1
|
Barachini S, Ghelardoni S, Varga ZV, Mehanna RA, Montt-Guevara MM, Ferdinandy P, Madonna R. Antineoplastic drugs inducing cardiac and vascular toxicity - An update. Vascul Pharmacol 2023; 153:107223. [PMID: 37678516 DOI: 10.1016/j.vph.2023.107223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/14/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
With the improvement in cancer prognosis due to advances in antitumor therapeutic protocols and new targeted and immunotherapies, we are witnessing a growing increase in survival, however, at the same timeincrease in morbidity among cancer survivors as a consequences of the increased cardiovascular adverse effects of antineoplastic drugs. Common cardiovascular complications of antineoplastic therapies may include cardiac complications such as arrhythmias, myocardial ischemia, left ventricular dysfunction culminating in heart failure as well as vascular complications including arterial hypertension, thromboembolic events, and accelerated atherosclerosis. The toxicity results from the fact that these drugs not only target cancer cells but also affect normal cells within the cardiovascular system. In this article, we review the clinical features and main mechanisms implicated in antineoplastic drug-induced cardiovascular toxicity, including oxidative stress, inflammation, immunothrombosis and growth factors-induced signaling pathways.
Collapse
Affiliation(s)
- Serena Barachini
- Department of Clinical and Experimental Medicine, Laboratory for Cell Therapy, University of Pisa, Pisa, Italy
| | - Sandra Ghelardoni
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Pisa, Italy
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary; MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Radwa A Mehanna
- Medical Physiology Department, Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Egypt
| | | | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, Pisa, Italy.
| |
Collapse
|
2
|
Caradonna E, Mormone E, Centritto EM, Mazzanti A, Papini S, Fanelli M, Petrella L, Petruzziello A, Farina MA, Farina E, Amato B, De Filippo CM, Vanoli E. Different methods of bone marrow harvesting influence cell characteristics and purity, affecting clinical outcomes. JVS Vasc Sci 2023; 4:100130. [PMID: 38058747 PMCID: PMC10696233 DOI: 10.1016/j.jvssci.2023.100130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/17/2023] [Indexed: 12/08/2023] Open
Abstract
Background Bone marrow (BM)-derived stem cells were implanted to induce angiogenesis in patients with no-option critical limb-threatening ischemia. Considering the potential for this therapy, conflicting results related to BM harvesting methods have been reported that could affect stem cell concentrations and quality. Methods A total of 75 patients with no-option critical limb-threatening ischemia were treated with BM implantation. For 58 patients, BM was harvested using a BM aspirate concentrate system (Harvest Technologies; group HT) with a standard aspiration needle, followed by an automated centrifugation process, to produce BM aspirate concentrate. For 17 patients, BM was harvested using the Marrow Cellution system (Aspire Medical Innovation; group MC). CD34+ cells/mL, CD117+ cells/mL, CD133+ cells/mL, CD309+ cells/mL, hematocrit, and BM purity were compared between the two BM preparations. Results The retrospective analysis of a subset group after adjustment for age shows that the quality of BM obtained using the Marrow Cellution system is better, in terms of purity, than the classic harvesting method before centrifugation. Harvested BM before centrifugation is characterized by a higher percentage of CD133+ cells compared with BM after centrifugation. In contrast, the MC aspirate had a larger amount of very small embryonic-like cells, as indicated by the higher percentage of CD133+, CD34+, and CD45- cells. These differences translated into an increased occurrence of leg amputations in group HT than in group MC and an increase in transcutaneous oxygen pressure in patients treated with BM aspirated using MC. Conclusions BM manipulation, such as centrifugation, affects the quality and number of stem cells, with detrimental consequences on clinical outcomes, as reflected by the different amputation rates between the two groups.
Collapse
Affiliation(s)
| | - Elisabetta Mormone
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | | | - Andrea Mazzanti
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Unit of Molecular Cardiology, ICS Maugeri, Pavia, Italy
| | - Stefano Papini
- Clinical and Research Laboratory, Gemelli Molise S.p.A., Campobasso, Italy
| | - Mara Fanelli
- Laboratorio di Diagnostica Molecolare, Gemelli Molise S.p.A., Campobasso, Italy
| | - Lella Petrella
- Laboratorio di Diagnostica Molecolare, Gemelli Molise S.p.A., Campobasso, Italy
| | - Arnolfo Petruzziello
- UOC Patologia Clinica, Dipartimento dei Servizi Sanitari, AORN CASERTA, Caserta, Italy
| | | | | | - Bruno Amato
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Emilio Vanoli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Cardiology Unit, Sacra Famiglia Hospital, Erba, Italy
| |
Collapse
|
3
|
Wang Y, Li Y, Liu D. Erythropoietin promoted intraplaque angiogenesis by PI3K/AKT/mTOR signaling pathway in atherosclerosis. Tissue Cell 2023; 82:102084. [PMID: 37060746 DOI: 10.1016/j.tice.2023.102084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND This study aimed to investigate role of erythropoietin in atherosclerosis and explore whether underlying mechanism is associated with PI3K/AKT/mTOR pathway. METHODS High-fat-diet-induced atherosclerosis model was established in apolipoprotein E knockout mice (C57BL/6 genetic background). Mice were randomly divided into the control group and the EPO group. Hematoxylin-eosin was performed for the determination of atherosclerotic lesions. The expression levels of related proteins were detected by western blot analysis. RESULTS Erythropoietin significantly enhanced the incidence of hemorrhage in atherosclerotic plaques compared with the control group. The proteins' expression signaling pathways (including PI3K, AKT, and mTOR) and angiogenesis-related proteins (VEGF, COX-2, and HIF-1α) were proved to be up-regulated by erythropoietin. Additionally, erythropoietin significantly enhanced the incidence of hemorrhage in the atherosclerotic plaques compared with the control group. The vitro experiments were conducted in macrophages at 21% O2 or 1% O2. The data showed that expression of p-PI3K, p-AKT, p-mTOR, VEGF, COX-2, and HIF-1α related proteins increased in 1% O2 group than 21% O2 group. Moreover, compared with control group, protein expression including p-PI3K, p-AKT, p-mTOR, VEGF, COX-2, and HIF-1α was markedly increased in EPO group, decreased in inhibitors group, and similar results were observed in EPO+ inhibitors group. CONCLUSION The present study demonstrated that erythropoietin might promote angiogenesis in atherosclerotic vulnerable by activating PI3K/AKT/mTOR signaling pathway in atherosclerotic, providing a novel therapeutic target for atherosclerotic targeted therapy.
Collapse
Affiliation(s)
- Ying Wang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, P.R. China.; Department of Cardiology, Hebei General Hospital, Shijiazhuang, P.R. China
| | - Yongjun Li
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, P.R. China.; Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China.
| | - Dongxia Liu
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, P.R. China
| |
Collapse
|
4
|
Chen L, Yu YN, Liu J, Chen YY, Wang B, Qi YF, Guan S, Liu X, Li B, Zhang YY, Hu Y, Wang Z. Modular networks and genomic variation during progression from stable angina pectoris through ischemic cardiomyopathy to chronic heart failure. Mol Med 2022; 28:140. [DOI: 10.1186/s10020-022-00569-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 11/04/2022] [Indexed: 11/28/2022] Open
Abstract
Abstract
Background
Analyzing disease–disease relationships plays an important role for understanding etiology, disease classification, and drug repositioning. However, as cardiovascular diseases with causative links, the molecular relationship among stable angina pectoris (SAP), ischemic cardiomyopathy (ICM) and chronic heart failure (CHF) is not clear.
Methods
In this study, by integrating the multi-database data, we constructed paired disease progression modules (PDPMs) to identified relationship among SAP, ICM and CHF based on module reconstruction pairs (MRPs) of K-value calculation (a Euclidean distance optimization by integrating module topology parameters and their weights) methods. Finally, enrichment analysis, literature validation and structural variation (SV) were performed to verify the relationship between the three diseases in PDPMs.
Results
Total 16 PDPMs were found with K > 0.3777 among SAP, ICM and CHF, in which 6 pairs in SAP–ICM, 5 pairs for both ICM–CHF and SAP–CHF. SAP–ICM was the most closely related by having the smallest average K-value (K = 0.3899) while the maximum is SAP–CHF (K = 0.4006). According to the function of the validation gene, inflammatory response were through each stage of SAP–ICM–CHF, while SAP–ICM was uniquely involved in fibrosis, and genes were related in affecting the upstream of PI3K–Akt signaling pathway. 4 of the 11 genes (FLT1, KDR, ANGPT2 and PGF) in SAP–ICM–CHF related to angiogenesis in HIF-1 signaling pathway. Furthermore, we identified 62.96% SVs were protein deletion in SAP–ICM–CHF, and 53.85% SVs were defined as protein replication in SAP–ICM, while ICM–CHF genes were mainly affected by protein deletion.
Conclusion
The PDPMs analysis approach combined with genomic structural variation provides a new avenue for determining target associations contributing to disease progression and reveals that inflammation and angiogenesis may be important links among SAP, ICM and CHF progression.
Collapse
|
5
|
Zhu QM, MacDonald BT, Mizoguchi T, Chaffin M, Leed A, Arduini A, Malolepsza E, Lage K, Kaushik VK, Kathiresan S, Ellinor PT. Endothelial ARHGEF26 is an angiogenic factor promoting VEGF signalling. Cardiovasc Res 2022; 118:2833-2846. [PMID: 34849650 PMCID: PMC9586566 DOI: 10.1093/cvr/cvab344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Indexed: 12/22/2022] Open
Abstract
AIMS Genetic studies have implicated the ARHGEF26 locus in the risk of coronary artery disease (CAD). However, the causal pathways by which DNA variants at the ARHGEF26 locus confer risk for CAD are incompletely understood. We sought to elucidate the mechanism responsible for the enhanced risk of CAD associated with the ARHGEF26 locus. METHODS AND RESULTS In a conditional analysis of the ARHGEF26 locus, we show that the sentinel CAD-risk signal is significantly associated with various non-lipid vascular phenotypes. In human endothelial cell (EC), ARHGEF26 promotes the angiogenic capacity, and interacts with known angiogenic factors and pathways. Quantitative mass spectrometry showed that one CAD-risk coding variant, rs12493885 (p.Val29Leu), resulted in a gain-of-function ARHGEF26 that enhances proangiogenic signalling and displays enhanced interactions with several proteins partially related to the angiogenic pathway. ARHGEF26 is required for endothelial angiogenesis by promoting macropinocytosis of Vascular Endothelial Growth Factor Receptor 2 (VEGFR2) on cell membrane and is crucial to Vascular Endothelial Growth Factor (VEGF)-dependent murine vessel sprouting ex vivo. In vivo, global or tissue-specific deletion of ARHGEF26 in EC, but not in vascular smooth muscle cells, significantly reduced atherosclerosis in mice, with enhanced plaque stability. CONCLUSIONS Our results demonstrate that ARHGEF26 is involved in angiogenesis signaling, and that DNA variants within ARHGEF26 that are associated with CAD risk could affect angiogenic processes by potentiating VEGF-dependent angiogenesis.
Collapse
Affiliation(s)
- Qiuyu Martin Zhu
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Bryan T MacDonald
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Taiji Mizoguchi
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Mark Chaffin
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Alison Leed
- Center for the Development of Therapeutics, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Alessandro Arduini
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
| | - Edyta Malolepsza
- Genomics Platform, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kasper Lage
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Virendar K Kaushik
- Center for the Development of Therapeutics, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Sekar Kathiresan
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Verve Therapeutics, Cambridge, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
6
|
Donis N, Jiang Z, D'Emal C, Hulin A, Debuisson M, Dulgheru R, Nguyen ML, Postolache A, Lallemand F, Coucke P, Martinive P, Herzog M, Pamart D, Terrell J, Pincemail J, Drion P, Delvenne P, Nchimi A, Lancellotti P, Oury C. Differential Biological Effects of Dietary Lipids and Irradiation on the Aorta, Aortic Valve, and the Mitral Valve. Front Cardiovasc Med 2022; 9:839720. [PMID: 35295264 PMCID: PMC8918952 DOI: 10.3389/fcvm.2022.839720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 01/12/2023] Open
Abstract
Aims Dietary cholesterol and palmitic acid are risk factors for cardiovascular diseases (CVDs) affecting the arteries and the heart valves. The ionizing radiation that is frequently used as an anticancer treatment promotes CVD. The specific pathophysiology of these distinct disease manifestations is poorly understood. We, therefore, studied the biological effects of these dietary lipids and their cardiac irradiation on the arteries and the heart valves in the rabbit models of CVD. Methods and Results Cholesterol-enriched diet led to the thickening of the aortic wall and the aortic valve leaflets, immune cell infiltration in the aorta, mitral and aortic valves, as well as aortic valve calcification. Numerous cells expressing α-smooth muscle actin were detected in both the mitral and aortic valves. Lard-enriched diet induced massive aorta and aortic valve calcification, with no detectable immune cell infiltration. The addition of cardiac irradiation to the cholesterol diet yielded more calcification and more immune cell infiltrates in the atheroma and the aortic valve than cholesterol alone. RNA sequencing (RNAseq) analyses of aorta and heart valves revealed that a cholesterol-enriched diet mainly triggered inflammation-related biological processes in the aorta, aortic and mitral valves, which was further enhanced by cardiac irradiation. Lard-enriched diet rather affected calcification- and muscle-related processes in the aorta and aortic valve, respectively. Neutrophil count and systemic levels of platelet factor 4 and ent-8-iso-15(S)-PGF2α were identified as early biomarkers of cholesterol-induced tissue alterations, while cardiac irradiation resulted in elevated levels of circulating nucleosomes. Conclusion Dietary cholesterol, palmitic acid, and cardiac irradiation combined with a cholesterol-rich diet led to the development of distinct vascular and valvular lesions and changes in the circulating biomarkers. Hence, our study highlights unprecedented specificities related to common risk factors that underlie CVD.
Collapse
Affiliation(s)
- Nathalie Donis
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Zheshen Jiang
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Céline D'Emal
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Alexia Hulin
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Margaux Debuisson
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Raluca Dulgheru
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Mai-Linh Nguyen
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Adriana Postolache
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | | | | | - Philippe Martinive
- Department Radiation Oncology, Institut Jules Bordet, Université Libre Bruxelles, Brussels, Belgium
| | - Marielle Herzog
- Belgian Volition Société à Responsabilité Limitée, Gembloux, Belgium
| | - Dorian Pamart
- Belgian Volition Société à Responsabilité Limitée, Gembloux, Belgium
| | - Jason Terrell
- Department of Oncology and Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Volition America, Austin, TX, United States
| | | | - Pierre Drion
- Experimental Surgery Unit, Centre de Recherche du Département de Chrirurgie, Groupe Interdisciplinaire de Géno-Protéomique Appliquée Institute, University of Liège, Liège, Belgium
| | - Philippe Delvenne
- Department of Pathology, Centre Hospitalier Universitaire of Liège, Liège, Belgium
- Laboratory of Experimental Pathology, GIGA Institute, University of Liège, Liège, Belgium
| | - Alain Nchimi
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| | - Patrizio Lancellotti
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
- Gruppo Villa Maria Care and Research, Maria Cecilia Hospital, Cotignola, Italy
- Anthea Hospital, Bari, Italy
| | - Cécile Oury
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, CHU Sart Tilman, Liège, Belgium
| |
Collapse
|
7
|
Fortini F, Vieceli Dalla Sega F, Marracino L, Severi P, Rapezzi C, Rizzo P, Ferrari R. Well-Known and Novel Players in Endothelial Dysfunction: Updates on a Notch(ed) Landscape. Biomedicines 2021; 9:biomedicines9080997. [PMID: 34440201 PMCID: PMC8393382 DOI: 10.3390/biomedicines9080997] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
Endothelial dysfunction characterizes every aspect of the so-called cardiovascular continuum, a series of events ranging from hypertension to the development of atherosclerosis and, finally, to coronary heart disease, thrombus formation, myocardial infarction, and heart failure. Endothelial dysfunction is the main prognostic factor for the progression of vascular disorders, which responds to drug intervention and lifestyle changes. Virtually all of the drugs used to prevent cardiovascular disorders, such as long-used and new antilipidemic agents and inhibitors of angiotensin enzyme (ACEi), exert an important effect on the endothelium. Endothelial dysfunction is a central feature of coronavirus disease -19 (COVID-19), and it is now clear that life-risk complications of the disease are prompted by alterations of the endothelium induced by viral infection. As a consequence, the progression of COVID-19 is worse in the subjects in whom endothelial dysfunction is already present, such as elderly, diabetic, obese, and hypertensive patients. Importantly, circulating biomarkers of endothelial activation and injury predict the severity and mortality of the disease and can be used to evaluate the efficacy of treatments. The purpose of this review is to provide updates on endothelial function by discussing its clinical relevance in the cardiovascular continuum, the latest insights from molecular and cellular biology, and their implications for clinical practice, with a focus on new actors, such as the Notch signaling and emerging therapies for cardiovascular disease.
Collapse
Affiliation(s)
- Francesca Fortini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
| | | | - Luisa Marracino
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Paolo Severi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Claudio Rapezzi
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (F.F.); (F.V.D.S.); (C.R.); (P.R.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (P.S.)
- Correspondence: ; Tel.: +39-053-229-3707
| |
Collapse
|
8
|
Associations of Variability in Metabolic Parameters with Lung Cancer: A Nationwide Population-Based Study. Cancers (Basel) 2021; 13:cancers13081982. [PMID: 33924149 PMCID: PMC8074362 DOI: 10.3390/cancers13081982] [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: 02/08/2021] [Revised: 03/27/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Lung cancer is currently the most common cancer worldwide. This study investigates whether visit-to-visit variability in metabolic parameters is associated with lung cancer risk. We found that a high variability in fasting blood glucose, systolic blood pressure, total cholesterol, and body weight were each associated with increased risk of lung cancer. A higher number of high-variability parameters were also associated with increased lung cancer risk. Further research is needed to examine whether reducing variability can lead to decreased lung cancer risk. Abstract We investigated whether visit-to-visit variability in metabolic parameters is associated with lung cancer risk. We used nationally representative data from the Korean National Health Insurance System, and 8,011,209 lung-cancer-free subjects who underwent over three health examinations from 2005 to 2010 were followed until 2017. Variability of fasting blood glucose, total cholesterol, systolic blood pressure, and body weight were measured by the variability independent of the mean, assessed by quartiles. There were 44,982 lung cancer events. The hazard ratio (HR) and 95% confidence interval (CI) for lung cancer risk was 1.07 (1.04, 1.10) for fasting blood glucose in the highest quartile, 1.08 (1.05, 1.10) for systolic blood pressure, 1.04 (1.01, 1.07) for weight, and 1.11 (1.08, 1.14) for total cholesterol. When comparing ≥3 vs. 0 high-variability metabolic parameters, the HR for lung cancer was 1.18 (95% CI, 1.14, 1.22). However, while ≥3 high-variability parameters showed an increased lung cancer risk in men (HR 1.26, 95% CI 1.21, 1.31), women did not show increased risk (HR 0.99, 95% CI 0.92, 1.06). High variability in each metabolic parameter, and a higher number of high-variability parameters, were associated with increased lung cancer risk.
Collapse
|
9
|
Dybas J, Chiura T, Marzec KM, Mak PJ. Probing Heme Active Sites of Hemoglobin in Functional Red Blood Cells Using Resonance Raman Spectroscopy. J Phys Chem B 2021; 125:3556-3565. [PMID: 33787265 PMCID: PMC8154613 DOI: 10.1021/acs.jpcb.1c01199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
![]()
The UV–vis absorption, Raman
imaging, and resonance Raman
(rR) spectroscopy methods were employed to study cyanohemoglobin (HbCN)
adducts inside living functional red blood cells (RBCs). The cyanide
ligands are especially optically sensitive probes of the active site
environment of heme proteins. The rR studies of HbCN and its isotopic
analogues (13CN–, C15N–, and 13C15N–), as well as a careful deconvolution of spectral data, revealed
that the ν(Fe–CN) stretching, δ(Fe–CN) bending,
and ν(C≡N) stretching modes occur at 454, 382, and 2123
cm–1, respectively. Interestingly, while the ν(Fe–CN)
modes exhibit the same frequencies in both the isolated and RBC-enclosed
hemoglobin molecules, small frequency differences are observed in
the δ(Fe–CN) bending modes and the values of their isotopic
shifts. These studies show that even though the overall tilted conformation
of the Fe–C≡N fragment in the isolated HbCN is preserved
in the HbCN enclosed within living cells, there is a small difference
in the degree of distortion of the Fe–C≡N fragment.
The slight changes in the ligand geometry can be reasonably attributed
to the high ordering and tight packing of Hb molecules inside RBCs.
Collapse
Affiliation(s)
- Jakub Dybas
- Chemistry Department, Saint Louis University, 3501 Laclede Avenue, Saint Louis 63103, Missouri, United States.,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzyńskiego Str., Krakow 30-348, Poland
| | - Tapiwa Chiura
- Chemistry Department, Saint Louis University, 3501 Laclede Avenue, Saint Louis 63103, Missouri, United States
| | - Katarzyna M Marzec
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzyńskiego Str., Krakow 30-348, Poland
| | - Piotr J Mak
- Chemistry Department, Saint Louis University, 3501 Laclede Avenue, Saint Louis 63103, Missouri, United States
| |
Collapse
|
10
|
Abstract
Cancer therapies can lead to a broad spectrum of cardiovascular complications. Among these, cardiotoxicities remain of prime concern, but vascular toxicities have emerged as the second most common group. The range of cancer therapies with a vascular toxicity profile and the clinical spectrum of vascular toxic effects are quite broad. Historically, venous thromboembolism has received the greatest attention but, over the past decade, the arterial toxic effects, which can present as acute vasospasm, acute thrombosis and accelerated atherosclerosis, of cancer therapies have gained greater recognition. This Review focuses on these types of cancer therapy-related arterial toxicity, including their mechanisms, and provides an update on venous thromboembolism and pulmonary hypertension associated with cancer therapies. Recommendations for the screening, treatment and prevention of vascular toxic effects of cancer therapies are outlined in the context of available evidence and society guidelines and consensus statements. The shift towards greater awareness of the vascular toxic effects of cancer therapies has further unveiled the urgent needs in this area in terms of defining best clinical practices. Well-designed and well-conducted clinical studies and registries are needed to more precisely define the incidence rates, risk factors, primary and secondary modes of prevention, and best treatment modalities for vascular toxicities related to cancer therapies. These efforts should be complemented by preclinical studies to outline the pathophysiological concepts that can be translated into the clinic and to identify drugs with vascular toxicity potential even before their widespread clinical use.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
11
|
Singh S, Nguyen H, Michels D, Bazinet H, Matkar PN, Liu Z, Esene L, Adam M, Bugyei‐Twum A, Mebrahtu E, Joseph J, Ehsan M, Chen HH, Qadura M, Singh KK. BReast CAncer susceptibility gene 2 deficiency exacerbates oxidized LDL-induced DNA damage and endothelial apoptosis. Physiol Rep 2020; 8:e14481. [PMID: 32638521 PMCID: PMC7340845 DOI: 10.14814/phy2.14481] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/18/2020] [Accepted: 05/15/2020] [Indexed: 12/24/2022] Open
Abstract
Mutations in the tumor suppressor gene BRCA2 (BReast CAncer susceptibility gene 2) predispose carriers to breast, ovarian, and other cancers. In response to DNA damage, BRCA2 participates in homology-directed DNA damage repair to maintain genome stability. Genome-wide association studies have identified an association between BRCA2 single nucleotide polymorphisms and plasma-lipid levels and lipid deregulation in humans. To date, DNA damage, apoptosis, and lipid deregulation are recognized as central pathways for endothelial dysfunction and atherosclerosis; however, the role of BRCA2 in endothelial dysfunction remains to be elucidated. To determine the role of BRCA2 in endothelial dysfunction, BRCA2 was silenced in human umbilical vein endothelial cells (ECs) and assessed for markers of DNA damage, apoptosis, and endothelial function following oxidized low-density lipoprotein (oxLDL) treatment. OxLDL was found to induce significant reactive oxygen species (ROS) production in BRCA2-silenced ECs. This increase in ROS production was associated with exacerbated DNA damage evidenced by increased expression and activation of DNA double-stranded break (DSB) marker γH2AX and reduced RAD51-foci formation-an essential regulator of DSB repair. Increased DSBs were associated with enhanced expression and activation of pro-apoptotic p53 and significant apoptosis in oxLDL-treated BRCA2-silenced ECs. Loss of BRCA2 in ECs was further associated with oxLDL-induced impaired tube-forming potential and eNOS expression. Collectively, the data reveals, for the first time, a novel role of BRCA2 as a regulator of EC survival and function in the setting of oxLDL treatment in vitro. Additionally, the data provide important clues regarding the potential susceptibility of BRCA2 mutation carriers to endothelial dysfunction, atherosclerosis, and other cardiovascular diseases.
Collapse
Affiliation(s)
- Shweta Singh
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Hien Nguyen
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Anatomy and Cell BiologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - David Michels
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Hannah Bazinet
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Pratiek N. Matkar
- Division of CardiologyKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Zongyi Liu
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Lilian Esene
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Mohamed Adam
- Division of CardiologyKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Antoinette Bugyei‐Twum
- Division of CardiologyKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Elizabeth Mebrahtu
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Jameela Joseph
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Department of BiologyUniversity of Western OntarioLondonONCanada
| | - Mehroz Ehsan
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Hao H. Chen
- Division of CardiologyKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Mohammad Qadura
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Vascular SurgeryKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s HospitalTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
| | - Krishna K. Singh
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Anatomy and Cell BiologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Vascular SurgeryKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s HospitalTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
- Pharmacology and ToxicologyUniversity of TorontoTorontoONCanada
| |
Collapse
|
12
|
Wang Y, Jiang C, Huang H, Liu N, Wang Y, Chen Z, Liang S, Wu M, Jiang Y, Wang X, Zhou T, Chen H, Zhang L, Li H. Correlation of Cerebral White Matter Lesions with Carotid Intraplaque Neovascularization assessed by Contrast-enhanced Ultrasound. J Stroke Cerebrovasc Dis 2020; 29:104928. [PMID: 32689582 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Carotid atherosclerotic plaque is closely associated with cerebral white matter lesions (WMLs), while intraplaque neovascularization (IPN) contributes significantly to arterial remodeling and plaque vulnerability. In this study, we aim to evaluate the correlation of carotid IPN with cerebral WMLs. METHODS The presence of IPN and WMLs were assessed by contrast-enhanced ultrasound (CEUS) and MRI respectively. IPN was evaluated utilizing semi-quantification visual grading scale and WMLs was divided according to Fazekas grading scale. We investigated the baseline data, Fazekas grades, and IPN grades among 269 participants. We explored the influences of each variable on Fazekas grades using ordinal logistic regression and evaluated the relationship between IPN grades and WMLs Fazekas grades. RESULTS Increased age (OR: 1.06, P<0.001), hypertension (OR: 2.17, P=0.002), cerebral infarction (OR: 1.74, P=0.046), and elevated carotid IPN grading were significantly associated with aggravated Fazekas grades (grade 2 or 3). To be specific, people having grade 3, 2, and 1 carotid IPN were 25.84 (P<0.001), 10.64 (P<0.001), and 5.96 (P=0.010) times as likely to have elevated Fazekas grades compared with those who having grade 0 carotid IPN. CONCLUSION Increased carotid IPN is independently correlated with aggravated cerebral WMLs.
Collapse
Affiliation(s)
- Yuxuan Wang
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Chao Jiang
- Department of Public Health, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Hui Huang
- Department of Ultrasound, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Niu Liu
- Department of Ultrasound, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yi Wang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhaoyao Chen
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Sen Liang
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Minghua Wu
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yajun Jiang
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaoxiao Wang
- GCP Center, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tingting Zhou
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Hu Chen
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lin Zhang
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Hui Li
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
13
|
Holm Nielsen S, Jonasson L, Kalogeropoulos K, Karsdal MA, Reese-Petersen AL, Auf dem Keller U, Genovese F, Nilsson J, Goncalves I. Exploring the role of extracellular matrix proteins to develop biomarkers of plaque vulnerability and outcome. J Intern Med 2020; 287:493-513. [PMID: 32012358 DOI: 10.1111/joim.13034] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/04/2019] [Accepted: 01/13/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease (CVD) is the most common cause of death in industrialized countries. One underlying cause is atherosclerosis, which is a systemic disease characterized by plaques of retained lipids, inflammatory cells, apoptotic cells, calcium and extracellular matrix (ECM) proteins in the arterial wall. The biologic composition of an atherosclerotic plaque determines whether the plaque is more or less vulnerable, that is prone to rupture or erosion. Here, the ECM and tissue repair play an important role in plaque stability, vulnerability and progression. This review will focus on ECM remodelling in atherosclerotic plaques, with focus on how ECM biomarkers might predict plaque vulnerability and outcome.
Collapse
Affiliation(s)
- S Holm Nielsen
- From the, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - L Jonasson
- Department of Medical and Health Sciences, Division of Cardiovascular Medicine, Linköping University, Linköping, Sweden
| | - K Kalogeropoulos
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - M A Karsdal
- From the, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark
| | | | - U Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - F Genovese
- From the, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark
| | - J Nilsson
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - I Goncalves
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
14
|
Geng T, Yan Y, Xu L, Cao M, Xu Y, Pu J, Yan JC. CD137 signaling induces macrophage M2 polarization in atherosclerosis through STAT6/PPARδ pathway. Cell Signal 2020; 72:109628. [PMID: 32247042 DOI: 10.1016/j.cellsig.2020.109628] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 11/28/2022]
Abstract
CD137 signaling plays an important role in the formation and development of atherosclerotic plaques. The purpose of the present study was to investigate the effects of CD137 signaling on macrophage polarization during atherosclerosis and to explore the underlying mechanisms. The effect of CD137 signaling on macrophage phenotype in atherosclerotic plaques was determined by intraperitoneal injection of agonist-CD137 recombinant protein in apolipoprotein E-deficient (ApoE-/-) mice, an established in vivo model of atherosclerosis. Murine peritoneal macrophages and RAW 264.7 cells were treated with AS1517499 and siPPARδ (peroxisome proliferator-activated receptor δ) to study the role of STAT6 (signal transducers and activators of transcription 6)/PPARδ signaling in CD137-induced M2 macrophage polarization in vitro. Results from both in vivo and in vitro experiments showed that CD137 signaling can transform macrophages into the M2 phenotype during the process of atherosclerotic plaque formation and regulate the angiogenic features of M2 macrophages. Furthermore, activation of the CD137 signaling pathway induces phosphorylation of STAT6 and enhances the expression of PPARδ. We further found that macrophage M2 polarization is reduced when the STAT6/PPARδ pathway is inhibited. Together, these data show a role for the STAT6/PPARδ signaling pathway in the CD137 signaling-induced M2 macrophage polarization pathway.
Collapse
Affiliation(s)
- Tianxin Geng
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province 212000, China
| | - Yang Yan
- Department of Cardiology, Ren Ji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200135, China
| | - Liangjie Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province 212000, China
| | - Mengfei Cao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province 212000, China
| | - Yu Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province 212000, China
| | - Jun Pu
- Department of Cardiology, Ren Ji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200135, China
| | - Jin Chuan Yan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province 212000, China.
| |
Collapse
|
15
|
Toya T, Sara JD, Corban MT, Taher R, Godo S, Herrmann J, Lerman LO, Lerman A. Assessment of peripheral endothelial function predicts future risk of solid-tumor cancer. Eur J Prev Cardiol 2020; 27:608-618. [PMID: 31668110 DOI: 10.1177/2047487319884246] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
AIMS Cardiovascular health metrics predict the risk not only of cardiovascular diseases but also of several types of cancers. Microvascular endothelial dysfunction can predict future cardiovascular adverse events, but the predictive value of microvascular endothelial dysfunction for future risk of solid-tumor cancer has not been characterized. METHODS A total of 488 patients who underwent microvascular endothelial function assessment using reactive hyperemia peripheral arterial tonometry were included in this study. Microvascular endothelial dysfunction was defined as a reactive hyperemia peripheral arterial tonometry index ≤2.0. RESULTS Of 221 patients with a baseline reactive hyperemia peripheral arterial tonometry index ≤2.0, 21 patients (9.5%) were diagnosed with incident solid-tumor cancer during follow-up, whereas of 267 patients with a baseline reactive hyperemia peripheral arterial tonometry index >2.0, 10 patients (3.7%) were diagnosed with incident solid-tumor cancer during follow-up (p = 0.009). Patients with a reactive hyperemia peripheral arterial tonometry index ≤2.0 had lower solid-tumor cancer-free survival compared to patients with a reactive hyperemia peripheral arterial tonometry index >2.0 (log-rank p = 0.017) (median follow-up 6.0 (3.0-9.1) years). Cox proportional hazard analyses showed that a reactive hyperemia peripheral arterial tonometry index ≤2.0 predicted the incidence of solid-tumor cancer, with a hazard ratio of 2.52 (95% confidence interval 1.17-5.45; p = 0.019) after adjusting for age, sex, and coronary artery disease, 2.83 (95% confidence interval 1.30-6.17; p = 0.009) after adjusting for diabetes mellitus, hypertension, smoking status, and body mass index >30 kg/m2, 2.79 (95% confidence interval 1.21-6.41; p = 0.016) after adjusting for fasting plasma glucose, systolic blood pressure, smoking status (current or former), and body mass index, and 2.43 (95% confidence interval 1.10-5.34; p = 0.028) after adjusting for Framingham risk score. CONCLUSION Microvascular endothelial dysfunction, as defined by a reactive hyperemia peripheral arterial tonometry index ≤2.0, was associated with a greater than two-fold increased risk of solid-tumor cancer. Microvascular endothelial dysfunction may be a useful marker to predict the future risk of solid-tumor cancer, in addition to its known ability to predict cardiovascular disease. Further research is necessary to develop adequate cancer screening strategies for patients with microvascular endothelial dysfunction.
Collapse
Affiliation(s)
- Takumi Toya
- Department of Cardiovascular Medicine, Mayo Clinic, USA
- Division of Cardiology, National Defense Medical College, Japan
| | | | | | - Riad Taher
- Department of Cardiovascular Medicine, Mayo Clinic, USA
| | - Shigeo Godo
- Department of Cardiovascular Medicine, Mayo Clinic, USA
| | | | | | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, USA
| |
Collapse
|
16
|
Peluso R, Caso F, Tasso M, Sabbatino V, Lupoli R, Dario Di Minno MN, Ursini F, Costa L, Scarpa R. Biomarkers of subclinical atherosclerosis in patients with psoriatic arthritis. Open Access Rheumatol 2019; 11:143-156. [PMID: 31388317 PMCID: PMC6607207 DOI: 10.2147/oarrr.s206931] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Psoriatic arthritis (PsA) is a chronic immune-mediated disease. It is associated with an increase in cardiovascular risk factors (obesity, hypertension, diabetes, and dyslipidemia), giving a higher risk of major adverse cardiovascular events. Patients with PsA have an increased incidence of subclinical atherosclerosis and endothelial dysfunction. The aim of this study is to perform a review of the biomarkers of subclinical atherosclerosis in patients with PsA. Methods: A search was performed in the electronic databases (PubMed, Web of Science, Scopus, and Embase) up until July 2017. Studies were considered if they included data on biomarkers of subclinical atherosclerosis in PsA, and each article was then reviewed for quality and clinical relevance. After completing the literature search, all screened literature was summarized and discussed in our study group (CaRRDs study group). Results: The initial search produced 532 abstracts, which were limited to 258 potentially relevant articles by preliminary review of the titles and by excluding review articles and case reports (n=274). A further 102 articles were deemed ineligible after examining the abstracts. Full texts of the remaining 156 articles were retrieved. Most articles were excluded because they were not relevant to the biomarkers of subclinical atherosclerosis in psoriasis and/or PsA. In the end, 54 articles were deemed eligible for this review. Conclusion: Patients with PsA showed more severe atherosclerotic disease compared with patients with only psoriasis. This may have been due to the higher systemic inflammatory burden from the combination of both diseases. In patients with PsA some molecules may be considered as markers of atherosclerotic disease, and their detection may be a prognostic marker, in addition to imaging procedures, for the development of atherosclerotic disease, and could be suitable for the management of patients with PsA.
Collapse
Affiliation(s)
- Rosario Peluso
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Francesco Caso
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Marco Tasso
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Vincenzo Sabbatino
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Roberta Lupoli
- Department of Clinical Medicine and Surgery, Division of Internal Medicine, Federico II University, Naples, Italy
| | | | - Francesco Ursini
- Internal Medicine Unit, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Luisa Costa
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Raffaele Scarpa
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| |
Collapse
|
17
|
Oren O, Herrmann J. Arterial events in cancer patients-the case of acute coronary thrombosis. J Thorac Dis 2018; 10:S4367-S4385. [PMID: 30701104 PMCID: PMC6328398 DOI: 10.21037/jtd.2018.12.79] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/14/2018] [Indexed: 12/21/2022]
Abstract
Patients with cancer are at high risk for both venous and arterial thrombotic complications. A variety of factors account for the greater thrombotic risk, including the underlying malignancy and numerous cancer-directed therapies. The occurrence of an acute thrombotic event in patients with cancer is associated with substantial morbidity and mortality. Acute coronary syndrome (ACS) represents a particularly important cardiovascular complication in cancer patients. With cardio-vascular risk factors becoming more prevalent in an aging cancer population that is surviving longer, questions pertaining to the appropriate management of vascular toxicity are likely to assume even greater value in the coming years. In this article, we review the current understanding of ACS in patients with cancer. The predisposition to thrombosis in a malignant host and the cancer treatments most commonly associated with vascular toxicity are reviewed. Risk prediction and management strategies are discussed, and discrepancies in the clinical evidence are highlighted.
Collapse
Affiliation(s)
- Ohad Oren
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
18
|
Reustle A, Torzewski M. Role of p38 MAPK in Atherosclerosis and Aortic Valve Sclerosis. Int J Mol Sci 2018; 19:ijms19123761. [PMID: 30486366 PMCID: PMC6321637 DOI: 10.3390/ijms19123761] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/16/2018] [Accepted: 11/22/2018] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis and aortic valve sclerosis are cardiovascular diseases with an increasing prevalence in western societies. Statins are widely applied in atherosclerosis therapy, whereas no pharmacological interventions are available for the treatment of aortic valve sclerosis. Therefore, valve replacement surgery to prevent acute heart failure is the only option for patients with severe aortic stenosis. Both atherosclerosis and aortic valve sclerosis are not simply the consequence of degenerative processes, but rather diseases driven by inflammatory processes in response to lipid-deposition in the blood vessel wall and the aortic valve, respectively. The p38 mitogen-activated protein kinase (MAPK) is involved in inflammatory signaling and activated in response to various intracellular and extracellular stimuli, including oxidative stress, cytokines, and growth factors, all of which are abundantly present in atherosclerotic and aortic valve sclerotic lesions. The responses generated by p38 MAPK signaling in different cell types present in the lesions are diverse and might support the progression of the diseases. This review summarizes experimental findings relating to p38 MAPK in atherosclerosis and aortic valve sclerosis and discusses potential functions of p38 MAPK in the diseases with the aim of clarifying its eligibility as a pharmacological target.
Collapse
Affiliation(s)
- Anna Reustle
- Dr. Margarete-Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany.
- University of Tuebingen, 72074 Tuebingen, Germany.
| | - Michael Torzewski
- Department of Laboratory Medicine and Hospital Hygiene, Robert Bosch-Hospital, 70376 Stuttgart, Germany.
| |
Collapse
|
19
|
Magnoni M, Ammirati E, Moroni F, Norata GD, Camici PG. Impact of Cardiovascular Risk Factors and Pharmacologic Treatments on Carotid Intraplaque Neovascularization Detected by Contrast-Enhanced Ultrasound. J Am Soc Echocardiogr 2018; 32:113-120.e6. [PMID: 30340893 DOI: 10.1016/j.echo.2018.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Indexed: 10/28/2022]
Abstract
BACKGROUND Neovascularization is a marker of plaque vulnerability that can be assessed noninvasively using contrast-enhanced ultrasound (CEUS). The presence and extent of plaque neovascularization and their relation to cardiovascular risk factors and treatments were assessed in asymptomatic patients with carotid stenosis of intermediate severity and no indication for revascularization. METHODS Sixty-six patients aged 69 ± 8 years (59% men) were prospectively enrolled. Plaque neovascularization was assessed using CEUS with sulfur hexafluoride contrast in each of the four carotid segments bilaterally (a total of 528 segments). In each plaque, the presence or absence of contrast enhancement was assessed semiquantitatively as CEUS grade 1 (no signal or signal confined to the adventitia and/or shoulder of the plaque) or CEUS grade 2 (signal within the plaque). RESULTS Plaques were detectable in 289 of 528 carotid segments (54.7%). CEUS grade 2 was present in at least one plaque in 48 of 66 patients (72.7%) and was not influenced by stenosis severity or morphology. The highest CEUS grade 2 prevalence was observed in patients with diabetes and the lowest in those treated with angiotensin-converting enzyme inhibitors and statins, especially when low-density lipoprotein cholesterol was <100 mg/dL. Patients with multiple CEUS grade 2 plaques (20 of 66 [30%]) had both higher low-density lipoprotein and higher C-reactive protein. CONCLUSION Intraplaque neovascularization is frequent in asymptomatic patients with intermediate carotid stenosis and is more prevalent in those with diabetes. Low-density lipoprotein cholesterol < 100 mg/dL and treatment with angiotensin-converting enzyme inhibitors seem to confer protection from neovascularization, although larger interventional studies are necessary to confirm these data.
Collapse
Affiliation(s)
- Marco Magnoni
- Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Enrico Ammirati
- Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy; De Gasperis Cardio Center, Niguarda Hospital, Milan, Italy
| | - Francesco Moroni
- Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giuseppe D Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paolo G Camici
- Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
20
|
Darjani A, Rafiei R, Shafaei S, Rafiei E, Eftekhari H, Alizade N, Gharaei nejad K, Rafiee B, Najirad S. Evaluation of Lipid Profile in Patients with Cherry Angioma: A Case-Control Study in Guilan, Iran. Dermatol Res Pract 2018; 2018:4639248. [PMID: 29861719 PMCID: PMC5971275 DOI: 10.1155/2018/4639248] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/04/2018] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Cherry angioma is the most common type of acquired cutaneous vascular proliferation which would increase with aging due to some angiogenic factors but the exact pathogenesis is unknown. Usually angiogenic factors are synthesized in human body to compensate occlusive effects of atherogenic agents such as serum lipids. Our hypothesis was that increased levels of these angiogenic factors could be a trigger for development of cherry angioma. This study has been designed to compare frequency of dyslipidemia in subjects with and without cutaneous cherry angioma. METHODS In this case-control study, 122 cases with cherry angioma and 122 control subjects without cherry angioma were enrolled. Demographic characteristics, number of the cherry angioma lesions, and serum lipid profile were collected for all subjects. The data was analyzed using SPSS 18 software. RESULTS Mean levels of the total cholesterol, triglyceride, low-density lipoprotein, and high-density lipoprotein were higher in patients with cherry angioma compared to control subjects in which differences were significant for total cholesterol, low-density lipoprotein, and triglyceride (P < 0.05) but not for high-density lipoprotein level. CONCLUSION Serum lipids may have a role in producing angiogenic factors and development of cherry angioma and it seems logical to evaluate lipid profile in these cases.
Collapse
Affiliation(s)
- Abbas Darjani
- Skin Research Center, Dermatology Department, Guilan University of Medical Sciences, Razi Hospital, Sardare Jangal Street, Rasht, Iran
| | - Rana Rafiei
- Fellowship of Dermatopathology, Skin Research Center, Dermatology Department, Guilan University of Medical Sciences, Razi Hospital, Sardare Jangal Street, Rasht, Iran
| | - Sareh Shafaei
- Skin Research Center, Dermatology Department, Guilan University of Medical Sciences, Razi Hospital, Sardare Jangal Street, Rasht, Iran
| | - Elahe Rafiei
- Razi Clinical Research Development Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Hojat Eftekhari
- Skin Research Center, Dermatology Department, Guilan University of Medical Sciences, Razi Hospital, Sardare Jangal Street, Rasht, Iran
| | - Narges Alizade
- Skin Research Center, Dermatology Department, Guilan University of Medical Sciences, Razi Hospital, Sardare Jangal Street, Rasht, Iran
| | - Kaveh Gharaei nejad
- Skin Research Center, Dermatology Department, Guilan University of Medical Sciences, Razi Hospital, Sardare Jangal Street, Rasht, Iran
| | - Behnam Rafiee
- Department of Pathology, NYU Winthrop Hospital, 222 Station Plaza, No. 620, Mineola, NY 11501, USA
| | - Sara Najirad
- Department of Internal Medicine, Nassau University Medical Center, 2201 Hempstead Turnpike, East Meadow, NY 11554, USA
| |
Collapse
|
21
|
Touyz RM, Herrmann J. Cardiotoxicity with vascular endothelial growth factor inhibitor therapy. NPJ Precis Oncol 2018; 2:13. [PMID: 30202791 PMCID: PMC5988734 DOI: 10.1038/s41698-018-0056-z] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis inhibitors targeting the vascular endothelial growth factor (VEGF) signaling pathway (VSP) have been important additions in the therapy of various cancers, especially renal cell carcinoma and colorectal cancer. Bevazicumab, the first VSP to receive FDA approval in 2004 targeting all circulating isoforms of VEGF-A, has become one of the best-selling drugs of all times. The second wave of tyrosine kinase inhibitors (TKIs), which target the intracellular site of VEGF receptor kinases, began with the approval of sorafenib in 2005 and sunitinib in 2006. Heart failure was subsequently noted, in 2-4% of patients on bevacizumab and in 3-8% of patients on VSP-TKIs. The very fact that the single-targeted monoclonal antibody bevacizumab can induce cardiotoxicity supports a pathomechanistic role for the VSP and the postulate of the "vascular" nature of VSP inhibitor cardiotoxicity. In this review we will outline this scenario in greater detail, reflecting on hypertension and coronary artery disease as risk factors for VSP inhibitor cardiotoxicity, but also similarities with peripartum and diabetic cardiomyopathy. This leads to the concept that any preexisting or coexisting condition that reduces the vascular reserve or utilizes the vascular reserve for compensatory purposes may pose a risk factor for cardiotoxicity with VSP inhibitors. These conditions need to be carefully considered in cancer patients who are to undergo VSP inhibitor therapy. Such vigilance is not to exclude patients from such prognostically extremely important therapy but to understand the continuum and to recognize and react to any cardiotoxicity dynamics early on for superior overall outcomes.
Collapse
Affiliation(s)
- Rhian M. Touyz
- Institute of Cardiovascular & Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
22
|
Guo M, Cai Y, Yao X, Li Z. Mathematical modeling of atherosclerotic plaque destabilization: Role of neovascularization and intraplaque hemorrhage. J Theor Biol 2018; 450:53-65. [PMID: 29704490 DOI: 10.1016/j.jtbi.2018.04.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/29/2018] [Accepted: 04/23/2018] [Indexed: 01/03/2023]
Abstract
Observational studies have identified angiogenesis from the adventitial vasa vasorum and intraplaque hemorrhage (IPH) as critical factors in atherosclerotic plaque progression and destabilization. Here we propose a mathematical model incorporating intraplaque neovascularization and hemodynamic calculation with plaque destabilization for the quantitative evaluation of the role of neoangiogenesis and IPH in the vulnerable atherosclerotic plaque formation. An angiogenic microvasculature is generated by two-dimensional nine-point discretization of endothelial cell proliferation and migration from the vasa vasorum. Three key cells (endothelial cells, smooth muscle cells and macrophages) and three key chemicals (vascular endothelial growth factors, extracellular matrix and matrix metalloproteinase) are involved in the plaque progression model, and described by the reaction-diffusion partial differential equations. The hemodynamic calculation of the microcirculation on the generated microvessel network is carried out by coupling the intravascular, interstitial and transvascular flow. The plasma concentration in the interstitial domain is defined as the description of IPH area according to the diffusion and convection with the interstitial fluid flow, as well as the extravascular movement across the leaky vessel wall. The simulation results demonstrate a series of pathophysiological phenomena during the vulnerable progression of an atherosclerotic plaque, including the expanding necrotic core, the exacerbated inflammation, the high microvessel density (MVD) region at the shoulder areas, the transvascular flow through the capillary wall and the IPH. The important role of IPH in the plaque destabilization is evidenced by simulations with varied model parameters. It is found that the IPH can significantly speed up the plaque vulnerability by increasing necrotic core and thinning fibrous cap. In addition, the decreased MVD and vessel permeability may slow down the process of plaque destabilization by reducing the IPH dramatically. We envision that the present model and its future advances can serve as a valuable theoretical platform for studying the dynamic changes in the microenvironment during the plaque destabilization.
Collapse
Affiliation(s)
- Muyi Guo
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yan Cai
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xinke Yao
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhiyong Li
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China; School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology, Brisbane, QLD 4001, Australia.
| |
Collapse
|
23
|
Sun H, Krauss RM, Chang JT, Teng BB. PCSK9 deficiency reduces atherosclerosis, apolipoprotein B secretion, and endothelial dysfunction. J Lipid Res 2018; 59:207-223. [PMID: 29180444 PMCID: PMC5794417 DOI: 10.1194/jlr.m078360] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/22/2017] [Indexed: 01/05/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) interacts directly with cytoplasmic apoB and prevents its degradation via the autophagosome/lysosome pathway. This process affects VLDL and LDL production and influences atherogenesis. Here, we investigated the molecular machinery by which PCSK9 modulates autophagy and affects atherogenesis. We backcrossed Pcsk9-/- mice with atherosclerosis-prone Ldlr-/-Apobec1-/- (LDb) mice to generate Ldlr-/-Apobec1-/-Pcsk9-/- (LTp) mice. Deletion of PCSK9 resulted in decreased hepatic apoB secretion, increased autophagic flux, and decreased plasma levels of IDL and LDL particles. The LDLs from LTp mice (LTp-LDLs) were less atherogenic and contained less cholesteryl ester and phospholipids than LDb-LDLs. Moreover LTp-LDLs induced lower endothelial expression of the genes encoding TLR2, Lox-1, ICAM-1, CCL2, CCL7, IL-6, IL-1β, Beclin-1, p62, and TRAF6 Collectively, these effects were associated with substantially less atherosclerosis development (>4-fold) in LTp mice. The absence of PCSK9 in LDb mice results in decreased lipid and apoB levels, fewer atherogenic LDLs, and marked reduction of atherosclerosis. The effect on atherogenesis may be mediated in part by the effects of modified LDLs on endothelial cell receptors and proinflammatory and autophagy molecules. These findings suggest that there may be clinical benefits of PCSK9 inhibition due to mechanisms unrelated to increased LDL receptor activity.
Collapse
Affiliation(s)
- Hua Sun
- Research Center for Human Genetics, Brown Foundation Institute of Molecular Medicine University of Texas Health Science Center at Houston, Houston, TX
| | | | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX
- University of Texas MD Anderson Cancer Center, University of Texas Health Science Center at Houston Graduate School of Biomedical Sciences, Houston, TX
| | - Ba-Bie Teng
- Research Center for Human Genetics, Brown Foundation Institute of Molecular Medicine University of Texas Health Science Center at Houston, Houston, TX
- University of Texas MD Anderson Cancer Center, University of Texas Health Science Center at Houston Graduate School of Biomedical Sciences, Houston, TX
| |
Collapse
|
24
|
Skoda M, Stangret A, Szukiewicz D. Fractalkine and placental growth factor: A duet of inflammation and angiogenesis in cardiovascular disorders. Cytokine Growth Factor Rev 2018; 39:116-123. [DOI: 10.1016/j.cytogfr.2017.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/11/2022]
|
25
|
Bonanad C, González-Parra E, Rivera R, Carrascosa J, Daudén E, Olveira A, Botella-Estrada R. Clinical, Diagnostic, and Therapeutic Implications in Psoriasis Associated With Cardiovascular Disease. ACTAS DERMO-SIFILIOGRAFICAS 2017. [DOI: 10.1016/j.adengl.2017.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
26
|
Bonanad C, González-Parra E, Rivera R, Carrascosa J, Daudén E, Olveira A, Botella-Estrada R. Implicaciones clínicas, diagnósticas y terapéuticas de la psoriasis y enfermedad cardiovascular. ACTAS DERMO-SIFILIOGRAFICAS 2017; 108:800-808. [DOI: 10.1016/j.ad.2016.12.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/01/2016] [Accepted: 12/06/2016] [Indexed: 12/16/2022] Open
|
27
|
Abstract
The introduction of molecularly targeted therapies with tyrosine kinase inhibitors has revolutionized cancer therapy and has contributed to a steady decline in cancer-related mortality since the late 1990s. However, not only cardiac but also vascular toxicity has been reported for these agents, some as expected on-target effects (e.g., VEGF receptor inhibitors) and others as unanticipated events (e.g., BCR-Abl inhibitors). A sound understanding of these cardiovascular toxic effects is critical to advance mechanistic insight into vascular disease and clinical care. From a conceptual standpoint, there might be value in defining type I (permanent) and type II (transient) vascular toxicity. This review will focus on the tyrosine kinase inhibitors in current clinical use and their associated vascular side effects.
Collapse
|
28
|
Demeure F, Bouzin C, Roelants V, Bol A, Verhelst R, Astarci P, Gerber BL, Pouleur AC, Pasquet A, de Meester C, Vanoverschelde JLJ, Vancraeynest D. Head-to-Head Comparison of Inflammation and Neovascularization in Human Carotid Plaques: Implications for the Imaging of Vulnerable Plaques. Circ Cardiovasc Imaging 2017; 10:CIRCIMAGING.116.005846. [PMID: 28487317 DOI: 10.1161/circimaging.116.005846] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/28/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Inflammation and intraplaque neovascularization are acknowledged to be 2 features of plaque vulnerability, although their temporal expression and their respective value in predicting clinical events are poorly understood. To determine their respective temporal associations, we conducted a comprehensive assessment of inflammation and intraplaque neovascularization in the carotid plaque of symptomatic and asymptomatic patients. METHODS AND RESULTS Thirty patients with severe carotid stenosis underwent 18F-fluorodeoxyglucose-positron emission tomography/computed tomographic imaging. Plaque 18F-fluorodeoxyglucose-uptake, indicative of inflammation, was measured by calculating the target:background ratio. The presence of intraplaque neovascularization during contrast-enhanced ultrasound was judged semiquantitatively; low-grade contrast enhancement (CE) suggested its absence, and high-grade CE, the presence of neovascularization. Carotid surgery was performed 1.6±1.8 days after completing both imaging modalities in all patients, and the presence of macrophages and neovessels was quantified by immunohistochemistry. We identified a significant correlation between the target:background ratio and macrophage quantification (R=0.78; P<0.001). The number of vessels was also significantly higher in carotid plaque with high-CE (P<0.001). Surprisingly, immunohistochemistry showed that high-CE and vessel number were neither associated with an elevated target:background ratio (P=0.28 and P=0.60, respectively) nor macrophage infiltration (P=0.59 and P=0.40, respectively). Finally, macrophage infiltration and target:background ratio were higher in the carotid plaque of symptomatic patients (P=0.021 and P=0.05, respectively), whereas CE grade and the presence of neovessels were not. CONCLUSIONS Inflammation and intraplaque neovascularization are not systematically associated in carotid plaques, suggesting a temporal separation between the 2 processes. Inflammation seems more pronounced when symptoms are present. These data highlight the challenges that face any imaging strategy designed to assess plaque vulnerability.
Collapse
Affiliation(s)
- Fabian Demeure
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Caroline Bouzin
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Véronique Roelants
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Anne Bol
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Robert Verhelst
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Parla Astarci
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Bernhard L Gerber
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Anne-Catherine Pouleur
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Agnès Pasquet
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Christophe de Meester
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - Jean-Louis J Vanoverschelde
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.)
| | - David Vancraeynest
- From the Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); Cardiovascular Department, Institut Cardiovasculaire, Cliniques Universitaires Saint-Luc, Brussels, Belgium (F.D., R.V., P.A., B.L.G., A.-C.P., A.P., C.d.M., J.-L.J.V., D.V.); IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (C.B.); Pôle d'Imagerie Médicale, Radiothérapie et Oncologie (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium (V.R., A.B.); and Division of Nuclear Medicine, Internal Medicine Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium (V.R., A.B.).
| |
Collapse
|
29
|
Formanowicz D, Radom M, Zawierucha P, Formanowicz P. Petri net-based approach to modeling and analysis of selected aspects of the molecular regulation of angiogenesis. PLoS One 2017; 12:e0173020. [PMID: 28253310 PMCID: PMC5333880 DOI: 10.1371/journal.pone.0173020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 02/14/2017] [Indexed: 11/18/2022] Open
Abstract
The functioning of both normal and pathological tissues depends on an adequate supply of oxygen through the blood vessels. A process called angiogenesis, in which new endothelial cells and smooth muscles interact with each other, forming new blood vessels either from the existing ones or from a primary vascular plexus, is particularly important and interesting, due to new therapeutic possibilities it offers. This is a multi-step and very complex process, so an accurate understanding of the underlying mechanisms is a significant task, especially in recent years, with the constantly increasing amount of new data that must be taken into account. A systems approach is necessary for these studies because it is not sufficient to analyze the properties of the building blocks separately and an analysis of the whole network of interactions is essential. This approach is based on building a mathematical model of the system, while the model is expressed in the formal language of a mathematical theory. Recently, the theory of Petri nets was shown to be especially promising for the modeling and analysis of biological phenomena. This analysis, based mainly on t-invariants, has led to a particularly important finding that a direct link (close connection) exist between transforming growth factor β1 (TGF-β1), endothelial nitric oxide synthase (eNOS), nitric oxide (NO), and hypoxia-inducible factor 1, the molecules that play a crucial roles during angiogenesis. We have shown that TGF-β1 may participate in the inhibition of angiogenesis through the upregulation of eNOS expression, which is responsible for catalyzing NO production. The results obtained in the previous studies, concerning the effects of NO on angiogenesis, have not been conclusive, and therefore, our study may contribute to a better understanding of this phenomenon.
Collapse
Affiliation(s)
- Dorota Formanowicz
- Department of Clinical Biochemistry and Laboratory Medicine, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznań, Poland
| | - Marcin Radom
- Institute of Computing Science, Poznan University of Technology, Piotrowo 2, 60-965 Poznań, Poland
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Z. Noskowskiego 12/14, 61-704 Poznań, Poland
| | - Piotr Zawierucha
- Department of Histology and Embryology, Poznan University of Medical Sciences, Świȩcickiego 6 St., 61-781 Poznań, Poland
- Department of Anatomy, Poznan University of Medical Sciences, Świȩcickiego 6, 61-781 Poznań, Poland
| | - Piotr Formanowicz
- Institute of Computing Science, Poznan University of Technology, Piotrowo 2, 60-965 Poznań, Poland
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Z. Noskowskiego 12/14, 61-704 Poznań, Poland
- * E-mail:
| |
Collapse
|
30
|
Luo JY, Li XM, Zhou Y, Zhao Q, Chen BD, Liu F, Chen XC, Zheng H, Ma YT, Gao XM, Yang YN. Mutant DD genotype of NFKB1 gene is associated with the susceptibility and severity of coronary artery disease. J Mol Cell Cardiol 2017; 103:56-64. [PMID: 28088561 DOI: 10.1016/j.yjmcc.2017.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/22/2016] [Accepted: 01/10/2017] [Indexed: 12/18/2022]
Abstract
Nuclear factor κappa B (NF-κB) is an important transcription factor in the development and progression of coronary artery disease (CAD). Recent evidence suggests that -94 ATTG ins/del mutant in the promoter of NFKB1 gene is an essential functional mutant. The present study demonstrated the frequencies of the del/del (DD) genotype and del (D) allele were significantly higher in CAD patients than in controls. CAD patients carrying mutant DD genotype had worse stenosis of diseased coronary arteries compared to those carrying ins/ins (II) or ins/del (ID) genotype. Plasma levels of endothelial nitric oxide synthase (eNOS) were lower, while inflammatory cytokine incnterlukin-6 (IL-6) was higher in CAD patients with DD genotype than those with II or ID genotype (both P<0.05). In vitro study showed that mutant human umbilical vein endothelial cells (DD genotype HUVECs) were more susceptible to H2O2-induced apoptosis, which was accompanied with a decreased Bcl-2 expression. Further, mutant HUVECs had lower eNOS but higher IL-6 mRNA levels and decreased phosphorylation of eNOS under H2O2-stimulation (both P<0.05). Compared to wild type cells (II genotype), significantly downregulated protein expression of total NF-κB p50 subunit were observed in mutant HUVECs with or without oxidative stress, and a lower expression of unclear p50 was associated with a decreased p50 nuclear translocation in mutant HUVECs versus wild type cells under H2O2-stimulation (both P<0.05). In conclusion, mutant DD genotype of NFKB1 gene is associated with the risk and severity of CAD. Dwonregulation of NF-κB p50 subunit leads to exacerbated endothelial dysfunction and apoptosis and enhanced inflammatory response that is the potential underlying mechanism.
Collapse
Affiliation(s)
- Jun-Yi Luo
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiao-Mei Li
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yun Zhou
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Qiang Zhao
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Bang-Dang Chen
- Xinjiang Key Laboratory of Cardiovascular Research, Urumqi, Xinjiang, China
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Research, Urumqi, Xinjiang, China
| | - Xiao-Cui Chen
- Xinjiang Key Laboratory of Cardiovascular Research, Urumqi, Xinjiang, China
| | - Hong Zheng
- Department of Anaesthesiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, Xinjiang, China; Department of Surgery, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Yi-Ning Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| |
Collapse
|
31
|
Ammirati E, Moroni F, Magnoni M, Di Terlizzi S, Villa C, Sizzano F, Palini A, Garlaschelli K, Tripiciano F, Scotti I, Catapano AL, Manfredi AA, Norata GD, Camici PG. Circulating CD14+ and CD14 highCD16- classical monocytes are reduced in patients with signs of plaque neovascularization in the carotid artery. Atherosclerosis 2016; 255:171-178. [PMID: 27751505 DOI: 10.1016/j.atherosclerosis.2016.10.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 09/16/2016] [Accepted: 10/04/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND AIMS Monocytes are known to play a key role in the initiation and progression of atherosclerosis and contribute to plaque destabilization through the generation of signals that promote inflammation and neoangiogenesis. In humans, studies investigating the features of circulating monocytes in advanced atherosclerotic lesions are lacking. METHODS Patients (mean age 69 years, 56% males) with intermediate asymptomatic carotid stenosis (40-70% in diameter) were evaluated for maximal stenosis in common carotid artery, carotid bulb and internal carotid artery, overall disease burden as estimated with total plaque area (TPA), greyscale and neovascularization in 244 advanced carotid plaques. Absolute counts of circulating CD14+ monocytes, of classical (CD14highCD16-), intermediate (CD14highCD16+) and non-classical (CD14lowCD16+) monocytes and HLA-DR+ median fluorescence intensity for each subset were evaluated with flow cytometry. RESULTS No correlation was found between monocytes and overall atherosclerotic burden, nor with high sensitivity C-reactive protein (hsCRP) or interleukin-6 (IL-6). In contrast, plaque signs of neovascularization were associated with significantly lower counts of circulating CD14+ monocytes (297 versus 350 cells/mm3, p = 0.039) and of classical monocytes (255 versus 310 cells/mm3, p = 0.029). CONCLUSIONS Neovascularized atherosclerotic lesions selectively associate with lower blood levels of CD14+ and CD14highCD16- monocytes independently of systemic inflammatory activity, as indicated by normal hsCRP levels. Whether the reduction of circulating CD14+ and CD14highCD16- monocytes is due to a potential redistribution of these cell types into active lesions remains to be explored.
Collapse
Affiliation(s)
- Enrico Ammirati
- Cardiothoracic Department, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy; De Gasperis Cardio Center, Niguarda Ca' Granda Hospital, Milan, Italy.
| | - Francesco Moroni
- Cardiothoracic Department, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy.
| | - Marco Magnoni
- Cardiothoracic Department, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy
| | - Simona Di Terlizzi
- FRACTAL - Flow cytometry Resource Advanced Cytometry Technical Applications Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Villa
- FRACTAL - Flow cytometry Resource Advanced Cytometry Technical Applications Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Federico Sizzano
- Nestlé Institute of Health Sciences, Biobanking & Flow Cytometry Core EPFL, Innovation Park Bâtiment H, Lausanne, Switzerland
| | - Alessio Palini
- Nestlé Institute of Health Sciences, Biobanking & Flow Cytometry Core EPFL, Innovation Park Bâtiment H, Lausanne, Switzerland
| | - Katia Garlaschelli
- Center SISA for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy
| | - Fernanda Tripiciano
- Hematology and Blood Transfusion Service, San Raffaele Scientific Institute, Milan, Italy
| | - Isabella Scotti
- Department of Rheumatology, Istituto Ortopedico Gaetano Pini, Milan, Italy
| | - Alberico Luigi Catapano
- IRCCS - Multimedica Hospital, Sesto San Giovanni, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Angelo A Manfredi
- Unit of Internal Medicine & Clinical Immunology, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy
| | - Giuseppe Danilo Norata
- Center SISA for the Study of Atherosclerosis, Bassini Hospital, Cinisello Balsamo, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Paolo G Camici
- Cardiothoracic Department, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy
| |
Collapse
|
32
|
Imaging inflammation and neovascularization in atherosclerosis: clinical and translational molecular and structural imaging targets. Curr Opin Cardiol 2016; 30:671-80. [PMID: 26398413 DOI: 10.1097/hco.0000000000000226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW The purpose of this study is to showcase advances in molecular imaging of atheroma biology in living individuals. RECENT FINDINGS F-fluorodeoxyglucose (FDG) PET/computed tomography (CT) continues to be the predominant molecular imaging approach for clinical applications, particularly in the large arterial beds. Recently, there has been significant progress in imaging of neovascularization and inflammation to delineate high-risk atheroma and to evaluate drug efficacy. In addition, new hardware detection technology and imaging agents are enabling in-vivo imaging of new targets on diverse imaging platforms. SUMMARY In this review, we present recent exciting developments in molecular and structural imaging of atherosclerotic plaque inflammation and neovascularization. Building upon prior studies, these advances develop key technology that will play an important role in propelling new diagnostic and therapeutic strategies identifying high-risk plaque phenotypes and assessing new plaque stabilization therapies in clinical trials.
Collapse
|
33
|
Whiteford JR, De Rossi G, Woodfin A. Mutually Supportive Mechanisms of Inflammation and Vascular Remodeling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:201-78. [PMID: 27572130 DOI: 10.1016/bs.ircmb.2016.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic inflammation is often accompanied by angiogenesis, the development of new blood vessels from existing ones. This vascular response is a response to chronic hypoxia and/or ischemia, but is also contributory to the progression of disorders including atherosclerosis, arthritis, and tumor growth. Proinflammatory and proangiogenic mediators and signaling pathways form a complex and interrelated network in these conditions, and many factors exert multiple effects. Inflammation drives angiogenesis by direct and indirect mechanisms, promoting endothelial proliferation, migration, and vessel sprouting, but also by mediating extracellular matrix remodeling and release of sequestered growth factors, and recruitment of proangiogenic leukocyte subsets. The role of inflammation in promoting angiogenesis is well documented, but by facilitating greater infiltration of leukocytes and plasma proteins into inflamed tissues, angiogenesis can also propagate chronic inflammation. This review examines the mutually supportive relationship between angiogenesis and inflammation, and considers how these interactions might be exploited to promote resolution of chronic inflammatory or angiogenic disorders.
Collapse
Affiliation(s)
- J R Whiteford
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - G De Rossi
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - A Woodfin
- Cardiovascular Division, King's College, University of London, London, United Kingdom.
| |
Collapse
|
34
|
Iliescu CA, Grines CL, Herrmann J, Yang EH, Cilingiroglu M, Charitakis K, Hakeem A, Toutouzas KP, Leesar MA, Marmagkiolis K. SCAI Expert consensus statement: Evaluation, management, and special considerations of cardio-oncology patients in the cardiac catheterization laboratory (endorsed by the cardiological society of india, and sociedad Latino Americana de Cardiologıa intervencionista). Catheter Cardiovasc Interv 2016; 87:E202-23. [PMID: 26756277 DOI: 10.1002/ccd.26379] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/28/2015] [Indexed: 12/24/2022]
Abstract
In the United States alone, there are currently approximately 14.5 million cancer survivors, and this number is expected to increase to 20 million by 2020. Cancer therapies can cause significant injury to the vasculature, resulting in angina, acute coronary syndromes (ACS), stroke, critical limb ischemia, arrhythmias, and heart failure, independently from the direct myocardial or pericardial damage from the malignancy itself. Consequently, the need for invasive evaluation and management in the cardiac catheterization laboratory (CCL) for such patients has been increasing. In recognition of the need for a document on special considerations for cancer patients in the CCL, the Society for Cardiovascular Angiography and Interventions (SCAI) commissioned a consensus group to provide recommendations based on the published medical literature and on the expertise of operators with accumulated experience in the cardiac catheterization of cancer patients.
Collapse
Affiliation(s)
- Cezar A Iliescu
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Cindy L Grines
- Detroit Medical Center, Cardiovascular Institute, Detroit, Michigan
| | - Joerg Herrmann
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Eric H Yang
- Division of Cardiology, University of California at Los Angeles, Los Angeles, California
| | - Mehmet Cilingiroglu
- School of Medicine, Arkansas Heart Hospital, Little Rock, Arkansas.,Department of Cardiology, Koc University, Istanbul, Turkey
| | | | - Abdul Hakeem
- Department of Cardiology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Massoud A Leesar
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Konstantinos Marmagkiolis
- Department of Cardiology, Citizens Memorial Hospital, Bolivar, Missouri.,Department of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
35
|
Shah RV, Murthy VL. Cardiac magnetic resonance detection of the human carotid: A new lens on neovascularization? Atherosclerosis 2015; 245:60-1. [PMID: 26708284 DOI: 10.1016/j.atherosclerosis.2015.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 12/01/2015] [Indexed: 11/19/2022]
Affiliation(s)
- Ravi V Shah
- Department of Medicine and Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Venkatesh L Murthy
- Departments of Radiology and Cardiology, University of Michigan at Ann Arbor, Ann Arbor, MI, USA
| |
Collapse
|
36
|
van Hinsbergh VWM, Eringa EC, Daemen MJAP. Neovascularization of the atherosclerotic plaque: interplay between atherosclerotic lesion, adventitia-derived microvessels and perivascular fat. Curr Opin Lipidol 2015; 26:405-11. [PMID: 26241102 DOI: 10.1097/mol.0000000000000210] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Neovascularization is a prominent feature in advanced human atherosclerotic plaques. This review surveys recent evidence for and remaining uncertainties regarding a role of neovascularization in atherosclerotic plaque progression. Specific emphasis is given to hypoxia, angiogenesis inhibition, and perivascular adipose tissue (PVAT). RECENT FINDINGS Immunohistochemical and imaging studies showed a strong association between hypoxia, inflammation and neovascularization, and the progression of the atherosclerotic plaque both in humans and mice. Whereas in humans, a profound invasion of microvessels from the adventitia into the plaque occurs, neovascularization in mice is found mainly (peri)adventitially. Influencing neovascularization in mice affected plaque progression, possibly by improving vessel perfusion, but supportive clinical data are not available. Whereas plaque neovascularization contributes to monocyte/macrophage accumulation in the plaque, lymphangiogenesis may facilitate egress of cells and waste products. A specific role for PVAT and its secreted factors is anticipated and wait further clinical evaluation. SUMMARY Hypoxia, inflammation, and plaque neovascularization are associated with plaque progression as underpinned by recent imaging data in humans. Recent studies provide new insights into modulation of adventitia-associated angiogenesis, PVAT, and plaque development in mice, but there is still a need for detailed information on modulating human plaque vascularization in patients.
Collapse
Affiliation(s)
- Victor W M van Hinsbergh
- aLaboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center bDepartment of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
37
|
Traditional Chinese medication Tongxinluo inhibits inflammatory angiogenesis via Bmx/NF- B/MAPK pathways. Eur Heart J Suppl 2015. [DOI: 10.1093/eurheartj/suv020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
38
|
Sadat U, Jaffer FA, van Zandvoort MAMJ, Nicholls SJ, Ribatti D, Gillard JH. Inflammation and neovascularization intertwined in atherosclerosis: imaging of structural and molecular imaging targets. Circulation 2014; 130:786-94. [PMID: 25156914 DOI: 10.1161/circulationaha.114.010369] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Umar Sadat
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.).
| | - Farouc A Jaffer
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Marc A M J van Zandvoort
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Stephen J Nicholls
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Domenico Ribatti
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| | - Jonathan H Gillard
- From the Cambridge Vascular Unit (U.S.) and University Department of Radiology (U.S., J.H.G.), Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, United Kingdom; Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, MA (F.A.J.); Advanced Microscopy Unit, Department of Genetics and Cell Biology-Molecular Cell Biology, Maastricht University, Maastricht, The Netherlands (M.A.M.J.v.Z.); Institute for Molecular Cardiovascular Research, Aachen University, Aachen, Germany (M.A.M.J.v.Z.); South Australian Health and Medical Research Institute and Heart Foundation Heart Health, University of Adelaide and Royal Adelaide Hospital, Adelaide, South Australia, Australia (S.J.N.); Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy (D.R.); and National Cancer Institute "Giovanni Paolo II," Bari, Italy (D.R.)
| |
Collapse
|
39
|
Herrmann J, Lerman A, Sandhu NP, Villarraga HR, Mulvagh SL, Kohli M. Evaluation and management of patients with heart disease and cancer: cardio-oncology. Mayo Clin Proc 2014; 89:1287-306. [PMID: 25192616 PMCID: PMC4258909 DOI: 10.1016/j.mayocp.2014.05.013] [Citation(s) in RCA: 277] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/17/2014] [Accepted: 05/14/2014] [Indexed: 01/17/2023]
Abstract
The care for patients with cancer has advanced greatly over the past decades. A combination of earlier cancer diagnosis and greater use of traditional and new systemic treatments has decreased cancer-related mortality. Effective cancer therapies, however, can result in short- and long-term comorbidities that can decrease the net clinical gain by affecting quality of life and survival. In particular, cardiovascular complications of cancer treatments can have a profound effect on the health of patients with cancer and are more common among those with recognized or unrecognized underlying cardiovascular diseases. A new discipline termed cardio-oncology has thus evolved to address the cardiovascular needs of patients with cancer and optimize their care in a multidisciplinary approach. This review provides a brief introduction and background on this emerging field and then focuses on its practical aspects including cardiovascular risk assessment and prevention before cancer treatment, cardiovascular surveillance and therapy during cancer treatment, and cardiovascular monitoring and management after cancer therapy. The content of this review is based on a literature search of PubMed between January 1, 1960, and February 1, 2014, using the search terms cancer, cardiomyopathy, cardiotoxicity, cardio-oncology, chemotherapy, heart failure, and radiation.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Internal Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN.
| | - Amir Lerman
- Department of Internal Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Nicole P Sandhu
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
| | - Hector R Villarraga
- Department of Internal Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Sharon L Mulvagh
- Department of Internal Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Manish Kohli
- Department of Oncology, Mayo Clinic, Rochester, MN
| |
Collapse
|
40
|
Gacche RN, Meshram RJ. Angiogenic factors as potential drug target: Efficacy and limitations of anti-angiogenic therapy. Biochim Biophys Acta Rev Cancer 2014; 1846:161-79. [DOI: 10.1016/j.bbcan.2014.05.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/05/2014] [Accepted: 05/07/2014] [Indexed: 12/17/2022]
|
41
|
Xu Y, An X, Guo X, Habtetsion TG, Wang Y, Xu X, Kandala S, Li Q, Li H, Zhang C, Caldwell RB, Fulton DJ, Su Y, Hoda MN, Zhou G, Wu C, Huo Y. Endothelial PFKFB3 plays a critical role in angiogenesis. Arterioscler Thromb Vasc Biol 2014; 34:1231-9. [PMID: 24700124 DOI: 10.1161/atvbaha.113.303041] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular cells, particularly endothelial cells, adopt aerobic glycolysis to generate energy to support cellular functions. The effect of endothelial glycolysis on angiogenesis remains unclear. 6-Phosphofructo-2-kinase/fructose-2, 6-bisphosphatase, isoform 3 (PFKFB3) is a critical enzyme for endothelial glycolysis. By blocking or deleting PFKFB3 in endothelial cells, we investigated the influence of endothelial glycolysis on angiogenesis both in vitro and in vivo. APPROACH AND RESULTS Under hypoxic conditions or after treatment with angiogenic factors, endothelial PFKFB3 was upregulated both in vitro and in vivo. The knockdown or overexpression of PFKFB3 suppressed or accelerated endothelial proliferation and migration in vitro, respectively. Neonatal mice from a model of oxygen-induced retinopathy showed suppressed neovascular growth in the retina when endothelial PFKFB3 was genetically deleted or when the mice were treated with a PFKFB3 inhibitor. In addition, tumors implanted in mice deficient in endothelial PFKFB3 grew more slowly and were provided with less blood flow. A lower level of phosphorylated protein kinase B was observed in PFKFB3-knockdown endothelial cells, which was accompanied by a decrease in intracellular lactate. The addition of lactate to PFKFB3-knockdown cells rescued the suppression of endothelial proliferation and migration. CONCLUSIONS The blockade or deletion of endothelial PFKFB3 decreases angiogenesis both in vitro and in vivo. Thus, PFKFB3 is a promising target for the reduction of endothelial glycolysis and its related pathological angiogenesis.
Collapse
Affiliation(s)
- Yiming Xu
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Xiaofei An
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Xin Guo
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Tsadik Ghebreamlak Habtetsion
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Yong Wang
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Xizhen Xu
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Sridhar Kandala
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Qinkai Li
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Honggui Li
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Chunxiang Zhang
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Ruth B Caldwell
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - David J Fulton
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Yunchao Su
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Md Nasrul Hoda
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Gang Zhou
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.)
| | - Chaodong Wu
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.).
| | - Yuqing Huo
- From the Vascular Biology Center, Department of Cellular Biology and Anatomy (Y.X., X.A., X.X., S.K., R.B.C., D.J.F., Y.H.), Cancer Center, Department of Medicine (T.G.H., G.Z.), and Department of Pharmacology and Toxicology (Y.W., Y.S.), Medical College of Georgia and Departments of Medical Laboratory, Imaging and Radiologic Sciences, and Neurology (M.N.H.), Georgia Regents University, Augusta; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China (X.A., Q.L., Y.H.); Department of Nutrition and Food Science, Texas A&M University, College Station (X.G., H.L., C.W.); and Department of Pharmacology, Rush Medical College, Rush University, Chicago, IL (C.Z.).
| |
Collapse
|
42
|
van den Oord SC, van der Burg J, Akkus Z, Bosch JG, van Domburg RT, Sijbrands EJ, van der Steen AF, Schinkel AF. Impact of gender on the density of intraplaque neovascularization: A quantitative contrast-enhanced ultrasound study. Atherosclerosis 2014; 233:461-466. [DOI: 10.1016/j.atherosclerosis.2013.12.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/10/2013] [Accepted: 12/29/2013] [Indexed: 11/30/2022]
|
43
|
Qiao Y, Zhang PJ, Lu XT, Sun WW, Liu GL, Ren M, Yan L, Zhang JD. Panax notoginseng saponins inhibits atherosclerotic plaque angiogenesis by down-regulating vascular endothelial growth factor and nicotinamide adenine dinucleotide phosphate oxidase subunit 4 expression. Chin J Integr Med 2014; 21:259-65. [PMID: 24599820 DOI: 10.1007/s11655-014-1832-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate the mechanism of Panax notoginseng saponins (PNS), an effective component extracted from Panax notoginseng, on atherosclerotic plaque angiogenesis in atherosclerosis-prone apolipoprotein E-knockout (ApoE-KO) mice fed with high-fat, high-cholesterol diet. METHODS Twenty ApoE-KO mice were divided into two groups, the model group and the PNS group. Ten normal C57BL/6J mice were used as a control group. PNS (60 mg/kg) was orally administered daily for 12 weeks in the PNS group. The ratio of plaque area to vessel area was examined by histological staining. The tissue sample of aortic root was used to detect the CD34 and vascular endothelial growth factor (VEGF) expression areas by immunohistochemistry. The expression of VEGF and nicotinamide adenine dinucleotide phosphate oxidase subunit 4 (NOX4) were measured by reverse transcription polymerase chain reaction and Western blotting respectively. RESULTS After treatment with PNS, the plaque areas were decreased (P<0.05). CD34 expressing areas and VEGF expression areas in plaques were significantly decreased (P<0.05). Meanwhile, VEGF and NOX4 mRNA expression were decreased after treatment with PNS. VEGF and NOX4 protein expression were also decreased by about 72% and 63%, respectively (P<0.01). CONCLUSION PNS, which decreases VEGF and NOX4 expression, could alleviate plaque angiogenesis and attenuate atherosclerosis.
Collapse
Affiliation(s)
- Yun Qiao
- Department of Traditional Chinese Medicine, Qilu Hospital, Shandong University, Jinan, 250012, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Mittal B, Mishra A, Srivastava A, Kumar S, Garg N. Matrix metalloproteinases in coronary artery disease. Adv Clin Chem 2014; 64:1-72. [PMID: 24938016 DOI: 10.1016/b978-0-12-800263-6.00001-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Matrix metalloproteinases (MMP) are a family of zinc-containing endoproteinases that degrade extracellular matrix (ECM) components. MMP have important roles in the development, physiology and pathology of cardiovascular system. Metalloproteases also play key roles in adverse cardiovascular remodeling, atherosclerotic plaque formation and plaque instability, vascular smooth muscle cell (SMC) migration and restenosis that lead to coronary artery disease (CAD), and progressive heart failure. The study of MMP in developing animal model cardiovascular systems has been helpful in deciphering numerous pathologic conditions in humans. Increased peripheral blood MMP-2 and MMP-9 in acute coronary syndrome (ACS) may be useful as noninvasive tests for detection of plaque vulnerability. MMP function can be modulated by certain pharmacological drugs that can be exploited for treatment of ACS. CAD is a polygenic disease and hundreds of genes contribute toward its predisposition. A large number of sequence variations in MMP genes have been identified. Case-control association studies have highlighted their potential association with CAD and its clinical manifestations. Although results thus far are inconsistent, meta-analysis has demonstrated that MMP-3 Glu45Lys and MMP-9 1562C/T gene polymorphisms were associated with CAD risk.
Collapse
|
45
|
Dol-Gleizes F, Delesque-Touchard N, Marès AM, Nestor AL, Schaeffer P, Bono F. A new synthetic FGF receptor antagonist inhibits arteriosclerosis in a mouse vein graft model and atherosclerosis in apolipoprotein E-deficient mice. PLoS One 2013; 8:e80027. [PMID: 24224032 PMCID: PMC3817113 DOI: 10.1371/journal.pone.0080027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/27/2013] [Indexed: 01/01/2023] Open
Abstract
Objective The role of fibroblast growth factors (FGFs) in the development of vascular diseases remains incompletely understood. The objective of this study was to examine the effects of a new small-molecule multi-FGF receptor blocker with allosteric properties, SSR128129E, on neointimal proliferation after a vein graft procedure in mice and on the development of atherosclerosis in atherosclerosis-prone apolipoprotein E (apoE)-deficient mice. Methods and Results Vein grafts were performed in 3 month-old male C57BL6 mice. Segments of the vena cava were interposed at the level of the carotid artery. In SSR128129E (50 mg/kg/d)-treated animals, a dramatic decrease in neointimal proliferation was observed 2 and 8 weeks after the graft (72.5 %, p<0.01, and 47.8 %, p<0.05, respectively). Four-week old male apoE-deficient mice were treated with SSR128129E (50 mg/kg/d) for 3 and 5 months in comparison with a control group. SSR128129E treatment resulted in a reduction of lesion size in the aortic sinus (16.4 % (ns) at 3 months and 42.9 % (p<0.01) at 5 months, without any change in serum lipids. SSR128129 significantly reduced FGFR2 mRNA levels in the aortic sinus (p<0.05, n=5-6), but did not affect the mRNA expression levels of other FGF receptors or ligands. Conclusion These studies indicate that FGFs have an important role in the development of vascular diseases like atherosclerosis and graft arteriosclerosis. These data suggest that inhibition of FGF receptors by compounds like SSR128129E might be useful as a new therapeutic approach for these vascular pathologies.
Collapse
|
46
|
Hugh J, Van Voorhees AS, Nijhawan RI, Bagel J, Lebwohl M, Blauvelt A, Hsu S, Weinberg JM. From the Medical Board of the National Psoriasis Foundation: The risk of cardiovascular disease in individuals with psoriasis and the potential impact of current therapies. J Am Acad Dermatol 2013; 70:168-77. [PMID: 24184141 DOI: 10.1016/j.jaad.2013.09.020] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 09/06/2013] [Accepted: 09/10/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Many studies have identified cardiovascular risk factors in patients with psoriasis. Some psoriasis therapies may increase cardiovascular disease (CVD) and others may decrease CVD. OBJECTIVE We reviewed the literature to define the impact of common psoriasis therapies on cardiovascular measures and outcomes. RESULTS Phototherapy has no major cardiovascular impact and may reduce levels of proinflammatory cytokines. Acitretin increases serum lipids and triglycerides, but has not been shown to increase cardiovascular risk. Cyclosporine A increases blood pressure, serum triglycerides, and total cholesterol. Methotrexate is associated with a decreased risk of CVD morbidity and mortality. Among the biologics, data for tumor necrosis factor inhibitors suggest an overall reduction in cardiovascular events. Most data on short-term ustekinumab use suggest no effect on major adverse cardiovascular events, however some authorities remain concerned. Nevertheless, ustekinumab use over a 4-year period shows a decrease in major adverse cardiovascular events when compared both with the general US population and with psoriatics in Great Britain. LIMITATIONS Most studies lack the power and randomization of large clinical trials and long-term follow-up periods. In addition, the increased risk of CVD associated with psoriasis itself is a confounding factor. CONCLUSION Some therapies for moderate to severe psoriasis, including methotrexate and tumor necrosis factor inhibitors, may reduce cardiovascular events in psoriatic patients. Ustekinumab appears to be neutral but there may be a long-term benefit. Appropriate patient counseling and selection and clinical follow-up are necessary to maximize safety with these agents. Further long-term study is necessary to quantify the benefits and risks associated with biologic therapies.
Collapse
Affiliation(s)
- Jeremy Hugh
- Department of Dermatology, St Luke's-Roosevelt Hospital Center, New York, New York
| | - Abby S Van Voorhees
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rajiv I Nijhawan
- Department of Dermatology, St Luke's-Roosevelt Hospital Center, New York, New York
| | - Jerry Bagel
- Psoriasis Treatment Center of Central New Jersey, East Windsor, New Jersey
| | - Mark Lebwohl
- Department of Dermatology, Mount Sinai School of Medicine, New York, New York
| | | | - Sylvia Hsu
- Department of Dermatology, Baylor College of Medicine, Houston, Texas
| | - Jeffrey M Weinberg
- Department of Dermatology, St Luke's-Roosevelt Hospital Center, New York, New York.
| |
Collapse
|
47
|
Yla-Herttuala S, Bentzon JF, Daemen M, Falk E, Garcia-Garcia HM, Herrmann J, Hoefer I, Jauhiainen S, Jukema JW, Krams R, Kwak BR, Marx N, Naruszewicz M, Newby A, Pasterkamp G, Serruys PWJC, Waltenberger J, Weber C, Tokgozoglu L. Stabilization of atherosclerotic plaques: an update. Eur Heart J 2013; 34:3251-8. [DOI: 10.1093/eurheartj/eht301] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
48
|
Hashemi S, Ramezani Tehrani F, Mehrabi Y, Azizi F. Hypertensive pregnancy disorders as a risk factor for future cardiovascular and metabolic disorders (Tehran Lipid and Glucose Study). J Obstet Gynaecol Res 2013; 39:891-7. [PMID: 23438323 DOI: 10.1111/j.1447-0756.2012.02069.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 09/18/2012] [Indexed: 12/01/2022]
Abstract
BACKGROUND Although hypertensive pregnancy disorders (HPD) are common worldwide, there is no consensus regarding their long-term cardiovascular and metabolic consequences. We aimed to ascertain the association between HPD and the development of cardiovascular and metabolic disturbances later in life. MATERIAL AND METHODS A cohort of 226 women with a history of HPD was selected as the case group, and a group of 226 age- and body-mass-index-matched women, with no history of HPD, was selected as controls. Both groups were selected from among participants of the Tehran Lipid and Glucose Study and were compared for concentrations of metabolic parameters and incidence of hypertension, type 2 diabetes mellitus (T2DM) and dyslipidemia over a follow up of 10 years. RESULTS Women with a history of HPD, compared with age- and body-mass-index-matched women without such a history, had an increased risk of cardiovascular and metabolic disorders, including a twofold increased risk for hypertension (95% confidence interval [CI]: 1.4-3.2), a threefold increased risk for T2DM (95% CI: 1.8-5.2) and a 1.3-fold increased risk for dyslipidemia (95% CI: 1.2-1.5). CONCLUSIONS Women with a history of hypertension in pregnancy are at increased risk for subsequent diagnosis of hypertension, dyslipidemia and T2DM later in life. These women may benefit from close monitoring and timely implementation of primary prevention measures of cardiovascular and metabolic risk factors and lifestyle modifications.
Collapse
Affiliation(s)
- Somayeh Hashemi
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, I.R. Iran
| | | | | | | |
Collapse
|
49
|
Purushothaman M, Krishnan P, Purushothaman KR, Baber U, Tarricone A, Perez JS, Wiley J, Kini A, Sharma SK, Fuster V, Moreno PR. Genotype-Dependent Impairment of Hemoglobin Clearance Increases Oxidative and Inflammatory Response in Human Diabetic Atherosclerosis. Arterioscler Thromb Vasc Biol 2012; 32:2769-75. [DOI: 10.1161/atvbaha.112.252122] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Objective—
Haptoglobin (Hp) protein is responsible for hemoglobin clearance after intra-plaque hemorrhage. Hp gene exists as Hp-1 and Hp-2 alleles and the phenotypes show important molecular heterogeneity. We tested the hypothesis that hemoglobin clearance may be deficient in diabetic atheroma from patients with Hp2-2, triggering increased oxidative, inflammatory, and angiogenic patterns compared with controls.
Methods and Results—
Forty patients with diabetes mellitus were genotyped and their peripheral plaques compared after atherectomy. Plaque hemorrhage, iron content, hemoglobin-binding protein CD163, and heme-oxygenase-1 were quantified. Oxidative, inflammatory, and angiogenic patterns were evaluated by measuring myeloperoxidase, interleukin-10, macrophages, vascular cell adhesion molecule-1, smooth muscle actin, and plaque neovascularization (CD34/CD31). Plaques with Hp2-2 (n=7) had increased hemorrhage (
P
<0.005), iron content (
P
<0.001), and reduced CD163 expression (
P
<0.002) compared with controls (n=14). Hp2-2 plaques had increased heme-oxygenase-1 protein (
P
<0.02), myeloperoxidase gene (
P
<0.05), and protein (
P
<0.0001). Anti-inflammatory interleukin-10 gene (
P
<0.04), and protein expressions (
P
<0.0001) were decreased in Hp2-2. Finally, macrophage (
P
<0.0001), vascular cell adhesion molecule-1 (
P
=0.001), smooth muscle actin (
P
=0.002) scores, and neovessels density (
P
<0.0001) were increased in Hp2-2.
Conclusion—
Genotype-dependent impairment of hemoglobin clearance after intra-plaque hemorrhage is associated with increased oxidative, inflammatory, and angiogenic response in human diabetic atherosclerosis.
Collapse
Affiliation(s)
- Meerarani Purushothaman
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - Prakash Krishnan
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - K-Raman Purushothaman
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - Usman Baber
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - Arthur Tarricone
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - Juan S. Perez
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - Jose Wiley
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - Annapoorna Kini
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - Samin K. Sharma
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - Valentin Fuster
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| | - Pedro R. Moreno
- From the Zena and Michael A. Weiner Cardiovascular Institute and the Marie-Josée and Henry R. Kravis Cardiovascular Health Center, Mount Sinai School of Medicine, New York, NY (M.P., P.K., K-R.P., U.B., A.T., J.S.P., J.W., A.K., S.K.S., V.F., P.R.M); and the Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (V.F.)
| |
Collapse
|
50
|
MicroRNAs in Vascular Biology. Int J Vasc Med 2012; 2012:794898. [PMID: 23056947 PMCID: PMC3463915 DOI: 10.1155/2012/794898] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 08/17/2012] [Accepted: 08/21/2012] [Indexed: 02/08/2023] Open
Abstract
Vascular inflammation is an important component of the pathophysiology of cardiovascular diseases, such as hypertension, atherosclerosis, and aneurysms. All vascular cells, including endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), and infiltrating cells, such as macrophages, orchestrate a series of pathological events. Despite dramatic improvements in the treatment of atherosclerosis, the molecular basis of vascular inflammation is not well understood. In the last decade, microRNAs (miRNAs) have been revealed as novel regulators of vascular inflammation. Each miRNAs suppresses a set of genes, forming complex regulatory network. This paper provides an overview of current advances that have been made in revealing the roles of miRNAs during vascular inflammation. Recent studies show that miRNAs not only exist inside cells but also circulate in blood. These circulating miRNAs are useful biomarkers for diagnosis of cardiovascular diseases. Furthermore, recent studies demonstrate that circulating miRNAs are delivered into certain recipient cells and act as messengers. These studies suggest that miRNAs provide new therapeutic opportunities.
Collapse
|