frequency (22 ± 2 vs. 16 ± 2 bursts/min, P = 0.036) and burst incidence (36 ± 3 vs. 25 ± 3 bursts/100 heart beats, P = 0.025) were higher in ERBP compared to controls. Cardiovagal BRS was blunted in ERBP compared to controls (13 ± 2 vs. 20 ± 3 msec/mmHg, P = 0.032), while there was no difference in sympathetic BRS between groups. Lp-PLA2 (169 ± 8 vs. 142 ± 9 nmol/min/mL, P = 0.020) and E-selectin (6.89 ± 0.6 vs. 4.45 ± 0.51 ng/mL, P = 0.004) were higher in ERBP versus controls. E-selectin (r = 0.501, P = 0.011) and ICAM-1 (r = 0.481, P = 0.015) were positively correlated with MSNA, while E-selectin was negatively correlated with cardiovagal BRS (r = -0.427, P = 0.030). These findings demonstrate that individuals with ERBP have SNS overactivity and impaired arterial BRS that are linked to biomarkers of vascular inflammation.
Collapse
Affiliation(s)
- Ida T. Fonkoue
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGeorgia
- Research Service LineAtlanta VA Healthcare SystemDecaturGeorgia
| | - Ngoc‐Anh Le
- Biomarker Core LaboratoryAtlanta VA Healthcare SystemDecaturGeorgia
| | - Melanie L. Kankam
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGeorgia
- Research Service LineAtlanta VA Healthcare SystemDecaturGeorgia
| | - Dana DaCosta
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGeorgia
- Research Service LineAtlanta VA Healthcare SystemDecaturGeorgia
| | - Toure N. Jones
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGeorgia
- Research Service LineAtlanta VA Healthcare SystemDecaturGeorgia
| | - Paul J. Marvar
- Department of Pharmacology and PhysiologyInstitute for NeuroscienceGeorge Washington UniversityWashingtonDistrict of Columbia
| | - Jeanie Park
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGeorgia
- Research Service LineAtlanta VA Healthcare SystemDecaturGeorgia
| |
Collapse
|
32
|
Jiang SJ, Tsai PI, Peng SY, Chang CC, Chung Y, Tsao HH, Huang HT, Chen SY, Hsu HJ. A potential peptide derived from cytokine receptors can bind proinflammatory cytokines as a therapeutic strategy for anti-inflammation. Sci Rep 2019; 9:2317. [PMID: 30783144 PMCID: PMC6381106 DOI: 10.1038/s41598-018-36492-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/22/2018] [Indexed: 01/19/2023] Open
Abstract
Chronic inflammation is a pivotal event in the pathogenesis of cardiovascular diseases, including atherosclerosis, restenosis, and coronary artery disease. The efficacy of current treatment or preventive strategies for such inflammation is still inadequate. Thus, new anti-inflammatory strategies are needed. In this study, based on molecular docking and structural analysis, a potential peptide KCF18 with amphiphilic properties (positively charged and hydrophobic residues) derived from the receptors of proinflammatory cytokines was designed to inhibit cytokine-induced inflammatory response. Simulations suggested that KCF18 could bind to cytokines simultaneously, and electrostatic interactions were dominant. Surface plasmon resonance detection showed that KCF18 bound to both tumor necrosis factor-α (TNF-α) and interleukin-6, which is consistent with MM/PBSA binding free energy calculations. The cell experiments showed that KCF18 significantly reduced the binding of proinflammatory cytokines to their cognate receptors, suppressed TNF-α mRNA expression and monocyte binding and transmigration, and alleviated the infiltration of white blood cells in a peritonitis mouse model. The designed peptide KCF18 could remarkably diminish the risk of vascular inflammation by decreasing plasma cytokines release and by directly acting on the vascular endothelium. This study demonstrated that a combination of structure-based in silico design calculations, together with experimental measurements can be used to develop potential anti-inflammatory agents.
Collapse
Affiliation(s)
- Shinn-Jong Jiang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Pei-I Tsai
- Department of Materials Science and Engineering, National Chiao-Tung University, Hsinchu, 30010, Taiwan
| | - Shih-Yi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Chun-Chun Chang
- Institute of Medical Sciences, Tzu Chi University, Hualien, 97004, Taiwan.,Department of Laboratory Medicine, Tzu Chi Medical Center, Hualien, 97004, Taiwan
| | - Yi Chung
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Hao-Hsiang Tsao
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Hsin-Ting Huang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - San-Yuan Chen
- Department of Materials Science and Engineering, National Chiao-Tung University, Hsinchu, 30010, Taiwan
| | - Hao-Jen Hsu
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan. .,Department of Life Sciences, Tzu Chi University, Hualien, 97004, Taiwan.
| |
Collapse
|
33
|
Bansal S, Kare PK, Tripathi AK, Madhu SV. Advanced Glycation End Products: A Potential Contributor of Oxidative Stress for Cardio-Vascular Problems in Diabetes. OXIDATIVE STRESS IN HEART DISEASES 2019:437-459. [DOI: 10.1007/978-981-13-8273-4_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
34
|
Involvement of heme oxygenase-1 induction in anti-vascular inflammation effects of Xanthoceras sorbifolia in human umbilical vein endothelial cells. J TRADIT CHIN MED 2018. [DOI: 10.1016/s0254-6272(18)30979-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
35
|
Chang WC, Yu YM, Cheng AC. Curcumin suppresses pro-inflammatory adhesion response in Human Umbilical Vein Endothelial Cells. J Food Biochem 2018. [DOI: 10.1111/jfbc.12623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Weng-Cheng Chang
- Department of Otolaryngology; Taichung Tzu Chi Hospital; Buddhist Tzu Chi Medical Foundation; Taiwan
| | - Ya-Mei Yu
- Department of Senior Citizen Service Management; National Taichung University of Science and Technology; Taichung Taiwan
| | - An-Chin Cheng
- Department of Nutrition and Health Sciences; Chang Jung Christian University; Tainan Taiwan
| |
Collapse
|
36
|
Bajaj HS, Ye C, Hanley AJ, Sermer M, Zinman B, Retnakaran R. Biomarkers of vascular injury and endothelial dysfunction after recent glucose intolerance in pregnancy. Diab Vasc Dis Res 2018; 15:449-457. [PMID: 29871496 DOI: 10.1177/1479164118779924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Women with gestational diabetes mellitus and milder gestational impaired glucose intolerance have elevated future risks of type 2 diabetes and cardiovascular disease. However, it is unclear whether they show postpartum evidence of vascular injury/dysfunction, an early event in the natural history of cardiovascular disease. METHODS In total, 337 women underwent a glucose challenge test and oral glucose tolerance test in pregnancy, yielding four gestational glucose tolerance groups: gestational diabetes mellitus, gestational impaired glucose intolerance, abnormal glucose challenge test with normal glucose tolerance on the oral glucose tolerance test and normal glucose challenge test with normal glucose tolerance. At 3 years postpartum, they underwent repeat oral glucose tolerance test (on which 69 women had pre-diabetes/diabetes) and measurement of the following serum markers of vascular injury/dysfunction: thrombomodulin, E-selectin, P-selectin, intercellular adhesion molecule-3 and vascular cell adhesion molecule-1. RESULTS At 3 years postpartum, mean adjusted vascular cell adhesion molecule-1 was the only vascular marker that differed across the previous gestational glucose tolerance groups. On multiple linear regression analysis, each strata of gestational dysglycaemia was an independent predictor of lower vascular cell adhesion molecule-1 at 3 years postpartum (gestational diabetes mellitus: p = 0.005; gestational impaired glucose intolerance: p = 0.003; abnormal glucose challenge test normal glucose tolerance: p = 0.0008), as was current pre-diabetes/diabetes ( p = 0.01). CONCLUSION Dysregulation of vascular cell adhesion molecule-1 may be an early event in the natural history of cardiovascular disease in women with recent glucose intolerance in pregnancy.
Collapse
MESH Headings
- Adult
- Antigens, CD/blood
- Biomarkers/blood
- Blood Glucose/metabolism
- Cardiovascular Diseases/blood
- Cardiovascular Diseases/diagnosis
- Cardiovascular Diseases/etiology
- Cardiovascular Diseases/physiopathology
- Cell Adhesion Molecules/blood
- Chi-Square Distribution
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/diagnosis
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes, Gestational/blood
- Diabetes, Gestational/diagnosis
- Diabetes, Gestational/physiopathology
- E-Selectin/blood
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Female
- Glucose Tolerance Test
- Humans
- Linear Models
- P-Selectin/blood
- Pregnancy
- Prognosis
- Prospective Studies
- Risk Factors
- Thrombomodulin/blood
- Time Factors
- Vascular Cell Adhesion Molecule-1/blood
Collapse
Affiliation(s)
- Harpreet S Bajaj
- 1 Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, ON, Canada
| | - Chang Ye
- 1 Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, ON, Canada
| | - Anthony J Hanley
- 1 Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, ON, Canada
- 2 Division of Endocrinology, University of Toronto, Toronto, ON, Canada
- 3 Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Mathew Sermer
- 4 Division of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada
| | - Bernard Zinman
- 1 Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, ON, Canada
- 2 Division of Endocrinology, University of Toronto, Toronto, ON, Canada
- 5 The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Ravi Retnakaran
- 1 Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, ON, Canada
- 2 Division of Endocrinology, University of Toronto, Toronto, ON, Canada
- 5 The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
37
|
Maggio ABR, Farpour-Lambert NJ, Aggoun Y, Galan K, Montecucco F, Mach F, Beghetti M. Serum cardiovascular risk biomarkers in pre-pubertal obese children. Eur J Clin Invest 2018; 48:e12995. [PMID: 29992540 DOI: 10.1111/eci.12995] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/09/2018] [Accepted: 07/09/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Childhood obesity is associated with premature cardiovascular complications. However, little is known about the effect of a family-based behavioural intervention on the relationship between arterial function, blood pressure and biomarkers in pre-pubertal children with obesity. DESIGN This was a single centre randomized controlled trial (RCT) including 74 children randomized to a 6-month behavioural intervention to treat obesity. In 48 children (13 controls and 35 interventions), we assessed: serum level of cytokine (CCL2), adiponectin, and neutrophil product (MMP-8), as well as carotid intima-media thickness, flow-mediated dilation (FMD), nitroglycerin-mediated dilation; arterial stiffness (incremental elastic modulus, Einc), pulse wave velocity (PWV), resting and 24-hour blood pressure (BP). RESULTS At baseline, resting systolic BP was positively associated with MMP-8 levels which was significantly higher in children with hypertension (P = 0.033). Biochemical markers were not related to endothelial function at baseline, but they globally increased after 6 months in the intervention group. The significant increase of CCL2 levels in the intervention group was associated with a decrease in diastolic BP. Furthermore, adiponectin change was positively related to a change in FMD and negatively to change in Einc and PWV. CONCLUSIONS The usefulness of serum biomarkers for the detection of cardiovascular diseases is not well established in children. In our population, MMP-8 concentration was higher in hypertensive children. Furthermore, behavioural interventions resulted in a paradoxical increase in some biomarkers in children, with potentially beneficial effects detected with CCL2 changes. Caution should be taken when using nonspecific serum biomarkers for the clinical monitoring of children with obesity.
Collapse
Affiliation(s)
- Albane B R Maggio
- Santé et Mouvement Consultation, Service of Paediatric Specialties, Department of Child and Adolescent, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| | - Nathalie J Farpour-Lambert
- Obesity Prevention and Care Program "Contrepoids", Service of Therapeutic Education for Chronic Diseases, Department of Community Medicine, Primary Care and Emergency, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
- Paediatric Sports Medicine Consultation, Service of General Paediatrics, Department of Child and Adolescent, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| | - Yacine Aggoun
- Paediatric Cardiology Unit, Service of Paediatric Specialties, Department of Child and Adolescent, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| | - Katia Galan
- Cardiology Division, Foundation for Medical Research, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
- Cardiology Service, Department of Internal Medicine, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - François Mach
- Cardiology Division, Foundation for Medical Research, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
- Cardiology Service, Department of Internal Medicine, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| | - Maurice Beghetti
- Paediatric Cardiology Unit, Service of Paediatric Specialties, Department of Child and Adolescent, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| |
Collapse
|
38
|
Research Progress on the Relationship between Atherosclerosis and Inflammation. Biomolecules 2018; 8:biom8030080. [PMID: 30142970 PMCID: PMC6163673 DOI: 10.3390/biom8030080] [Citation(s) in RCA: 547] [Impact Index Per Article: 78.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/03/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease; unstable atherosclerotic plaque rupture, vascular stenosis, or occlusion caused by platelet aggregation and thrombosis lead to acute cardiovascular disease. Atherosclerosis-related inflammation is mediated by proinflammatory cytokines, inflammatory signaling pathways, bioactive lipids, and adhesion molecules. This review discusses the effects of inflammation and the systemic inflammatory signaling pathway on atherosclerosis, the role of related signaling pathways in inflammation, the formation of atherosclerosis plaques, and the prospects of treating atherosclerosis by inhibiting inflammation.
Collapse
|
39
|
Elli A, Traversi L, Ponticelli C. Cardiovascular Risk Factors in Renal Transplant Recipients. Int J Artif Organs 2018. [DOI: 10.1177/039139880002301102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- A. Elli
- Nephrology and Dialysis Division, Maggiore Policlinico Hospital, IRCCS, Milano - Italy
| | - L. Traversi
- Nephrology and Dialysis Division, Maggiore Policlinico Hospital, IRCCS, Milano - Italy
| | - C. Ponticelli
- Nephrology and Dialysis Division, Maggiore Policlinico Hospital, IRCCS, Milano - Italy
| |
Collapse
|
40
|
Isasi CR, Strizich GM, Kaplan R, Daviglus ML, Sotres-Alvarez D, Vidot DC, Llabre MM, Talavera G, Carnethon MR. The association of cardiorespiratory fitness with cardiometabolic factors, markers of inflammation, and endothelial dysfunction in Latino youth: findings from the Hispanic Community Children's Health Study/Study of Latino Youth. Ann Epidemiol 2018; 28:583-589.e3. [PMID: 29548689 DOI: 10.1016/j.annepidem.2018.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 02/01/2018] [Accepted: 02/12/2018] [Indexed: 10/18/2022]
Abstract
PURPOSE To evaluate the relationship of cardiorespiratory fitness (CRF) with cardiovascular disease risk factors and a biomarker of endothelial dysfunction (e-selectin) among Hispanic/Latino youth. METHODS The study included 1380 Hispanic/Latino youths (8-16 years old) from the Hispanic Community Children's Health Study/Study of Latino Youth that enrolled from four cities (Bronx, Chicago, Miami, and San Diego). CRF was assessed by a 3-minute step test that uses postexercise heart rate to estimate maximal oxygen uptake. Regression models assessed differences in cardiometabolic markers across quartiles of CRF, adjusting for potential confounders. RESULTS CRF was higher among boys (mean: 57.6 mL per kg/min, 95% confidence interval, 56.8-58.4) compared to girls (mean: 54.7 mL per kg/min, 95% confidence interval, 53.9-55.5). Higher levels of CRF were associated with more favorable levels of cardiometabolic, inflammation, and endothelial dysfunction factors (P-values <.001) and independently of physical activity and sedentary time. Compared to the lowest quartile of CRF, the odds of having greater than or equal to two cardiovascular disease risk factors was lower at higher quartiles of CRF, after adjustment for potential confounders. CONCLUSIONS Among Hispanic/Latino youth, CRF appears to be a strong protective factor for endothelial dysfunction and cardiometabolic risk factors. Strategies to improve CRF may be a useful approach for improving cardiovascular health in youth.
Collapse
Affiliation(s)
- Carmen R Isasi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY.
| | - Garrett M Strizich
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Robert Kaplan
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Martha L Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago
| | - Daniela Sotres-Alvarez
- Collaborative Studies Coordinating Center, Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill
| | - Denise C Vidot
- Department of Psychology, University of Miami Miller School of Medicine, Miami, FL
| | - Maria M Llabre
- Department of Psychology, University of Miami, Miami, FL
| | - Gregory Talavera
- Institute for Behavioral and Community Health, Graduate School of Public Health, San Diego State University, San Diego, CA
| | - Mercedes R Carnethon
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
41
|
Kibel A, Selthofer-Relatic K, Drenjancevic I, Bacun T, Bosnjak I, Kibel D, Gros M. Coronary microvascular dysfunction in diabetes mellitus. J Int Med Res 2017; 45:1901-1929. [PMID: 28643578 PMCID: PMC5805190 DOI: 10.1177/0300060516675504] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 09/30/2016] [Indexed: 12/16/2022] Open
Abstract
The significance, mechanisms and consequences of coronary microvascular dysfunction associated with diabetes mellitus are topics into which we have insufficient insight at this time. It is widely recognized that endothelial dysfunction that is caused by diabetes in various vascular beds contributes to a wide range of complications and exerts unfavorable effects on microcirculatory regulation. The coronary microcirculation is precisely regulated through a number of interconnected physiological processes with the purpose of matching local blood flow to myocardial metabolic demands. Dysregulation of this network might contribute to varying degrees of pathological consequences. This review discusses the most important findings regarding coronary microvascular dysfunction in diabetes from pre-clinical and clinical perspectives.
Collapse
Affiliation(s)
- Aleksandar Kibel
- Department for Heart and Vascular
Diseases, Clinic of Internal Medicine, Osijek University Hospital, Osijek,
Croatia
- Department of Physiology and Immunology,
Faculty of Medicine, University of Osijek, Croatia
| | - Kristina Selthofer-Relatic
- Department for Heart and Vascular
Diseases, Clinic of Internal Medicine, Osijek University Hospital, Osijek,
Croatia
- Department of Internal Medicine, Faculty
of Medicine, University of Osijek, Osijek, Croatia
| | - Ines Drenjancevic
- Department of Physiology and Immunology,
Faculty of Medicine, University of Osijek, Croatia
| | - Tatjana Bacun
- Department of Internal Medicine, Faculty
of Medicine, University of Osijek, Osijek, Croatia
- Department of Endocrinology, Clinic of
Internal Medicine, Osijek University Hospital, Osijek, Croatia
| | - Ivica Bosnjak
- Department for Heart and Vascular
Diseases, Clinic of Internal Medicine, Osijek University Hospital, Osijek,
Croatia
| | - Dijana Kibel
- Department of Physiology and Immunology,
Faculty of Medicine, University of Osijek, Croatia
- Department of Diagnostic and
Interventional Radiology, Osijek University Hospital, Osijek, Croatia
| | - Mario Gros
- Department of Physiology and Immunology,
Faculty of Medicine, University of Osijek, Croatia
- Department of Diagnostic and
Interventional Radiology, Osijek University Hospital, Osijek, Croatia
| |
Collapse
|
42
|
Lai KSP, Liu CS, Rau A, Lanctôt KL, Köhler CA, Pakosh M, Carvalho AF, Herrmann N. Peripheral inflammatory markers in Alzheimer's disease: a systematic review and meta-analysis of 175 studies. J Neurol Neurosurg Psychiatry 2017; 88:876-882. [PMID: 28794151 DOI: 10.1136/jnnp-2017-316201] [Citation(s) in RCA: 321] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/23/2017] [Accepted: 07/17/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Increasing evidence suggests that inflammation is involved in Alzheimer's disease (AD) pathology. This study quantitatively summarised the data on peripheral inflammatory markers in patients with AD compared with healthy controls (HC). METHODS Original reports containing measurements of peripheral inflammatory markers in AD patients and HC were included for meta-analysis. Standardised mean differences were calculated using a random effects model. Meta-regression and exploration of heterogeneity was performed using publication year, age, gender, Mini-Mental State Examination (MMSE) scores, plasma versus serum measurements and immunoassay type. RESULTS A total of 175 studies were combined to review 51 analytes in 13 344 AD and 12 912 HC patients. Elevated peripheral interleukin (IL)-1β, IL-2, IL-6, IL-18, interferon-γ, homocysteine, high-sensitivity C reactive protein, C-X-C motif chemokine-10, epidermal growth factor, vascular cell adhesion molecule-1, tumour necrosis factor (TNF)-α converting enzyme, soluble TNF receptors 1 and 2, α1-antichymotrypsin and decreased IL-1 receptor antagonist and leptin were found in patients with AD compared with HC. IL-6 levels were inversely correlated with mean MMSE scores. CONCLUSIONS These findings suggest that AD is accompanied by a peripheral inflammatory response and that IL-6 may be a useful biological marker to correlate with the severity of cognitive impairment. Further studies are needed to determine the clinical utility of these markers.
Collapse
Affiliation(s)
- Ka Sing P Lai
- Neuropsychopharmacology Research Group, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Celina S Liu
- Neuropsychopharmacology Research Group, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Allison Rau
- Neuropsychopharmacology Research Group, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Krista L Lanctôt
- Neuropsychopharmacology Research Group, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Departments of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Cristiano A Köhler
- Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Maureen Pakosh
- Toronto Rehabilitation Institute, University Health Network, Toronto, Ontario, Canada
| | - André F Carvalho
- Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Nathan Herrmann
- Neuropsychopharmacology Research Group, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Departments of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Malerba M, Nardin M, Radaeli A, Montuschi P, Carpagnano GE, Clini E. The potential role of endothelial dysfunction and platelet activation in the development of thrombotic risk in COPD patients. Expert Rev Hematol 2017; 10:821-832. [PMID: 28693343 DOI: 10.1080/17474086.2017.1353416] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Despite lack of knowledge in the field, several studies have underlined the role of endothelium dysfunction and platelet activation as significant players in the development and progression of chronic obstructive pulmonary disease (COPD). Indeed, endothelium plays a crucial role in vascular homeostasis and impairment, due to the inflammation process enhanced by smoking. Chronic inflammation and endothelial dysfunction have been proved to drive platelet activity. Consequently, thrombotic risk is enhanced in COPD, and might explain the higher percentage of cardiovascular death in such patients. Areas covered: This review aims to clarify the role of endothelium function and platelet hyper-activity as the pathophysiological mechanisms of the increased thrombotic risk in COPD. Expert commentary: In COPD patients, chronic inflammation does not impact only on lung parenchyma, but potentially involves all systems, including the endothelium of blood vessels. Impaired endothelium has several consequences, such as reduced vasodilatation capacity, enhanced blood coagulation, and increased platelet activation resulting in higher risk of thrombosis in COPD patients. Endothelium dysfunction and platelet activation are potential targets of therapy in patients with COPD aiming to reduce their risk of cardiovascular events.
Collapse
Affiliation(s)
- Mario Malerba
- a Department of Internal Medicine , University of Brescia and ASST Spedali Civili , Brescia , Italy
| | - Matteo Nardin
- a Department of Internal Medicine , University of Brescia and ASST Spedali Civili , Brescia , Italy
| | | | - Paolo Montuschi
- c Department of Pharmacology, Faculty of Medicine , University Hospital Agostino Gemelli Catholic University of the Sacred Heart, Pharmacology , Rome , Italy
| | - Giovanna E Carpagnano
- d Department of Medical and Surgical Sciences , Institute of Respiratory Diseases, University of Foggia , Foggia , Italy
| | - Enrico Clini
- e Department of Medical and Surgical Sciences , University of Modena-Reggio Emilia , Modena , Italy
| |
Collapse
|
44
|
Yoon CY, Steffen LM, Gross MD, Launer LJ, Odegaard A, Reiner A, Sanchez O, Yaffe K, Sidney S, Jacobs DR. Circulating Cellular Adhesion Molecules and Cognitive Function: The Coronary Artery Risk Development in Young Adults Study. Front Cardiovasc Med 2017; 4:37. [PMID: 28596958 PMCID: PMC5442165 DOI: 10.3389/fcvm.2017.00037] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/05/2017] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Higher circulating concentrations of cellular adhesion molecules (CAMs) can be used as markers of endothelial dysfunction. Given that the brain is highly vascularized, we assessed whether endothelial function is associated with cognitive performance. METHOD Within the Coronary Artery Risk Development in Young Adults (CARDIA) Study, excluding N = 54 with stroke before year 25, we studied CAMs among N = 2,690 black and white men and women in CARDIA year 7 (1992-1993, ages 25-37) and N = 2,848 in CARDIA year 15 (2000-2001, ages 33-45). We included subjects with levels of circulating soluble CAMs measured in year 7 or 15 and cognitive function testing in year 25 (2010-2011, ages 43-55). Using multiple regression analysis, we evaluated the association between CAMs and year 25 cognitive test scores: Rey Auditory Verbal Learning Test (RAVLT, memory), Digit Symbol Substitution Test (DSST, speed of processing), and the Stroop Test (executive function). RESULT All CAM concentrations were greater in year 15 vs. year 7. Adjusting for age, race, sex, education, smoking, alcohol, diet, physical activity, participants in the fourth vs. the first quartile of CARDIA year 7 of circulating intercellular adhesion molecule-1 (ICAM-1) scored worse on RAVLT, DSST, and Stroop Test (p ≤ 0.05) in CARDIA year 25. Other CAMs showed little association with cognitive test scores. Findings were similar for ICAM-1 assessed at year 15. Adjustment for possibly mediating physical factors attenuated the findings. CONCLUSION Higher circulating ICAM-1 at average ages 32 and 40 was associated with lower cognitive skills at average age 50. The study is consistent with the hypothesis that endothelial dysfunction is associated with worse short-term memory, speed of processing, and executive function.
Collapse
Affiliation(s)
- Cynthia Yursun Yoon
- Division of Epidemiology and Community Heath, University of Minnesota, Minneapolis, MN, USA
| | - Lyn M. Steffen
- Division of Epidemiology and Community Heath, University of Minnesota, Minneapolis, MN, USA
| | - Myron D. Gross
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Lenore J. Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, MD, USA
| | - Andrew Odegaard
- Department of Epidemiology, University of California, Irvine, CA, USA
| | - Alexander Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Otto Sanchez
- Division of Renal Diseases and Hypertension, University of Minnesota, Minneapolis, MN, USA
| | - Kristine Yaffe
- Department of Psychiatry, University of California, San Francisco, CA, USA
| | - Stephen Sidney
- Division of Research, Kaiser Permanente Oakland, Oakland, CA, USA
| | - David R. Jacobs
- Division of Epidemiology and Community Heath, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
45
|
Ozderya A, Aydin K, Temizkan S, Dogru Abbasoglu S, Vural P, Altuntas Y. High circulating levels of sICAM-1 and sVCAM-1 in the patients with Hashimoto's thyroiditis. Endocr Res 2017; 42:110-116. [PMID: 27366829 DOI: 10.1080/07435800.2016.1201100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To investigate whether soluble intercellular adhesion molecule-1 (sICAM-1) and soluble vascular cell adhesion molecule-1 (sVCAM-1) levels are increased in euthyroid patients with Hashimoto's thyroiditis (HT) and whether they are associated with thyroid autoimmunity and metabolic parameters. DESIGN Cross-sectional. SUBJECTS AND METHODS In total, 80 euthyroid patients with HT and 80 age- and body mass index (BMI)-matched control participants were included. Serum sICAM-1, sVCAM-1, free triiodothyronine (fT3), free thyroxine (fT4), thyroid-stimulating hormone (TSH), thyroid peroxidase antibody (anti-TPO), thyroglobulin antibody (anti-TG), fasting blood glucose (FBG), insulin, and lipid levels and homeostasis model assessment for insulin resistance (HOMA-IR) were assessed in all participants. RESULTS The patients with HT had significantly higher levels of sICAM-1 and sVCAM-1 than controls (both p < 0.001). The difference was sustained after adjustment for TSH and levothyroxine use. Regression analysis demonstrated that sICAM-1 was related to anti-TPO (p < 0.001), and sVCAM-1 was related to both anti-TPO and-TG (p < 0.001 and p = 0.03, respectively); this relationship was sustained after adjustment for age and BMI. Although FBG and HOMA-IR were higher in the HT group, logistic regression analysis revealed that there was no effect of anti-TPO, anti-TG, sICAM-1, sVCAM-1, and C-reactive protein (CRP) on the occurrence of high FBG and high HOMA-IR. CONCLUSION sICAM-1 and sVCAM-1 levels were significantly elevated in the patients with euthyroid HT and correlated closely with thyroid autoimmunity. However, soluble adhesion molecules had no relation with glucose metabolism parameters in the HT patients.
Collapse
Affiliation(s)
- Aysenur Ozderya
- a Department of Endocrinology and Metabolism , Kartal Dr. Lutfi Kirdar Training and Research Hospital , Istanbul , Turkey
| | - Kadriye Aydin
- a Department of Endocrinology and Metabolism , Kartal Dr. Lutfi Kirdar Training and Research Hospital , Istanbul , Turkey
| | - Sule Temizkan
- a Department of Endocrinology and Metabolism , Kartal Dr. Lutfi Kirdar Training and Research Hospital , Istanbul , Turkey
| | - Semra Dogru Abbasoglu
- b Department of Biochemistry, Istanbul University , Istanbul Faculty of Medicine , Istanbul , Turkey
| | - Pervin Vural
- b Department of Biochemistry, Istanbul University , Istanbul Faculty of Medicine , Istanbul , Turkey
| | - Yuksel Altuntas
- c Department of Endocrinology and Metabolism , Sisli Hamidiye Etfal Training and Research Hospital , Istanbul , Turkey
| |
Collapse
|
46
|
Parrinello CM, Hua S, Carnethon MR, Gallo LC, Hudson BI, Goldberg RB, Delamater AM, Kaplan RC, Isasi CR. Associations of hyperglycemia and insulin resistance with biomarkers of endothelial dysfunction in Hispanic/Latino youths: Results from the Hispanic Community Children's Health Study/Study of Latino Youth (SOL Youth). J Diabetes Complications 2017; 31:836-842. [PMID: 28242270 PMCID: PMC6119470 DOI: 10.1016/j.jdiacomp.2017.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 01/22/2017] [Accepted: 01/26/2017] [Indexed: 12/14/2022]
Abstract
AIMS We hypothesized that Hispanic/Latino youth at high risk for diabetes would have elevated biomarkers of endothelial dysfunction. METHODS Among 1316 children 8-16years old from the Study of Latino Youth (SOL Youth), we used Poisson regression to obtain prevalence ratios (PRs) and 95% CIs for the cross-sectional association of quartiles of fasting glucose, HbA1c, and insulin resistance with E-selectin and plasminogen activator inhibitor-1 (PAI-1) levels above the median (≥48.1 and ≥2.02ng/mL, respectively). RESULTS Levels of E-selectin and PAI-1 were higher in children who were obese or had higher levels of hs-CRP (p<0.05). Insulin resistance was independently associated with higher levels of PAI-1 (adjusted PR and 95% CI for the highest versus lowest quartile (Q4 vs Q1): 2.25 [1.64, 3.09]). We found stronger evidence of associations of insulin resistance with higher levels of PAI-1 among boys as compared with girls (p-interaction = 0.10). CONCLUSIONS Insulin resistance was associated with endothelial dysfunction, as measured by higher levels of PAI-1, in Hispanic/Latino youth. These biomarkers may be useful in risk stratification and prediction of diabetes and cardiovascular disease in high-risk youth.
Collapse
Affiliation(s)
- Christina M Parrinello
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Simin Hua
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mercedes R Carnethon
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Linda C Gallo
- Department of Psychology, San Diego State University, San Diego, CA, USA
| | - Barry I Hudson
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Ronald B Goldberg
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alan M Delamater
- Department of Pediatrics, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Carmen R Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
47
|
Nygaard H, Falch GS, Whist JE, Hollan I, Ellefsen S, Holmboe-Ottesen G, Rønnestad BR, Høstmark AT. Acute effects of post-absorptive and postprandial moderate exercise on markers of inflammation in hyperglycemic individuals. Eur J Appl Physiol 2017; 117:787-794. [PMID: 28255731 DOI: 10.1007/s00421-017-3576-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/16/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE Systemic inflammation is involved in the development of several diseases, including cardiovascular disease and type 2 diabetes. It is known that vigorous exercise affects systemic inflammation, but less is known about exercise at lower intensities. Hyperglycemia can also entail pro-inflammatory responses; however, postprandial hyperglycemia is blunted if the meal is followed by exercise. Hypotheses were: (1) moderate physical exercise acutely affects levels of C-reactive protein (CRP) and serum soluble vascular cell adhesion molecule 1 (sVCAM-1) in hyperglycemic individuals and (2) the effect depends on whether the activity is performed in a post-absorptive or postprandial state. METHODS Twelve participants diagnosed with hyperglycemia, but not using anti-diabetic medication, underwent three test days in a randomized cross-over study; 1 control day without exercise, 1 day with 60 min of treadmill walking ending 30 min before breakfast, and 1 day with an identical bout of activity 30 min after the start of breakfast. Food intake was strictly standardized and venous blood for CRP, and sVCAM-1 analysis was sampled at standardized timepoints during the first 3.5 h after breakfast and once 24 h later. RESULTS Merged data from the two exercise days showed that sVCAM-1 increased from baseline (4 ± 16 ng/mL) compared to the control condition (-28 ± 47 ng/mL, ES = 0.7, p = 0.024). There was no statistically significant difference in changes in sVCAM-1 levels between the two exercise test days. Exercise did not affect CRP values. CONCLUSION Moderate exercise increases sVCAM-1 in hyperglycemic individuals, whereas it does not affect CRP.
Collapse
Affiliation(s)
- Håvard Nygaard
- Section for Sport Science, Lillehammer University College, PB 952, 2604, Lillehammer, Norway.
| | | | - Jon Elling Whist
- Department of Medical Biochemistry, Innlandet Hospital Trust, PB 990, 2629, Lillehammer, Norway
- Department of Research, Innlandet Hospital Trust, Brumunddal, Norway
| | - Ivana Hollan
- Hospital for Rheumatic Diseases, Margrethe Grundtvigs veg 6, 2609, Lillehammer, Norway
- Department of Research, Innlandet Hospital Trust, Brumunddal, Norway
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Stian Ellefsen
- Section for Sport Science, Lillehammer University College, PB 952, 2604, Lillehammer, Norway
| | - Gerd Holmboe-Ottesen
- Department of Community Medicine, Institute of Health and Society, University of Oslo, PB 1130 Blindern, 0318, Oslo, Norway
| | - Bent R Rønnestad
- Section for Sport Science, Lillehammer University College, PB 952, 2604, Lillehammer, Norway
| | - Arne T Høstmark
- Department of Community Medicine, Institute of Health and Society, University of Oslo, PB 1130 Blindern, 0318, Oslo, Norway
| |
Collapse
|
48
|
Kunutsor SK, Bakker SJL, Dullaart RPF. Soluble Vascular Cell Adhesion Molecules May be Protective of Future Cardiovascular Disease Risk: Findings from the PREVEND Prospective Cohort Study. J Atheroscler Thromb 2017; 24:804-818. [PMID: 28202840 PMCID: PMC5556188 DOI: 10.5551/jat.38836] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim: Soluble cell adhesion molecules, such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1, E-selectin, and P-selectin, have been suggested to be associated with cardiovascular disease (CVD) risk; however, the nature and magnitude of the association between VCAM-1 and CVD risk is uncertain. We aimed to assess the association of VCAM-1 with CVD risk and determine its potential utility for CVD risk prediction. Methods: VCAM-1 concentrations were measured at baseline in the PREVEND prospective study of 2,638 participants. Hazard ratios (95% confidence intervals [CI]) and measures of risk discrimination for CVD (e.g., C-index) and reclassification (i.e., net reclassification improvement) of participants were assessed. Results: During a median follow-up of 9.9 years, 614 CVD events occurred. Plasma VCAM-1 was weakly associated with several cardiovascular risk markers. In analyses adjusted for established cardiovascular risk factors, the hazard ratio (95% CI) for CVD per 1 standard deviation increase in loge VCAM-1 was 0.91 (0.84–0.99; P = 0.020), which remained consistent after additional adjustment for body mass index, alcohol consumption, triglycerides, renal function, and C-reactive protein; hazard ratio (95% CI) 0.89 (0.82–0.97; P = 0.006). Comparing the top versus bottom quintiles of VCAM-1 levels, the corresponding adjusted hazard ratios were 0.74 (0.57–0.96; P = 0.023) and 0.70 (0.54–0.91; P = 0.007) respectively. Adding VCAM-1 to a CVD risk prediction model containing conventional risk factors did not improve the C-index or net reclassification. Conclusions: Plasma VCAM-1 is inversely and independently associated with CVD. However, VCAM-1 provides no significant improvement in CVD risk assessment beyond conventional CVD risk factors.
Collapse
Affiliation(s)
| | - Stephan J L Bakker
- Department of Nephrology Medicine, University of Groningen and University Medical Center Groningen.,Top Institute Food and Nutrition
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen and University Medical Center Groningen
| |
Collapse
|
49
|
Rallidis LS, Kolomvotsou A, Lekakis J, Farajian P, Vamvakou G, Dagres N, Zolindaki M, Efstathiou S, Anastasiou-Nana M, Zampelas A. Short-term effects of Mediterranean-type diet intervention on soluble cellular adhesion molecules in subjects with abdominal obesity. Clin Nutr ESPEN 2016; 17:38-43. [PMID: 28361746 DOI: 10.1016/j.clnesp.2016.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 07/25/2016] [Accepted: 11/28/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND & AIMS Abdominal obesity (AO) is associated with increased risk for cardiovascular disease and with increased production of adhesion molecules. The present work examined the effect of a Mediterranean-style diet on soluble cellular adhesion molecules in individuals with AO. METHODS Ninety subjects with AO without cardiovascular disease or diabetes mellitus were randomly allocated to the intervention or control group and were instructed to follow a Mediterranean-style diet for two months. Intervention group followed a specific relevant food plan with close dietetic supervision and provision of basic foods. Soluble intercellular adhesion molecule-1 (sICAM-1), soluble vascular cell adhesion molecule-1 (sVCAM-1), sP and sE-selectin, C-reactive protein (CRP) and interleukin-6 (IL-6) were measured. RESULTS Subjects in the intervention group increased their intake of total fat, monounsaturated fatty acids, dietary fiber, vitamin C, and alcohol compared to controls, while decreased their intake of saturated fat. Although there was a significant decrease in CRP, sP-selectin and in sE-selectin in the intervention group, and an increase in sVCAM-1 in the control group, between-group analysis showed no statistically significant differences. There were also no significant changes in sICAM-1, and IL-6 levels after intervention. CONCLUSIONS Mediterranean-type diet for two months combined with close dietetic supervision showed a beneficial tendency towards the down-regulation of some markers of vascular inflammation, although the comparison between groups after the intervention did not reach statistical significance. A longer period of dietary intervention may be required to further support these changes.
Collapse
Affiliation(s)
- Loukianos S Rallidis
- Second Department of Cardiology, Attikon Hospital, School of Medicine, University of Athens, Greece
| | - Anastasia Kolomvotsou
- Unit of Human Nutrition, Department of Food Science and Human Nutrition, Agricultural University of Athens, Greece
| | - John Lekakis
- Second Department of Cardiology, Attikon Hospital, School of Medicine, University of Athens, Greece
| | - Paul Farajian
- Unit of Human Nutrition, Department of Food Science and Human Nutrition, Agricultural University of Athens, Greece
| | - Georgia Vamvakou
- Second Department of Cardiology, Attikon Hospital, School of Medicine, University of Athens, Greece
| | - Nikolaos Dagres
- Second Department of Cardiology, Attikon Hospital, School of Medicine, University of Athens, Greece
| | - Maria Zolindaki
- Biochemistry Laboratory, General Hospital of Nikea, Piraeus, Greece
| | | | - Maria Anastasiou-Nana
- Second Department of Cardiology, Attikon Hospital, School of Medicine, University of Athens, Greece
| | - Antonis Zampelas
- Unit of Human Nutrition, Department of Food Science and Human Nutrition, Agricultural University of Athens, Greece.
| |
Collapse
|
50
|
Daan NMP, Koster MPH, de Wilde MA, Dalmeijer GW, Evelein AMV, Fauser BCJM, de Jager W. Biomarker Profiles in Women with PCOS and PCOS Offspring; A Pilot Study. PLoS One 2016; 11:e0165033. [PMID: 27806063 PMCID: PMC5091782 DOI: 10.1371/journal.pone.0165033] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 10/05/2016] [Indexed: 11/22/2022] Open
Abstract
Objective To study metabolic/inflammatory biomarker risk profiles in women with PCOS and PCOS offspring. Design Cross-sectional comparison of serum biomarkers. Setting University Medical Center Utrecht. Patients Hyperandrogenic PCOS women (HA-PCOS, n = 34), normoandrogenic PCOS women (NA-PCOS, n = 34), non-PCOS reference population (n = 32), PCOS offspring (n = 14, age 6–8 years), and a paedriatic reference population (n = 30). Main Outcome Measure(s) Clustering profile of adipocytokines (IL-1b, IL-6, IL-13, IL-17, IL-18, TNF-α, adiponectin, adipsin, leptin, chemerin, resistin, RBP4, DPP-IV/sCD26, CCL2/MCP-1), growth factors (PIGF, VEGF, sVEGF-R1), soluble cell adhesion molecules (sICAM-1/sCD54, sVCAM-1/sCD106), and other inflammatory related proteases (MMP-9, S100A8, Cathepsin S). Differences in median biomarker concentrations between groups, and associations with the free androgen index (FAI; Testosterone/SHBG x100). Results The cluster analysis identified leptin, RBP-4, DPP-IV and adiponectin as potential discriminative markers for HA-PCOS with a specifically strong correlation in cases with increased BMI. Leptin (R2 = 0.219) and adiponectin (R2 = 0.182) showed the strongest correlation with the FAI. When comparing median protein concentrations adult PCOS women with or without hyperandrogenemia, the most profound differences were observed for leptin (P < 0.001), DPP-IV (P = 0.005), and adiponectin (P < 0.001). Adjusting for age, BMI and multiple testing attenuated all differences. In PCOS offspring, MMP-9 (P = 0.001) and S100A8 (P < 0.001) concentrations were significantly higher compared to a healthy matched reference population, even after correcting for age and BMI and adjustment for multiple testing. Conclusion In this preliminary investigation we observed significant differences in adipocytokines between women with or without hyperandrogenic PCOS and non-PCOS controls, mostly influenced by BMI. Leptin and adiponectin showed the strongest correlation with the FAI in adult women with PCOS. In PCOS offspring other inflammatory biomarkers (MMP-9, S100A8) were increased, suggesting that these children may exhibit increased chronic low-grade inflammation. Additional research is required to confirm results of the current exploratory investigation.
Collapse
Affiliation(s)
- Nadine M. P. Daan
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
- * E-mail:
| | - Maria P. H. Koster
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marlieke A. de Wilde
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Gerdien W. Dalmeijer
- Julius Centre for Health Sciences and Primary care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Annemieke M. V. Evelein
- Julius Centre for Health Sciences and Primary care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bart C. J. M. Fauser
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wilco de Jager
- Laboratory of Translational Immunology, Division of Pediatrics, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| |
Collapse
|