1
|
Hamam O, Gudenkauf J, Moustafa R, Cho A, Montes D, Sharara M, Moustafa A, Radmard M, Nabi M, Chen K, Sepehri S, Shin C, Mazumdar I, Kim M, Mohseni A, Malhotra A, Romero J, Yedavalli V. Hypoperfusion Intensity Ratio as an Indirect Imaging Surrogate in Patients With Anterior Circulation Large-Vessel Occlusion and Association of Baseline Characteristics With Poor Collateral Status. J Am Heart Assoc 2024; 13:e030897. [PMID: 39158547 PMCID: PMC11963918 DOI: 10.1161/jaha.123.030897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 11/15/2023] [Indexed: 08/20/2024]
Abstract
BACKGROUND Collateral status (CS) plays a crucial role in infarct growth rate, risk of postthrombectomy hemorrhage, and overall clinical outcomes in patients with acute ischemic stroke (AIS) secondary to anterior circulation large-vessel occlusions (LVOs). Hypoperfusion intensity ratio has been previously validated as an indirect noninvasive pretreatment imaging biomarker of CS. In addition to imaging, derangements in admission laboratory findings can also influence outcomes in patients with AIS-LVO. Therefore, our study aims to assess the relationship between admission laboratory findings, baseline characteristics, and CS, as assessed by hypoperfusion intensity ratio in patients with AIS-LVO. METHODS AND RESULTS In this retrospective study, consecutive patients presenting with AIS secondary to anterior circulation LVO who underwent pretreatment computed tomography perfusion were included. The computed tomography perfusion data processed by RAPID (Ischema View, Menlo Park, CA) generated the hypoperfusion intensity ratio. Binary logistic regression models were used to assess the relationship between patients' baseline characteristics, admission laboratory findings, and poor CS. A total of 221 consecutive patients with AIS-LVO between January 2017 and September 2022 were included in our study (mean±SD age, 67.0±15.8 years; 119 men [53.8%]). Multivariable logistic regression showed that patients with AIS caused by cardioembolic and cryptogenic causes (adjusted odds ratio [OR], 2.67; 95% CI, 1.20-5.97; P=0.016), those who presented with admission National Institutes of Health Stroke Scale score ≥12 (adjusted OR, 3.12; 95% CI, 1.61-6.04; P=0.001), and male patients (adjusted OR, 2.06; 95% CI, 1.13-3.77; P=0.018) were associated with poor CS. CONCLUSIONS Stroke caused by cardioembolic or cryptogenic causes, admission National Institutes of Health Stroke Scale score of ≥12, and male sex were associated with poor CS, as defined by hypoperfusion intensity ratio in the patients with AIS-LVO.
Collapse
Affiliation(s)
- Omar Hamam
- Department of Radiology, Massachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Julie Gudenkauf
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | - Rawan Moustafa
- Department of Cardiovascular MedicineRobert Wood Johnson Medical SchoolNew BrunswickNJ
- School of Arts and SciencesRutgers University‐NewarkNewarkNJ
| | - Andrew Cho
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | - Daniel Montes
- Radiology ResidentUniversity of Colorado, Anschutz Medical CampusAuroraCO
| | | | - Abdallah Moustafa
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | - Mahla Radmard
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | - Mehreen Nabi
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | - Kevin Chen
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | - Sadra Sepehri
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | | | - Ishan Mazumdar
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | - Minsoo Kim
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | - Alireza Mohseni
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| | | | - Javier Romero
- Department of Radiology, Massachusetts General HospitalHarvard Medical SchoolBostonMA
| | - Vivek Yedavalli
- Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD
| |
Collapse
|
2
|
Salim HA, Hamam O, Parilday G, Moustafa RA, Ghandour S, Rutgers M, Sharara M, Cho A, Mazumdar I, Radmard M, Shin C, Montes D, Malhotra A, Romero JM, Yedavalli V. Relative Cerebral Blood Flow as an Indirect Imaging Surrogate in Patients With Anterior Circulation Large Vessel Occlusion and Association of Baseline Characteristics With Poor Collateral Status. J Am Heart Assoc 2024; 13:e034581. [PMID: 39158542 PMCID: PMC11963935 DOI: 10.1161/jaha.124.034581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND In acute ischemic stroke (AIS), collateral status (CS) is an important predictor of favorable outcomes in patients with AIS. Among quantitative cerebral perfusion parameters, relative cerebral blood flow (rCBF) is considered an accurate perfusion-based indicator of CS. This study investigated the relationship between admission laboratory values, baseline characteristics, and CS as assessed by rCBF in patients with AIS-large vessel occlusion. METHODS AND RESULTS In this retrospective multicenter study, consecutive patients presenting with AIS secondary to anterior circulation large vessel occlusion who underwent pretreatment computed tomography perfusion were included. The computed tomography perfusion data processed by RAPID (IschemaView, Menlo Park, CA) generated the rCBF. Binary logistic regression models assessed the relationship between patients' baseline characteristics, admission laboratory values, and poor CS. The primary outcome measure was the presence of poor CS, which was defined as rCBF <38% at a lesion size ≥27 mL. Between January 2017 and September 2022, there were 221 consecutive patients with AIS-large vessel occlusion included in our study (mean age 67.0±15.8 years, 119 men [53.8%]). Logistic regression showed that male sex (odds ratio [OR], 2.98 [1.59-5.59]; P=0.001), chronic kidney disease (OR, 5.18 [2.44-11.0]; P<0.001), admission National Institutes of Health Stroke Scale score ≥12 (OR, 5.17 [2.36-11.36]; P<0.001), and systolic blood pressure <140 (OR, 2.00 [1.07-3.76]; P=0.030) were associated with poor CS. CONCLUSIONS Higher stroke severity on admission with National Institutes of Health Stroke Scale score ≥12, systolic blood pressure <140, chronic kidney disease, and male sex are statistically significantly associated with poor CS in patients with AIS due to anterior circulation large vessel occlusion as defined by rCBF <38%.
Collapse
Affiliation(s)
| | | | | | | | | | - Moustafa Rutgers
- Rutgers University School of Arts and SciencesNew BrunswickNJUSA
| | | | - Andrew Cho
- Johns Hopkins University School of MedicineBaltimoreMDUSA
| | - Ishan Mazumdar
- Johns Hopkins University School of MedicineBaltimoreMDUSA
| | | | | | - Daniel Montes
- University of ColoradoAnschutz Medical CampusAuroraCAUSA
| | | | | | | |
Collapse
|
3
|
McCallinhart PE, Chade AR, Bender SB, Trask AJ. Expanding landscape of coronary microvascular disease in co-morbid conditions: Metabolic disease and beyond. J Mol Cell Cardiol 2024; 192:26-35. [PMID: 38734061 PMCID: PMC11340124 DOI: 10.1016/j.yjmcc.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Coronary microvascular disease (CMD) and impaired coronary blood flow control are defects that occur early in the pathogenesis of heart failure in cardiometabolic conditions, prior to the onset of atherosclerosis. In fact, recent studies have shown that CMD is an independent predictor of cardiac morbidity and mortality in patients with obesity and metabolic disease. CMD is comprised of functional, structural, and mechanical impairments that synergize and ultimately reduce coronary blood flow in metabolic disease and in other co-morbid conditions, including transplant, autoimmune disorders, chemotherapy-induced cardiotoxicity, and remote injury-induced CMD. This review summarizes the contemporary state-of-the-field related to CMD in metabolic and these other co-morbid conditions based on mechanistic data derived mostly from preclinical small- and large-animal models in light of available clinical evidence and given the limitations of studying these mechanisms in humans. In addition, we also discuss gaps in current understanding, emerging areas of interest, and opportunities for future investigations in this field.
Collapse
Affiliation(s)
- Patricia E McCallinhart
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America
| | - Alejandro R Chade
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, United States of America; Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States of America
| | - Shawn B Bender
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, United States of America.
| | - Aaron J Trask
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States of America.
| |
Collapse
|
4
|
Bartkowiak K, Bartkowiak M, Jankowska-Steifer E, Ratajska A, Kujawa M, Aniołek O, Niderla-Bielińska J. Metabolic Syndrome and Cardiac Vessel Remodeling Associated with Vessel Rarefaction: A Possible Underlying Mechanism May Result from a Poor Angiogenic Response to Altered VEGF Signaling Pathways. J Vasc Res 2024; 61:151-159. [PMID: 38615659 DOI: 10.1159/000538361] [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: 11/15/2023] [Accepted: 03/09/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Elevated mortality rates in patients with metabolic syndrome (MetS) are partly due to adverse remodeling of multiple organs, which may lead to cardiovascular disease, nonalcoholic fatty liver disease, kidney failure, or other conditions. MetS symptoms, such as obesity, hypertension, hyperglycemia, dyslipidemia, associated with insulin and leptin resistance, are recognized as major cardiovascular risk factors that adversely affect the heart. SUMMARY Pathological cardiac remodeling is accompanied by endothelial cell dysfunction which may result in diminished coronary flow, dysregulated oxygen demand/supply balance, as well as vessel rarefaction. The reduced number of vessels and delayed or inhibited formation of collaterals after myocardial infarction in MetS heart may be due to unfavorable changes in endothelial cell metabolism but also to altered expression of vascular endothelial growth factor molecules, their receptors, and changes in signal transduction from the cell membrane, which severely affect angiogenesis. KEY MESSAGES Given the established role of cardiac vessel endothelial cells in maintaining tissue homeostasis, defining the molecular background underlying vessel dysfunction associated with impaired angiogenesis is of great importance for future therapeutic purposes. Therefore, the aim of this paper was to present current information regarding vascular endothelial growth factor signaling in the myocardium of MetS individuals.
Collapse
Affiliation(s)
- Krzysztof Bartkowiak
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Bartkowiak
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| | - Anna Ratajska
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland
| | - Marek Kujawa
- Department of Histology and Embryology, Faculty of Medicine, Lazarski University, Warsaw, Poland
| | - Olga Aniołek
- Department of Histology and Embryology, Faculty of Medicine, Lazarski University, Warsaw, Poland
| | | |
Collapse
|
5
|
Bhalla SR, Riu F, Machado MJC, Bates DO. Measurement of Revascularization in the Hind Limb After Experimental Ischemia in Mice. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2441:105-113. [PMID: 35099732 DOI: 10.1007/978-1-0716-2059-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Peripheral vascular disease is a major cause of morbidity and mortality, and is a consequence of impaired blood flow to the limbs. This arises due to the inability of the tissue to develop sufficiently functional collateral vessel circulation to overcome occluded arteries, or microvascular impairment. The mouse hind limb model of hind limb ischemia can be used to investigate the impact of different treatment modalities, behavioral changes, or genetic knockout. Here we described the model in detail, providing examples of adverse events, and details of ex vivo analysis of blood vessel density.
Collapse
Affiliation(s)
- Sohni Ria Bhalla
- Tumour and Vascular Biology Laboratories, Centre for Cancer Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Federica Riu
- Tumour and Vascular Biology Laboratories, Centre for Cancer Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Maria J C Machado
- Tumour and Vascular Biology Laboratories, Centre for Cancer Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - David O Bates
- Tumour and Vascular Biology Laboratories, Centre for Cancer Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK.
| |
Collapse
|
6
|
The essential role for endothelial cell sprouting in coronary collateral growth. J Mol Cell Cardiol 2022; 165:158-171. [PMID: 35074317 PMCID: PMC8940680 DOI: 10.1016/j.yjmcc.2022.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/11/2022] [Accepted: 01/16/2022] [Indexed: 12/11/2022]
Abstract
RATIONALE Coronary collateral growth is a natural bypass for ischemic heart diseases. It offers tremendous therapeutic benefit, but the process of coronary collateral growth isincompletely understood due to limited preclinical murine models that would enable interrogation of its mechanisms and processes via genetic modification and lineage tracing. Understanding the processes by which coronary collaterals develop can unlock new therapeutic strategies for ischemic heart disease. OBJECTIVE To develop a murine model of coronary collateral growth by repetitive ischemia and investigate whether capillary endothelial cells could contribute to the coronary collateral formation in an adult mouse heart after repetitive ischemia by lineage tracing. METHODS AND RESULTS A murine model of coronary collateral growth was developed using short episodes of repetitive ischemia. Repetitive ischemia stimulation resulted in robust collateral growth in adult mouse hearts, validated by high-resolution micro-computed tomography. Repetitive ischemia-induced collateral formation compensated ischemia caused by occlusion of the left anterior descending artery. Cardiac function improved during ischemia after repetitive ischemia, suggesting the improvement of coronary blood flow. A capillary-specific Cre driver (Apln-CreER) was used for lineage tracing capillary endothelial cells. ROSA mT/mG reporter mice crossed with the Apln-CreER transgene mice underwent a 17 days' repetitive ischemia protocol for coronary collateral growth. Two-photon and confocal microscopy imaging of heart slices revealed repetitive ischemia-induced coronary collateral growth initiated from sprouting Apelin+ endothelial cells. Newly formed capillaries in the collateral-dependent zone expanded in diameter upon repetitive ischemia stimulation and arterialized with smooth muscle cell recruitment, forming mature coronary arteries. Notably, pre-existing coronary arteries and arterioles were not Apelin+, and all Apelin+ collaterals arose from sprouting capillaries. Cxcr4, Vegfr2, Jag1, Mcp1, and Hif1⍺ mRNA levels in the repetitive ischemia-induced hearts were also upregulated at the early stage of coronary collateral growth, suggesting angiogenic signaling pathways are activated for coronary collaterals formation during repetitive ischemia. CONCLUSIONS We developed a murine model of coronary collateral growth induced by repetitive ischemia. Our lineage tracing study shows that sprouting endothelial cells contribute to coronary collateral growth in adult mouse hearts. For the first time, sprouting angiogenesis is shown to give rise to mature coronary arteries in response to repetitive ischemia in the adult mouse hearts.
Collapse
|
7
|
Shao MJ, Luo JY, Shi J, Liu F, Shan CF, Luo F, Yu XL, Zhao Q, Tian T, Li XM, Yang YN. Association of Visceral Obesity-Related Indices With Coronary Collateralization in Patients With Chronic Total Occlusion. Front Cardiovasc Med 2021; 8:742855. [PMID: 34746259 PMCID: PMC8566720 DOI: 10.3389/fcvm.2021.742855] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/24/2021] [Indexed: 01/20/2023] Open
Abstract
Background: Obesity is an independent risk factor for cardiovascular disease. We investigated whether and to what extent visceral obesity-related indices were associated with coronary collateralization (CC) in chronic total occlusion (CTO) patients. Methods: This retrospective cohort study involved 1,008 consecutive patients with CTO who underwent CTO-percutaneous coronary artery intervention (PCI). CC was graded according to the Rentrop scoring system. Data on demographic and clinical characteristics were collected by cardiovascular doctors. Logistic regression, receiver operating characteristic (ROC) curve and Kaplan-Meier analyses were performed to assess the predictive value of visceral obesity-related indices for CC. Results: Overall, 1,008 inpatients were assigned to the poor CC group (n = 592) and good CC group (n = 416). In multivariate-adjusted logistic regression analyses, all visceral obesity-related indices (P-value < 0.001) were significantly associated with CC. After ROC analysis and the Delong test, the Chinese visceral adiposity index (CVAI) had the largest area under the curve (AUC) of 0.741 (0.711–0.771). Further analysis revealed that CVAI quartile remained a risk factor for poor CC in all groups, CVAI was associated with a 1.018-fold higher risk of poor CC (OR = 1.018, 95% CI: 1.014–1.021, P < 0.001). Individuals in the top CVAI quartile group had the highest risk of poor CC (OR = 10.657, 95% CI: 6.492–17.493, P < 0.001). Subgroup analyses showed similar results, and CVAI quartile remained a risk factor for poor CC. Moreover, increased CVAI predicted poor prognosis in CTO patients. Conclusion: In summary, this study indicated that all the increased visceral obesity-related indices were significantly associated with increased poor CC risk. After adjusting for potential risks, CVAI had the best performance for estimating CC and predicting prognosis in CTO patients.
Collapse
Affiliation(s)
- Meng-Jiao Shao
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jun-Yi Luo
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jia Shi
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chun-Fang Shan
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fan Luo
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiao-Lin Yu
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Qian Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ting Tian
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiao-Mei Li
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yi-Ning Yang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
8
|
A novel selective PPARα modulator, pemafibrate promotes ischemia-induced revascularization through the eNOS-dependent mechanisms. PLoS One 2020; 15:e0235362. [PMID: 32584895 PMCID: PMC7316279 DOI: 10.1371/journal.pone.0235362] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/13/2020] [Indexed: 12/14/2022] Open
Abstract
Objective Cardiovascular disease is a leading cause of death worldwide. Obesity-related metabolic disorders including dyslipidemia cause impaired collateralization under ischemic conditions, thereby resulting in exacerbated cardiovascular dysfunction. Pemafibrate is a novel selective PPARα modulator, which has been reported to improve atherogenic dyslipidemia, in particular, hypertriglyceridemia and low HDL-cholesterol. Here, we investigated whether pemafibrate modulates the revascularization process in a mouse model of hindlimb ischemia. Methods and results Male wild-type (WT) mice were randomly assigned to two groups, normal diet or pemafibrate admixture diet from the ages of 6 weeks. After 4 weeks, mice were subjected to unilateral hindlimb surgery to remove the left femoral artery and vein. Pemafibrate treatment enhanced blood flow recovery and capillary formation in ischemic limbs of mice, which was accompanied by enhanced phosphorylation of endothelial nitric oxide synthase (eNOS). Treatment of cultured endothelial cells with pemafibrate resulted in increased network formation and migratory activity, which were blocked by pretreatment with the NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME). Pemafibrate treatment also increased plasma levels of the PPARα-regulated gene, fibroblast growth factor (FGF) 21 in WT mice. Systemic administration of adenoviral vectors expressing FGF21 (Ad-FGF21) to WT mice enhanced blood flow recovery, capillary density and eNOS phosphorylation in ischemic limbs. Treatment of cultured endothelial cells with FGF21 protein led to increases in endothelial cell network formation and migration, which were canceled by pretreatment with L-NAME. Furthermore, administration of pemafibrate or Ad-FGF21 had no effects on blood flow in ischemic limbs in eNOS-deficient mice. Conclusion These data suggest that pemafibrate can promote revascularization in response to ischemia, at least in part, through direct and FGF21-mediated modulation of endothelial cell function. Thus, pemafibrate could be a potentially beneficial drug for ischemic vascular disease.
Collapse
|
9
|
Elevated GTP Cyclohydrolase I Pathway in Endothelial Progenitor Cells of Overweight Premenopausal Women. Cardiol Res Pract 2020; 2020:5914916. [PMID: 32377428 PMCID: PMC7199531 DOI: 10.1155/2020/5914916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/15/2019] [Indexed: 11/17/2022] Open
Abstract
Background/Aims. Sexual differences exist in endothelial progenitor cells (EPCs), and various cardiovascular risk factors are associated with the preservation of endothelial function in premenopausal women. However, it is unclear whether differences in endothelial function and circulating EPCs exist between overweight premenopausal women and age-matched men. Methods. We compared EPC counting and functions in normal-weight and overweight premenopausal women and men, evaluated endothelial function in each group, and detected the expression of the guanosine triphosphate cyclohydrolase I (GTPCH I) pathway. Results. The number of EPCs was lower in the male group than in the female group, regardless of normal-weight or overweight status, and there was no significant difference between the different weight groups among females or males. Endothelial function and EPC migration and proliferation were preserved in overweight premenopausal women compared with overweight men as were nitric oxide (NO) levels in plasma and secreted by EPCs. Endothelial function, the circulating EPC population, and NO levels were not different between normal-weight and overweight premenopausal women. Flow-mediated dilatation was significantly correlated with EPC function, plasma NO levels, and EPC-secreted NO. Conclusions. This investigation provides the first evidence for sex-based differences in EPC activity and endothelial function in overweight middle-aged individuals; these differences are associated with alterations in NO production and may partly occur through downregulation of the GTPCH I pathway. The present results provide new insights into the mechanism underlying the preserved endothelial function in overweight premenopausal women and may uncover a potential therapeutic target for endothelial repair in overweight population.
Collapse
|
10
|
Sahinturk Y, Kucukseymen S, Avci R, Akarsu A, Yolcular BO, Koker G, Tokuc A, Bayar N, Arslan S. Visceral fat index: a novel predictor for coronary collateral circulation. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2020; 64:150-158. [PMID: 32236316 PMCID: PMC10118954 DOI: 10.20945/2359-3997000000218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/30/2019] [Indexed: 11/23/2022]
Abstract
Objective This study was designed to investigate the role of visceral adiposity along with other clinical parameters in predicting poor coronary collateral circulation (CCC) among patients with severe obstructive coronary artery disease (CAD). Subjects and methods A total of 135 patients with severe obstructive CAD and good (n = 70) or poor (n = 65) CCC were included. Data on angiographically detected CCC, the quality criteria for CCC (Rentrop scores) and visceral fat index (VFI) obtained via bioelectrical impedance were compared between good and poor CCC groups. Independent predictors of poor CCC, the correlation between VFI and Rentrop score and the role of VFI in the identification of CCC were analyzed. Results A significant negative correlation was noted between VFI and Rentrop scores (r = -0.668, < 0.001). The presence of hypertension (OR 4.244, 95% CI 1.184 to 15.211, p = 0.026) and higher VFI (OR 1.955, 95% CI 1.342 to 2.848, p < 0.001) were shown to be independent predictors of an increased risk for poor CCC. ROC analysis revealed a VFI > 9 (AUC [area under the curve] (95% CI): 0.898 (0.834-0.943), p < 0.0001) to be a potential predictor of poor CCC with a sensitivity of 95.38% and specificity of 85.71%. Conclusion In conclusion, our findings revealed comorbid hypertension and higher VFI to significantly predict the risk of poor CCC in patients with severe obstructive CAD.
Collapse
Affiliation(s)
- Yasin Sahinturk
- Departments of Internal Medicine, Training and Research Hospital, University of Health Sciences Antalya, Antalya, Turkey
| | - Selcuk Kucukseymen
- Departments of Cardiology, Training and Research Hospital, University of Health Sciences Antalya, Antalya, Turkey
| | - Rauf Avci
- Departments of Cardiology, Training and Research Hospital, University of Health Sciences Antalya, Antalya, Turkey
| | - Ayse Akarsu
- Departments of Internal Medicine, Training and Research Hospital, University of Health Sciences Antalya, Antalya, Turkey
| | | | - Gokhan Koker
- Departments of Internal Medicine, Training and Research Hospital, University of Health Sciences Antalya, Antalya, Turkey
| | - Abdullah Tokuc
- Departments of Internal Medicine, Training and Research Hospital, University of Health Sciences Antalya, Antalya, Turkey
| | - Nermin Bayar
- Departments of Cardiology, Training and Research Hospital, University of Health Sciences Antalya, Antalya, Turkey
| | - Sakir Arslan
- Departments of Cardiology, Training and Research Hospital, University of Health Sciences Antalya, Antalya, Turkey
| |
Collapse
|
11
|
Jamaiyar A, Juguilon C, Dong F, Cumpston D, Enrick M, Chilian WM, Yin L. Cardioprotection during ischemia by coronary collateral growth. Am J Physiol Heart Circ Physiol 2018; 316:H1-H9. [PMID: 30379567 DOI: 10.1152/ajpheart.00145.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ischemic heart diseases (IHD) cause millions of deaths around the world annually. While surgical and pharmacological interventions are commonly used to treat patients with IHD, their efficacy varies from patient to patient and is limited by the severity of the disease. One promising, at least theoretically, approach for treating IHD is induction of coronary collateral growth (CCG). Coronary collaterals are arteriole-to-arteriole anastomoses that can undergo expansion and remodeling in the setting of coronary disease when the disease elicits myocardial ischemia and creates a pressure difference across the collateral vessel that creates unidirectional flow. Well-developed collaterals can restore blood flow in the ischemic area of the myocardium and protect the myocardium at risk. Moreover, such collaterals are correlated to reduced mortality and infarct size and better cardiac function during occlusion of coronary arteries. Therefore, understanding the process of CCG is highly important as a potentially viable treatment of IHD. While there are several excellent review articles on this topic, this review will provide a unified overview of the various aspects related to CCG as well as an update of the advancements in the field. We also call for more detailed studies with an interdisciplinary approach to advance our knowledge of CCG. In this review, we will describe growth of coronary collaterals, the various factors that contribute to CCG, animal models used to study CCG, and the cardioprotective effects of coronary collaterals during ischemia. We will also discuss the impairment of CCG in metabolic syndrome and the therapeutic potentials of CCG in IHD.
Collapse
Affiliation(s)
- Anurag Jamaiyar
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University , Kent, Ohio
| | - Cody Juguilon
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Devan Cumpston
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Molly Enrick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| |
Collapse
|
12
|
Mazidi M, Rezaie P, Kengne AP, Stathopoulou MG, Azimi-Nezhad M, Siest S. VEGF, the underlying factor for metabolic syndrome; fact or fiction? Diabetes Metab Syndr 2017; 11 Suppl 1:S61-S64. [PMID: 28040466 DOI: 10.1016/j.dsx.2016.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 12/08/2016] [Indexed: 01/20/2023]
Abstract
Metabolic syndrome (MetS) is currently diagnosed by the co-presence of at least three of the five following abnormalities: abdominal obesity, dysglycaemia, elevated serum triglycerides, low high-density cholesterol (HDL) and finally elevated blood pressure. Metabolic syndrome increases the risk of developing cardiovascular disease and diabetes. This review is on the associations between MetS and vascular endothelial growth factor (VEGF). VEGF induces migration and proliferation of endothelial cells (ECs), increases vascular permeability and has a role in tumor growth, adipose tissue expansion, age-related macular degeneration and diabetic retinopathy. Circulating levels of VEGFs are elevated in obese individuals and it has also been suggested that VEGF is secreted from adipose tissues, especially from intra-abdominal adipose tissue. There is abundant evidence to support that poor glycemic control in diabetic patients is associated with increased plasma VEGF, which in turn may cause hypertension and several vascular complications in diabetic patients. Circulating VEGF levels are increased in children and young adults with type 1 diabetes mellitus and middle-aged diabetic patients with proliferative retinopathy. It has been revealed that plasma VEGF increases in patients with hyperlipidemia and may trigger the development of atherosclerosis. It can be concluded that there is a positive association between VEGF and components of MetS. Because of the importance of this relationship, more investigations are needed in this field.
Collapse
Affiliation(s)
- Mohsen Mazidi
- Institute of Genetics and Developmental Biology, International College, University of Chinese Academy of Science (IC-UCAS), West Beichen Road, Chaoyang, China; Key State Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| | - Peyman Rezaie
- Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - A P Kengne
- Non-Communicable Disease Research Unit, South African Medical Research Council and University of Cape Town, Cape Town, South Africa
| | - Maria G Stathopoulou
- UMR INSERM U 1122, IGE-PCV "Interactions Gène-Environnement en Physiopathologie CardioVasculaire ", Université de Lorraine, Nancy, France
| | - Mohsen Azimi-Nezhad
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran; Neyshabur University of Medical Sciences, Neyshabur, Iran.
| | - Sophie Siest
- UMR INSERM U 1122, IGE-PCV "Interactions Gène-Environnement en Physiopathologie CardioVasculaire ", Université de Lorraine, Nancy, France
| |
Collapse
|
13
|
Sen T, Astarcioglu MA, Beton O, Asarcikli LD, Kilit C. Which Coronary Lesions Are More Prone to Cause Acute Myocardial Infarction? Arq Bras Cardiol 2017; 108:149-153. [PMID: 28099589 PMCID: PMC5344660 DOI: 10.5935/abc.20170003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 09/13/2016] [Indexed: 11/27/2022] Open
Abstract
Background According to common belief, most myocardial infarctions (MIs) are due to the
rupture of nonsevere, vulnerable plaques with < 70% obstruction. Data
from recent trials challenge this belief, suggesting that the risk of
coronary occlusion is, in fact, much higher after severe stenosis. The aim
of this study was to investigate whether or not acute ST-elevation MIs
result from high-grade stenoses by evaluating the presence of coronary
collateral circulation (CCC). Methods We retrospectively included 207 consecutive patients who had undergone
primary percutaneous coronary intervention for acute ST-elevation MI.
Collateral blood flow distal to the culprit lesion was assessed by two
investigators using the Rentrop scoring system. Results Out of the 207 patients included in the study, 153 (73.9%) had coronary
collateral vessels (Rentrop 1-3). The Rentrop scores were 0, 1, 2, and 3 in
54 (26.1%), 50 (24.2%), 51 (24.6%), and 52 (25.1%) patients, respectively.
Triglycerides, mean platelet volume (MPV), white cell (WBC) count, and
neutrophil count were significantly lower in the group with good collateral
vessels (p = 0.013, p = 0.002, p = 0.003, and p = 0.021, respectively). Conclusion More than 70% of the patients with acute MI had CCC with Rentrop scores of
1-3 during primary coronary angiography. This shows that most cases of acute
MI in our study originated from underlying high-grade stenoses, challenging
the common believe. Higher serum triglycerides levels, greater MPV, and
increased WBC and neutrophil counts were independently associated with
impaired development of collateral vessels.
Collapse
Affiliation(s)
- Taner Sen
- Dumlupinar University Kutahya Evliya Celebi Education and Research Hospital, Kutahya - Turkey
| | - Mehmet Ali Astarcioglu
- Dumlupinar University Kutahya Evliya Celebi Education and Research Hospital, Kutahya - Turkey
| | | | | | - Celal Kilit
- Dumlupinar University Kutahya Evliya Celebi Education and Research Hospital, Kutahya - Turkey
| |
Collapse
|
14
|
Amoah V, Wrigley B, Holroyd E, Smallwood A, Armesilla AL, Nevill A, Cotton J. Vascular endothelial growth factor and hypoxia-inducible factor-1α gene polymorphisms and coronary collateral formation in patients with coronary chronic total occlusions. SAGE Open Med 2016; 4:2050312116654403. [PMID: 27621802 PMCID: PMC5006805 DOI: 10.1177/2050312116654403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/10/2016] [Indexed: 11/30/2022] Open
Abstract
Introduction: We evaluated the association between two single nucleotide polymorphisms of the vascular endothelial growth factor gene and one of the hypoxia-inducible factor-1α gene and the degree of coronary collateral formation in patients with a coronary chronic total occlusion. Methods: Totally, 98 patients with symptomatic coronary artery disease and a chronic total occlusion observed during coronary angiography were recruited. Genotyping of two vascular endothelial growth factor promoter single nucleotide polymorphisms (−152G>A and −165C>T) and the C1772T single nucleotide polymorphism of hypoxia-inducible factor-1α were performed using polymerase chain reaction and restriction fragment length polymorphism analysis. The presence and extent of collateral vessel filling was scored by blinded observers using the Rentrop grade. Results: We found no association between the vascular endothelial growth factor −152G>A, −165C>T and hypoxia-inducible factor-1α −1772C>T with the presence and filling of coronary collateral vessels. A history of percutaneous coronary intervention and transient ischaemic attack/cerebrovascular accident were associated with the presence of enhanced collateral vessel formation following binary logistic regression analysis. Conclusion: The study findings suggest that coronary collateral formation is not associated with the tested polymorphic variants of vascular endothelial growth factor and hypoxia-inducible factor-1α in patients with symptomatic coronary artery disease and the presence of a chronic total occlusion.
Collapse
Affiliation(s)
- Vincent Amoah
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Benjamin Wrigley
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Eric Holroyd
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Andrew Smallwood
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Angel L Armesilla
- Research Institute in Healthcare Science, School of Pharmacy, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - Alan Nevill
- Faculty of Education, Health and Wellbeing, University of Wolverhampton, Wolverhampton, UK
| | - James Cotton
- Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
- James Cotton, Department of Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, West Midlands WV10 0QP, UK.
| |
Collapse
|
15
|
DiStasi MR, Mund JA, Bohlen HG, Miller SJ, Ingram DA, Dalsing MC, Unthank JL. Impaired compensation to femoral artery ligation in diet-induced obese mice is primarily mediated via suppression of collateral growth by Nox2 and p47phox. Am J Physiol Heart Circ Physiol 2015; 309:H1207-17. [PMID: 26297224 DOI: 10.1152/ajpheart.00180.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023]
Abstract
The present study was undertaken to establish the role of NADPH oxidase (Nox) in impaired vascular compensation to arterial occlusion that occurs in the presence of risk factors associated with oxidative stress. Diet-induced obese (DIO) mice characterized by multiple comorbidities including diabetes and hyperlipidemia were used as a preclinical model. Arterial occlusion was induced by distal femoral artery ligation in lean and DIO mice. Proximal collateral arteries were identified as the site of major (∼70%) vascular resistance to calf perfusion by distal arterial pressures, which decreased from ∼80 to ∼30 mmHg with ligation in both lean and DIO mice. Two weeks after ligation, significant vascular compensation occurred in lean but not DIO mice as evidenced by increased perfusion (147 ± 48% vs. 49 ± 29%) and collateral diameter (151 ± 30% vs. 44 ± 17%). Vascular mRNA expression of p22(phox), Nox2, Nox4, and p47(phox) were all increased in DIO mice. Treatment of DIO mice with either apocynin or Nox2ds-tat or with whole body ablation of either Nox2 or p47(phox) ameliorated the impairment in both collateral growth and hindlimb perfusion. Multiparametric flow cytometry analysis demonstrated elevated levels of circulating monocytes in DIO mice without impaired mobilization and demargination after femoral artery ligation. These results establish collateral resistance as the major limitation to calf perfusion in this preclinical model, demonstrate than monocyte mobilization and demarginatin is not suppressed, implicate Nox2-p47(phox) interactions in the impairment of vascular compensation to arterial occlusion in DIO mice, and suggest that selective Nox component suppression/inhibition may be effective as either primary or adjuvant therapy for claudicants.
Collapse
Affiliation(s)
- Matthew R DiStasi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Julie A Mund
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - H Glenn Bohlen
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Steven J Miller
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana; Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - David A Ingram
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana; and Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Michael C Dalsing
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Joseph L Unthank
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana; Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana;
| |
Collapse
|
16
|
Söğüt E, Kadı H, Karayakalı M, Mertoğlu C. The association of plasma vitamin A and E levels with coronary collateral circulation. Atherosclerosis 2015; 239:547-51. [PMID: 25728388 DOI: 10.1016/j.atherosclerosis.2015.02.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/15/2015] [Accepted: 02/14/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To investigate if plasma levels of vitamin A and E have an association with coronary collateral development. METHODS A total of 189 patients who underwent coronary angiography and had total occlusion in at least one major epicardial coronary artery were enrolled in the study. To classify coronary collateral circulation (CCC), the Rentrop scoring system was used. Patients were classified as having poor CCC (Rentrop grades 0-1) or good CCC (Rentrop grades 2-3), and all patients were also screened for hypertension, hypercholesterolemia, diabetes, and smoking history. RESULTS There were no differences in plasma vitamin A and E levels between the two groups (vitamin A: 2.37 ± 0.65 vs. 2.35 ± 0.78, p = 0.253; vitamin E: 47.1 ± 12.8 vs. 44.6 ± 15.1, p = 0.082), and plasma vitamin A and E levels were not associated with CCC. Serum high-sensitivity C-reactive protein (hs-CRP) levels were significantly higher in patients with poor CCC (4.68 ± 2.52 vs. 3.89 ± 1.78, p = 0.001). The higher frequency of diabetes and higher serum hs-CRP levels were found to be an independent predictor for poor CCC (odds ratio = 2.44, p = 0.006; odds ratio = 1.24, p = 0.007, respectively). And a higher frequency of total occluded RCA was found to be a positive predictor for good CCC (odds ratio = 2.36, p = 0.06) in a multivariate logistic regression analysis. CONCLUSIONS We found that serum hs-CRP levels, presence of diabetes, and total occlusion of RCA have an effect on coronary collateral development. We found no correlation between plasma vitamin A and E levels and CCC.
Collapse
Affiliation(s)
- Erkan Söğüt
- Izmir Kâtip Çelebi University, Faculty of Medicine, Department of Biochemistry, Izmir, Turkey.
| | - Hasan Kadı
- Gaziosmanpaşa University, Faculty of Medicine, Department of Cardiology, Tokat, Turkey
| | - Metin Karayakalı
- Gaziosmanpaşa University, Faculty of Medicine, Department of Cardiology, Tokat, Turkey
| | - Cuma Mertoğlu
- Gaziosmanpaşa University, Faculty of Medicine, Department of Biochemistry, Tokat, Turkey
| |
Collapse
|
17
|
Metformin stimulates ischemia-induced revascularization through an eNOS dependent pathway in the ischemic hindlimb mice model. J Vasc Surg 2015; 61:489-96. [DOI: 10.1016/j.jvs.2013.09.061] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/08/2013] [Accepted: 09/17/2013] [Indexed: 11/24/2022]
|
18
|
Ngo DTM, Farb MG, Kikuchi R, Karki S, Tiwari S, Bigornia SJ, Bates DO, LaValley MP, Hamburg NM, Vita JA, Hess DT, Walsh K, Gokce N. Antiangiogenic actions of vascular endothelial growth factor-A165b, an inhibitory isoform of vascular endothelial growth factor-A, in human obesity. Circulation 2014; 130:1072-80. [PMID: 25116954 DOI: 10.1161/circulationaha.113.008171] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Experimental studies suggest that visceral adiposity and adipose tissue dysfunction play a central role in obesity-related cardiometabolic complications. Impaired angiogenesis in fat has been implicated in the development of adipose tissue hypoxia, capillary rarefaction, inflammation, and metabolic dysregulation, but pathophysiological mechanisms remain unknown. In this study, we examined the role of a novel antiangiogenic isoform of vascular endothelial growth factor-A (VEGF-A), VEGF-A165b, in human obesity. METHODS AND RESULTS We biopsied paired subcutaneous and visceral adipose tissue in 40 obese subjects (body mass index, 45±8 kg/m(2); age, 45±11 years) during bariatric surgery and characterized depot-specific adipose tissue angiogenic capacity using an established ex vivo assay. Visceral adipose tissue exhibited significantly blunted angiogenic growth compared with subcutaneous fat (P<0.001) that was associated with marked tissue upregulation of VEGF-A165b (P=0.004). The extent of VEGF-A165b expression correlated negatively with angiogenic growth (r=-0.6, P=0.006). Although recombinant VEGF-A165b significantly impaired angiogenesis, targeted inhibition of VEGF-A165b with neutralizing antibody stimulated fat pad neovascularization and restored VEGF receptor activation. Blood levels of VEGF-A165b were significantly higher in obese subjects compared with lean control subjects (P=0.02), and surgical weight loss induced a marked decline in serumVEGF-A165b (P=0.003). CONCLUSIONS We demonstrate that impaired adipose tissue angiogenesis is associated with overexpression of a novel antiangiogenic factor, VEGF-A165b, that may play a pathogenic role in human adiposopathy. Moreover, systemic upregulation of VEGF-A165b in circulating blood may have wider-ranging implications beyond the adipose milieu. VEGF-A165b may represent a novel area of investigation to gain further understanding of mechanisms that modulate the cardiometabolic consequences of obesity.
Collapse
Affiliation(s)
- Doan T M Ngo
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Melissa G Farb
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Ryosuke Kikuchi
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Shakun Karki
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Stephanie Tiwari
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Sherman J Bigornia
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - David O Bates
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Michael P LaValley
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Naomi M Hamburg
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Joseph A Vita
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Donald T Hess
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Kenneth Walsh
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.)
| | - Noyan Gokce
- From the Evans Department of Medicine and Whitaker Cardiovascular Institute (D.T.M.N., M.G.F., R.K., S.K., S.T., S.J.B., N.M.H., J.A.V., K.W., N.G.) and Department of General Surgery (D.T.H.), Boston University School of Medicine, Boston, MA; Microvascular Research Laboratories, School of Physiology and Pharmacology, Bristol Heart Institute, School of Veterinary Science, University of Bristol, Bristol, UK (D.O.B.); and Department of Biostatistics, Boston University School of Public Health, Boston, MA (M.P.L.).
| |
Collapse
|
19
|
Bhatt H, Kochar S, Htun WW, Julliard K, Fernaine G. Coronary Collateral Circulation and Cardiovascular Risk Factors: Is There a Paradox? Angiology 2014; 66:588-94. [PMID: 25092680 DOI: 10.1177/0003319714545342] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We sought to determine the association of major cardiovascular risk factors and other comorbidities with the presence or absence of coronary collateral (CC) circulation. All electronic medical records from 2010 to 2011 were retrospectively reviewed. A total of 563 patients were divided into 2 groups: CC present (180) and CC absent (383). Smoking (P = .012, odds ratio [OR] 1.58), hypercholesterolemia (P = .001, OR 2.21), and hypertension (P = .034, OR 1.75) were associated with the presence of CC. Increasing body mass index (BMI, P = .001) and decreasing estimated glomerular filtration rate (eGFR, P = .042) were associated with the absence of CC. On multivariable linear regression analysis, hypercholesterolemia (P = .001, OR 2.28), BMI (P = .012, OR 0.77), and eGFR (P = .001, OR 0.70) were found to be independently associated with CC. Our findings will help predict patient populations more likely to have presence or absence of CC circulation.
Collapse
Affiliation(s)
- Hemal Bhatt
- Department of Internal Medicine, Lutheran Medical Center, Brooklyn, NY, USA
| | - Suzi Kochar
- Department of Internal Medicine, Lutheran Medical Center, Brooklyn, NY, USA
| | - Wah Wah Htun
- Department of Internal Medicine, Lutheran Medical Center, Brooklyn, NY, USA
| | - Kell Julliard
- Department of Internal Medicine, Lutheran Medical Center, Brooklyn, NY, USA
| | - George Fernaine
- Department of Cardiology, Lutheran Medical Center, Brooklyn, NY, USA
| |
Collapse
|
20
|
Tanaka M, Fukui M, Okada H, Senmaru T, Asano M, Akabame S, Yamazaki M, Tomiyasu KI, Oda Y, Hasegawa G, Toda H, Nakamura N. Low serum bilirubin concentration is a predictor of chronic kidney disease. Atherosclerosis 2014; 234:421-5. [PMID: 24763407 DOI: 10.1016/j.atherosclerosis.2014.03.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/13/2014] [Accepted: 03/17/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Chronic kidney disease (CKD) is a worldwide public health problem. It is very important to identify the factors that affect CKD. Previous studies have reported that serum bilirubin concentration was positively correlated with renal function in a cross-sectional study. The aim of this study was to investigate the relationship between serum bilirubin concentration and the progression of CKD. METHODS A cohort study was performed on a consecutive series of 2784 subjects without CKD, defined as estimated glomerular filtration rate (eGFR) < 60 ml/min/1.73 m(2), at baseline. We analyzed the relationship between serum total bilirubin concentration at baseline and new-onset CKD in the general population. RESULTS We followed the subjects for a median period of 7.7 years. There were 1157 females and 1627 males, and 231 females and 370 males developed CKD during this period. Multiple Cox regression analyses revealed that serum total bilirubin concentration (hazard ratio (HR) per 1.0 μmol/L increase 0.97 (95% CI 0.95-0.99), P = 0.0084) in addition to age, gamma-glutamyl transpeptidase (GGT), uric acid (UA), creatinine and medication for hypertension in men and serum total bilirubin concentration (HR per 1.0 μmol/L increase 0.96 (95% CI 0.93-1.00), P = 0.0309) in addition to age, GGT, alanine aminotransferase, UA, creatinine and medication for dyslipidemia in women were independent predictors of new-onset CKD, after adjusting for confounders. CONCLUSION Our study demonstrated that serum total bilirubin concentration could be a novel risk factor for the progression of CKD, defined as eGFR <60 ml/min/1.73 m(2), in the general population.
Collapse
Affiliation(s)
- Muhei Tanaka
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan.
| | - Hiroshi Okada
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan
| | - Takafumi Senmaru
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan
| | - Mai Asano
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan
| | | | - Masahiro Yamazaki
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan
| | | | - Yohei Oda
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan
| | - Goji Hasegawa
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan
| | - Hitoshi Toda
- Department of Internal Medicine, Oike Clinic, Kyoto, Japan
| | - Naoto Nakamura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan
| |
Collapse
|
21
|
Ohashi K, Enomoto T, Joki Y, Shibata R, Ogura Y, Kataoka Y, Shimizu Y, Kambara T, Uemura Y, Yuasa D, Matsuo K, Hayakawa S, Hiramatsu-Ito M, Murohara T, Ouchi N. Neuron-derived neurotrophic factor functions as a novel modulator that enhances endothelial cell function and revascularization processes. J Biol Chem 2014; 289:14132-44. [PMID: 24706764 DOI: 10.1074/jbc.m114.555789] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Strategies to stimulate revascularization are valuable for cardiovascular diseases. Here we identify neuron-derived neurotrophic factor (NDNF)/epidermacan as a secreted molecule that is up-regulated in endothelial cells in ischemic limbs of mice. NDNF was secreted from cultured human endothelial cells, and its secretion was stimulated by hypoxia. NDNF promoted endothelial cell network formation and survival in vitro through activation of Akt/endothelial NOS (eNOS) signaling involving integrin αvβ3. Conversely, siRNA-mediated knockdown of NDNF in endothelial cells led to reduction of cellular responses and basal Akt signaling. Intramuscular overexpression of NDNF led to enhanced blood flow recovery and capillary density in ischemic limbs of mice, which was accompanied by enhanced phosphorylation of Akt and eNOS. The stimulatory actions of NDNF on perfusion recovery in ischemic muscles of mice were abolished by eNOS deficiency or NOS inhibition. Furthermore, siRNA-mediated reduction of NDNF in muscles of mice resulted in reduction of perfusion recovery and phosphorylation of Akt and eNOS in response to ischemia. Our data indicate that NDNF acts as an endogenous modulator that promotes endothelial cell function and ischemia-induced revascularization through eNOS-dependent mechanisms. Thus, NDNF can represent a therapeutic target for the manipulation of ischemic vascular disorders.
Collapse
Affiliation(s)
- Koji Ohashi
- From the Department of Molecular Cardiology and
| | - Takashi Enomoto
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yusuke Joki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Rei Shibata
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yasuhiro Ogura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yoshiyuki Kataoka
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Takahiro Kambara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yusuke Uemura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Daisuke Yuasa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kazuhiro Matsuo
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Satoko Hayakawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Mizuho Hiramatsu-Ito
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | | |
Collapse
|
22
|
Doronzo G, Viretto M, Barale C, Russo I, Mattiello L, Anfossi G, Trovati M. Oleic acid increases synthesis and secretion of VEGF in rat vascular smooth muscle cells: role of oxidative stress and impairment in obesity. Int J Mol Sci 2013; 14:18861-80. [PMID: 24065093 PMCID: PMC3794811 DOI: 10.3390/ijms140918861] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/05/2013] [Accepted: 09/05/2013] [Indexed: 12/30/2022] Open
Abstract
Obesity is characterized by poor collateral vessel formation, a process involving vascular endothelial growth factor (VEGF) action on vascular smooth muscle cells (VSMC). Free fatty acids are involved in the pathogenesis of obesity vascular complications, and we have aimed to clarify whether oleic acid (OA) enhances VEGF synthesis/secretion in VSMC, and whether this effect is impaired in obesity. In cultured aortic VSMC from lean and obese Zucker rats (LZR and OZR, respectively) we measured the influence of OA on VEGF-A synthesis/secretion, signaling molecules and reactive oxygen species (ROS). In VSMC from LZR we found the following: (a) OA increases VEGF-A synthesis/secretion by a mechanism blunted by inhibitors of Akt, mTOR, ERK-1/2, PKC-beta, NADPH-oxidase and mitochondrial electron transport chain complex; (b) OA activates the above mentioned signaling pathways and increases ROS; (c) OA-induced activation of PKC-beta enhances oxidative stress, which activates signaling pathways responsible for the increased VEGF synthesis/secretion. In VSMC from OZR, which present enhanced baseline oxidative stress, the above mentioned actions of OA on VEGF-A, signaling pathways and ROS are impaired: this impairment is reproduced in VSMC from LZR by incubation with hydrogen peroxide. Thus, in OZR chronically elevated oxidative stress causes a resistance to the action on VEGF that OA exerts in LZR by increasing ROS.
Collapse
Affiliation(s)
- Gabriella Doronzo
- Internal Medicine and Metabolic Disease Unit, Department of Clinical and Biological Sciences of the University of Turin, San Luigi Gonzaga Hospital, Orbassano (Turin) 10043, Italy.
| | | | | | | | | | | | | |
Collapse
|
23
|
Pung YF, Sam WJ, Stevanov K, Enrick M, Chen CL, Kolz C, Thakker P, Hardwick JP, Chen YR, Dyck JRB, Yin L, Chilian WM. Mitochondrial oxidative stress corrupts coronary collateral growth by activating adenosine monophosphate activated kinase-α signaling. Arterioscler Thromb Vasc Biol 2013; 33:1911-9. [PMID: 23788766 DOI: 10.1161/atvbaha.113.301591] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Our goal was to determine the mechanism by which mitochondrial oxidative stress impairs collateral growth in the heart. APPROACH AND RESULTS Rats were treated with rotenone (mitochondrial complex I inhibitor that increases reactive oxygen species production) or sham-treated with vehicle and subjected to repetitive ischemia protocol for 10 days to induce coronary collateral growth. In control rats, repetitive ischemia increased flow to the collateral-dependent zone; however, rotenone treatment prevented this increase suggesting that mitochondrial oxidative stress compromises coronary collateral growth. In addition, rotenone also attenuated mitochondrial complex I activity and led to excessive mitochondrial aggregation. To further understand the mechanistic pathway(s) involved, human coronary artery endothelial cells were treated with 50 ng/mL vascular endothelial growth factor, 1 µmol/L rotenone, and rotenone/vascular endothelial growth factor for 48 hours. Vascular endothelial growth factor induced robust tube formation; however, rotenone completely inhibited this effect (P<0.05 rotenone versus vascular endothelial growth factor treatment). Inhibition of tube formation by rotenone was also associated with significant increase in mitochondrial superoxide generation. Immunoblot analyses of human coronary artery endothelial cells with rotenone treatment showed significant activation of adenosine monophosphate activated kinase (AMPK)-α and inhibition of mammalian target of rapamycin and p70 ribosomal S6 kinase. Activation of AMPK-α suggested impairments in energy production, which was reflected by decrease in O2 consumption and bioenergetic reserve capacity of cultured cells. Knockdown of AMPK-α (siRNA) also preserved tube formation during rotenone, suggesting the negative effects were mediated by the activation of AMPK-α. Conversely, expression of a constitutively active AMPK-α blocked tube formation. CONCLUSIONS We conclude that activation of AMPK-α during mitochondrial oxidative stress inhibits mammalian target of rapamycin signaling, which impairs phenotypic switching necessary for the growth of blood vessels.
Collapse
Affiliation(s)
- Yuh Fen Pung
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Dodd T, Wiggins L, Hutcheson R, Smith E, Musiyenko A, Hysell B, Russell JC, Rocic P. Impaired coronary collateral growth in the metabolic syndrome is in part mediated by matrix metalloproteinase 12-dependent production of endostatin and angiostatin. Arterioscler Thromb Vasc Biol 2013; 33:1339-49. [PMID: 23599440 DOI: 10.1161/atvbaha.113.301533] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We have previously shown that transient coronary artery occlusion stimulated coronary collateral growth (CCG) in healthy (Sprague Dawley) but not in metabolic syndrome (JCR:LA-cp [JCR] ) rats. Here, we sought to determine whether matrix metalloproteinases (MMPs) negatively regulate CCG in the metabolic syndrome via release of endostatin and angiostatin. APPROACH AND RESULTS Rats underwent transient, repetitive left anterior descending occlusion and resultant myocardial ischemia (RI) for 0 to 10 days. CCG was measured in the collateral-dependent and normal zones using microspheres, MMP activation by Western blot, and endostatin and angiostatin by ELISA on days 0, 3, 6, 9, or 10 of RI. Endostatin and angiostatin were increased in JCR but not in Sprague Dawley rats on days 6 and 9 of RI. Increased endostatin and angiostatin correlated with increased MMP12 (≈ 4-fold) activation in JCR but not in Sprague Dawley rats on days 6 and 9 of RI. Inhibition of MMP12 in JCR rats nearly completely blocked endostatin (≈ 85%) and angiostatin (≈ 90%) generation and significantly improved CCG (collateral-dependent zone flow was ≈ 66% of normal zone flow versus ≈ 12% for JCR RI). CONCLUSIONS Compromised CCG in the metabolic syndrome is, in large part, because of increased MMP12 activation and consequent increased generation of endostatin and angiostatin, which inhibits late-stage collateral remodeling.
Collapse
Affiliation(s)
- Tracy Dodd
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Jesmin S, Akter S, Rahman MM, Islam MM, Islam AMS, Sultana SN, Mowa CN, Yamaguchi N, Okazaki O, Satoru K, Kimura S, Hiroe M, Mizutani T, Moroi M. Disruption of components of vascular endothelial growth factor angiogenic signalling system in metabolic syndrome. Findings from a study conducted in rural Bangladeshi women. Thromb Haemost 2013; 109:696-705. [PMID: 23364276 DOI: 10.1160/th12-09-0654] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 01/09/2013] [Indexed: 01/01/2023]
Abstract
Metabolic syndrome (MetS) is associated with impaired angiogenesis, a process that is chiefly regulated by vascular endothelial growth factor (VEGF) upon binding to its specific receptors, VEGF-R1 and VEGF-R2. The purpose of the present study was to assess trends or patterns in plasma levels of VEGF and its soluble receptors in subjects with (MetS) or without (non-MetS) MetS; and further examine their association with clinical or metabolic parameters using a subpopulation of South Asian country. A total of 1,802 rural Bangladeshi women aged ≥15 years were studied using a population-based cross-sectional survey. Plasma levels of VEGF were found to be significantly increased (MetS vs. non-MetS: 483.9 vs. 386.9, p<0.001), whereas, the soluble forms of VEGF receptors, sVEGF-R1 and sVEGF-R2, were significantly decreased in subjects with Mets (sVEGF-R1, MetS vs. non-MetS: 512.5 vs. 631.3, p<0.001; sVEGF-R2, MetS vs. non-MetS: 9,302.8 vs. 9,787.4, p=0.004). After adjustment for age and all potential variables, multiple regression analysis revealed that plasma levels of VEGF had significant positive association with blood glucose (p = 0.019) and body mass index (p = 0.007). We also found that mean plasma levels of VEGF increased in direct proportion to levels of MetS components. The present study is the first ever to demonstrate a positive association between trends in levels of plasma VEGF and MetS using a large sample size from South Asia. The association between plasma VEGF and MetS needs further investigations in order to clearly decipher the clinical predictive value and accuracy of plasma VEGF in MetS.
Collapse
Affiliation(s)
- Subrina Jesmin
- Subrina Jesmin, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Teunissen PF, Horrevoets AJ, van Royen N. The coronary collateral circulation: Genetic and environmental determinants in experimental models and humans. J Mol Cell Cardiol 2012; 52:897-904. [DOI: 10.1016/j.yjmcc.2011.09.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/25/2011] [Accepted: 09/12/2011] [Indexed: 12/27/2022]
|
27
|
Nifedipine ameliorates ischemia-induced revascularization in diet-induced obese mice. Am J Hypertens 2012; 25:401-6. [PMID: 22223040 DOI: 10.1038/ajh.2011.239] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Obesity is a risk factor for the development of cardiovascular diseases that are associated with impaired angiogenesis. Nifedipine, a calcium-channel blocker, has a number of blood pressure (BP)-independent effects as well, such as improving endothelial function and decreasing oxidative stress. Here, we investigated whether nifedipine could improve the angiogenic responses in a diet-induced obese (DIO) model. METHODS DIO was induced by allowing 8-week-old C57BL/6J mice ad libitum access to a high-fat/high-sucrose (HF/HS) diet. Mice were randomly divided into two groups that were fed either the HF/HS or normal chow. At the age of 12 weeks, the animals were treated/not treated with nifedipine admixed with food at a concentration of 0.001%. Then, 1 week later, the mice were subjected to unilateral hind limb surgery. RESULTS Angiogenic repair of the ischemic hind limb was impaired in the DIO mice as compared with that in the control mice as evaluated by laser Doppler blood flowmetry (LDBF) and capillary density analysis. Treatment with nifedipine accelerated angiogenic repair in the DIO mice to a level equal to that seen in the control mice. DIO mice showed increased reactive oxygen species (ROS) production after hind limb ischemia. The number of endothelial progenitor cells (EPCs), which contribute to blood vessel formation, was also significantly lower in these mice. Nifedipine treatment ameliorated the oxidative status and increased the number of EPCs in the DIO mice. CONCLUSIONS Our observations demonstrated that DIO impaired revascularization in response to tissue ischemia. Nifedipine ameliorated obesity-impaired revascularization through suppressing oxidative stress and enhancing the number of EPCs.
Collapse
|
28
|
Erdogan T, Ciçek Y, Kocaman SA, Canga A, Cetin M, Durakoglugil E, Satiroglu O, Temiz A, Ergül E, Sahin I, Bostan M. Increased serum bilirubin level is related to good collateral development in patients with chronic total coronary occlusion. Intern Med 2012; 51:249-55. [PMID: 22293798 DOI: 10.2169/internalmedicine.51.6417] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Bilirubin is a bile pigment with potent anti-oxidant properties; in previous studies it has been reported to be negatively associated with coronary artery disease (CAD). Although elevated serum bilirubin concentrations may protect against endothelial dysfunction, it is not clear whether higher serum bilirubin levels (SBLs) in physiological ranges may work in favor of good collateral development in patients with chronic total coronary occlusion (CTO). METHODS The study population consisted of patients who underwent coronary angiography with a suspicion of CAD. Patients who had CTO in at least one major coronary artery were included. Coronary angiograms of 179 eligible patients from our database were reanalyzed and 110 of them had good collateral development and 69 had poor collateral development according to the Cohen-Rentrop method. RESULTS Patients with good collateral development had a lower fasting plasma glucose level (FPG) (p=0.024) and higher SBLs in comparison to patients with poor collateral development (p<0.001). The number of CTO vessels (p=0.013) and left ventricular ejection fraction (EF%) were higher in good collateral group than poor collateral group (p=0.017). In multivariate analysis, FPG negatively (odds ratio [OR]: 0.981, p=0.003) and SBL (OR for per 0.1-mg/dL increase: 1.832, p<0.001) and the number of CTO vessels (OR: 5.642, p=0.007) were positively related to coronary collateral development. CONCLUSION This study suggests that higher bilirubin levels within relatively normal ranges were related with favorable coronary collateral growth in patients with CTO. SBL may be responsible for the difference in coronary collateral vessel development among different patients with coronary artery disease. The anti-inflammatory and anti-oxidant properties of bilirubin may mediate this effect.
Collapse
Affiliation(s)
- Turan Erdogan
- Department of Cardiology, Rize University Medical Faculty, Turkey
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Maruyama S, Shibata R, Kikuchi R, Izumiya Y, Rokutanda T, Araki S, Kataoka Y, Ohashi K, Daida H, Kihara S, Ogawa H, Murohara T, Ouchi N. Fat-derived factor omentin stimulates endothelial cell function and ischemia-induced revascularization via endothelial nitric oxide synthase-dependent mechanism. J Biol Chem 2011; 287:408-417. [PMID: 22081609 DOI: 10.1074/jbc.m111.261818] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Obesity-related diseases are associated with vascular dysfunction and impaired revascularization. Omentin is a fat-derived secreted protein, which is down-regulated in association with obese complications. Here, we investigated whether omentin modulates endothelial cell function and revascularization processes in vitro and in vivo. Systemic delivery of an adenoviral vector expressing omentin (Ad-omentin) enhanced blood flow recovery and capillary density in ischemic limbs of wild-type mice in vivo, which were accompanied by increased phosphorylation of Akt and endothelial nitric oxide synthase (eNOS). In cultured human umbilical vein endothelial cells (HUVECs), a physiological concentration of recombinant omentin protein increased differentiation into vascular-like structures and decreased apoptotic activity under conditions of serum starvation. Treatment with omentin protein stimulated the phosphorylation of Akt and eNOS in HUVECs. Inhibition of Akt signaling by treatment with dominant-negative Akt or LY294002 blocked the stimulatory effects of omentin on differentiation and survival of HUVECs and reversed omentin-stimulated eNOS phosphorylation. Pretreatment with the NOS inhibitor also reduced the omentin-induced increase in HUVEC differentiation and survival. Omentin protein also stimulated the phosphorylation of AMP-activated protein kinase in HUVECs. Transduction with dominant-negative AMP-activated protein kinase diminished omentin-induced phosphorylation of Akt and omentin-stimulated increase in HUVEC differentiation and survival. Of importance, in contrast to wild-type mice, systemic administration of Ad-omentin did not affect blood flow in ischemic muscle in eNOS-deficient mice in vivo. These data indicate that omentin promotes endothelial cell function and revascularization in response to ischemia through its ability to stimulate an Akt-eNOS signaling pathway.
Collapse
Affiliation(s)
- Sonomi Maruyama
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Department of Cardiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Rei Shibata
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Ryosuke Kikuchi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yasuhiro Izumiya
- Department of Cardiology, Kumamoto University School of Medicine, Kumamoto 860-8556, Japan
| | - Taku Rokutanda
- Department of Cardiology, Kumamoto University School of Medicine, Kumamoto 860-8556, Japan
| | - Satoshi Araki
- Department of Cardiology, Kumamoto University School of Medicine, Kumamoto 860-8556, Japan
| | - Yoshiyuki Kataoka
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Koji Ohashi
- Department of Molecular Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hiroyuki Daida
- Department of Cardiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Shinji Kihara
- Department of Biomedical Informatics, Osaka University Graduate School of Medicine, 565-0871 Osaka, Japan
| | - Hisao Ogawa
- Department of Cardiology, Kumamoto University School of Medicine, Kumamoto 860-8556, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Noriyuki Ouchi
- Department of Molecular Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
30
|
Dodd T, Jadhav R, Wiggins L, Stewart J, Smith E, Russell JC, Rocic P. MMPs 2 and 9 are essential for coronary collateral growth and are prominently regulated by p38 MAPK. J Mol Cell Cardiol 2011; 51:1015-25. [PMID: 21884701 DOI: 10.1016/j.yjmcc.2011.08.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 08/11/2011] [Accepted: 08/15/2011] [Indexed: 12/19/2022]
Abstract
Transient, repetitive ischemia (RI) stimulates coronary collateral growth (CCG) in normal, healthy (SD) rats, which requires p38 MAPK activation. In contrast, RI does not induce CCG in the metabolic syndrome (JCR) rats, which is associated with lack of p38 MAPK activation. The functional consequences of p38 MAPK activation in CCG remain unknown. Theoretically, effective collateral growth would require extracellular matrix remodeling; however, direct assessment as well as identification of proteases responsible for this degradation are lacking. In this study, we investigated the role of p38 MAPK in the regulation of matrix metalloproteinases 2 and 9 (MMPs 2 and 9) and their requirement for CCG in SD vs. JCR rats. The rats underwent the RI protocol (8 LAD occlusions, 40s each, every 20min, in 8h cycles for 0, 3, 6, or 9days). MMP expression was measured in the ischemic, collateral-dependent zone (CZ) and the normal zone (NZ) by Western blot, and MMP activity by zymography. Expression and activation of MMP 2 and 9 were significantly increased (~3.5 fold) on day 3 of RI in the CZ of SD rats. In vivo p38 MAPK inhibition completely blocked RI-induced MMP 2 and 9 expression and activation. MMP activation correlated with increased degradation of components of the basement membrane and the vascular elastic laminae: elastin (~3 fold), laminin (~3 fold) and type IV collagen (~2 fold). This was blocked by MMP 2 and 9 inhibition, which also abolished RI-induced CCG. In contrast, in JCR rats, RI did not induce expression or activation of MMP 2 or 9 and there was no associated degradation of elastin, laminin or type IV collagen. In conclusion, MMP 2 and 9 activation is essential for CCG and is mediated, in part, by p38 MAPK. Furthermore, compromised CCG in the metabolic syndrome may be partially due to the lack of p38 MAPK-dependent activation of MMP 2 and 9 and resultant decreased extracellular matrix degradation.
Collapse
Affiliation(s)
- Tracy Dodd
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Ege MR, Guray U. Comment on: the association between metabolic syndrome and infarct size in patients with acute myocardial infarction. Scandinavian Journal of Clinical and Laboratory Investigation 2010; 70:592. [PMID: 20942740 DOI: 10.3109/00365513.2010.525254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Li P, Shibata R, Maruyama S, Kondo M, Ohashi K, Ouchi N, Murohara T. Fenofibrate promotes ischemia-induced revascularization through the adiponectin-dependent pathway. Am J Physiol Endocrinol Metab 2010; 299:E560-6. [PMID: 20663986 DOI: 10.1152/ajpendo.00284.2010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Recent clinical trials demonstrated that PPARα agonist fenofibrate reduces cardiovascular events, including limb amputation in people with type 2 diabetes. Here, we investigated whether fenofibrate modulates the revascularization process in a mouse model of hindlimb ischemia. Treatment with fenofibrate led to acceleration of revascularization of ischemic hindlimb relative to the contralatereal limb in wild-type (WT) mice, as measured by laser Doppler blood flow and capillary density analyses. Treatment of WT mice with fenofibrate increased the serum levels of adiponectin, which has protective actions on the vasculature. Of importance, fenofibrate had no effects on the revascularization in ischemic limbs of adiponectin-deficient (APN-KO) mice. Fenofibrate stimulated the phosphorylation of AMPK and eNOS in the ischemic muscles in WT mice but not in APN-KO mice. AMPK inhibitor compound C suppressed fenofibrate-induced increase in limb perfusion and AMPK phosphorylation in ischemic muscle in WT mice without affecting adiponectin levels. NOS inhibitor l-NAME also blocked the increased blood flow of ischemic limbs in fenofibrate-treated WT mice. Our observations suggest that fenofibrate could promote revascularization in response to ischemia through adiponectin-dependent AMPK signaling.
Collapse
Affiliation(s)
- Ping Li
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
MacEneaney OJ, Kushner EJ, Westby CM, Cech JN, Greiner JJ, Stauffer BL, DeSouza CA. Endothelial progenitor cell function, apoptosis, and telomere length in overweight/obese humans. Obesity (Silver Spring) 2010; 18:1677-82. [PMID: 20057362 DOI: 10.1038/oby.2009.494] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Excess adiposity is associated with increased cardiovascular morbidity and mortality. Endothelial progenitor cells (EPCs) play an important role in vascular repair. We tested the hypothesis that increased adiposity is associated with EPC dysfunction, characterized by diminished capacity to release angiogenic cytokines, increased apoptotic susceptibility, reduced cell migration, and shorter telomere length. A total of 67 middle-aged and older adults (42-67 years) were studied: 25 normal weight (normal weight; BMI: 18.5-24.9 kg/m(2)) and 42 overweight/obese (overweight/obese; BMI: 25.0-34.9 kg/m(2)). Cells with phenotypic EPC characteristics were isolated from peripheral blood. EPC release of vascular endothelial growth factor (VEGF) and granulocyte colony-stimulating factor (G-CSF) was determined in the absence and presence of phytohemagglutinin (10 microg/ml). Intracellular active caspase-3 and cytochrome c concentrations were determined by immunoassay. Migratory activity of EPCs in response to VEGF (2 ng/ml) and stromal cell-derived factor-1alpha (SDF-1alpha; 10 ng/ml) was determined by Boyden chamber. Telomere length was assessed by Southern hybridization. Phytohemagglutinin-stimulated release of VEGF (90.6 +/- 7.6 vs. 127.2 +/- 11.6 pg/ml) and G-CSF (896.1 +/- 77.4 vs. 1,176.3 +/- 126.3 pg/ml) was ~25% lower (P < 0.05) in EPCs from overweight/obese vs. normal weight subjects. Staurosporine induced a ~30% greater (P < 0.05) increase in active caspase-3 in EPCs from overweight/obese (2.8 +/- 0.2 ng/ml) compared with normal weight (2.2 +/- 0.2) subjects. There were no significant differences in EPC migration to either VEGF or SDF-1alpha. Telomere length did not differ between groups. These results indicate that increased adiposity adversely affects the ability of EPCs to release proangiogenic cytokines and resist apoptosis, potentially compromising their reparative potential.
Collapse
Affiliation(s)
- Owen J MacEneaney
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Ege MR, Altay H. The impact of body mass index on clinical outcomes after acute myocardial infarction. Int J Cardiol 2010; 145:539. [PMID: 20554335 DOI: 10.1016/j.ijcard.2010.04.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 04/20/2010] [Indexed: 10/19/2022]
|
35
|
Steiner S, Schueller PO, Schulze V, Strauer BE. Occurrence of Coronary Collateral Vessels in Patients With Sleep Apnea and Total Coronary Occlusion. Chest 2010; 137:516-20. [DOI: 10.1378/chest.09-1136] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
36
|
Abstract
Arterial lumen narrowing and vascular occlusion is the actual cause of morbidity and mortality in atherosclerotic disease. Collateral artery formation (arteriogenesis) refers to an active remodelling of non-functional vascular anastomoses to functional collateral arteries, capable to bypass the site of obstruction and preserve the tissue that is jeopardized by ischaemia. Hemodynamic forces such as shear stress and wall stress play a pivotal role in collateral artery formation, accompanied by the expression of various cytokines and invasion of circulating leucocytes. Arteriogenesis hence represents an important compensatory mechanism for atherosclerotic vessel occlusion. As arteriogenesis mostly occurs when lumen narrowing by atherosclerotic plaques takes place, presence of cardiovascular risk factors (e.g. hypertension, hypercholesterolaemia and diabetes) is highly likely. Risk factors for atherosclerotic disease affect collateral artery growth directly and indirectly by altering hemodynamic forces or influencing cellular function and proliferation. Adequate collateralization varies significantly among atherosclerotic patients, some profit from the presence of extensive collateral networks, whereas others do not. Cardiovascular risk factors could increase the risk of adverse cardiovascular events in certain patients because of the reduced protection through an alternative vascular network. Likewise, drugs primarily thought to control cardiovascular risk factors might contribute or counteract collateral artery growth. This review summarizes current knowledge on the influence of cardiovascular risk factors and the effects of cardiovascular medication on the development of collateral vessels in experimental and clinical studies.
Collapse
Affiliation(s)
- D de Groot
- Laboratory of Experimental Cardiology, UMC Utrecht, the Netherlands
| | | | | |
Collapse
|
37
|
Abstract
Insulin is a vascular hormone, able to influence vascular cell responses. In this review, we consider the insulin actions on vascular endothelium and on vascular smooth muscle cells (VSMC) both in physiological conditions and in the presence of insulin resistance. In particular, we focus the relationships between activation of insulin signalling pathways of phosphatidylinositol-3 kinase (PI3-K) and mitogen-activated protein kinase (MAPK) and the different vascular actions of insulin, with a particular attention to the insulin ability to activate the pathway nitric oxide (NO)/cyclic GMP/PKG via PI3-K, owing to the peculiar relevance of NO in vascular biology. We also discuss the insulin actions mediated by the MAPK pathway (such as endothelin-1 synthesis and secretion and VSMC proliferation and migration) and by the interactions between the two pathways, both in insulin-sensitive and in insulin-resistant states. Finally, we consider the influence of free fatty acids, cytokines and endothelin on vascular insulin resistance.
Collapse
Affiliation(s)
- Giovanni Anfossi
- Internal Medicine University Unit, San Luigi Gonzaga Faculty of Medicine and Department of Clinical and Biological Sciences, Turin University, San Luigi Gonzaga Hospital, 10043 Orbassano, Turin, Italy
| | | | | | | |
Collapse
|
38
|
Yun J, Rocic P, Pung YF, Belmadani S, Carrao ACR, Ohanyan V, Chilian WM. Redox-dependent mechanisms in coronary collateral growth: the "redox window" hypothesis. Antioxid Redox Signal 2009; 11:1961-74. [PMID: 19416057 PMCID: PMC2848513 DOI: 10.1089/ars.2009.2476] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review addresses the complexity of coronary collateral growth from the aspect of redox signaling and introduces the concept of a "redox window" in the context of collateral growth. In essence, the redox window constitutes a range in the redox state of cells, which not only is permissive for the actions of growth factors but also amplifies their actions. The interactions of redox-dependent signaling with growth factors are well established through the actions of many redox-dependent kinases (e.g., Akt and p38 mitogen-activated protein kinase). The initial changes in cellular redox can be induced by a variety of events, from the oxidative burst during reperfusion after ischemia, to recruitment of various types of inflammatory cells capable of producing reactive oxygen species. Any event that "upsets" the normal redox equilibrium is capable of amplifying growth. However, extremes of the redox window, oxidative and reductive stresses, are associated with diminished growth-factor signaling and reduced activation of redox-dependent kinases. This concept of a redox window helps to explain why the clinical trials aimed at stimulating coronary collateral growth, the "therapeutic angiogenesis trials," failed. However, understanding of redox signaling in the context of coronary collateral growth could provide new paradigms for stimulating collateral growth in patients.
Collapse
Affiliation(s)
- June Yun
- Department of Integrative Medical Sciences, Northeastern Ohio Universities College of Medicine, Rootstown, Ohio 44272, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Wada H, Satoh N, Kitaoka S, Ono K, Morimoto T, Kawamura T, Nakano T, Fujita M, Kita T, Shimatsu A, Hasegawa K. Soluble VEGF receptor-2 is increased in sera of subjects with metabolic syndrome in association with insulin resistance. Atherosclerosis 2009; 208:512-7. [PMID: 19695569 DOI: 10.1016/j.atherosclerosis.2009.07.045] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 07/22/2009] [Accepted: 07/23/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Metabolic syndrome (MetS) is associated with impaired angiogenesis. Vascular endothelial growth factor (VEGF) plays a key role in angiogenesis through binding to its specific receptor, VEGF receptor-2 (VEGFR-2), whereas the expression of VEGF and VEGFR-2 in the myocardium of insulin-resistant rats is down-regulated. Soluble VEGF receptor-1 (sVEGFR-1) and -2 (sVEGFR-2) have been reported to inhibit angiogenesis both in vitro and in vivo. However, the balance between circulating levels of VEGF and its soluble receptors, which may reflect and/or affect cardiovascular VEGF signaling, in subjects with MetS is unknown. METHODS AND RESULTS We carried out a cross-sectional study including 272 consecutive, apparently healthy subjects who were not receiving any drugs. Plasma levels of VEGF and serum levels of its soluble receptors were determined using enzyme-linked immunosorbent assays. VEGF and sVEGFR-1 levels did not differ between subjects with and those without MetS. However, sVEGFR-2 levels were significantly increased in MetS compared with non-MetS subjects. Stepwise regression analysis revealed that HOMA-IR was the strongest independent determinant of the sVEGFR-2 level. Accordingly, the mean sVEGFR-2 levels increased in proportion to both the accumulation of components of MetS and quartile of HOMA-IR. Interestingly, multiple regression analyses revealed that independent determinants of VEGF were the body mass index and blood pressure, whereas, in contrast, those of sVEGFR-2 were HOMA-IR and high-sensitivity C-reactive protein. CONCLUSIONS The correlation of sVEGFR-2 with insulin resistance supports the need for further investigations to define the clinical utility and predictive value of serum sVEGFR-2 levels in cardiovascular dysfunction in MetS.
Collapse
Affiliation(s)
- Hiromichi Wada
- Division of Translational Research, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto 612-8555, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
MacEneaney OJ, Kushner EJ, Van Guilder GP, Greiner JJ, Stauffer BL, DeSouza CA. Endothelial progenitor cell number and colony-forming capacity in overweight and obese adults. Int J Obes (Lond) 2009; 33:219-25. [PMID: 19079361 PMCID: PMC2643316 DOI: 10.1038/ijo.2008.262] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate whether adiposity influences endothelial progenitor cell (EPC) number and colony-forming capacity. DESIGN Cross-sectional study of normal weight, overweight and obese adult humans. PARTICIPANTS Sixty-seven sedentary adults (aged 45-65 years): 25 normal weight (body mass index (BMI) or=30 kg/m(2); 18 males/6 females). All participants were non-smokers and free of overt cardiometabolic disease. MEASUREMENTS Peripheral blood samples were collected and circulating EPC number was assessed by flow cytometry. Putative EPCs were defined as CD45(-)/CD34(+)/VEGFR-2(+)/CD133(+) or CD45(-)/CD34(+) cells. EPC colony-forming capacity was measured in vitro using a colony-forming unit (CFU) assay. RESULTS Number of circulating putative EPCs (either CD45(-)/CD34(+)/VEGFR-2(+)/CD133(+) or CD45(-)/CD34(+) cells) was lower (P<0.05) in obese (0.0007+/-0.0001%; 0.050+/-0.006%) compared with overweight (0.0016+/-0.0004%; 0.089+/-0.019%) and normal weight (0.0015+/-0.0003%; 0.082+/-0.008%) adults. There were no differences in EPC number between the overweight and normal weight groups. EPC colony formation was significantly less in the obese (6+/-1) and overweight (4+/-1) compared with normal weight (9+/-2) adults. CONCLUSION These results indicate that: (1) the number of circulating EPCs is lower in obese compared with overweight and normal weight adults; and (2) EPC colony-forming capacity is blunted in overweight and obese adults compared with normal weight adults. Impairments in EPC number and function may contribute to adiposity-related cardiovascular risk.
Collapse
Affiliation(s)
- O J MacEneaney
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, CO 80309, USA
| | | | | | | | | | | |
Collapse
|
41
|
Kondo M, Shibata R, Miura R, Shimano M, Kondo K, Li P, Ohashi T, Kihara S, Maeda N, Walsh K, Ouchi N, Murohara T. Caloric restriction stimulates revascularization in response to ischemia via adiponectin-mediated activation of endothelial nitric-oxide synthase. J Biol Chem 2009; 284:1718-24. [PMID: 18990685 PMCID: PMC2615500 DOI: 10.1074/jbc.m805301200] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 11/05/2008] [Indexed: 01/09/2023] Open
Abstract
Caloric restriction (CR) can extend longevity and modulate the features of obesity-related metabolic and vascular diseases. However, the functional roles of CR in regulation of revascularization in response to ischemia have not been examined. Here we investigated whether CR modulates vascular response by employing a murine hindlimb ischemia model. Wild-type (WT) mice were randomly divided into two groups that were fed either ad libitum (AL) or CR (65% of the diet consumption of AL). Four weeks later, mice were subjected to unilateral hindlimb ischemic surgery. Body weight of WT mice fed CR (CR-WT) was decreased by 26% compared with WT mice fed AL (AL-WT). Revascularization of ischemic hindlimb relative to the contralateral limb was accelerated in CR-WT compared with AL-WT as evaluated by laser Doppler blood flow and capillary density analyses. CR-WT mice had significantly higher plasma levels of the fat-derived hormone adiponectin compared with AL-WT mice. In contrast to WT mice, CR did not affect the revascularization of ischemic limbs of adiponectin-deficient (APN-KO) mice. CR stimulated the phosphorylation of endothelial nitric-oxide synthase (eNOS) in the ischemic limbs of WT mice. CR increased plasma adiponectin levels in eNOS-KO mice but did not stimulate limb perfusion in this strain. CR-WT mice showed enhanced phosphorylation of AMP-activated protein kinase (AMPK) in ischemic muscle, and administration of AMPK inhibitor compound C abolished CR-induced increase in limb perfusion and eNOS phosphorylation in WT mice. Our observations indicate that CR can promote revascularization in response to tissue ischemia via an AMPK-eNOS-dependent mechanism that is mediated by adiponectin.
Collapse
Affiliation(s)
- Megumi Kondo
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Birhan Yilmaz M. Being obese with poor coronary collaterals in cardiogenic shock. Int J Cardiol 2008; 129:133; author reply 135. [PMID: 17692971 DOI: 10.1016/j.ijcard.2007.06.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Accepted: 06/30/2007] [Indexed: 11/29/2022]
|
44
|
Zhou X, Bohlen HG, Miller SJ, Unthank JL. NAD(P)H oxidase-derived peroxide mediates elevated basal and impaired flow-induced NO production in SHR mesenteric arteries in vivo. Am J Physiol Heart Circ Physiol 2008; 295:H1008-H1016. [PMID: 18599598 DOI: 10.1152/ajpheart.00114.2008] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) and reactive oxygen species (ROS) have fundamentally important roles in the regulation of vascular tone and remodeling. Although arterial disease and endothelial dysfunction alter NO and ROS levels to impact vasodilation and vascular structure, direct measurements of these reactive species under in vivo conditions with flow alterations are unavailable. In this study, in vivo measurements of NO and H2O2 were made on mesenteric arteries to determine whether antioxidant therapies could restore normal NO production in spontaneously hypertensive rats (SHR). Flow was altered from approximately 50-200% of control in anesthetized Wistar-Kyoto rats (WKY) and SHR by selective placement of microvascular clamps on adjacent arteries while NO and H2O2 were directly measured with microelectrodes. Relative to WKY, SHR had significantly increased baseline NO and H2O2 concentrations (2,572 +/- 241 vs. 1,059 +/- 160 nM, P < 0.01; and 26 +/- 7 vs. 7 +/- 1 microM, P < 0.05, respectively). With flow elevation, H2O2 but not NO increased in SHR; NO but not H2O2 was elevated in WKY. Apocynin and polyethylene-glycolated catalase decreased baseline SHR NO and H2O2 to WKY levels and restored flow-mediated NO production. Suppression of NAD(P)H oxidase with gp91ds-tat decreased SHR H2O2 to WKY levels. Addition of topical H2O2 to increase peroxide to the basal concentration measured in SHR elevated WKY NO to levels observed in SHR. The results support the hypothesis that increased vascular peroxide in SHR is primarily derived from NAD(P)H oxidase and increases NO concentration to levels that cannot be further elevated with increased flow. Short-term and even acute administration of antioxidants are able to restore normal flow-mediated NO signaling in young SHR.
Collapse
Affiliation(s)
- Xiaosun Zhou
- Department of Surgery, Indiana University Medical Center, 1001 West Tenth St., Indianapolis, IN 46202-2879, USA
| | | | | | | |
Collapse
|
45
|
Sasmaz H, Mehmet Birhan Y. Coronary Collaterals in Obese Patients: Impact of Metabolic Syndrome. Angiology 2008; 60:164-8. [DOI: 10.1177/0003319708316007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesity is a growing pandemic. Among obese patients with significant coronary artery stenosis, development of coronary collaterals was investigated. Consecutive 104 obese patients with stable angina pectoris were enrolled. Coronary collaterals were assessed according to the Cohen and Rentrop grading system and classified into 2 as those with poor collaterals (grade 0–1, Group 1) and those with good collaterals (grade 2–3, Group 2). Group 1 had higher body mass index, shorter duration of angina pectoris than Group 2. Poor collaterals were present in 82.3% and 59.5% of patients with and without ( P = .019) metabolic syndrome, respectively. Metabolic syndrome score (sum of each component) was found to be negatively correlated with Rentrop score ( r = −691; P < .001). After controlling for symptom duration and body mass index, metabolic syndrome kept independent association with poor collaterals among obese patients ( P = .043, B = 1.8). Metabolic syndrome appears to influence the development of coronary collaterals among obese patients with stable coronary artery disease.
Collapse
Affiliation(s)
- Hatice Sasmaz
- Department of Cardiology, Yuksek Ihtisas Education and Research Hospital, Cardiology Clinic, Ankara
| | - Yilmaz Mehmet Birhan
- Department of Cardiology, Cumhuriyet University School of Medicine, Sivas, Turkey
| |
Collapse
|
46
|
Ouchi N, Walsh K. Adiponectin as an anti-inflammatory factor. Clin Chim Acta 2007; 380:24-30. [PMID: 17343838 PMCID: PMC2755046 DOI: 10.1016/j.cca.2007.01.026] [Citation(s) in RCA: 631] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2006] [Revised: 01/13/2007] [Accepted: 01/21/2007] [Indexed: 12/26/2022]
Abstract
Obesity is characterized by low-grade systemic inflammation. Adiponectin is an adipose tissue-derived hormone, which is downregulated in obesity. Adiponectin displays protective actions on the development of various obesity-linked diseases. Several clinical studies demonstrate the inverse relationship between plasma adiponectin levels and several inflammatory markers including C-reactive protein. Adiponectin attenuates inflammatory responses to multiple stimuli by modulating signaling pathways in a variety of cell types. The anti-inflammatory properties of adiponectin may be a major component of its beneficial effects on cardiovascular and metabolic disorders including atherosclerosis and insulin resistance. In this review, we focus on the role of adiponectin in regulation of inflammatory response and discuss its potential as an anti-inflammatory marker.
Collapse
Affiliation(s)
- Noriyuki Ouchi
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, 715 Albany Street, W611, Boston, MA 02118, USA.
| | | |
Collapse
|
47
|
Hopkins TA, Ouchi N, Shibata R, Walsh K. Adiponectin actions in the cardiovascular system. Cardiovasc Res 2006; 74:11-8. [PMID: 17140553 PMCID: PMC1858678 DOI: 10.1016/j.cardiores.2006.10.009] [Citation(s) in RCA: 234] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Revised: 10/04/2006] [Accepted: 10/16/2006] [Indexed: 11/29/2022] Open
Abstract
Obesity is strongly associated with the pathogenesis of type 2 diabetes, hypertension, and cardiovascular disease. Levels of the hormone adiponectin are downregulated in obese individuals, and several experimental studies show that adiponectin protects against the development of various obesity-related metabolic and cardiovascular diseases. Adiponectin exhibits favorable effects on atherogenesis, endothelial function, and vascular remodeling by modulation of signaling cascades in cells of the vasculature. More recent findings have shown that adiponectin directly affects signaling in cardiac cells and is beneficial in the setting of pathological cardiac remodeling and acute cardiac injury. Several of these effects of adiponectin have been attributed to the activation of the 5' AMP-activated protein kinase signaling cascade and other signaling proteins. This review will discuss the epidemiological and experimental studies that have elucidated the role of adiponectin in a variety of cardiovascular diseases.
Collapse
Affiliation(s)
- Teresa A Hopkins
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | |
Collapse
|
48
|
Abstract
Adiponectin is an adipose tissue-derived plasma protein which has a reduced concentration in subjects with obesity-related diseases. Adiponectin has antidiabetic and anti-inflammatory characteristics, which lead to beneficial actions on various obesity-linked complications. Recent experimental findings have shown that adiponectin contributes to protection against cardiac remodelling after pressure overload and cardiac injury following ischaemia-reperfusion. Thus, adiponectin could emerge as a potential cardioprotective agent for the treatment of several pathological heart conditions.
Collapse
Affiliation(s)
- Noriyuki Ouchi
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, 715 Albany Street, W611, Boston, MA 02118, USA
| | | | | |
Collapse
|
49
|
Ouchi N, Shibata R, Walsh K. Cardioprotection by adiponectin. Trends Cardiovasc Med 2006; 16:141-6. [PMID: 16781946 PMCID: PMC2749293 DOI: 10.1016/j.tcm.2006.03.001] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 03/01/2006] [Accepted: 03/02/2006] [Indexed: 01/11/2023]
Abstract
Obesity-related disorders are closely associated with the pathogenesis of cardiovascular disease. Adiponectin is a circulating adipose tissue-derived hormone that is down-regulated in obese individuals. Hypoadiponectinemia has been identified as an independent risk factor for type 2 diabetes, coronary artery disease, and hypertension, and experimental studies show that adiponectin plays a protective role in the development of insulin resistance, atherosclerosis, and inflammation. More recent findings have shown that adiponectin directly affects signaling in myocardial cells and exerts beneficial actions on the heart after pressure overload and ischemia-reperfusion injury. This review focuses on the role of adiponectin in the regulation of myocardial remodeling and acute cardiac injury.
Collapse
Affiliation(s)
- Noriyuki Ouchi
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Rei Shibata
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kenneth Walsh
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
50
|
Li W, Shen W, Gill R, Corbly A, Jones B, Belagaje R, Zhang Y, Tang S, Chen Y, Zhai Y, Wang G, Wagle A, Hui K, Westmore M, Hanson J, Chen YF, Simons M, Singh J. High-Resolution Quantitative Computed Tomography Demonstrating Selective Enhancement of Medium-Size Collaterals by Placental Growth Factor-1 in the Mouse Ischemic Hindlimb. Circulation 2006; 113:2445-53. [PMID: 16702473 DOI: 10.1161/circulationaha.105.586818] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The process of arteriogenesis after occlusion of a major artery is poorly understood. We have used high-resolution microcomputed tomography (mu-CT) imaging to define the arteriogenic response in the mouse model of hindlimb ischemia and to examine the effect of placental growth factor-1 (PlGF-1) on this process. METHODS AND RESULTS After common femoral artery ligation, mu-CT imaging demonstrated formation of collateral vessels originating near the ligation site in the upper limb and connecting to the ischemic calf muscle region. Three-dimensional mu-CT and quantitative image analysis revealed changes in the number of segments and the segmental volume of vessels, ranging from 8 to 160 microm in diameter. The medium-size vessels (48 to 160 microm) comprising 85% of the vascular volume were the major contributor (188%) to the change in vascular volume in response to ischemia. Intramuscular injections of Ad-PlGF-1 significantly increased Sca1+ cells in the circulation, alpha-actin-stained vessels, and perfusion of the ischemic hindlimb. These effects were predominantly associated with an increase in vascular volume contributed by the medium-size (96 to 144 microm) vessels as determined by mu-CT. CONCLUSIONS High-resolution mu-CT delineated the formation of medium-size collaterals representing a major vascular change that contributed to the restoration of vascular volume after ischemia. This effect is selectively potentiated by PlGF-1. Such selective enhancement of arteriogenesis by therapeutically administered PlGF-1 demonstrates a desirable biological activity for promoting the growth of functionally relevant vasculature.
Collapse
Affiliation(s)
- Weiming Li
- Eli Lilly and Co, Indianapolis, IN 46285, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|