1
|
Irqsusi M, Rodepeter FR, Günther M, Kirschbaum A, Vogt S. Matrix metalloproteinases and their tissue inhibitors as indicators of aortic aneurysm and dissection development in extracellular matrix remodeling. World J Exp Med 2025; 15:100166. [DOI: 10.5493/wjem.v15.i2.100166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/04/2025] [Accepted: 01/15/2025] [Indexed: 04/16/2025] Open
Abstract
Aneurysms and dissections represent some of the most serious cardiovascular diseases. The prevailing theory posits that mechanical overloading of the vessel wall is the underlying cause. Inspired by Barkhordarian et al, the authors present matrix metalloproteinases (MMPs) and their inhibitors in immunohistological analyses as contributing factors in the pathophysiology of aortic aneurysms (AA). Data analysis of MMP-1, MMP-9, tissue inhibitors of metalloproteinases (TIMPs), including TIMP-1 and TIMP-2 expression reveals a varied distribution between the adventitia and media and a non-uniform expression of the investigated markers. These elements, as key components of the extracellular matrix (ECM), indicate that the formation of AA is not solely driven by endoluminal pressure loading of the aortic wall. Instead, degenerative processes within ECM elements contribute significantly. Importantly, AA do not necessarily imply dissection. Tissue destruction, allowing blood flow entry, arises from reduced oxygen supply to the media, primarily due to incomplete capillarization or neocapillarization.
Collapse
Affiliation(s)
- Marc Irqsusi
- Department of Heart Surgery, Universitätsklinikum Marburg and Gießen GmbH, Marburg 35043, Hesse, Germany
| | - Fiona R Rodepeter
- Institute of Pathology, Philipps-University Marburg, Marburg 35043, Hesse, Germany
| | - Madeline Günther
- Department of Heart Surgery, Cardiovascular Research Laboratory, Philipps-University Marburg, Marburg 35043, Hesse, Germany
| | - Andreas Kirschbaum
- Department of Visceral Surgery, University Hospital Giessen and Marburg GmbH, Marburg 35043, Hesse, Germany
| | - Sebastian Vogt
- Department of Heart Surgery, Philipps-University Marburg, Marburg 35043, Hesse, Germany
| |
Collapse
|
2
|
Sonaglioni A, Caminati A, Nicolosi GL, Muti-Schünemann GEU, Lombardo M, Harari S. Echocardiographic Assessment of Biventricular Mechanics in Patients with Mild-to-Moderate Idiopathic Pulmonary Fibrosis: A Systematic Review and Meta-Analysis. J Clin Med 2025; 14:714. [PMID: 39941384 PMCID: PMC11818070 DOI: 10.3390/jcm14030714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Over the last few years, a few imaging studies have performed conventional transthoracic echocardiography (TTE) implemented with speckle tracking echocardiography (STE) for the assessment of biventricular mechanics in patients with non-advanced idiopathic pulmonary fibrosis (IPF). This systematic review and meta-analysis aimed at evaluating the overall effect of mild-to-moderate IPF on the main indices of biventricular systolic function assessed by TTE and STE. Methods: All imaging studies assessing right ventricular (RV)-global longitudinal strain (GLS), left ventricular (LV)-GLS, tricuspid annular plane systolic excursion (TAPSE), and left ventricular ejection fraction (LVEF) in IPF patients vs. healthy controls, selected from PubMed, Scopus, and EMBASE databases, were included. Continuous data (RV-GLS, LV-GLS, TAPSE, and LVEF) were pooled as standardized mean differences (SMDs) comparing the IPF group with healthy controls. The SMD of RV-GLS was calculated using the random-effect model, whereas the SMDs of LV-GLS, TAPSE, and LVEF were calculated using the fixed-effect model. Results: The full texts of 6 studies with 255 IPF patients and 195 healthy controls were analyzed. Despite preserved TAPSE and LVEF, both RV-GLS and LV-GLS were significantly, although modestly, reduced in the IPF patients vs. the controls. The SMD was large (-1.01, 95% CI -1.47, -0.54, p < 0.001) for RV-GLS, medium (-0.62, 95% CI -0.82, -0.42, p < 0.001) for LV-GLS, small (-0.42, 95% CI -0.61, -0.23, p < 0.001) for TAPSE, and small and not statistically significant (-0.20, 95% CI -0.42, 0.03, p = 0.09) for LVEF assessment. Between-study heterogeneity was high for the studies assessing RV-GLS (I2 = 80.5%), low-to-moderate for those evaluating LV-GLS (I2 = 41.7%), and low for those measuring TAPSE (I2 = 16.4%) and LVEF (I2 = 7.63%). The Egger's test yielded a p-value of 0.60, 0.11, 0.31, and 0.68 for the RV-GLS, LV-GLS, TAPSE, and LVEF assessment, respectively, indicating no publication bias. On meta-regression analysis, none of the moderators was significantly associated with effect modification for RV-GLS (all p > 0.05). The sensitivity analysis supported the robustness of the results. Conclusions: RV-GLS impairment is an early marker of subclinical myocardial dysfunction in mild-to-moderate IPF. STE should be considered for implementation in clinical practice for early detection of RV dysfunction in IPF patients without advanced lung disease.
Collapse
Affiliation(s)
| | - Antonella Caminati
- Semi-Intensive Care Unit, Division of Pneumology, IRCCS MultiMedica, 20123 Milan, Italy; (A.C.); (S.H.)
| | | | | | | | - Sergio Harari
- Semi-Intensive Care Unit, Division of Pneumology, IRCCS MultiMedica, 20123 Milan, Italy; (A.C.); (S.H.)
- Department of Clinical Sciences and Community Health, Università di Milano, 20122 Milan, Italy
| |
Collapse
|
3
|
Rostamzadeh F, Joukar S, Yeganeh-Hajahmadi M. The role of Klotho and sirtuins in sleep-related cardiovascular diseases: a review study. NPJ AGING 2024; 10:43. [PMID: 39358364 PMCID: PMC11447243 DOI: 10.1038/s41514-024-00165-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 08/07/2024] [Indexed: 10/04/2024]
Abstract
The prevalence of sleep disorders has been reported from 1.6% to 56.0%, worldwide. Sleep deprivation causes cardiovascular diseases (CVDs) including atherosclerosis, vascular aging, hypertension, heart dysfunction, reduced heart rate variability, and cardiac arrhythmia. Reduced tissue oxygen causes various CVDs by activating pro-inflammatory factors and increasing oxidative stress. Sleep disorders are more important and prevalent in older people and cause more severe cardiovascular complications. On the other hand, the reduction of Klotho level, an age-dependent protein whose expression decreases with age, is associated with age-related diseases. Sirtuins, class III histone deacetylases, also are among the essential factors in postponing cellular aging and increasing the lifespan of organisms, and they do this by regulating different pathways in the cell. Sirtuins and Klotho play an important role in the pathophysiology of CVDS and both have anti-oxidative stress and anti-inflammatory activity. Studies have shown that the levels of Klotho and sirtuins are altered in sleep disorders. In this article, alterations of Klotho and sirtuins in sleep disorders and in the development of sleep-related CVDs were reviewed and the possible signaling pathways were discussed. The inclusion criteria were studies with keywords of different types of sleep disorders and CVDs, klotho, SIRT1-7, and sirtuins in PubMed, Scopus, Embase، Science Direct، Web of Sciences and Google Scholar by the end of 2023. The studies revealed there is a bidirectional relationship between sleep disorders and the serum and tissue levels of Klotho and sirtuins and sleep related-CVDs.
Collapse
Affiliation(s)
- Farzaneh Rostamzadeh
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Siyavash Joukar
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Cardiovascular Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mahboobeh Yeganeh-Hajahmadi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
4
|
Krenytska D, Strubchevska K, Kozyk M, Vovk T, Halenova T, Kot L, Raksha N, Savchuk O, Falalyeyeva T, Tsyryuk O, Ostapchenko L. Circulating levels of inflammatory cytokines and angiogenesis-related growth factors in patients with osteoarthritis after COVID-19. Front Med (Lausanne) 2023; 10:1168487. [PMID: 37484856 PMCID: PMC10358362 DOI: 10.3389/fmed.2023.1168487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/25/2023] [Indexed: 07/25/2023] Open
Abstract
Background The disease COVID-19, caused by SARS-CoV-2 infection, has a systemic effect and is associated with a number of pathophysiological mechanisms that mobilize a wide range of biomolecules. Cytokines and growth factors (GFs) are critical regulators of tissue damage or repair in osteoarthritis (OA) and are being recognized as key players in the pathogenesis of COVID-19. A clear understanding of the long-term consequences of SARS-CoV-2 infection, especially in patients with concomitant chronic diseases, is limited and needs to be elucidated. The study aimed to evaluate the degree of inflammation and levels of pro-angiogenic and hypoxic factors, as well as heat shock proteins HSP60 and HSP70 in plasma, of patients with OA after recovery from COVID-19. Methods The research involved patients of an orthopedic specialty clinic aged 39 to 80 diagnosed with knee OA. All examined patients were divided into three groups: the Control group included conditionally healthy donors, group OA included patients with knee OA mainly stage II or III and the group of OA and COVID-19 included patients with OA who had COVID-19. The plasma levels of pro-inflammatory molecules IL-1β, IL-6, TNF-α, NF-κB, angiogenic factors VEGF, FGF-2, PDGF, hypoxic factor HIF-1α and molecular chaperones HSP60 and HSP70 were measured by enzyme-linked immunosorbent assay. Results The study showed that in both groups of patients, with OA and convalescent COVID-19, there was an increase in the plasma level of IL-1β and a decrease in TNF-α and NF-κB levels when compared to healthy controls. Systemic deregulation of the cytokine profile was accompanied by reduction in plasma levels of pro-angiogenic growth factors, most pronounced in cases of VEGF and PDGF. This analysis did not reveal any significant difference in the plasma level of HIF-1α. A decrease in the level of stress protein HSP60 in the blood of patients with OA, as well as those patients who have had SARS-CoV-2 infection, has been established. Conclusion The results suggest the potential role pro-inflammatory cytokines and angiogenesis-related growth factors in pathogenesis of both joint pathologies and long-term systemic post-COVID-19 disorders.
Collapse
Affiliation(s)
- Daryna Krenytska
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | | | - Marko Kozyk
- William Beaumont Hospital, Royal Oak, MI, United States
| | - Tetiana Vovk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetiana Halenova
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Larysa Kot
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Nataliia Raksha
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olexii Savchuk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetyana Falalyeyeva
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olena Tsyryuk
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Liudmyla Ostapchenko
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| |
Collapse
|
5
|
Salyha N, Oliynyk I. Hypoxia modeling techniques: A review. Heliyon 2023; 9:e13238. [PMID: 36718422 PMCID: PMC9877323 DOI: 10.1016/j.heliyon.2023.e13238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/08/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Hypoxia is the main cause and effect of a large number of diseases, including the most recent one facing the world, the coronavirus disease (COVID-19). Hypoxia is divided into short-term, long-term, and periodic, it can be the result of diseases, climate change, or living and traveling in the high mountain regions of the world. Since each type of hypoxia can be a cause and a consequence of various physiological changes, the methods for modeling these hypoxias are also different. There are many techniques for modeling hypoxia under experimental conditions. The most common animal for modeling hypoxia is a rat. Hypoxia models (hypoxia simulations) in rats are a tool to study the effect of various conditions on the oxygen supply of the body. These models can provide a necessary information to understand hypoxia and also provide effective treatment, highlighting the importance of various reactions of the body to hypoxia. The main parameters when choosing a model should be reproducibility and the goal that the scientist wants to achieve. Hypoxia in rats can be reproduced both ways exogenously and endogenously. The reason for writing this review was the aim to systematize the models of rats available in the literature in order to facilitate their selection by scientists. The relative strengths and limitations of each model need to be identified and understood in order to evaluate the information obtained from these models and extrapolate these results to humans to develop the necessary generalizations. Despite these problems, animal models have been and remain vital to understanding the mechanisms involved in the development and progression of hypoxia. The eligibility criteria for the selected studies was a comprehensive review of the methods and results obtained from the studies. This made it possible to make generalizations and give recommendations on the application of these methods. The review will assist scientists in choosing an appropriate hypoxia simulation method, as well as assist in interpreting the results obtained with these methods.
Collapse
Affiliation(s)
- Nataliya Salyha
- Institute of Animal Biology NAAS, Lviv, Ukraine,Corresponding author
| | | |
Collapse
|
6
|
Tissue oxygenation stabilizes neovessels and mitigates hemorrhages in human atherosclerosis-induced angiogenesis. Angiogenesis 2023; 26:63-76. [PMID: 35947328 DOI: 10.1007/s10456-022-09851-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/21/2022] [Indexed: 11/01/2022]
Abstract
Progression of atherosclerosis is associated with a maladaptive form of angiogenesis which contributes to intraplaque hemorrhage and plaque disruption. Hypoxia has been implicated in mechanisms of angiogenic neovessel fragility and atherosclerotic plaque destabilization. We used ex vivo and in vivo models to characterize the effect of oxygen (O2) on the formation, stability and tendency to bleed of human plaque-induced neovessels. Plaque explants potently stimulated the ex vivo angiogenic response of rat aortic rings at atmospheric O2 levels. Severe hypoxia (1% O2) inhibited plaque-induced angiogenesis and pericyte recruitment causing neovessel breakdown, whereas increasing O2 levels dose dependently enhanced pericyte numbers and neovessel stability. Plaque fragments implanted subcutaneously with or without aortic rings in SCID mice stimulated the host angiogenic response with plaques causing minimal or no hemorrhages and plaques co-implanted with aortic rings causing marked hemorrhages. Plaque/aortic ring-induced hemorrhages were reduced in mice exposed to moderate hyperoxia (50% O2). Hyperoxia downregulated expression of the hypoxia-sensitive genes Ca9, Ca12 and VegfA and increased influx into implants of mesenchymal cells reactive for the pericyte marker NG2. In both ex vivo and in vivo models, O2 promoted expression of vasostabilizing genes required for pericyte recruitment (Angpt1, Pdgfb), basement membrane assembly (Col4A1), and tight junction formation (Cldn5 and/or Ocln). Our results suggest that formation of neovessels that are stable, pericyte-coated, and resistant to bleeding requires adequate tissue oxygenation. Understanding the mechanisms by which O2 stabilizes neovessels and mitigates neovessel bleeding may lead to new therapies for the prevention of atherosclerosis complications.
Collapse
|
7
|
Wang JY, Liu K, Wang YB, Deng YB, Sun J. Effects of norepinephrine on plaque hypoxia in atherosclerotic rabbits. Front Cardiovasc Med 2023; 10:1121413. [PMID: 36873394 PMCID: PMC9974659 DOI: 10.3389/fcvm.2023.1121413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Background Hypoxia plays a vital role throughout the whole process of atherosclerotic vulnerable plaque formation, which may be induced by a reduced oxygen supply. The vasa vasorum can be affected by norepinephrine (NE) and cause a reduced oxygen supply, ultimately leading to plaque hypoxia. This study aimed to investigate the effects of norepinephrine, which can increase the tension of the vasa vasorum, on plaque hypoxia, evaluated by contrast-enhanced ultrasound imaging. Methods Atherosclerosis (AS) was induced in New Zealand white rabbits by a combination of a cholesterol-rich diet and aortic balloon dilation. After the atherosclerotic model was well established, NE was intravenously administered three times per day for 2 weeks. Contrast-enhanced ultrasound (CEUS) and immunohistochemistry staining were performed to evaluate the expression of hypoxia-inducible factor alpha (HIF-α) and vascular endothelial growth factor (VEGF) in atherosclerotic plaques. Results The plaque blood flow decreased after long-term norepinephrine administration. The expression of HIF-α and VEGF in atherosclerotic plaques concentrated in the outer medial layers increased, which indicated that NE might cause plaque hypoxia by contraction of the vasa vasorum. Conclusion Apparent hypoxia of atherosclerotic plaques after long-term NE administration was mainly caused by decreased plaque blood flow due to the contraction of the vasa vasorum and high blood pressure.
Collapse
Affiliation(s)
- Jia-Yu Wang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Liu
- Department of Medical Ultrasound, The Third People's Hospital of Hubei, Wuhan, China
| | - Yu-Bo Wang
- Department of Medical Ultrasound, Maternal and Child Healthcare Hospital of Hubei, Wuhan, China
| | - You-Bin Deng
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Sun
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Locatelli F, Del Vecchio L. Hypoxia-Inducible Factor-Prolyl Hydroxyl Domain Inhibitors: From Theoretical Superiority to Clinical Noninferiority Compared with Current ESAs? J Am Soc Nephrol 2022; 33:1966-1979. [PMID: 36041790 PMCID: PMC9678041 DOI: 10.1681/asn.2022040413] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Anemia is a common complication of chronic kidney disease; it is mainly treated with erythropoiesis-stimulating agents (ESAs) and iron. Experimental studies extensively investigated the mechanisms involved in the body's response to hypoxia and led to the discovery of the hypoxia-inducible factor (HIF) pathway and the enzymes regulating its function. HIF-prolyl-hydroxyl domain (PHD) inhibitors are a new class of oral drugs developed to treat anemia in chronic kidney disease. By inhibiting the function of PHD enzymes, they mimic the exposure to moderate hypoxia and stimulate the production of endogenous erythropoietin and very likely increase iron availability. Some data also suggest that their efficacy and, consequently, dose needs are less influenced by inflammation than ESAs. Overall, data from phases 2 and 3 clinical development showed efficacy in anemia correction and maintenance for all of the class molecules compared with placebo (superiority) or erythropoiesis-stimulating agents (noninferiority). Three molecules, roxadustat, vadadustat, and daprodustat, underwent extensive clinical investigation to assess their safety on hard cardiovascular end points, mortality, and special interest events (including cancer and thrombosis). Aside from vadadustat in the nondialysis population, at the prespecified primary analyses, all three molecules met the noninferiority margin for the risk of major cardiovascular events compared with erythropoiesis-stimulating agents or placebo. The reason for this discrepancy is difficult to explain. Other safety signals came from secondary analyses of some of the other randomized clinical trials, including a higher incidence of thrombosis. A more extensive clinical experience with post-marketing data on hard safety issues is needed to define better when and how to use HIF-PHD inhibitors compared with already available ESAs.
Collapse
Affiliation(s)
- Francesco Locatelli
- Department of Nephrology and Dialysis, Alessandro Manzoni Hospital (past Director) ASST Lecco, Lecco, Italy
| | - Lucia Del Vecchio
- Department of Nephrology and Dialysis, Sant’Anna Hospital, ASST Lariana, Como, Italy
| |
Collapse
|
9
|
Sonaglioni A, Caminati A, Nicolosi GL, Lombardo M, Harari S. Incremental prognostic value of arterial elastance in mild-to-moderate idiopathic pulmonary fibrosis. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2022; 38:1473-1485. [PMID: 35103898 DOI: 10.1007/s10554-022-02541-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/25/2022] [Indexed: 11/05/2022]
Abstract
Previous reports suggested that poor pulmonary function was associated with increased arterial elastance (Ea) in patients with chronic obstructive pulmonary disease and systemic sclerosis. The mechanisms connecting pulmonary function and Ea have not yet been accurately studied in patients with idiopathic pulmonary fibrosis (IPF). The present study was designed to assess Ea in IPF patients without chronic severe pulmonary hypertension and to determine its prognostic role over a medium-term follow-up. This retrospective study included 60 consecutive patients with mild-to-moderate IPF (73.8 ± 6.6 years, 75% males) and 60 controls matched by age, sex and cardiovascular risk factors. All patients underwent physical examination, spirometry, blood tests, modified Haller index (MHI, chest transverse diameter over the distance between sternum and spine) assessment, conventional transthoracic echocardiography implemented with speckle tracking analysis of left atrial positive global strain (LA-GSA+ ) and finally carotid Doppler ultrasonography, at basal evaluation. The effective arterial elastance index (EaI) was calculated as the ratio of end-systolic pressure to stroke volume index. During follow-up period, we evaluated the composite endpoint of (1) pulmonary or cardiovascular hospitalizations; (2) all-cause mortality. At baseline, EaI was significantly higher in IPF patients than controls (4.1 ± 1.3 vs 3.5 ± 1.0 mmHg/ml/m2, p = 0.01). EaI was strongly correlated to the following variables: C-reactive protein (CRP) (r = 0.86), forced vital capacity (FVC) (r = - 0.91), E/e' ratio (r = 0.91), LA-GSA+ (r = - 0.92), common carotid artery-cross sectional area (CCA-CSA) (r = 0.89) and MHI (r = 0.86), in IPF patients. Mean follow-up time was 2.4 ± 1.3 years. During follow-up, 12 patients died and 17 were hospitalized due to major adverse clinical events. At univariate Cox analysis, CRP (HR 1.51, 95% CI 1.25-1.82), FVC (HR 0.88, 95% CI 0.85-0.91), LA-GSA+ (HR 0.85, 95% CI 0.77-0.94), CCA-CSA (HR 1.12, 95% CI 1.03-1.22) and EaI (HR 2.43, 95% CI 1.75-3.37) were significantly associated with outcome. At multivariate Cox analysis, only EaI (HR 1.60, 95% CI 1.03-2.50) retained statistical significance. An EaI ≥ 4 mmHg/ml/m2 showed 100% sensitivity and 99.4% specificity for predicting outcome (AUC = 0.98). In patients with mild-to-moderate IPF, an EaI ≥ 4 mmHg/ml/m2 is a negative prognostic factor over a medium-term follow-up.
Collapse
Affiliation(s)
- Andrea Sonaglioni
- Division of Cardiology, Ospedale San Giuseppe MultiMedica IRCCS, Via San Vittore 12, 20123, Milan, Italy
| | - Antonella Caminati
- Division of Pneumology, Semi-Intensive Care Unit, Ospedale San Giuseppe MultiMedica IRCCS, Via San Vittore 12, 20123, Milan, Italy.
| | - Gian Luigi Nicolosi
- Division of Cardiology, Policlinico San Giorgio, Via Agostino Gemelli 10, 33170, Pordenone, Italy
| | - Michele Lombardo
- Division of Cardiology, Ospedale San Giuseppe MultiMedica IRCCS, Via San Vittore 12, 20123, Milan, Italy
| | - Sergio Harari
- Division of Pneumology, Semi-Intensive Care Unit, Ospedale San Giuseppe MultiMedica IRCCS, Via San Vittore 12, 20123, Milan, Italy
- Department of Clinical Sciences and Community Health, Università Di Milano, Milan, Italy
| |
Collapse
|
10
|
Thomas C, Leleu D, Masson D. Cholesterol and HIF-1α: Dangerous Liaisons in Atherosclerosis. Front Immunol 2022; 13:868958. [PMID: 35386720 PMCID: PMC8977597 DOI: 10.3389/fimmu.2022.868958] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
HIF-1α exerts both detrimental and beneficial actions in atherosclerosis. While there is evidence that HIF-1α could be pro-atherogenic within the atheromatous plaque, experimental models of atherosclerosis suggest a more complex role that depends on the cell type expressing HIF-1α. In atheroma plaques, HIF-1α is stabilized by local hypoxic conditions and by the lipid microenvironment. Macrophage exposure to oxidized LDLs (oxLDLs) or to necrotic plaque debris enriched with oxysterols induces HIF-1α -dependent pathways. Moreover, HIF-1α is involved in many oxLDL-induced effects in macrophages including inflammatory response, angiogenesis and metabolic reprogramming. OxLDLs activate toll-like receptor signaling pathways to promote HIF-1α stabilization. OxLDLs and oxysterols also induce NADPH oxidases and reactive oxygen species production, which subsequently leads to HIF-1α stabilization. Finally, recent investigations revealed that the activation of liver X receptor, an oxysterol nuclear receptor, results in an increase in HIF-1α transcriptional activity. Reciprocally, HIF-1α signaling promotes triglycerides and cholesterol accumulation in macrophages. Hypoxia and HIF-1α increase the uptake of oxLDLs, promote cholesterol and triglyceride synthesis and decrease cholesterol efflux. In conclusion, the impact of HIF-1α on cholesterol homeostasis within macrophages and the feedback activation of the inflammatory response by oxysterols via HIF-1α could play a deleterious role in atherosclerosis. In this context, studies aimed at understanding the specific mechanisms leading to HIF-1α activation within the plaque represents a promising field for research investigations and a path toward development of novel therapies.
Collapse
Affiliation(s)
- Charles Thomas
- Univ. Bourgogne Franche-Comté, LNC UMR1231, Dijon, France.,INSERM, LNC UMR1231, Dijon, France.,LipSTIC LabEx, Dijon, France
| | - Damien Leleu
- Univ. Bourgogne Franche-Comté, LNC UMR1231, Dijon, France.,INSERM, LNC UMR1231, Dijon, France.,LipSTIC LabEx, Dijon, France.,CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, Dijon, France
| | - David Masson
- Univ. Bourgogne Franche-Comté, LNC UMR1231, Dijon, France.,INSERM, LNC UMR1231, Dijon, France.,LipSTIC LabEx, Dijon, France.,CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, Dijon, France
| |
Collapse
|
11
|
Barnes LA, Mesarwi OA, Sanchez-Azofra A. The Cardiovascular and Metabolic Effects of Chronic Hypoxia in Animal Models: A Mini-Review. Front Physiol 2022; 13:873522. [PMID: 35432002 PMCID: PMC9008331 DOI: 10.3389/fphys.2022.873522] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Animal models are useful to understand the myriad physiological effects of hypoxia. Such models attempt to recapitulate the hypoxemia of human disease in various ways. In this mini-review, we consider the various animal models which have been deployed to understand the effects of chronic hypoxia on pulmonary and systemic blood pressure, glucose and lipid metabolism, atherosclerosis, and stroke. Chronic sustained hypoxia (CSH)-a model of chronic lung or heart diseases in which hypoxemia may be longstanding and persistent, or of high altitude, in which effective atmospheric oxygen concentration is low-reliably induces pulmonary hypertension in rodents, and appears to have protective effects on glucose metabolism. Chronic intermittent hypoxia (CIH) has long been used as a model of obstructive sleep apnea (OSA), in which recurrent airway occlusion results in intermittent reductions in oxyhemoglobin saturations throughout the night. CIH was first shown to increase systemic blood pressure, but has also been associated with other maladaptive physiological changes, including glucose dysregulation, atherosclerosis, progression of nonalcoholic fatty liver disease, and endothelial dysfunction. However, models of CIH have generally been implemented so as to mimic severe human OSA, with comparatively less focus on milder hypoxic regimens. Here we discuss CSH and CIH conceptually, the effects of these stimuli, and limitations of the available data.
Collapse
Affiliation(s)
- Laura A. Barnes
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Omar A. Mesarwi
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Ana Sanchez-Azofra
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
- Servicio de Neumología, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
12
|
Hypoxia inducible factor 1α inhibitor PX-478 reduces atherosclerosis in mice. Atherosclerosis 2022; 344:20-30. [PMID: 35121387 PMCID: PMC8885973 DOI: 10.1016/j.atherosclerosis.2022.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Hypoxia inducible factor 1α (HIF1α) plays a critical role in atherosclerosis as demonstrated in endothelial-targeted HIF1α -deficient mice. However, it has not been shown if specific pharmacological inhibitors of HIF1α can be used as potential drugs for atherosclerosis. PX-478 is a selective inhibitor of HIF1α, which was used to reduce cancer and obesity in animal models. Here, we tested whether PX-478 can be used to inhibit atherosclerosis. METHODS We first tested PX-478 in human aortic endothelial cells (HAEC) and found that it significantly inhibited expression of HIF1α and its targets, including Collagen I. Next, two independent atherosclerosis models, C57BL/6 mice treated with AAV-PCSK9 and ApoE-/- mice, were used to test the efficacy of PX-478. Both mouse models were fed a Western diet for 3 months with bi-weekly treatment with PX-478 (40 mg/kg) or saline. RESULTS PX-478 treatment reduced atherosclerotic plaque burden in the aortic trees in both mouse models, while plaque burden in the aortic sinus was reduced in the AAV-PCSK9 mouse model, but not in the ApoE-/- mice. Russell-Movat's Pentachrome and Picrosirius Red staining showed a significant reduction in extracellular matrix remodeling and collagen maturation, respectively, in the PX-478-treated mice. As expected, PX-478 treatment reduced diet-induced weight-gain and abdominal adipocyte hypertrophy. Interestingly, PX-478 reduced plasma LDL cholesterol by 69% and 30% in AAV-PCSK9 and ApoE-/- mice, respectively. To explore the cholesterol-lowering mechanisms, we carried out an RNA sequencing study using the liver tissues from the ApoE-/- mouse study. We found 450 genes upregulated and 381 genes downregulated by PX-478 treatment in the liver. Further, gene ontology analysis showed that PX-478 treatment upregulated fatty acid and lipid catabolic pathways, while downregulating lipid biosynthesis and plasma lipoprotein particle remodeling processes. Of interest, Cfd, Elovl3, and Insig2 were some of the most downregulated genes by PX-478, and have been implicated in fat storage, fatty acid elongation, and cholesterol metabolism. The downregulation of Cfd, Elovl3, and Insig2 was further validated by qPCR in the liver tissues of ApoE-/- mice treated with PX-478. CONCLUSIONS These results suggest that PX-478 is a potential anti-atherogenic drug, which targets vascular endothelium and hepatic cholesterol pathways.
Collapse
|
13
|
Zhaojun LMDP, Cuiqin SMS, Qingqing CMB, Lei SMB, Xianghong LMDP, Lianfang DMDP. Shear Wave Dispersion Imaging for Measuring Carotid Elasticity and Viscosity. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2022. [DOI: 10.37015/audt.2021.200071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
14
|
Sánchez E, Santos MD, Nuñez-Garcia M, Bueno M, Sajoux I, Yeramian A, Lecube A. Randomized Clinical Trial to Evaluate the Morphological Changes in the Adventitial Vasa Vasorum Density and Biological Markers of Endothelial Dysfunction in Subjects with Moderate Obesity Undergoing a Very Low-Calorie Ketogenic Diet. Nutrients 2021; 14:33. [PMID: 35010908 PMCID: PMC8746664 DOI: 10.3390/nu14010033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022] Open
Abstract
Weight loss after bariatric surgery decreases the earlier expansion of the adventitial vasa vasorum (VV), a biomarker of early atheromatous disease. However, no data are available regarding weight loss achieved by very low calorie ketogenic diets (VLCKD) on VV and lipid-based atherogenic indices. A randomized clinical trial was performed to examine changes in adventitial VV density in 20 patients with moderate obesity who underwent a 6-month very low calorie ketogenic diet (VLCKD, 600-800 kcal/day), and 10 participants with hypocaloric diet based on the Mediterranean Diet (MedDiet, estimated reduction of 500 kcal on the usual intake). Contrast-enhanced carotid ultrasound was used to assess the VV. Body composition analysis was also used. The atherogenic index of plasma (log (triglycerides to high-density lipoprotein cholesterol ratio)) and the triglyceride-glucose index were calculated. Serum concentrations of soluble intercellular adhesion molecule 1 (sICAM-1), and soluble vascular cell adhesion molecule 1 (sVCAM-1) were measured. The impact of weight on quality of life-lite (IWQOL-Lite) questionnaire was administered. Participants of intervention groups displayed a similar VV values. Significant improvements of BMI (-5.3 [-6.9 to -3.6] kg/m2, p < 0.001), total body fat (-7.0 [-10.7 to -3.3] %, p = 0.003), and IWQOL-Lite score (-41.4 [-75.2 to -7.6], p = 0.027) were observed in VLCKD group in comparison with MedDiet group. Although after a 6-months follow-up period VV density (mean, right and left sides) did not change significantly in any group, participants in the VLCKD exhibited a significantly decrease both in their atherogenic index of plasma and serum concentration of sICAM-1. A 6-month intervention with VLCKD do not impact in the density of the adventitial VV in subjects with moderate obesity, but induces significant changes in markers of endothelial dysfunction and CV risk.
Collapse
Affiliation(s)
- Enric Sánchez
- Obesity, Diabetes and Metabolism (ODIM) Research Group, IRBLleida, University of Lleida, 25198 Lleida, Spain; (E.S.); (M.-D.S.); (M.B.)
| | - Maria-Dolores Santos
- Obesity, Diabetes and Metabolism (ODIM) Research Group, IRBLleida, University of Lleida, 25198 Lleida, Spain; (E.S.); (M.-D.S.); (M.B.)
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
| | | | - Marta Bueno
- Obesity, Diabetes and Metabolism (ODIM) Research Group, IRBLleida, University of Lleida, 25198 Lleida, Spain; (E.S.); (M.-D.S.); (M.B.)
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
| | - Ignacio Sajoux
- Pronokal Group, 08009 Barcelona, Spain; (M.N.-G.); (I.S.)
| | - Andree Yeramian
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
| | - Albert Lecube
- Obesity, Diabetes and Metabolism (ODIM) Research Group, IRBLleida, University of Lleida, 25198 Lleida, Spain; (E.S.); (M.-D.S.); (M.B.)
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, 25198 Lleida, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
15
|
Ielciu I, Filip GA, Oniga I, Olah NK, Bâldea I, Olteanu D, Burtescu RF, Turcuș V, Sevastre-Berghian AC, Benedec D, Hanganu D. Oxidative Stress and DNA Lesion Reduction of a Polyphenolic Enriched Extract of Thymus marschallianus Willd. in Endothelial Vascular Cells Exposed to Hyperglycemia. PLANTS (BASEL, SWITZERLAND) 2021; 10:plants10122810. [PMID: 34961280 PMCID: PMC8708594 DOI: 10.3390/plants10122810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/06/2021] [Accepted: 12/15/2021] [Indexed: 05/04/2023]
Abstract
The present study aimed to compare two polyphenolic-enriched extracts obtained from the Thymus marschallianus Willd. (Lamiaceae) species, harvested from culture (TMCE in doses of 0.66 μg GAE/mL and 0.066 μg GAE/mL) and from spontaneous flora (TMSE in doses of 0.94 μg GAE/mL and 0.094 μg GAE/mL) by assessing their biological effects on human umbilical vein endothelial cells (HUVECs) exposed to normoglycemic (137 mmol/L glucose) and hyperglycemic conditions (200 mmol/L glucose). Extracts were obtained by solid phase extraction (SPE) and analyzed by chromatographical (HPLC-DAD) and spectrophotometrical methods. Their effects on hyperglycemia were evaluated by the quantification of oxidative stress and NF-ĸB, pNF-ĸB, HIF-1α, and γ-H2AX expressions. The HPLC-DAD analysis highlighted significant amounts of rosmarinic acid (ranging between 0.18 and 1.81 mg/g dry extract), luteolin (ranging between 2.04 and 17.71 mg/g dry extract), kaempferol (ranging between 1.85 and 7.39 mg/g dry extract), and apigenin (ranging between 4.97 and 65.67 mg/g dry extract). Exposure to hyperglycemia induced oxidative stress and the activation of NF-ĸ increased the expression of HIF-1α and produced DNA lesions. The polyphenolic-enriched extracts proved a significant reduction of oxidative stress and γ-H2AX formation and improved the expression of HIF-1α, suggesting their protective role on endothelial cells in hyperglycemia. The tested extracts reduced the total NF-ĸB expression and diminished its activation in hyperglycemic conditions. The obtained results bring evidence for the use of the polyphenolic-enriched extracts of T. marschallianus as adjuvants in hyperglycemia.
Collapse
Affiliation(s)
- Irina Ielciu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania;
| | - Gabriela Adriana Filip
- Department of Physiology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (I.B.); (D.O.); (A.C.S.-B.)
- Correspondence: (G.A.F.); (I.O.)
| | - Ilioara Oniga
- Department of Pharmacognosy, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania; (D.B.); (D.H.)
- Correspondence: (G.A.F.); (I.O.)
| | - Neli-Kinga Olah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Vasile Goldiş” Western University of Arad, 310414 Arad, Romania;
- PlantExtrakt Ltd., Rădaia, 407059 Cluj-Napoca, Romania;
| | - Ioana Bâldea
- Department of Physiology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (I.B.); (D.O.); (A.C.S.-B.)
| | - Diana Olteanu
- Department of Physiology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (I.B.); (D.O.); (A.C.S.-B.)
| | | | - Violeta Turcuș
- Department of Botany, Faculty of Medicine, “Vasile Goldiş” Western University of Arad, 310414 Arad, Romania;
| | - Alexandra C. Sevastre-Berghian
- Department of Physiology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (I.B.); (D.O.); (A.C.S.-B.)
| | - Daniela Benedec
- Department of Pharmacognosy, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania; (D.B.); (D.H.)
| | - Daniela Hanganu
- Department of Pharmacognosy, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania; (D.B.); (D.H.)
| |
Collapse
|
16
|
Wu Y, Cai F, Lu Y, Hu Y, Wang Q. lncRNA RP11-531A24.3 inhibits the migration and proliferation of vascular smooth muscle cells by downregulating ANXA2 expression. Exp Ther Med 2021; 22:1439. [PMID: 34721681 DOI: 10.3892/etm.2021.10874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 07/16/2021] [Indexed: 12/19/2022] Open
Abstract
A complete understanding of the behavioral influence and phenotypic transition of vascular smooth muscle cells, as well as the effects of the characteristics of these cells on the physiological and pathological processes of atherosclerosis, is crucial if new therapeutic targets for atherosclerosis are to be identified. In the present study, the long non-coding RNA RP11-531A24.3 was identified to be expressed at low levels in plaque tissues through screening a microarray for differentially expressed genes. The functional experimental results suggested that RP11-531A24.3 reduced the viability and inhibited the migration of human aortic vascular smooth muscle cells (HA-VSMCs). RNA antisense purification-mass spectrometry was used to identify the RNA-binding proteins (RBPs) for RP11-531A24.3. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis indicated that the pathway with the highest degree of association with RP11-531A24.3 RBPs was related to cell migration. The reduced migration and viability mediated by RP11-531A24.3 overexpression was more significantly suppressed after annexin 2 (ANXA2) depletion in RP11-531A24.3-overexpressing HA-VSMCs. Culture of HA-VSMCs under hypoxic conditions (1% O2) reduced the expression of RP11-531A24.3, and enhanced the protein expression of ANXA2 and HIF-1α, while knockdown of ANXA2 downregulated the protein expression of HIF-1α. These results suggested that RP11-531A24.3 regulated the proliferation and migration of HA-VSMCs through ANXA2 expression, and hypoxia may be an external factor in the regulation of RP11-531A24.3 and its downstream targets.
Collapse
Affiliation(s)
- Yilin Wu
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Fen Cai
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Clinical Laboratory, Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou, Guangdong 510800, P.R. China
| | - Yuanbin Lu
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yanwei Hu
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Qian Wang
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
17
|
Knutson AK, Williams AL, Boisvert WA, Shohet RV. HIF in the heart: development, metabolism, ischemia, and atherosclerosis. J Clin Invest 2021; 131:137557. [PMID: 34623330 DOI: 10.1172/jci137557] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The heart forms early in development and delivers oxygenated blood to the rest of the embryo. After birth, the heart requires kilograms of ATP each day to support contractility for the circulation. Cardiac metabolism is omnivorous, utilizing multiple substrates and metabolic pathways to produce this energy. Cardiac development, metabolic tuning, and the response to ischemia are all regulated in part by the hypoxia-inducible factors (HIFs), central components of essential signaling pathways that respond to hypoxia. Here we review the actions of HIF1, HIF2, and HIF3 in the heart, from their roles in development and metabolism to their activity in regeneration and preconditioning strategies. We also discuss recent work on the role of HIFs in atherosclerosis, the precipitating cause of myocardial ischemia and the leading cause of death in the developed world.
Collapse
|
18
|
Yücel C, Gürsoy M, Ketenciler S, Tenekeciğil A, Kızıltan F, Kayalar N. Is Biochemical Follow Up Possible in Peripheral Arterial Disease Treatment: Hypoxia Inducible Factor-1 Alpha? Vasc Specialist Int 2021; 37:29. [PMID: 34429387 PMCID: PMC8385279 DOI: 10.5758/vsi.210011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/30/2021] [Accepted: 07/17/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose The hypoxia inducible factor (HIF)-1 is a dimeric protein complex that plays an integral role in the body’s response to hypoxia. This study aimed to analyze the regulation of HIF-1α following vascular and/or endovascular surgery in peripheral arterial disease (PAD) patients. Methods Materials and A total of 40 patients with PAD (≥Rutherford category 3) were included in this prospective study. The mean age was 61.9±9.2 years. Open surgery was performed in 16 patients, and endovascular intervention was performed in 34 patients. At preoperative (T1), postoperative day 1 (T2), and month 3 (T3), the serum HIF-1α levels were checked using the ELISA technique. Results At T3, the ankle-brachial index was significantly higher than the preoperative value (P<0.001). Serum HIF-1α levels at T1, T2, and T3 were 2.0±1.7 ng/mL, 1.9±1.7 ng/mL, and 1.6±1.4 ng/mL, respectively. Serum HIF-1α levels between T1 and T3 and between T2 and T3 were significantly different (P<0.05). The preoperative HIF-1α levels were lowest in iliac lesions compared to femoropopliteal or tibial lesions. Conclusion The HIF-1α levels were decreased in all patients on postoperative days, T2 and T3, compared with the preoperative values. Our results indicated that HIF-1α may be a surrogate marker after revascularization in patients with PAD. Further studies are needed to analyze the sensitivity, specificity, and cut-off values of HIF-1α in patients with PAD.
Collapse
Affiliation(s)
- Cihan Yücel
- Department of Cardiovascular Surgery, Prof Dr Cemil Tascıoglu City Hospital, İstanbul, Turkey
| | - Mete Gürsoy
- Department of Cardiovascular Surgery, Prof Dr Cemil Tascıoglu City Hospital, İstanbul, Turkey
| | - Serkan Ketenciler
- Department of Cardiovascular Surgery, Prof Dr Cemil Tascıoglu City Hospital, İstanbul, Turkey
| | - Aslıhan Tenekeciğil
- Medical Laboratory, Bağcılar Training and Research Hospital, İstanbul, Turkey
| | - Feryaz Kızıltan
- Department of Cardiovascular Surgery, Prof Dr Cemil Tascıoglu City Hospital, İstanbul, Turkey
| | - Nihan Kayalar
- Department of Cardiovascular Surgery, Prof Dr Cemil Tascıoglu City Hospital, İstanbul, Turkey
| |
Collapse
|
19
|
Liu YH, Guo C, Sun YQ, Li Q. Polymorphisms in HIF-1a gene are not associated with diabetic retinopathy in China. World J Diabetes 2021; 12:1304-1311. [PMID: 34512895 PMCID: PMC8394233 DOI: 10.4239/wjd.v12.i8.1304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/09/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND It has been reported that vascular endothelial growth factor (VEGF) is a susceptibility gene for both type 2 diabetes mellitus (T2DM) and diabetic retinopathy (DR). In response to hypoxia, VEGF mRNA levels are increased, which is mainly mediated by the binding of hypoxia-inducible factor-1 (HIF-1) and hypoxia response element upstream of the transcriptional start site of VEGF. Therefore, HIF-1a is supposed to be involved in pathology of DR.
AIM To investigate whether the polymorphisms in HIF-1a gene are associated with DR.
METHODS Two hundred and ninety-nine type 2 diabetic patients (128 males and 171 females) and 144 healthy volunteers were recruited. Mean age was 56.04 ± 21.05 years. According to the results of fundus fluorescein angiography and examination of ophthalmoscopy, patients were divided into two groups, DNR group (diabetes without retinopathy) and DR group (diabetes with retinopathy). There are 150 cases in DNR group and 149 cases in DR group. Two single nucleotide polymorphisms (SNP) of the HIF-1a gene were tested using matrix-assisted laser desorption/Ionization time of flight mass spectrometry. The frequency of genotypes and alleles, and odds ratio were measured.
RESULTS The mean age of the cases with diabetes was 55.84 ± 3.66 years, the mean age of the cases with DR was 55.97 ± 4.66 years and that of controls was 56.32 ± 4.70 years. Two variations were found in 76 patients. Rs11549465 is the change of C-T base, rs11549467 is the change of G-A base. The rs11549467 G/A genotype was 5.33% in diabetes and 6.04% in DR patients, respectively. The rs11549465 C/T genotype was 10% and 12.75% in patients with diabetes and DR. The rs11549467 A allele frequencies and rs11549465 T frequencies was similar to that of controls. Paired SNP linkage disequilibrium analysis indicated that rs11549467 was in linkage disequilibrium with rs11549465. Haplotype association analysis denoted that the haplotype association exhibited similar distribution in the patients compared to the normal controls.
CONCLUSION This study suggests that there is no relationship between the genetic variations of HIF1a and diabetes or DR.
Collapse
Affiliation(s)
- Yue-Hong Liu
- Department of Endocrinology, Hainan Cancer Hospital, Haikou 570312, Hainan Province, China
| | - Chang Guo
- Department of Endocrinology, Shenzhen University General Hospital, Shenzhen 518055,Guangdong Province, China
| | - Yi-Qiong Sun
- Department of Endocrinology, Shenzhen University General Hospital, Shenzhen 518055,Guangdong Province, China
| | - Qiang Li
- Department of Endocrinology, Shenzhen University General Hospital, Shenzhen 518055,Guangdong Province, China
| |
Collapse
|
20
|
Turpin C, Catan A, Meilhac O, Bourdon E, Canonne-Hergaux F, Rondeau P. Erythrocytes: Central Actors in Multiple Scenes of Atherosclerosis. Int J Mol Sci 2021; 22:ijms22115843. [PMID: 34072544 PMCID: PMC8198892 DOI: 10.3390/ijms22115843] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/16/2022] Open
Abstract
The development and progression of atherosclerosis (ATH) involves lipid accumulation, oxidative stress and both vascular and blood cell dysfunction. Erythrocytes, the main circulating cells in the body, exert determinant roles in the gas transport between tissues. Erythrocytes have long been considered as simple bystanders in cardiovascular diseases, including ATH. This review highlights recent knowledge concerning the role of erythrocytes being more than just passive gas carriers, as potent contributors to atherosclerotic plaque progression. Erythrocyte physiology and ATH pathology is first described. Then, a specific chapter delineates the numerous links between erythrocytes and atherogenesis. In particular, we discuss the impact of extravasated erythrocytes in plaque iron homeostasis with potential pathological consequences. Hyperglycaemia is recognised as a significant aggravating contributor to the development of ATH. Then, a special focus is made on glycoxidative modifications of erythrocytes and their role in ATH. This chapter includes recent data proposing glycoxidised erythrocytes as putative contributors to enhanced atherothrombosis in diabetic patients.
Collapse
Affiliation(s)
- Chloé Turpin
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Aurélie Catan
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | - Olivier Meilhac
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Centre Hospitalier Universitaire de La Réunion, 97400 Saint Denis, France
| | - Emmanuel Bourdon
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
| | | | - Philippe Rondeau
- Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), INSERM, UMR 1188, Université de La Réunion, 97400 Saint Denis, France; (C.T.); (A.C.); (O.M.); (E.B.)
- Correspondence: ; Tel.: +262(0)-2-62-93-88-43; Fax: +262-(0)-2-62-93-88-01
| |
Collapse
|
21
|
Li M, Qi Z, Zhang J, Zhu K, Wang Y. Effect and Mechanism of Si-Miao-Yong-An on Vasa Vasorum Remodeling in ApoE -/- Mice with Atherosclerosis Vulnerable Plague. Front Pharmacol 2021; 12:634611. [PMID: 33935723 PMCID: PMC8080061 DOI: 10.3389/fphar.2021.634611] [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: 11/28/2020] [Accepted: 01/26/2021] [Indexed: 01/20/2023] Open
Abstract
Objective: To observe the effect of Si-Miao-Yong-An (SMYA) on atherosclerosis (AS) vulnerable plaques, and to further explore the mechanism by vasa vasorum (VV) angiogenesis and maturation as an entry point. Methods: SPF-class healthy male ApoE−/− mice were randomized into model group, simvastatin group and SMYA group, and C57BL/6 mice were used as the control group. After 8 weeks of intervention, the pathological morphology of plaque was observed by HE staining; the VV density in plaque and aortic adventitia were observed by immunohistochemistry; VV maturation was measured by double-labelling immunofluorescence; the critical proteins of HIF-1α-Apelin/APJ and Ang-1/Tie signal pathways were detected by western blotting. Results: SMYA decreased the plaque area and the ratio of plaque to lumen area; increased the minimum thickness of fibrous cap and its effect was greater than simvastatin. SMYA suppressed the VV neovascularization; promoted smooth muscle cells recruitment and VV maturation, which maintained plaque stability; its effect was obviously superior to simvastatin. SMYA deceased the expression of HIF-1α, Apelin, APJ, Phospho-MEK1/2 (Ser217/221), Phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204), Phospho-p70 S6 Kinase (Thr421/Ser424), Ang-2 and Tie-2; it also increased the expression of Ang-1, Phospho-Akt (Ser473), Phospho-FOXO1 (Ser256) and Survivin. Conclusions: SMYA can decrease the AS plaque area in ApoE−/− mice, suppress the VV neovascularization and promote the VV maturation, and stabilize AS vulnerable plaque. The mechanism could be regulating the HIF-1α-Apelin/APJ and Ang-1/Tie signal pathways.
Collapse
Affiliation(s)
- Meng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhongwen Qi
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ke Zhu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yueyao Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
22
|
Liu J, Wei E, Wei J, Zhou W, Webster KA, Zhang B, Li D, Zhang G, Wei Y, Long Y, Qi X, Zhang Q, Xu D. MiR-126-HMGB1-HIF-1 Axis Regulates Endothelial Cell Inflammation during Exposure to Hypoxia-Acidosis. DISEASE MARKERS 2021; 2021:4933194. [PMID: 34970357 PMCID: PMC8714334 DOI: 10.1155/2021/4933194] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/20/2021] [Indexed: 02/05/2023]
Abstract
Crosstalk between molecular regulators miR-126, hypoxia-inducible factor 1-alpha (HIF-1-α), and high-mobility group box-1 (HMGB1) contributes to the regulation of inflammation and angiogenesis in multiple physiological and pathophysiological settings. Here, we present evidence of an overriding role for miR-126 in the regulation of HMGB1 and its downstream proinflammatory effectors in endothelial cells subjected to hypoxia with concurrent acidosis (H/A). Methods. Primary mouse endothelial cells (PMEC) were exposed to hypoxia or H/A to simulate short or chronic low-flow ischemia, respectively. RT-qPCR quantified mRNA transcripts, and proteins were measured by western blot. ROS were quantified by fluorogenic ELISA and luciferase reporter assays employed to confirm an active miR-126 target in the HMGB1 3'UTR. Results. Enhanced expression of miR-126 in PMECs cultured under neutral hypoxia was suppressed under H/A, whereas the HMGB1 expression increased sequentially under both conditions. Enhanced expression of HMGB1 and downstream inflammation markers was blocked by the premiR-126 overexpression and optimized by antagomiR. Compared with neutral hypoxia, H/A suppressed the HIF-1α expression independently of miR-126. The results show that HMGB1 and downstream effectors are optimally induced by H/A relative to neutral hypoxia via crosstalk between hypoxia signaling, miR-126, and HIF-1α, whereas B-cell lymphoma 2(Bcl2), a HIF-1α, and miR-126 regulated gene expressed optimally under neutral hypoxia. Conclusion. Inflammatory responses of ECs to H/A are dynamically regulated by the combined actions of hypoxia, miR-126, and HIF-1α on the master regulator HMGB1. The findings may be relevant to vascular diseases including atherosclerotic occlusion and interiors of plaque where coexisting hypoxia and acidosis promote inflammation as a defining etiology.
Collapse
Affiliation(s)
- Jinxue Liu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Eileen Wei
- Gulliver High School, Miami, FL 33156, USA
| | - Jianqin Wei
- Department of Medicine Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Wei Zhou
- Department of Ophthalmology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Keith A. Webster
- Integene International, LLC, Miami, FL 33137, USA
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
- Everglades Biopharma, LLC, Houston, TX 77030, USA
| | - Bin Zhang
- Department of Cardiology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Dong Li
- Department of Intensive Care Unit and Clinical Experimental Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Gaoxing Zhang
- Department of Cardiology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Yidong Wei
- Department of Surgery, Youjiang Medical University for Nationalities, Chengxiang Rd, Baise, Guangxi 533000, China
| | - Yusheng Long
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
- Department of Cardiology, Guangdong Cardiovascular Institute and Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiuyu Qi
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
- Department of Cardiology, Guangdong Cardiovascular Institute and Shantou University Medical College, Shantou 515041, China
| | - Qianhuan Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
| | - Dingli Xu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
23
|
Zhu D, Kang W, Zhang S, Qiao X, Liu J, Liu C, Lu H. Effect of mandibular advancement device treatment on HIF-1α, EPO and VEGF in the myocardium of obstructive sleep apnea-hypopnea syndrome rabbits. Sci Rep 2020; 10:13261. [PMID: 32764565 PMCID: PMC7414037 DOI: 10.1038/s41598-020-70238-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 07/22/2020] [Indexed: 11/09/2022] Open
Abstract
The aim of this study was to investigate the effects of mandibular advancement device (MAD) therapy for obstructive sleep apnea-hypopnea syndrome (OSAHS) on hypoxia-inducible factor-1α (HIF-1α), erythropoietin (EPO) and vascular endothelial growth factor (VEGF) in myocardial tissue. New Zealand rabbits were used to develop OSAHS and MAD models. Cone beam computed tomography (CBCT) of the upper airway and polysomnography (PSG) recordings were performed with the animals in the supine position. All of the animals were induced to sleep in a supine position for 4-6 h each day and were observed continuously for 8 weeks. The myocardial tissue of the three groups was dissected to measure the expression of HIF-1α, EPO and VEGF. The results showed that there was higher expression of HIF-1α, EPO and VEGF in the OSAHS group than those in the MAD and control groups. MAD treatment significantly downregulated the expression of HIF-1α, EPO and VEGF in the OSAHS animals. We concluded that MAD treatment could significantly downregulate the increased expression of HIF-1α, EPO and VEGF in OSAHS rabbits, improving their myocardial function.
Collapse
Affiliation(s)
- Dechao Zhu
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University & Hebei Key Laboratory of Stomatology, No. 383, East Zhongshan Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Wenjing Kang
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University & Hebei Key Laboratory of Stomatology, No. 383, East Zhongshan Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Shilong Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University & Hebei Key Laboratory of Stomatology, No. 383, East Zhongshan Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Xing Qiao
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University & Hebei Key Laboratory of Stomatology, No. 383, East Zhongshan Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Jie Liu
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University & Hebei Key Laboratory of Stomatology, No. 383, East Zhongshan Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Chunyan Liu
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University & Hebei Key Laboratory of Stomatology, No. 383, East Zhongshan Road, Shijiazhuang, 050017, Hebei, People's Republic of China.
| | - Haiyan Lu
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University & Hebei Key Laboratory of Stomatology, No. 383, East Zhongshan Road, Shijiazhuang, 050017, Hebei, People's Republic of China.
| |
Collapse
|
24
|
Tarbell J, Mahmoud M, Corti A, Cardoso L, Caro C. The role of oxygen transport in atherosclerosis and vascular disease. J R Soc Interface 2020; 17:20190732. [PMID: 32228404 PMCID: PMC7211472 DOI: 10.1098/rsif.2019.0732] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/11/2020] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis and vascular disease of larger arteries are often associated with hypoxia within the layers of the vascular wall. In this review, we begin with a brief overview of the molecular changes in vascular cells associated with hypoxia and then emphasize the transport mechanisms that bring oxygen to cells within the vascular wall. We focus on fluid mechanical factors that control oxygen transport from lumenal blood flow to the intima and inner media layers of the artery, and solid mechanical factors that influence oxygen transport to the adventitia and outer media via the wall's microvascular system-the vasa vasorum (VV). Many cardiovascular risk factors are associated with VV compression that reduces VV perfusion and oxygenation. Dysfunctional VV neovascularization in response to hypoxia contributes to plaque inflammation and growth. Disturbed blood flow in vascular bifurcations and curvatures leads to reduced oxygen transport from blood to the inner layers of the wall and contributes to the development of atherosclerotic plaques in these regions. Recent studies have shown that hypoxia-inducible factor-1α (HIF-1α), a critical transcription factor associated with hypoxia, is also activated in disturbed flow by a mechanism that is independent of hypoxia. A final section of the review emphasizes hypoxia in vascular stenting that is used to enlarge vessels occluded by plaques. Stenting can compress the VV leading to hypoxia and associated intimal hyperplasia. To enhance oxygen transport during stenting, new stent designs with helical centrelines have been developed to increase blood phase oxygen transport rates and reduce intimal hyperplasia. Further study of the mechanisms controlling hypoxia in the artery wall may contribute to the development of therapeutic strategies for vascular diseases.
Collapse
Affiliation(s)
- John Tarbell
- Biomedical Engineering Department, The City College of New York, New York, NY, USA
| | - Marwa Mahmoud
- Biomedical Engineering Department, The City College of New York, New York, NY, USA
| | - Andrea Corti
- Biomedical Engineering Department, The City College of New York, New York, NY, USA
| | - Luis Cardoso
- Biomedical Engineering Department, The City College of New York, New York, NY, USA
| | - Colin Caro
- Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
25
|
Alique M, Sánchez-López E, Bodega G, Giannarelli C, Carracedo J, Ramírez R. Hypoxia-Inducible Factor-1α: The Master Regulator of Endothelial Cell Senescence in Vascular Aging. Cells 2020; 9:cells9010195. [PMID: 31941032 PMCID: PMC7016968 DOI: 10.3390/cells9010195] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/07/2020] [Accepted: 01/11/2020] [Indexed: 12/11/2022] Open
Abstract
Aging is one of the hottest topics in biomedical research. Advances in research and medicine have helped to preserve human health, leading to an extension of life expectancy. However, the extension of life is an irreversible process that is accompanied by the development of aging-related conditions such as weakness, slower metabolism, and stiffness of vessels. It also debated that aging can be considered an actual disease with aging-derived comorbidities, including cancer or cardiovascular disease. Currently, cardiovascular disorders, including atherosclerosis, are considered as premature aging and represent the first causes of death in developed countries, accounting for 31% of annual deaths globally. Emerging evidence has identified hypoxia-inducible factor-1α as a critical transcription factor with an essential role in aging-related pathology, in particular, regulating cellular senescence associated with cardiovascular aging. In this review, we will focus on the regulation of senescence mediated by hypoxia-inducible factor-1α in age-related pathologies, with particular emphasis on the crosstalk between endothelial and vascular cells in age-associated atherosclerotic lesions. More specifically, we will focus on the characteristics and mechanisms by which cells within the vascular wall, including endothelial and vascular cells, achieve a senescent phenotype.
Collapse
Affiliation(s)
- Matilde Alique
- Departamento Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud (IRYCIS), Universidad de Alcalá, Alcalá de Henares, 28805 Madrid, Spain;
- Correspondence: (M.A.); (J.C.); Tel.: +34-91-885-6436 (M.A.); +34-91-394-5005 (J.C.)
| | - Elsa Sánchez-López
- Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, CA 92037, USA;
| | - Guillermo Bodega
- Departamento de Biomedicina y Biotecnología, Facultad de Biología, Química y Ciencias Ambientales, Universidad de Alcalá, Alcalá de Henares, 28805 Madrid, Spain;
| | - Chiara Giannarelli
- Cardiovascular Research Center, Institute for Genomics and Multiscale Biology, New York, NY 10029, USA;
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Julia Carracedo
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), 28041 Madrid, Spain
- Correspondence: (M.A.); (J.C.); Tel.: +34-91-885-6436 (M.A.); +34-91-394-5005 (J.C.)
| | - Rafael Ramírez
- Departamento Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud (IRYCIS), Universidad de Alcalá, Alcalá de Henares, 28805 Madrid, Spain;
| |
Collapse
|
26
|
Zhao Q, Sun D, Li Y, Qin J, Yan J. Integrated analyses of lncRNAs microarray profiles and mRNA-lncRNA coexpression in smooth muscle cells under hypoxic and normoxic conditions. Biosci Rep 2019; 39:BSR20181783. [PMID: 30850398 PMCID: PMC6443952 DOI: 10.1042/bsr20181783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 03/03/2019] [Accepted: 03/06/2019] [Indexed: 01/23/2023] Open
Abstract
Hypoxia may cause abnormal proliferation and migration of the vascular smooth muscle cells (VSMCs) from the media to the intima. This contributes to vessel narrowing and accelerates the process of atherosclerosis. The association of the aberrant expression of long noncoding RNAs (lncRNAs) with the development and progression of atherosclerosis is well known; however, it is not well investigated in hypoxic VSMCs. Using a microarray approach, we identified 1056 and 2804 differentially expressed lncRNAs and mRNAs, respectively, in hypoxic and normoxic mouse aorta smooth muscle (MOVAS) cells. Of them, we randomly chose several lncRNAs and validated the microarray data using the quantitative PCR (qPCR) assay. Advanced bioinformatics analyses indicated that the up-regulated mRNAs were mainly involved in inflammatory responses, lipid metabolism, clearance of amyloid-β peptide, citrate cycle (TCA cycle), TGF-β signaling, and chemokine signaling. The down-regulated mRNAs were mainly involved in the apoptosis pathway, glycerolipid metabolism, Wnt signaling pathway, and MAPK signaling pathway. The constructed coexpression network indicated interactions between 87 lncRNAs and ten mRNAs. In addition, we demonstrated that the silence of lncRNA NONMMUT002434 expression could abrogate the migration and proliferation of smooth muscle cells dramatically. Our data provide comprehensive evidence on the differential expression of lncRNAs and mRNAs in hypoxic MOVAS cells, which may be valuable biomarkers for atherosclerotic diseases, and thereby facilitating diagnosis of atherosclerosis.
Collapse
Affiliation(s)
- Qinshuo Zhao
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Dating Sun
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yuanyuan Li
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Jin Qin
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - JiangTao Yan
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
- Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
27
|
López-Cano C, Rius F, Sánchez E, Gaeta AM, Betriu À, Fernández E, Yeramian A, Hernández M, Bueno M, Gutiérrez-Carrasquilla L, Dalmases M, Lecube A. The influence of sleep apnea syndrome and intermittent hypoxia in carotid adventitial vasa vasorum. PLoS One 2019; 14:e0211742. [PMID: 30721271 PMCID: PMC6363284 DOI: 10.1371/journal.pone.0211742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/18/2019] [Indexed: 12/11/2022] Open
Abstract
Subjects with sleep apnea-hypopnea syndrome (SAHS) show an increased carotid intima-media thickness. However, no data exist about earlier markers of atheromatous disease, such as the proliferation and expansion of the adventitial vasa vasorum (VV) to the avascular intima in this setting. Our aim was to assess carotid VV density and its relationship with sleep parameters in a cohort of obese patients without prior vascular events. A total of 55 subjects evaluated for bariatric surgery were prospectively recruited. A non-attended respiratory polygraphy was performed. The apnea-hypopnea index (AHI) and the cumulative percentage of time spent with oxygen saturation below 90% (CT90) were assessed. Serum concentrations of soluble intercellular adhesion molecule 1, P-selectin, lipocalin-2 and soluble vascular cell adhesion molecule 1 (sVCAM-1) were measured. Contrast-enhanced carotid ultrasound was used to assess the VV density. Patients with SAHS (80%) showed a higher adventitial VV density (0.801±0.125 vs. 0.697±0.082, p = 0.005) and higher levels of sVCAM-1 (745.2±137.8 vs. 643.3±122.7 ng/ml, p = 0.035) than subjects with an AHI lower than 10 events/hour. In addition, a positive association exist between mean VV density and AHI (r = 0.445, p = 0.001) and CT90 (r = 0.399, p = 0.005). Finally, in the multiple linear regression analysis, female sex, fasting plasma glucose and AHI (but not CT90) were the only variables independently associated with the mean adventitial VV density (R2 = 0.327). In conclusion, a high VV density is present in obese subjects with SAHS, and chronic intermittent hypoxia is pointed as an independent risk factor for the development of this early step of atheromatous disease.
Collapse
Affiliation(s)
- Carolina López-Cano
- Endocrinology and Nutrition Department, University Hospital Arnau de Vilanova, Obesity, Diabetes and Metabolism (ODIM) research group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Ferran Rius
- Endocrinology and Nutrition Department, University Hospital Arnau de Vilanova, Obesity, Diabetes and Metabolism (ODIM) research group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Enric Sánchez
- Endocrinology and Nutrition Department, University Hospital Arnau de Vilanova, Obesity, Diabetes and Metabolism (ODIM) research group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Anna Michela Gaeta
- Respiratory Department, University Hospital Arnau de Vilanova-Santa María, Translational Research in Respiratory Medicine, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Àngels Betriu
- Unit for the Detection and Treatment of Atherothrombotic Diseases (UDETMA V&R), University Hospital Arnau de Vilanova, Vascular and Renal Translational Research Group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Elvira Fernández
- Unit for the Detection and Treatment of Atherothrombotic Diseases (UDETMA V&R), University Hospital Arnau de Vilanova, Vascular and Renal Translational Research Group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Andree Yeramian
- Endocrinology and Nutrition Department, University Hospital Arnau de Vilanova, Obesity, Diabetes and Metabolism (ODIM) research group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Marta Hernández
- Endocrinology and Nutrition Department, University Hospital Arnau de Vilanova, Obesity, Diabetes and Metabolism (ODIM) research group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Marta Bueno
- Endocrinology and Nutrition Department, University Hospital Arnau de Vilanova, Obesity, Diabetes and Metabolism (ODIM) research group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Liliana Gutiérrez-Carrasquilla
- Endocrinology and Nutrition Department, University Hospital Arnau de Vilanova, Obesity, Diabetes and Metabolism (ODIM) research group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
| | - Mireia Dalmases
- Respiratory Department, University Hospital Arnau de Vilanova-Santa María, Translational Research in Respiratory Medicine, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Albert Lecube
- Endocrinology and Nutrition Department, University Hospital Arnau de Vilanova, Obesity, Diabetes and Metabolism (ODIM) research group, IRBLleida, University of Lleida, Lleida, Catalonia, Spain
- Centro de Investigación en Red en Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- * E-mail:
| |
Collapse
|
28
|
Cui J, Zhang F, Cao W, Wang Y, Liu J, Liu X, Chen T, Li L, Tian J, Yu B. Erythropoietin alleviates hyperglycaemia-associated inflammation by regulating macrophage polarization via the JAK2/STAT3 signalling pathway. Mol Immunol 2018; 101:221-228. [DOI: 10.1016/j.molimm.2018.05.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 12/23/2022]
|
29
|
Loesch A, Dashwood MR. Vasa vasorum inside out/outside in communication: a potential role in the patency of saphenous vein coronary artery bypass grafts. J Cell Commun Signal 2018; 12:631-643. [PMID: 30078142 PMCID: PMC6235771 DOI: 10.1007/s12079-018-0483-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 07/30/2018] [Indexed: 01/08/2023] Open
Abstract
The saphenous vein (SV) is the most commonly used conduit for revascularization in patients undergoing coronary artery bypass surgery (CABG). The patency rate of this vessel is inferior to the internal thoracic artery (ITA). In the majority of CABG procedures the ITA is removed with its outer pedicle intact whereas the (human) SV (hSV) is harvested with pedicle removed. The vasa vasorum, a microvessel network providing the adventitia and media with oxygen and nutrients, is more pronounced and penetrates deeper towards the lumen in veins than in arteries. When prepared in conventional CABG the vascular trauma caused when removing the hSV pedicle damages the vasa vasorum, a situation affecting transmural flow potentially impacting on graft performance. In patients, where the hSV is harvested with pedicle intact, the vasa vasorum is preserved and transmural blood flow restored at graft insertion and completion of CABG. By maintaining blood supply to the hSV wall, apart from oxygen and nutrients, the vasa vasorum may also transport factors potentially beneficial to graft performance. Studies, using either corrosion casts or India ink, have shown the course of vasa vasorum in animal SV as well as in hSV. In addition, there is some evidence that vasa vasorum of hSV terminate in the vessel lumen based on ex vivo perfusion, histological and ultrastructural studies. This review describes the preparation of the hSV as a bypass conduit in CABG and its performance compared with the ITA as well as how and why its patency might be improved by harvesting with minimal trauma in a way that preserves an intact vasa vasorum.
Collapse
Affiliation(s)
- Andrzej Loesch
- Centre for Rheumatology and Connective Tissue Diseases, Division of Medicine, University College London Medical School, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK.
| | - Michael R Dashwood
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London Medical School, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK.
| |
Collapse
|
30
|
Chen R, Huang Z, Wang J, Chen X, Fu Y, Wang W. Silent Information Regulator 1 Negatively Regulates Atherosclerotic Angiogenesis via Mammalian Target of Rapamycin Complex 1 Signaling Pathway. Am J Med Sci 2018; 356:168-176. [PMID: 30219159 DOI: 10.1016/j.amjms.2018.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND This study aimed to investigate the interactions between silent information regulator 1 (SIRT1) and mammalian target of rapamycin (mTOR) in intraplaque angiogenesis and their potential mechanisms through in vivo and in vitro studies. METHODS An atherosclerosis model was established in 12 rabbits on a high-cholesterol diet. The rabbits were equally divided into 3 groups: a control group (high-lipid diet), RAP group (high-lipid diet supplemented with rapamycin) and RAP + NAM group (high-lipid diet supplemented with rapamycin and nicotinamide). At the end of 4 weeks, the area of plaques in the aorta was determined and the protein expression of CD31 and vascular endothelial growth factor (VEGF) was detected through hematoxylin and eosin staining and immunohistochemical staining, respectively. For in vitro study, a hypoxia model was established in human umbilical vein endothelial cells (HUVECs) by using the chemical method (CoCl2). The MTT assay, scratch assay and tube formation assay were performed to evaluate the proliferation and angiogenesis abilities of HUVECs. Reverse transcription polymerase chain reaction was used to examine the mRNA levels of SIRT1, hypoxia-inducible factor-1α (HIF-1α), mTOR and p70 ribosomal S6 kinase (p70S6K). Western blotting was used to examine the protein levels of SIRT1, HIF-1α, mTOR, p-mTOR, p-raptor and p-p70S6K. RESULTS The results of the in vivo study indicated a significant inhibitory effect of rapamycin on plaque size and intraplaque angiogenesis (0.05 ± 0.02mm2 versus 5.44 ± 0.50mm2, P < 0.05). This effect was attenuated by nicotinamide (0.76 ± 0.15mm2 versus 0.05 ± 0.02mm2, P < 0.05). Compared with the RAP group, CD31- and VEGF-positive vessels were abundant in the RAP + NAM group. The RAP group showed lower expression of p-mTOR, p-p70S6K and HIF-1α than did the control group (P < 0.05), whereas the RAP + NAM group showed slightly higher expression of these factors than did the RAP group (P < 0.05). Furthermore, in vitro studies revealed that the inhibitory effect of rapamycin on the angiogenic ability of HUVECs and its significant inhibitory effects on the protein level of HIF-1α and the phosphorylation of proteins involved in the mTORC1 pathway, including mTOR, raptor and p70S6K (P < 0.05), were enhanced by cotreatment with SRT1720 and rapamycin (P < 0.05). In contrast to mTOR and SIRT1, the mRNA levels of p70S6K and HIF-1α were reduced by rapamycin (P < 0.05) and further reduced by cotreatment with SRT1720 and rapamycin. CONCLUSIONS The study results indicate that SIRT1 might negatively regulate atherosclerotic angiogenesis via mTORC1 and HIF-1α signaling pathway and cointervention of SIRT1 and mTOR may serve as a crucial therapeutic strategy in cardiovascular medicine.
Collapse
Affiliation(s)
- Runtai Chen
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Zhenchun Huang
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Junyi Wang
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Xiaoying Chen
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Yucai Fu
- Laboratory of Cell Senescence, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Wei Wang
- Department of Cardiology of the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China.
| |
Collapse
|
31
|
Heikal L, Ghezzi P, Mengozzi M, Ferns G. Assessment of HIF-1α expression and release following endothelial injury in-vitro and in-vivo. Mol Med 2018; 24:22. [PMID: 30134815 PMCID: PMC6016879 DOI: 10.1186/s10020-018-0026-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/07/2018] [Indexed: 11/10/2022] Open
Abstract
Background Endothelial injury is an early and enduring feature of cardiovascular disease. Inflammation and hypoxia may be responsible for this, and are often associated with the up-regulation of several transcriptional factors that include Hypoxia Inducible Factor-1 (HIF-1). Although it has been reported that HIF-1α is detectable in plasma, it is known to be unstable. Our aim was to optimize an assay for HIF-1α to be applied to in vitro and in vivo applications, and to use this assay to assess the release kinetics of HIF-1α following endothelial injury. Methods An ELISA for the measurement of HIF-1α in cell-culture medium and plasma was optimized, and the assay was used to determine the best conditions for sample collection and storage. The results of the ELISA were validated using Western blotting and immunohistochemistry (IHC). In vitro, a standardized injury was produced in a monolayer of rat aortic endothelial cells (RAECs) and intracellular HIF-1α was measured at intervals over 24 h. In vivo, a rat angioplasty model was used. The right carotid artery was injured using a 2F Fogarty balloon catheter. HIF-1α was measured in the plasma and in the arterial tissue (0, 1, 2, 3 and 5 days post injury). Results The HIF-1α ELISA had a limit of detection of 2.7 pg/mL and was linear up to 1000 pg/ mL. Between and within-assay, the coefficient of variation values were less than 15%. HIF-1α was unstable in cell lysates and plasma, and it was necessary to add a protease inhibitor immediately after collection, and to store samples at -80 °C prior to analysis. The dynamics of HIF-1α release were different for the in vitro and in vivo models. In vitro, HIF-1α reached maximum concentrations approximately 2 h post injury, whereas peak values in plasma and tissues occurred approximately 2 days post injury, in the balloon injury model. Conclusion HIF-1α can be measured in plasma, but this requires careful sample collection and storage. The carotid artery balloon injury model is associated with the transient release of HIF-1α into the circulation that probably reflects the hypoxia induced in the artery wall. Electronic supplementary material The online version of this article (10.1186/s10020-018-0026-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lamia Heikal
- Brighton and Sussex Medical School Department of Clinical and experimental investigation, University of Sussex, Falmer East Sussex, Brighton, BN1 9PS, UK
| | - Pietro Ghezzi
- Brighton and Sussex Medical School Department of Clinical and experimental investigation, University of Sussex, Falmer East Sussex, Brighton, BN1 9PS, UK
| | - Manuela Mengozzi
- Brighton and Sussex Medical School Department of Clinical and experimental investigation, University of Sussex, Falmer East Sussex, Brighton, BN1 9PS, UK
| | - Gordon Ferns
- Brighton and Sussex Medical School Department of Clinical and experimental investigation, University of Sussex, Falmer East Sussex, Brighton, BN1 9PS, UK. .,Brighton and Sussex Medical School Department of Medical Education, Mayfield House, Falmer East Sussex, Brighton, BN1 9PH, UK.
| |
Collapse
|