201
|
Henein MY, Vancheri S, Longo G, Vancheri F. The Impact of Mental Stress on Cardiovascular Health-Part II. J Clin Med 2022; 11:4405. [PMID: 35956022 PMCID: PMC9369438 DOI: 10.3390/jcm11154405 10.3390/jcm11154405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 03/27/2025] Open
Abstract
Endothelial dysfunction is one of the earliest manifestations of atherosclerosis, contributing to its development and progression. Mental stress induces endothelial dysfunction through increased activity of the sympathetic nervous system, release of corticotropin-releasing hormone from the hypothalamus, inhibition of nitric oxide (NO) synthesis by cortisol, and increased levels of pro-inflammatory cytokines. Mental-stress-induced increased output of the sympathetic nervous system and concomitant withdrawal of the parasympathetic inflammatory reflex results in systemic inflammation and activation of a neural-hematopoietic-arterial axis. This includes the brainstem and subcortical regions network, bone marrow activation, release of leukocytes into the circulation and their migration to the arterial wall and atherosclerotic plaques. Low-grade, sterile inflammation is involved in all steps of atherogenesis, from coronary plaque formation to destabilisation and rupture. Increased sympathetic tone may cause arterial smooth-muscle-cell proliferation, resulting in vascular hypertrophy, thus contributing to the development of hypertension. Emotional events also cause instability of cardiac repolarisation due to brain lateralised imbalance of cardiac autonomic nervous stimulation, which may lead to asymmetric repolarisation and arrhythmia. Acute emotional stress can also provoke severe catecholamine release, leading to direct myocyte injury due to calcium overload, known as myocytolysis, coronary microvascular vasoconstriction, and an increase in left ventricular afterload. These changes can trigger a heart failure syndrome mimicking acute myocardial infarction, characterised by transient left ventricular dysfunction and apical ballooning, known as stress (Takotsubo) cardiomyopathy. Women are more prone than men to develop mental-stress-induced myocardial ischemia (MSIMI), probably reflecting gender differences in brain activation patterns during mental stress. Although guidelines on CV prevention recognise psychosocial factors as risk modifiers to improve risk prediction and decision making, the evidence that their assessment and treatment will prevent CAD needs further evaluation.
Collapse
Affiliation(s)
- Michael Y. Henein
- Institute of Public Health and Clinical Medicine, Umea University, 90187 Umea, Sweden;
- Brunel University, Middlesex, London UB8 3PH, UK
- St. George’s University, London SW17 0RE, UK
| | - Sergio Vancheri
- Radiology Department, I.R.C.C.S. Policlinico San Matteo, 27100 Pavia, Italy;
| | - Giovanni Longo
- Cardiovascular and Interventional Department, S. Elia Hospital, 93100 Caltanissetta, Italy;
| | - Federico Vancheri
- Department of Internal Medicine, S. Elia Hospital, 93100 Caltanissetta, Italy
| |
Collapse
|
202
|
Henein MY, Vancheri S, Longo G, Vancheri F. The Impact of Mental Stress on Cardiovascular Health—Part II. J Clin Med 2022; 11:jcm11154405. [PMID: 35956022 PMCID: PMC9369438 DOI: 10.3390/jcm11154405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 12/03/2022] Open
Abstract
Endothelial dysfunction is one of the earliest manifestations of atherosclerosis, contributing to its development and progression. Mental stress induces endothelial dysfunction through increased activity of the sympathetic nervous system, release of corticotropin-releasing hormone from the hypothalamus, inhibition of nitric oxide (NO) synthesis by cortisol, and increased levels of pro-inflammatory cytokines. Mental-stress-induced increased output of the sympathetic nervous system and concomitant withdrawal of the parasympathetic inflammatory reflex results in systemic inflammation and activation of a neural–hematopoietic–arterial axis. This includes the brainstem and subcortical regions network, bone marrow activation, release of leukocytes into the circulation and their migration to the arterial wall and atherosclerotic plaques. Low-grade, sterile inflammation is involved in all steps of atherogenesis, from coronary plaque formation to destabilisation and rupture. Increased sympathetic tone may cause arterial smooth-muscle-cell proliferation, resulting in vascular hypertrophy, thus contributing to the development of hypertension. Emotional events also cause instability of cardiac repolarisation due to brain lateralised imbalance of cardiac autonomic nervous stimulation, which may lead to asymmetric repolarisation and arrhythmia. Acute emotional stress can also provoke severe catecholamine release, leading to direct myocyte injury due to calcium overload, known as myocytolysis, coronary microvascular vasoconstriction, and an increase in left ventricular afterload. These changes can trigger a heart failure syndrome mimicking acute myocardial infarction, characterised by transient left ventricular dysfunction and apical ballooning, known as stress (Takotsubo) cardiomyopathy. Women are more prone than men to develop mental-stress-induced myocardial ischemia (MSIMI), probably reflecting gender differences in brain activation patterns during mental stress. Although guidelines on CV prevention recognise psychosocial factors as risk modifiers to improve risk prediction and decision making, the evidence that their assessment and treatment will prevent CAD needs further evaluation.
Collapse
Affiliation(s)
- Michael Y. Henein
- Institute of Public Health and Clinical Medicine, Umea University, 90187 Umea, Sweden;
- Brunel University, Middlesex, London UB8 3PH, UK
- St. George’s University, London SW17 0RE, UK
| | - Sergio Vancheri
- Radiology Department, I.R.C.C.S. Policlinico San Matteo, 27100 Pavia, Italy;
| | - Giovanni Longo
- Cardiovascular and Interventional Department, S. Elia Hospital, 93100 Caltanissetta, Italy;
| | - Federico Vancheri
- Department of Internal Medicine, S. Elia Hospital, 93100 Caltanissetta, Italy
- Correspondence:
| |
Collapse
|
203
|
Kuczmarski AV, Welti LM, Moreau KL, Wenner MM. ET-1 as a Sex-Specific Mechanism Impacting Age-Related Changes in Vascular Function. FRONTIERS IN AGING 2022; 2:727416. [PMID: 35822003 PMCID: PMC9261354 DOI: 10.3389/fragi.2021.727416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/13/2021] [Indexed: 01/30/2023]
Abstract
Aging is a primary risk factor for cardiovascular disease (CVD), which is the leading cause of death in developed countries. Globally, the population of adults over the age of 60 is expected to double by the year 2050. CVD prevalence and mortality rates differ between men and women as they age in part due to sex-specific mechanisms impacting the biological processes of aging. Measures of vascular function offer key insights into cardiovascular health. Changes in vascular function precede changes in CVD prevalence rates in men and women and with aging. A key mechanism underlying these changes in vascular function is the endothelin (ET) system. Studies have demonstrated sex and sex hormone effects on endothelin-1 (ET-1), and its receptors ETA and ETB. However, with aging there is a dysregulation of this system resulting in an imbalance between vasodilation and vasoconstriction. Thus, ET-1 may play a role in the sex differences observed with vascular aging. While most research has been conducted in pre-clinical animal models, we describe more recent translational data in humans showing that the ET system is an important regulator of vascular dysfunction with aging and acts through sex-specific ET receptor mechanisms. In this review, we present translational evidence (cell, tissue, animal, and human) that the ET system is a key mechanism regulating sex-specific changes in vascular function with aging, along with therapeutic interventions to reduce ET-mediated vascular dysfunction associated with aging. More knowledge on the factors responsible for the sex differences with vascular aging allow for optimized therapeutic strategies to attenuate CVD risk in the expanding aging population.
Collapse
Affiliation(s)
- Andrew V Kuczmarski
- University of Delaware, Kinesiology and Applied Physiology, Newark, DE, United States
| | - Laura M Welti
- University of Delaware, Kinesiology and Applied Physiology, Newark, DE, United States
| | - Kerrie L Moreau
- University of Colorado, Anschutz Medical Campus, Aurora, CO, United States.,Denver Veterans Administrative Medical Center, Geriatric Research Education and Clinical Center, Aurora, CO, United States
| | - Megan M Wenner
- University of Delaware, Kinesiology and Applied Physiology, Newark, DE, United States
| |
Collapse
|
204
|
Jurko T, Mestanik M, Mestanikova A, Zeleňák K, Jurko A. Early Signs of Microvascular Endothelial Dysfunction in Adolescents with Newly Diagnosed Essential Hypertension. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071048. [PMID: 35888136 PMCID: PMC9321176 DOI: 10.3390/life12071048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022]
Abstract
Endothelial dysfunction represents one of the key pathomechanisms in many diseases, including hypertension. Peripheral arterial tonometry (PAT) evaluates the functional status of microvascular endothelium and offers a biomarker of early, potentially reversible, vascular damage. This study aimed to assess endothelial function using conventional and novel indices of PAT in pediatric hypertensives. As such, 100 adolescents with normal blood pressure, and essential and white-coat hypertension were examined using EndoPAT 2000. Conventional reactive hyperemia index (RHI) and novel indices of hyperemic response, including the area under the curve of hyperemic response (AUC), were evaluated. AUC was the only parameter sensitive to the effect of hypertension, with significantly lower values in essential hypertensives compared to normotensives and white-coat hypertensives (p = 0.024, p = 0.032, respectively). AUC was the only parameter significantly correlating with mean ambulatory monitored blood pressure (r = −0.231, p = 0.021). AUC showed a significant negative association with age (p = 0.039), but a significant positive association with pubertal status indexed by plasma levels of dehydroepiandrosterone (p = 0.027). This is the first study reporting early signs of microvascular endothelial dysfunction evaluated using PAT in adolescents with newly diagnosed essential hypertension. Detailed analysis of hyperemic response using overall magnitude indexed by AUC provided a more robust method compared to the conventional evaluation of RHI.
Collapse
Affiliation(s)
- Tomas Jurko
- Clinic of Neonatology, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, University Hospital Martin, Kollarova 2, 03659 Martin, Slovakia;
| | - Michal Mestanik
- Pediatric Cardiology Clinic, Kollarova 13, 03601 Martin, Slovakia; (M.M.); (A.M.)
| | - Andrea Mestanikova
- Pediatric Cardiology Clinic, Kollarova 13, 03601 Martin, Slovakia; (M.M.); (A.M.)
| | - Kamil Zeleňák
- Clinic of Radiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Kollarova 2, 03659 Martin, Slovakia;
| | - Alexander Jurko
- Pediatric Cardiology Clinic, Kollarova 13, 03601 Martin, Slovakia; (M.M.); (A.M.)
- Correspondence:
| |
Collapse
|
205
|
Jamialahmadi T, Alidadi M, Atkin SL, Kroh M, Almahmeed W, Moallem SA, Al-Rasadi K, Rodriguez JH, Santos RD, Ruscica M, Sahebkar A. Effect of Bariatric Surgery on Flow-Mediated Vasodilation as a Measure of Endothelial Function: A Systematic Review and Meta-Analysis. J Clin Med 2022; 11:4054. [PMID: 35887817 PMCID: PMC9323618 DOI: 10.3390/jcm11144054] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/20/2022] [Accepted: 06/23/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives. Flow mediated vasodilation (FMD) is a marker of endothelial function and its decline is related to increased cardiovascular risk. This systematic review and meta-analysis evaluated the impact of bariatric surgery on FMD. Materials and methods. A systematic literature search in PubMed, Scopus, Embase, and Web of Science was performed to 1 May 2021. Meta-analysis was performed using Comprehensive Meta-Analysis (CMA) V2 software. All types of bariatric surgery were considered, with the inclusion that FMD had to have been tested before and after the surgical procedure. Meta-analysis was carried out using a random-effects model and the generic inverse variance approach. The leave-one-out approach was used for sensitivity analysis. To assess metabolic parameter confounders, a weighted random-effects meta-regression was used. Results. A meta-analysis and a systematic review of 23 studies (n = 891 individuals) demonstrated improvement in FMD following bariatric surgery (weighted mean difference (WMD): 5.867, 95% CI: 4.481, 7.252, p < 0.001; I2: 96.70). Iteratively removing each item in the meta-analysis did not result in a significant alteration in the pooled estimate of effect size. There was an improvement in FMD for up to 6 months following bariatric surgery in a meta-analysis from 7 trials that included 356 subjects (WMD: 5.248, 95% CI: 2.361, 8.135, p < 0.001; I2: 98.18). The meta-analysis from 9 trials (n = 414 subjects) showed an improvement in FMD 6 to 12 months after bariatric surgery (WMD: 5.451, 95% CI: 3.316, 7.587, p < 0.001; I2: 94.18). The meta-analysis from 10 trials (n = 414 subjects) demonstrated an improvement in FMD 12 months after bariatric surgery (WMD: 2.401, 95% CI: 0.944, 3.859, p = 0.001; I2: 88.35). Random-effects meta-regression did not show any association between the alteration in FMD and percent body mass index (BMI) change (slope: 0.0258; 95% CI: −0.323, 0.375; p = 0.884), or changes in blood pressure; however, there was an association between the changes in FMD and the duration of follow-up (slope: −0.106; 95% CI: −0.205, −0.008; p = 0.033) with greater changes in FMD after 12 months. Conclusions. Bariatric surgery significantly improved FMD that increased with time, and the resultant improvement in endothelial function was independent of weight loss or a reduction in blood pressure.
Collapse
Affiliation(s)
- Tannaz Jamialahmadi
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran;
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran;
| | - Mona Alidadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran;
| | - Stephen L. Atkin
- School of Postgraduate Studies and Research, RCSI Medical University of Bahrain, Busaiteen 15503, Bahrain;
| | - Matthew Kroh
- Digestive Disease and Surgery Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA;
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi P.O. Box 124140, United Arab Emirates;
| | - Seyed Adel Moallem
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq;
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalid Al-Rasadi
- Medical Research Centre, Sultan Qaboos University, Muscat P.O. Box 373, Oman;
| | - John H. Rodriguez
- Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi P.O. Box 112412, United Arab Emirates;
| | - Raul D. Santos
- Lipid Clinic Heart Institute (Incor), University of São Paulo, Medical School Hospital, São Paulo 01000, Brazil;
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20100 Milan, Italy;
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
206
|
Krajina I, Stupin A, Šola M, Mihalj M. Oxidative Stress Induced by High Salt Diet—Possible Implications for Development and Clinical Manifestation of Cutaneous Inflammation and Endothelial Dysfunction in Psoriasis vulgaris. Antioxidants (Basel) 2022; 11:antiox11071269. [PMID: 35883760 PMCID: PMC9311978 DOI: 10.3390/antiox11071269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/19/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Although oxidative stress is recognized as an important effector mechanism of the immune system, uncontrolled formation of reactive oxygen and nitrogen species promotes excessive tissue damage and leads to disease development. In view of this, increased dietary salt intake has been found to damage redox systems in the vessel wall, resulting in endothelial dysfunction associated with NO uncoupling, inflammation, vascular wall remodeling and, eventually, atherosclerosis. Several studies have reported increased systemic oxidative stress accompanied by reduced antioxidant capacity following a high salt diet. In addition, vigorous ionic effects on the immune mechanisms, such as (trans)differentiation of T lymphocytes are emerging, which together with the evidence of NaCl accumulation in certain tissues warrants a re-examination of the data derived from in vitro research, in which the ionic influence was excluded. Psoriasis vulgaris (PV), as a primarily Th17-driven inflammatory skin disease with proven inflammation-induced accumulation of sodium chloride in the skin, merits our interest in the role of oxidative stress in the pathogenesis of PV, as well as in the possible beneficial effects that could be achieved through modulation of dietary salt intake and antioxidant supplementation.
Collapse
Affiliation(s)
- Ivana Krajina
- Department of Dermatology and Venereology, Osijek University Hospital, J. Huttlera 4, HR-31000 Osijek, Croatia;
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Ana Stupin
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia;
- Institute and Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Marija Šola
- Department of Dermatology and Venereology, Osijek University Hospital, J. Huttlera 4, HR-31000 Osijek, Croatia;
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
- Correspondence: (M.Š.); (M.M.); Tel.: +385-31-512-800 (M.M.)
| | - Martina Mihalj
- Department of Dermatology and Venereology, Osijek University Hospital, J. Huttlera 4, HR-31000 Osijek, Croatia;
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia;
- Institute and Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
- Correspondence: (M.Š.); (M.M.); Tel.: +385-31-512-800 (M.M.)
| |
Collapse
|
207
|
Non-Lipid Effects of PCSK9 Monoclonal Antibodies on Vessel Wall. J Clin Med 2022; 11:jcm11133625. [PMID: 35806908 PMCID: PMC9267174 DOI: 10.3390/jcm11133625] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 12/07/2022] Open
Abstract
Elevated low density lipoprotein (LDL) cholesterol and lipoprotein(a) (Lp(a)) levels have an important role in the development and progression of atherosclerosis, followed by cardiovascular events. Besides statins and other lipid-modifying drugs, PCSK9 monoclonal antibodies are known to reduce hyperlipidemia. PCSK9 monoclonal antibodies decrease LDL cholesterol levels through inducing the upregulation of the LDL receptors and moderately decrease Lp(a) levels. In addition, PCSK9 monoclonal antibodies have shown non-lipid effects. PCSK9 monoclonal antibodies reduce platelet aggregation and activation, and increase platelet responsiveness to acetylsalicylic acid. Evolocumab as well as alirocumab decrease an incidence of venous thromboembolism, which is associated with the decrease of Lp(a) values. Besides interweaving in haemostasis, PCSK9 monoclonal antibodies play an important role in reducing the inflammation and improving the endothelial function. The aim of this review is to present the mechanisms of PCSK9 monoclonal antibodies on the aforementioned risk factors.
Collapse
|
208
|
Ardakani MV, Parviz S, Ghadimi E, Zamani Z, Salehi M, Firoozabadi MA, Mortazavi SMJ. Concomitant septic arthritis of the hip joint and femoral head avascular necrosis in patients with recent COVID-19 infection: a cautionary report. J Orthop Surg Res 2022; 17:302. [PMID: 35668523 PMCID: PMC9168355 DOI: 10.1186/s13018-022-03192-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
Purpose At present, concomitant avascular necrosis (AVN) of femoral head and septic arthritis (SA) as a sequel of COVID-19 infection has yet not been documented. By large-scale use of life-saving corticosteroids (CS) in COVID-19 cases, our aim is to warn of the occurrence of hip joint infection in these patients.
Methods We report a series of five cases in which patients developed septic arthritis concomitant with AVN after being treated for COVID-19 infection. The mean dose of prednisolone used in these cases was 1695.2 mg. The time period of onset of hip symptoms in our cases from the beginning of the COVID-19 infection was 56 days in the first case, 43 days in the second case, 30 days in the third case, 29 days in the fourth case and 50 days in the last case, with an average time of 41.6 days. All patients underwent surgery depending on the extent of articular cartilage damage by direct anterior approach. Results Clinical and laboratory symptoms improved significantly in all patients. The mean visual analogue pain score of the patients decreased from 9.4 (9–10) before surgery to 2.8 (1–4) after 1 week of operation. Conclusion In any patient with the history of COVID-19 infection specially those who have been treated with corticosteroid as one of the medications prescribed during the disease, any joint symptom specially in the hips should draw our attention to the joint infection, and with timely diagnosis and surgery, their hip joint can be saved.
Collapse
Affiliation(s)
- Mohammad Vahedian Ardakani
- Joint Reconstruction Research Centre, Tehran University of Medical Sciences, End of Keshavarz Blvd, Tehran, 1419733141, Iran
| | - Sara Parviz
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Department of Radiology, Medical Imaging Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Ghadimi
- Joint Reconstruction Research Centre, Tehran University of Medical Sciences, End of Keshavarz Blvd, Tehran, 1419733141, Iran
| | - Zahra Zamani
- Community Medicine Department, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Salehi
- Imam Khomeini Hospital Complex, Infectious Diseases Department, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ayati Firoozabadi
- Joint Reconstruction Research Centre, Tehran University of Medical Sciences, End of Keshavarz Blvd, Tehran, 1419733141, Iran
| | - S M Javad Mortazavi
- Joint Reconstruction Research Centre, Tehran University of Medical Sciences, End of Keshavarz Blvd, Tehran, 1419733141, Iran.
| |
Collapse
|
209
|
Galatou E, Mourelatou E, Hatziantoniou S, Vizirianakis IS. Nonalcoholic Steatohepatitis (NASH) and Atherosclerosis: Explaining Their Pathophysiology, Association and the Role of Incretin-Based Drugs. Antioxidants (Basel) 2022; 11:1060. [PMID: 35739957 PMCID: PMC9220192 DOI: 10.3390/antiox11061060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/17/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the most severe manifestation of nonalcoholic fatty liver disease (NAFLD), a common complication of type 2 diabetes, and may lead to cirrhosis and hepatocellular carcinoma. Oxidative stress and liver cell damage are the major triggers of the severe hepatic inflammation that characterizes NASH, which is highly correlated with atherosclerosis and coronary artery disease. Regarding drug therapy, research on the role of GLP-1 analogues and DPP4 inhibitors, novel classes of antidiabetic drugs, is growing. In this review, we outline the association between NASH and atherosclerosis, the underlying molecular mechanisms, and the effects of incretin-based drugs, especially GLP-1 RAs, for the therapeutic management of these conditions.
Collapse
Affiliation(s)
- Eleftheria Galatou
- Department of Life & Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417 Nicosia, Cyprus;
| | - Elena Mourelatou
- Department of Life & Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417 Nicosia, Cyprus;
| | - Sophia Hatziantoniou
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece;
| | - Ioannis S. Vizirianakis
- Department of Life & Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417 Nicosia, Cyprus;
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
210
|
Tong KL, Tan KE, Lim YY, Tien XY, Wong PF. CircRNA-miRNA interactions in atherogenesis. Mol Cell Biochem 2022; 477:2703-2733. [PMID: 35604519 DOI: 10.1007/s11010-022-04455-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/27/2022] [Indexed: 11/30/2022]
Abstract
Atherosclerosis is the major cause of coronary artery disease (CAD) which includes unstable angina, myocardial infarction, and heart failure. The onset of atherogenesis, a process of atherosclerotic lesion formation in the intima of arteries, is driven by lipid accumulation, a vicious cycle of reactive oxygen species (ROS)-induced oxidative stress and inflammatory reactions leading to endothelial cell (EC) dysfunction, vascular smooth muscle cell (VSMC) activation, and foam cell formation which further fuel plaque formation and destabilization. In recent years, there is a surge in the number of publications reporting the involvement of circular RNAs (circRNAs) in the pathogenesis of cardiovascular diseases, cancers, and metabolic syndromes. These studies have advanced our understanding on the biological functions of circRNAs. One of the most common mechanism of action of circRNAs reported is the sponging of microRNAs (miRNAs) by binding to the miRNAs response element (MRE), thereby indirectly increases the transcription of their target messenger RNAs (mRNAs). Individual networks of circRNA-miRNA-mRNA associated with atherogenesis have been extensively reported, however, there is a need to connect these findings for a complete overview. This review aims to provide an update on atherogenesis-related circRNAs and analyze the circRNA-miRNA-mRNA interactions in atherogenesis. The atherogenic mechanisms and clinical relevance of each atherogenesis-related circRNA were systematically discussed for better understanding of the knowledge gap in this area.
Collapse
Affiliation(s)
- Kind-Leng Tong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ke-En Tan
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yat-Yuen Lim
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Xin-Yi Tien
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
211
|
Endothelial Dysfunction Induced by Extracellular Neutrophil Traps Plays Important Role in the Occurrence and Treatment of Extracellular Neutrophil Traps-Related Disease. Int J Mol Sci 2022; 23:ijms23105626. [PMID: 35628437 PMCID: PMC9147606 DOI: 10.3390/ijms23105626] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 01/27/2023] Open
Abstract
Many articles have demonstrated that extracellular neutrophil traps (NETs) are often described as part of the antibacterial function. However, since the components of NETs are non-specific, excessive NETs usually cause inflammation and tissue damage. Endothelial dysfunction (ED) caused by NETs is the major focus of tissue damage, which is highly related to many inflammatory diseases. Therefore, this review summarizes the latest advances in the primary and secondary mechanisms between NETs and ED regarding inflammation as a mediator. Moreover, the detailed molecular mechanisms with emphasis on the disadvantages from NETs are elaborated: NETs can use its own enzymes, release particles as damage-associated molecular patterns (DAMPs) and activate the complement system to interact with endothelial cells (ECs), drive ECs damage and eventually aggravate inflammation. In view of the role of NETs-induced ED in different diseases, we also discussed possible molecular mechanisms and the treatments of NETs-related diseases.
Collapse
|
212
|
Khanna NN, Maindarkar M, Saxena A, Ahluwalia P, Paul S, Srivastava SK, Cuadrado-Godia E, Sharma A, Omerzu T, Saba L, Mavrogeni S, Turk M, Laird JR, Kitas GD, Fatemi M, Barqawi AB, Miner M, Singh IM, Johri A, Kalra MM, Agarwal V, Paraskevas KI, Teji JS, Fouda MM, Pareek G, Suri JS. Cardiovascular/Stroke Risk Assessment in Patients with Erectile Dysfunction-A Role of Carotid Wall Arterial Imaging and Plaque Tissue Characterization Using Artificial Intelligence Paradigm: A Narrative Review. Diagnostics (Basel) 2022; 12:1249. [PMID: 35626404 PMCID: PMC9141739 DOI: 10.3390/diagnostics12051249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The role of erectile dysfunction (ED) has recently shown an association with the risk of stroke and coronary heart disease (CHD) via the atherosclerotic pathway. Cardiovascular disease (CVD)/stroke risk has been widely understood with the help of carotid artery disease (CTAD), a surrogate biomarker for CHD. The proposed study emphasizes artificial intelligence-based frameworks such as machine learning (ML) and deep learning (DL) that can accurately predict the severity of CVD/stroke risk using carotid wall arterial imaging in ED patients. METHODS Using the PRISMA model, 231 of the best studies were selected. The proposed study mainly consists of two components: (i) the pathophysiology of ED and its link with coronary artery disease (COAD) and CHD in the ED framework and (ii) the ultrasonic-image morphological changes in the carotid arterial walls by quantifying the wall parameters and the characterization of the wall tissue by adapting the ML/DL-based methods, both for the prediction of the severity of CVD risk. The proposed study analyzes the hypothesis that ML/DL can lead to an accurate and early diagnosis of the CVD/stroke risk in ED patients. Our finding suggests that the routine ED patient practice can be amended for ML/DL-based CVD/stroke risk assessment using carotid wall arterial imaging leading to fast, reliable, and accurate CVD/stroke risk stratification. SUMMARY We conclude that ML and DL methods are very powerful tools for the characterization of CVD/stroke in patients with varying ED conditions. We anticipate a rapid growth of these tools for early and better CVD/stroke risk management in ED patients.
Collapse
Affiliation(s)
- Narendra N. Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi 110076, India;
| | - Mahesh Maindarkar
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India; (M.M.); (S.P.)
- Stroke Monitoring and Diagnostic Division, AtheroPoint, Roseville, CA 95661, USA;
| | - Ajit Saxena
- Department of Urology, Indraprastha APOLLO Hospitals, New Delhi 110076, India;
| | - Puneet Ahluwalia
- Max Institute of Cancer Care, Max Super Specialty Hospital, New Delhi 110017, India;
| | - Sudip Paul
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India; (M.M.); (S.P.)
| | - Saurabh K. Srivastava
- College of Computing Sciences and IT, Teerthanker Mahaveer University, Moradabad 244001, India;
| | - Elisa Cuadrado-Godia
- Department of Neurology, Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain;
| | - Aditya Sharma
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22908, USA;
| | - Tomaz Omerzu
- Department of Neurology, University Medical Centre Maribor, 2000 Maribor, Slovenia; (T.O.); (M.T.)
| | - Luca Saba
- Department of Radiology, University of Cagliari, 09124 Cagliari, Italy;
| | - Sophie Mavrogeni
- Cardiology Clinic, Onassis Cardiac Surgery Centre, 176 74 Athens, Greece;
| | - Monika Turk
- Department of Neurology, University Medical Centre Maribor, 2000 Maribor, Slovenia; (T.O.); (M.T.)
| | - John R. Laird
- Heart and Vascular Institute, Adventist Health St. Helena, St. Helena, CA 94574, USA;
| | - George D. Kitas
- Academic Affairs, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, UK;
- Arthritis Research UK Epidemiology Unit, Manchester University, Manchester M13 9PL, UK
| | - Mostafa Fatemi
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, NY 55905, USA;
| | - Al Baha Barqawi
- Division of Urology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Martin Miner
- Men’s Health Centre, Miriam Hospital Providence, Providence, RI 02906, USA;
| | - Inder M. Singh
- Stroke Monitoring and Diagnostic Division, AtheroPoint, Roseville, CA 95661, USA;
| | - Amer Johri
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | | | - Vikas Agarwal
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India;
| | - Kosmas I. Paraskevas
- Department of Vascular Surgery, Central Clinic of Athens, 106 80 Athens, Greece;
| | - Jagjit S. Teji
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA;
| | - Mostafa M. Fouda
- Department of Electrical and Computer Engineering, Idaho State University, Pocatello, ID 83209, USA;
| | - Gyan Pareek
- Minimally Invasive Urology Institute, Brown University, Providence, RI 02912, USA;
| | - Jasjit S. Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint, Roseville, CA 95661, USA;
| |
Collapse
|
213
|
Dindas F, Koyuncu I, Ocek L, Ozdemir AV, Yılmaz H, Abacıoglu OO, Yıldırım A, Yenercag M, Dogdus M. Association of serum elabela levels with carotid artery stenosis in patients with non-cardioembolic ischemic stroke. Biomark Med 2022; 16:623-631. [PMID: 35549392 DOI: 10.2217/bmm-2021-0865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Elabela (ELA) is a peptide of the apelinergic system and is known to play a role in endothelial homeostasis and vascular pathobiology. In this study, the relationship between carotid artery stenosis, which is the main culprit, and ELA level in patients with non-cardioembolic ischemic stroke was investigated. Materials & methods: Cross-sectional observation included two groups of 40 patients with critical carotid artery stenosis and 40 patients with age-sex matched noncritical carotid artery stenosis. Results: ELA levels were significantly higher in the noncritical stenosis group. ELA had a significantly moderate negative correlation with the carotid score (r = -0.334, p = 0.003), maximal carotid plaque length (r = -0.413, p < 0.001) and degree of stenosis (r = -0.397, p < 0.001). Conclusions: There is a significant inverse correlation between critical carotid artery disease and ELA level in patients with non-cardioembolic ischemic stroke.
Collapse
Affiliation(s)
- Ferhat Dindas
- Department of Cardiology, Usak University, Training & Research Hospital, Usak, 64000, Turkey
| | - Ilhan Koyuncu
- Department of Cardiology, Usak University, Training & Research Hospital, Usak, 64000, Turkey
| | - Levent Ocek
- Department of Neurology, University of Health Sciences Izmir Tepecik Training & Research Hospital, Izmir, 35460, Turkey
| | - Ali V Ozdemir
- Department of Biochemistry, Usak University, Training & Research Hospital, Usak, 64000, Turkey
| | - Hakan Yılmaz
- Department of Radiology, Usak University, Training & Research Hospital, Usak, 64000, Turkey
| | - Ozge O Abacıoglu
- University of Health Sciences, Adana Health Practice & Research Center, Adana, 01000, Turkey
| | - Arafat Yıldırım
- University of Health Sciences, Adana Health Practice & Research Center, Adana, 01000, Turkey
| | - Mustafa Yenercag
- Department of Cardiology, Ordu University, Training & Research Hospital, Ordu, 52200, Turkey
| | - Mustafa Dogdus
- Department of Cardiology, Usak University, Training & Research Hospital, Usak, 64000, Turkey
| |
Collapse
|
214
|
Sara JDS, Toya T, Ahmad A, Clark MM, Gilliam WP, Lerman LO, Lerman A. Mental Stress and Its Effects on Vascular Health. Mayo Clin Proc 2022; 97:951-990. [PMID: 35512885 PMCID: PMC9058928 DOI: 10.1016/j.mayocp.2022.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 01/13/2023]
Abstract
Coronary artery disease continues to be a major cause of morbidity and mortality despite significant advances in risk stratification and management. This has prompted the search for alternative nonconventional risk factors that may provide novel therapeutic targets. Psychosocial stress, or mental stress, has emerged as an important risk factor implicated in a higher incidence of cardiovascular events, and although our understanding of this far ranging and interesting phenomenon has developed greatly over recent times, there is still much to be learned regarding how to measure mental stress and how it may impact physical health. With the current coronavirus disease 2019 global pandemic and its incumbent lockdowns and social distancing, understanding the potentially harmful biological effects of stress related to life-changing events and social isolation has become even more important. In the current review our multidisciplinary team discusses stress from a psychosocial perspective and aims to define psychological stress as rigorously as possible; discuss the pathophysiologic mechanisms by which stress may mediate cardiovascular disease, with a particular focus to its effects on vascular health; outline existing methods and approaches to quantify stress by means of a vascular biomarker; outline the mechanisms whereby psychosocial stressors may have their pathologic effects ultimately transduced to the vasculature through the neuroendocrine immunologic axis; highlight areas for improvement to refine existing approaches in clinical research when studying the consequences of psychological stress on cardiovascular health; and discuss evidence-based therapies directed at reducing the deleterious effects of mental stress including those that target endothelial dysfunction. To this end we searched PubMed and Google Scholar to identify studies evaluating the relationship between mental or psychosocial stress and cardiovascular disease with a particular focus on vascular health. Search terms included "myocardial ischemia," "coronary artery disease," "mental stress," "psychological stress," "mental∗ stress∗," "psychologic∗ stress∗," and "cardiovascular disease∗." The search was limited to studies published in English in peer-reviewed journals between 1990 and the present day. To identify potential studies not captured by our database search strategy, we also searched studies listed in the bibliography of relevant publications and reviews.
Collapse
Key Words
- cad, coronary artery disease
- cbt, cognitive behavioral therapy
- cvd, cardiovascular disease
- fmd, flow-mediated dilatation
- il, interleukin
- mi, myocardial infarction
- ms, mental stress
- msimi, mental stress induced myocardial ischemia
- pat, peripheral arterial tonometry
- ped, peripheral endothelial dysfunction
- pet, positron emission tomography
- rh, reactive hyperemia
- ses, socioeconomic status
- tnf, tumor necrosis factor
- vsmc, vascular smooth muscle cells
Collapse
Affiliation(s)
| | - Takumi Toya
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Ali Ahmad
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Matthew M Clark
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Wesley P Gilliam
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Lliach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
215
|
Benincasa G, Coscioni E, Napoli C. Cardiovascular risk factors and molecular routes underlying endothelial dysfunction: Novel opportunities for primary prevention. Biochem Pharmacol 2022; 202:115108. [DOI: 10.1016/j.bcp.2022.115108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/23/2022]
|
216
|
Markousis-Mavrogenis G, Bacopoulou F, Mavragani C, Voulgari P, Kolovou G, Kitas GD, Chrousos GP, Mavrogeni SI. Coronary microvascular disease: The "Meeting Point" of Cardiology, Rheumatology and Endocrinology. Eur J Clin Invest 2022; 52:e13737. [PMID: 34939183 DOI: 10.1111/eci.13737] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Exertional chest pain/dyspnea or chest pain at rest are the main symptoms of coronary artery disease (CAD), which are traditionally attributed to insufficiency of the epicardial coronary arteries. However, 2/3 of women and 1/3 of men with angina and 10% of patients with acute myocardial infarction have no evidence of epicardial coronary artery stenosis in X-ray coronary angiography. In these cases, coronary microvascular disease (CMD) is the main causative factor. AIMS To present the pathophysiology of CMD in Cardiology, Rheumatology and Endocrinology. MATERIALS-METHODS The pathophysiology of CMD in Cardiology, Rheumatology and Endocrinology was evaluated. It includes impaired microvascular vasodilatation, which leads to inability of the organism to deal with myocardial oxygen needs and, hence, development of ischemic pain. CMD, observed in inflammatory autoimmune rheumatic and endocrine/metabolic disorders, brings together Cardiology, Rheumatology and Endocrinology. Causative factors include persistent systemic inflammation and endocrine/metabolic abnormalities influencing directly the coronary microvasculature. In the past, the evaluation of microcirculation was feasible only with the use of invasive techniques, such as coronary flow reserve assessment. Currently, the application of advanced imaging modalities, such as cardiovascular magnetic resonance (CMR), can evaluate CMD non-invasively and without ionizing radiation. RESULTS CMD may present with a variety of symptoms with 1/3 to 2/3 of them expressed as typical chest pain in effort, more commonly found in women during menopause than in men. Atypical presentation includes chest pain at rest or exertional dyspnea,but post exercise symptoms are not uncommon. The treatment with nitrates is less effective in CMD, because their vasodilator action in coronary micro-circulation is less pronounced than in the epicardial coronary arteries. DISCUSSION Although both classic and new medications have been used in the treatment of CMD, there are still many questions regarding both the pathophysiology and the treatment of this disorder. The potential effects of anti-rheumatic and endocrine medications on the evolution of CMD need further evaluation. CONCLUSION CMD is a multifactorial disease leading to myocardial ischemia/fibrosis alone or in combination with epicardial coronary artery disease. Endothelial dysfunction/vasospasm, systemic inflammation, and/or neuroendocrine activation may act as causative factors and bring Cardiology, Rheumatology and Endocrinology together. Currently, the application of advanced imaging modalities, and specifically CMR, allows reliable assessment of the extent and severity of CMD. These measurements should not be limited to "pure cardiac patients", as it is known that CMD affects the majority of patients with autoimmune rheumatic and endocrine/metabolic disorders.
Collapse
Affiliation(s)
| | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health and Precision Medicine, UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | - Clio Mavragani
- Pathophysiology Department, University of Athens, Athens, Greece
| | | | - Genovefa Kolovou
- Onassis Cardiac Surgery Hospital, Athens, Greece.,Epidemiology Department, University of Manchester, Manchester, UK
| | - George D Kitas
- Epidemiology Department, University of Manchester, Manchester, UK
| | - George P Chrousos
- University Research Institute of Maternal and Child Health and Precision Medicine, UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | | |
Collapse
|
217
|
Signaling cascades in the failing heart and emerging therapeutic strategies. Signal Transduct Target Ther 2022; 7:134. [PMID: 35461308 PMCID: PMC9035186 DOI: 10.1038/s41392-022-00972-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic heart failure is the end stage of cardiac diseases. With a high prevalence and a high mortality rate worldwide, chronic heart failure is one of the heaviest health-related burdens. In addition to the standard neurohormonal blockade therapy, several medications have been developed for chronic heart failure treatment, but the population-wide improvement in chronic heart failure prognosis over time has been modest, and novel therapies are still needed. Mechanistic discovery and technical innovation are powerful driving forces for therapeutic development. On the one hand, the past decades have witnessed great progress in understanding the mechanism of chronic heart failure. It is now known that chronic heart failure is not only a matter involving cardiomyocytes. Instead, chronic heart failure involves numerous signaling pathways in noncardiomyocytes, including fibroblasts, immune cells, vascular cells, and lymphatic endothelial cells, and crosstalk among these cells. The complex regulatory network includes protein-protein, protein-RNA, and RNA-RNA interactions. These achievements in mechanistic studies provide novel insights for future therapeutic targets. On the other hand, with the development of modern biological techniques, targeting a protein pharmacologically is no longer the sole option for treating chronic heart failure. Gene therapy can directly manipulate the expression level of genes; gene editing techniques provide hope for curing hereditary cardiomyopathy; cell therapy aims to replace dysfunctional cardiomyocytes; and xenotransplantation may solve the problem of donor heart shortages. In this paper, we reviewed these two aspects in the field of failing heart signaling cascades and emerging therapeutic strategies based on modern biological techniques.
Collapse
|
218
|
Slaninova N, Bryjova I, Lasota Z, Richterova R, Kubicek J, Augustynek M, Seal A, Krejcar O, Proto A. Thrombotic and Atherogenetic Predisposition in Polyglobulic Donors. Biomedicines 2022; 10:biomedicines10040888. [PMID: 35453637 PMCID: PMC9027744 DOI: 10.3390/biomedicines10040888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 02/04/2023] Open
Abstract
This work analyses the results of research regarding the predisposition of genetic hematological risks associated with secondary polyglobulia. The subjects of the study were selected based on shared laboratory markers and basic clinical symptoms. JAK2 (Janus Kinase 2) mutation negativity represented the common genetic marker of the subjects in the sample of interest. A negative JAK2 mutation hypothetically excluded the presence of an autonomous myeloproliferative disease at the time of detection. The parameters studied in this work focused mainly on thrombotic, immunological, metabolic, and cardiovascular risks. The final goal of the work was to discover the most significant key markers for the diagnosis of high-risk patients and to exclude the less important or only complementary markers, which often represent a superfluous economic burden for healthcare institutions. These research results are applicable as a clinical guideline for the effective diagnosis of selected parameters that demonstrated high sensitivity and specificity. According to the results obtained in the present research, groups with a high incidence of mutations were evaluated as being at higher risk for polycythemia vera disease. It was not possible to clearly determine which of the patients examined had a higher risk of developing the disease as different combinations of mutations could manifest different symptoms of the disease. In general, the entire study group was at risk for manifestations of polycythemia vera disease without a clear diagnosis. The group with less than 20% incidence appeared to be clinically insignificant for polycythemia vera testing and thus there is a potential for saving money in mutation testing. On the other hand, the JAK V617F (somatic mutation of JAK2) parameter from this group should be investigated as it is a clear exclusion or confirmation of polycythemia vera as the primary disease.
Collapse
Affiliation(s)
- Nikola Slaninova
- Department of Cybernetics and Biomedical Engineering, VŠB—Technical University of Ostrava, 17. listopadu 2172/15, 708 00 Ostrava–Poruba, Czech Republic; (N.S.); (I.B.); (M.A.); (A.P.)
| | - Iveta Bryjova
- Department of Cybernetics and Biomedical Engineering, VŠB—Technical University of Ostrava, 17. listopadu 2172/15, 708 00 Ostrava–Poruba, Czech Republic; (N.S.); (I.B.); (M.A.); (A.P.)
| | - Zenon Lasota
- Blood Donor Center, tr. T. G. Masaryka 495, 738 01 Frydek-Mistek, Czech Republic; (Z.L.); (R.R.)
| | - Radmila Richterova
- Blood Donor Center, tr. T. G. Masaryka 495, 738 01 Frydek-Mistek, Czech Republic; (Z.L.); (R.R.)
| | - Jan Kubicek
- Department of Cybernetics and Biomedical Engineering, VŠB—Technical University of Ostrava, 17. listopadu 2172/15, 708 00 Ostrava–Poruba, Czech Republic; (N.S.); (I.B.); (M.A.); (A.P.)
- Correspondence:
| | - Martin Augustynek
- Department of Cybernetics and Biomedical Engineering, VŠB—Technical University of Ostrava, 17. listopadu 2172/15, 708 00 Ostrava–Poruba, Czech Republic; (N.S.); (I.B.); (M.A.); (A.P.)
| | - Ayan Seal
- Department of Computer Science & Engineering, PDPM Indian Institute of Information Technology, Design and Manufacturing, Jabalpur 482005, India;
- Center for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kralove, Hradecka 1249, 500 03 Hradec Kralove, Czech Republic;
| | - Ondrej Krejcar
- Center for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kralove, Hradecka 1249, 500 03 Hradec Kralove, Czech Republic;
| | - Antonino Proto
- Department of Cybernetics and Biomedical Engineering, VŠB—Technical University of Ostrava, 17. listopadu 2172/15, 708 00 Ostrava–Poruba, Czech Republic; (N.S.); (I.B.); (M.A.); (A.P.)
| |
Collapse
|
219
|
Rapid shear stress-dependent ENaC membrane insertion is mediated by the endothelial glycocalyx and the mineralocorticoid receptor. Cell Mol Life Sci 2022; 79:235. [PMID: 35397686 PMCID: PMC8995297 DOI: 10.1007/s00018-022-04260-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/02/2022] [Accepted: 03/18/2022] [Indexed: 02/08/2023]
Abstract
The contribution of the shear stress-sensitive epithelial Na+ channel (ENaC) to the mechanical properties of the endothelial cell surface under (patho)physiological conditions is unclear. This issue was addressed in in vivo and in vitro models for endothelial dysfunction. Cultured human umbilical vein endothelial cells (HUVEC) were exposed to laminar (LSS) or non-laminar shear stress (NLSS). ENaC membrane insertion was quantified using Quantum-dot-based immunofluorescence staining and the mechanical properties of the cell surface were probed with the Atomic Force Microscope (AFM) in vitro and ex vivo in isolated aortae of C57BL/6 and ApoE/LDLR-/- mice. Flow- and acetylcholine-mediated vasodilation was measured in vivo using magnetic resonance imaging. Acute LSS led to a rapid mineralocorticoid receptor (MR)-dependent membrane insertion of ENaC and subsequent stiffening of the endothelial cortex caused by actin polymerization. Of note, NLSS stress further augmented the cortical stiffness of the cells. These effects strongly depend on the presence of the endothelial glycocalyx (eGC) and could be prevented by functional inhibition of ENaC and MR in vitro endothelial cells and ex vivo endothelial cells derived from C57BL/6, but not ApoE/LDLR-/- vessel. In vivo In C57BL/6 vessels, ENaC- and MR inhibition blunted flow- and acetylcholine-mediated vasodilation, while in the dysfunctional ApoE/LDLR-/- vessels, this effect was absent. In conclusion, under physiological conditions, endothelial ENaC, together with the glycocalyx, was identified as an important shear stress sensor and mediator of endothelium-dependent vasodilation. In contrast, in pathophysiological conditions, ENaC-mediated mechanotransduction and endothelium-dependent vasodilation were lost, contributing to sustained endothelial stiffening and dysfunction.
Collapse
|
220
|
Rocco E, Grimaldi MC, Maino A, Cappannoli L, Pedicino D, Liuzzo G, Biasucci LM. Advances and Challenges in Biomarkers Use for Coronary Microvascular Dysfunction: From Bench to Clinical Practice. J Clin Med 2022; 11:2055. [PMID: 35407662 PMCID: PMC8999821 DOI: 10.3390/jcm11072055] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/27/2022] [Accepted: 04/02/2022] [Indexed: 02/01/2023] Open
Abstract
Coronary microvascular dysfunction (CMD) is related to a broad variety of clinical scenarios in which cardiac microvasculature is morphologically and functionally affected, and it is associated with impaired responses to vasoactive stimuli. Although the prevalence of CMD involves about half of all patients with chronic coronary syndromes and more than 20% of those with acute coronary syndrome, the diagnosis of CMD is often missed, leading to the underestimation of its clinical importance. The established and validated techniques for the measurement of coronary microvascular function are invasive and expensive. An ideal method to assess endothelial dysfunction should be accurate, non-invasive, cost-effective and accessible. There are varieties of biomarkers available, potentially involved in microvascular disease, but none have been extensively validated in this heterogeneous clinical population. The investigation of potential biomarkers linked to microvascular dysfunction might improve the assessment of the diagnosis, risk stratification, disease progression and therapy response. This review article offers an update about traditional and novel potential biomarkers linked to CMD.
Collapse
Affiliation(s)
- Erica Rocco
- Department of Medical-Surgical Sciences and Biotechnologies, Cardiology Unit, ICOT Hospital, Sapienza University of Rome, 04110 Latina, Italy;
| | - Maria Chiara Grimaldi
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Alessandro Maino
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
| | - Luigi Cappannoli
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
| | - Daniela Pedicino
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Luigi Marzio Biasucci
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
221
|
Peracaula M, Torres D, Poyatos P, Luque N, Rojas E, Obrador A, Orriols R, Tura-Ceide O. Endothelial Dysfunction and Cardiovascular Risk in Obstructive Sleep Apnea: A Review Article. Life (Basel) 2022; 12:537. [PMID: 35455027 PMCID: PMC9025914 DOI: 10.3390/life12040537] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/23/2022] [Accepted: 03/31/2022] [Indexed: 11/16/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a respiratory condition during sleep caused by repeated pauses in breathing due to upper airway obstruction. It is estimated that OSA affects 30% of the population, but only 10% are well diagnosed due to the absence of a well-defined symptomatology and poor screening tools for early diagnosis. OSA is associated to an endothelial dysfunction inducing several biological responses such as hypoxia, hypercapnia and oxidative stress, among others. OSA also triggers respiratory, nervous, metabolic, humoral and immunity system activations that increase the possibility of suffering a cardiovascular (CV) disease. In this review, we expose different studies that show the relationship between OSA and endothelial dysfunction and its association with CV pathologies like hypertension, and we define the most well-known treatments and their limitations. Additionally, we describe the potential future directions in OSA research, and we report clinical features such as endothelial progenitor cell alterations that could act as biomarkers for the development of new diagnostic tools and target therapies.
Collapse
Affiliation(s)
- Miriam Peracaula
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (M.P.); (D.T.); (P.P.); (N.L.); (E.R.); (A.O.)
| | - Daniela Torres
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (M.P.); (D.T.); (P.P.); (N.L.); (E.R.); (A.O.)
| | - Paula Poyatos
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (M.P.); (D.T.); (P.P.); (N.L.); (E.R.); (A.O.)
| | - Neus Luque
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (M.P.); (D.T.); (P.P.); (N.L.); (E.R.); (A.O.)
| | - Eric Rojas
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (M.P.); (D.T.); (P.P.); (N.L.); (E.R.); (A.O.)
| | - Anton Obrador
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (M.P.); (D.T.); (P.P.); (N.L.); (E.R.); (A.O.)
| | - Ramon Orriols
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (M.P.); (D.T.); (P.P.); (N.L.); (E.R.); (A.O.)
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital of Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190 Girona, Spain; (M.P.); (D.T.); (P.P.); (N.L.); (E.R.); (A.O.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
222
|
Toll-Like Receptors/TNF-α Pathway Crosstalk and Impact on Different Sites of Recurrent Myocardial Infarction in Elderly Patients. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1280350. [PMID: 35425840 PMCID: PMC9005286 DOI: 10.1155/2022/1280350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/17/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022]
Abstract
Background Recurrent myocardial infarction is associated with increased mortality. Risk and predictive factors of recurrent myocardial infarction in elderly patients after coronary stenting are not well known. This research sought to investigate the effects of proinflammatory cytokines and toll-like receptor on recurrent myocardial infarction after coronary stenting in elderly patients. Methods We measured the levels of toll-like receptor 2 (TLR2), toll-like receptor 3 (TLR3), toll-like receptor 4 (TLR4), tumor necrosis factor-α (TNF-α), soluble tumor necrosis factor-α receptor-1 (sTNFR-1), soluble tumor necrosis factor-α receptor-2 (sTNFR-2), endothelial progenitor cells (EPCs), and vascular endothelial growth factor (VEGF) in elderly patients with recurrent myocardial infarction and assessed the changes of proinflammatory cytokines and toll-like receptors in elderly patients with recurrent myocardial infarction after coronary stenting. Results Levels of TLR2, TLR3, TLR4, TNF-α, sTNFR-1, and sTNFR-2 were remarkably increased (P < 0.001), and EPCs and VEGF were remarkably lowered (P < 0.001) in the elderly patients with recurrent myocardial infarction after coronary stent implantation. Increased expressions of proinflammatory cytokines and toll-like receptors induced recurrent myocardial infarction after coronary stenting. Elevated expressions of proinflammatory cytokines and toll-like receptors may be used to identify elderly patients who have an increased risk of developing recurrent myocardial infarction after coronary stenting. Conclusion The increase levels of proinflammatory cytokines and toll-like receptors were associated with recurrent myocardial infarction after coronary stenting. Increased expressions of proinflammatory cytokines and toll-like receptors may be clinically useful biomarkers for predicting recurrent myocardial infarction in the elderly patients after coronary stent implantation.
Collapse
|
223
|
Knowles KA, Stewart KJ, Tejan J, Ouyang P, Ratchford EV, Sullam L, Magliato K, Whitt MD, Silber HA. A novel operator-independent noninvasive device for assessing arterial reactivity. IJC HEART & VASCULATURE 2022; 39:100960. [PMID: 35402694 PMCID: PMC8984635 DOI: 10.1016/j.ijcha.2022.100960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/21/2021] [Accepted: 01/18/2022] [Indexed: 11/23/2022]
Abstract
Background Endothelial dysfunction is associated with increased risk of cardiovascular disease (CVD). Currently available noninvasive methods of measuring endothelial function have limitations. We tested a novel device that provides an automated measurement of the difference between baseline and post-ischemic, hyperemia-induced, brachial arterial compliance, a phenomenon known to be endothelium-dependent. The association between the calculated index, Flow-mediated Compliance Response (FCR), and established CVD risk indices was determined. Methods Adults with CVD risk factors or known coronary artery disease (CAD) were enrolled. Framingham Risk Score (FRS) was calculated and presence of metabolic syndrome (MetSyn) was assessed. Carotid artery plaques were identified by ultrasound. Cardiorespiratory fitness was assessed by 6-minute walk test (6MWT). FCR was measured using the device. Results Among 135 participants, mean age 49.3 +/- 17.9 years, characteristics included: 48% female, 7% smokers, 7% CAD, 10% type 2 diabetes, 34% MetSyn, and 38% with carotid plaque. Those with MetSyn had 24% lower FCR than those without (p < 0.001). Lower FCR was associated with higher FRS percentile (r = -0.29, p < 0.001), more MetSyn factors (r = -0.30, p < 0.001), more carotid plaques (r = -0.22, p = 0.01), and lower 6MWT (r = 0.34, p < 0.0001). Conclusion FCR, an index of arterial reactivity obtained automatically using a novel, operator-independent device, was inversely associated with established CVD risk indices, increased number of carotid plaques, and lower cardiorespiratory fitness. Whether measuring FCR could play a role in screening for CVD risk and assessing whether endothelial function changes in response to treatments aimed at CVD risk reduction, warrants further study.
Collapse
Affiliation(s)
- Kellen A. Knowles
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kerry J. Stewart
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Joseph Tejan
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Pamela Ouyang
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elizabeth V. Ratchford
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laura Sullam
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kathy Magliato
- Division of Cardiothoracic Surgery, Providence St. John’s Health Center, Santa Monica, CA, United States
| | - Michael D. Whitt
- Department of Biomedical Engineering, California Polytechnic State University, San Luis Obispo, CA, United States
| | - Harry A. Silber
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Corresponding author at: Cardiology, Suite 2400, 301 Building, Johns Hopkins Bayview Medical Center, 4940 Eastern Avenue, Baltimore, MD 21224, United States.
| |
Collapse
|
224
|
Haidara MA, Al-Ani B, Bin-Jaliah I, Shams Eldeen AM, Morsy MD. Vanadyl sulphate ameliorates biomarkers of endothelial injury and coagulation and thrombosis in a rat model of hyperglycaemia. Arch Physiol Biochem 2022; 128:447-454. [PMID: 31774317 DOI: 10.1080/13813455.2019.1691602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/14/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND We sought to determine whether the insulin mimicking agent, vanadyl sulphate (Van) can inhibit biomarkers of endothelial injury and coagulation and thrombosis induced by a moderate level of hyperglycaemia. MATERIAL AND METHODS Hyperglycaemia was induced in rats by a single injection of streptozotocin (STZ, 50 mg/kg) two weeks after being fed on a high-fat diet (model group). The treatment group started Van (20 mg/kg/day) treatment one-week post STZ injection and continued on Van until being sacrificed at week 10. RESULTS Administration of Van to the model group significantly (p < .05) ameliorated dyslipidemia and biomarkers of inflammation (TNF-α, IL-6, and hsCRP) and endothelial injury (E-selectin, P-selectin, sICAM-1, sVCAM-1, and ET-1). Van also significantly inhibited hyperglycaemia-induced blood levels of coagulation (vWF) and thrombosis (PAI-1 and fibrinogen) biomarkers. CONCLUSIONS Vanadyl sulphate effectively suppresses hyperglycaemia-induced endothelial injury, coagulation and thrombosis, which is associated with the inhibition of inflammation and dyslipidemia.
Collapse
Affiliation(s)
- Mohamed A Haidara
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Bahjat Al-Ani
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ismaeel Bin-Jaliah
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Asmaa M Shams Eldeen
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - M D Morsy
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Physiology, College of Medicine, Menoufia University, Shibin el Kom, Egypt
| |
Collapse
|
225
|
Whole Mount Preparation of Mouse Aorta for Confocal Microscopy Studies of the Intima. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:597-610. [PMID: 35237991 DOI: 10.1007/978-1-0716-1924-7_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Confocal imaging of the mouse aorta is a powerful, indispensable technique for the study of cardiovascular pathology ex vivo. Whole mount en face preparations allow visualization of wide areas of the luminal vessel surface, thus enabling a thorough analysis of multiple cellular and structural features of the endothelial cell-rich intimal layer. This method is a suitable tool for the study of endothelial cell dysfunction and leukocyte infiltration, both of which contribute to the onset of pathological vascular conditions such as atherosclerosis. This chapter provides a complete guide on how to perfuse-fix mouse aorta, dissect the vessel, immunostain target proteins, and carry out en face confocal image acquisition and analysis.
Collapse
|
226
|
Budoff MJ, Lakshmanan S, Toth PP, Hecht HS, Shaw LJ, Maron DJ, Michos ED, Williams KA, Nasir K, Choi AD, Chinnaiyan K, Min J, Blaha M. Cardiac CT angiography in current practice: An American society for preventive cardiology clinical practice statement ✰. Am J Prev Cardiol 2022; 9:100318. [PMID: 35146468 PMCID: PMC8802838 DOI: 10.1016/j.ajpc.2022.100318] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/29/2022] Open
Abstract
In this clinical practice statement, we represent a summary of the current evidence and clinical applications of cardiac computed tomography (CT) in evaluation of coronary artery disease (CAD), from an expert panel organized by the American Society for Preventive Cardiology (ASPC), and appraises the current use and indications of cardiac CT in clinical practice. Cardiac CT is emerging as a front line non-invasive diagnostic test for CAD, with evidence supporting the clinical utility of cardiac CT in diagnosis and prevention. CCTA offers several advantages beyond other testing modalities, due to its ability to identify and characterize coronary stenosis severity and pathophysiological changes in coronary atherosclerosis and stenosis, aiding in early diagnosis, prognosis and management of CAD. This document further explores the emerging applications of CCTA based on functional assessment using CT derived fractional flow reserve, peri‑coronary inflammation and artificial intelligence (AI) that can provide personalized risk assessment and guide targeted treatment. We sought to provide an expert consensus based on the latest evidence and best available clinical practice guidelines regarding the role of CCTA as an essential tool in cardiovascular prevention - applicable to risk assessment and early diagnosis and management, noting potential areas for future investigation.
Collapse
Affiliation(s)
- Matthew J. Budoff
- Division of Cardiology, Lundquist Institute at Harbor-UCLA, Torrance CA, USA
| | - Suvasini Lakshmanan
- Division of Cardiology, Lundquist Institute at Harbor-UCLA, Torrance CA, USA
| | - Peter P. Toth
- CGH Medical Center, Sterling, IL and Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Harvey S. Hecht
- Department of Medicine, Mount Sinai Medical Center, New York, NY
| | - Leslee J. Shaw
- Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David J. Maron
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Erin D. Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kim A. Williams
- Division of Cardiology, Rush University Medical Center, Chicago IL
| | - Khurram Nasir
- Cardiovascular Prevention and Wellness, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX
| | - Andrew D. Choi
- Division of Cardiology and Department of Radiology, The George Washington University School of Medicine, Washington, DC, USA
| | - Kavitha Chinnaiyan
- Division of Cardiology, Department of Medicine, Beaumont Hospital, Royal Oak, MI
| | - James Min
- Chief Executive Officer Cleerly Inc., New York, NY
| | - Michael Blaha
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
227
|
Zhong P, Li Z, Lin Y, Peng Q, Huang M, Jiang L, Li C, Kuang Y, Cui S, Yu D, Yu H, Yang X. Retinal microvasculature impairments in patients with coronary artery disease: An optical coherence tomography angiography study. Acta Ophthalmol 2022; 100:225-233. [PMID: 33629471 DOI: 10.1111/aos.14806] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/15/2021] [Accepted: 02/02/2021] [Indexed: 02/05/2023]
Abstract
PURPOSE To investigate the association between retinal microvasculature and the presence and severity of coronary artery disease (CAD) using optical coherence tomography angiography (OCTA). METHODS The cross-sectional study was conducted in Guangdong Provincial People's Hospital, China. Retinal microvasculature parameters were measured by OCTA of the optic disc, including the vessel density (VD) and retinal nerve fibre thickness of the radial peripapillary capillary. In terms of the entire macula, VD of the superficial capillary plexus (SCP), deep capillary plexus (DCP) and foveal density (FD-300) were included. The Gensini score was used to evaluate the severity of coronary artery obstructive lesions in CAD patients. RESULTS A total of 410 participants (270 CAD patients and 140 controls) were included. Overall, participants showed significantly greater odds of having CAD in the lower versus higher VD for mean SCP, OR = 2.33 (95% CI 1.49-3.65); in the parafoveal SCP, OR = 2.68 (95% CI 1.70-4.23); and in the perifoveal SCP, OR = 2.36 (95% CI 1.49-3.72). Additionally, participants showed significantly greater odds of having CAD in the lower versus higher VD for mean DCP, OR = 4.04 (95% CI 2.53-6.45); in the parafoveal DCP, OR = 4.08 (95% CI 2.54-6.55); and in the perifoveal DCP, OR = 3.88 (95% CI 2.43-6.19). Among CAD patients, lower VD of DCP was associated with significantly greater adjusted Gensini scores (p = 0.004 for mean DCP; p = 0.035 for parafoveal DCP; p = 0.006 for perifoveal DCP). CONCLUSIONS SCP and DCP were found to be associated with the presence of CAD among the whole population, while DCP was found to be associated with Gensini scores in CAD patients. Retinal microvasculature was associated with the presence and severity of coronary artery stenosis in CAD patients.
Collapse
Affiliation(s)
- Pingting Zhong
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Zhixi Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yingwen Lin
- Shantou University Medical College, Shantou, China
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qingsheng Peng
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Manqing Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lei Jiang
- Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Cong Li
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Yu Kuang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shirong Cui
- Department of Statistics, University of California, Davis, CA, USA
| | - Danqing Yu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaohong Yang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
228
|
Ataie Z, Fatehi-Hassanabad Z, Nakhaee S, Foadoddini M, Farrokhfall K. Sex-specific endothelial dysfunction induced by high-cholesterol diet in rats: The role of protein tyrosine kinase and nitric oxide. Nutr Metab Cardiovasc Dis 2022; 32:745-754. [PMID: 35144857 DOI: 10.1016/j.numecd.2021.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/28/2021] [Accepted: 11/15/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis is a chronic process playing a crucial role in the pathogenesis of cardiovascular disease. Sex-specific differences in the incidence of atherosclerosis indicate that estrogen has a protective effect on the cardiovascular disease. However, the role of sex on endothelium responses in animal models of high cholesterol (HC) diet-induced atherosclerosis has not been fully investigated. This study was aimed to investigate vascular responses in HC-fed rats. METHODS AND RESULTS Male and female Sprague rats (12-week-old) were treated with either a standard diet (n = 12 of each sex) or an HC enriched diet (n = 12 of each sex) containing 2% cholesterol for 24 weeks. HC treated animals (both sexes) showed increased levels of total cholesterol, LDL-cholesterol, triglyceride and blood pressure (BP) compared to control rats. While the BP of control rats (both sexes) was increased following aminoguanidine administration (AG, 100 mg/kg i.p.), it was not changed in HC animals (both sexes). The hypotensive effect of acetylcholine was significantly impaired in male HC-treated rats. In vitro experiments demonstrated that aortic rings from HC group (both sexes) had an increased contractile response to phenylephrine and a decreased vasodilatory response to acetylcholine. The vasorelaxant effect of acetylcholine in HC rats (only male) was improved by applying 10-5 M genistein (tyrosine kinase inhibitor) or AG. CONCLUSION HC diet alters endothelium function through Nitric oxide (NO) and tyrosine kinase pathways in male rats.
Collapse
Affiliation(s)
- Zomorrod Ataie
- Health Clinical Science Research Center, Zahedan Branch, Islamic Azad University, Zahedan, Iran; Student Research Committee, Islamic Azad University, Zahedan Branch, Zahedan, Iran
| | | | - Samaneh Nakhaee
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), Birjand, Iran
| | - Mohsen Foadoddini
- Cardiovascular Disease Research Center, Department of Physiology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Khadijeh Farrokhfall
- Cardiovascular Disease Research Center, Department of Physiology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
229
|
Ito N, Hishikari K, Yoshikawa H, Yano H, Iiya M, Murai T, Hikita H, Takahashi A, Yonetsu T, Sasano T. Elevated Remnant Lipoprotein Related to Adverse Events in Patients with Chronic Limb Threatening Ischemia after Endovascular Therapy for Below the Knee Lesions. Ann Vasc Surg 2022; 84:314-321. [DOI: 10.1016/j.avsg.2022.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 12/01/2022]
|
230
|
Chen XH, Tan Y, Yu S, Lu L, Deng Y. Pinitol Protects Against Ox-Low-Density Lipoprotein-Induced Endothelial Inflammation and Monocytes Attachment. J Cardiovasc Pharmacol 2022; 79:368-374. [PMID: 34861664 DOI: 10.1097/fjc.0000000000001190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/06/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Atherosclerosis is a cardiovascular disease that affects a majority of people around the world at old age. Atherosclerosis is slow to develop and challenging to treat. Endothelial dysfunction caused by oxidative stress, inflammation, and other pathological factors drives the process of atherogenesis. LOX-1 is one of the main scavenging receptors for oxidized low-density lipoprotein (ox-LDL) and contributes to atherogenesis by inducing overproduction of reactive oxygen species, increased expression of proinflammatory cytokines, and secretion of cellular adhesion molecules. In addition, activation of LOX-1 inhibits the expression of KLF2, a key protective factor against atherosclerosis. In this study, we investigated the effects of pinitol, and naturally occurring cyclic polyol, on endothelial dysfunction induced by ox-LDL. Our findings show that pinitol revealed a good safety profile, as evidenced by reducing lactate dehydrogenase release in human aortic endothelial cells. In our experiments, pinitol reduced the production of reactive oxygen species and expression of IL-6 and monocyte chemoattractant protein-1 induced by ox-LDL. Pinitol also significantly reduced the attachment of THP-1 monocytes to endothelial cells via downregulation of vascular cellular adhesion molecule-1 and E-selectin. Importantly, we found that pinitol reduced the expression of LOX-1 induced by ox-LDL and rescued the expression of KLF2, which is dependent on ERK5 expression. Together, our findings provide notable evidence that pinitol may have potential implication in the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xiao-Hong Chen
- Department of Neurology, Nanchang First Hospital, Nanchang, Jiangxi, China
| | | | | | | | | |
Collapse
|
231
|
Gunter S, Michel FS, Fourie SS, Singh M, le Roux R, Manilall A, Mokotedi LP, Millen AME. The effect of TNF-α inhibitor treatment on microRNAs and endothelial function in collagen induced arthritis. PLoS One 2022; 17:e0264558. [PMID: 35213638 PMCID: PMC8880872 DOI: 10.1371/journal.pone.0264558] [Citation(s) in RCA: 3] [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: 09/05/2021] [Accepted: 02/11/2022] [Indexed: 11/18/2022] Open
Abstract
Chronic inflammation causes dysregulated expression of microRNAs. Aberrant microRNA expression is associated with endothelial dysfunction. In this study we determined whether TNF-α inhibition impacted the expression of miRNA-146a-5p and miRNA-155-5p, and whether changes in the expression of these miRNAs were related to inflammation-induced changes in endothelial function in collagen-induced arthritis (CIA). Sixty-four Sprague-Dawley rats were divided into control (n = 24), CIA (n = 24) and CIA+etanercept (n = 16) groups. CIA and CIA+etanercept groups were immunized with bovine type-II collagen, emulsified in incomplete Freund’s adjuvant. Upon signs of arthritis, the CIA+etanercept group received 10mg/kg of etanercept intraperitoneally, every three days. After six weeks of treatment, mesenteric artery vascular reactivity was assessed using wire-myography. Serum concentrations of TNF-α, C-reactive protein, interleukin-6, vascular adhesion molecule-1 (VCAM-1) and pentraxin-3 (PTX-3) were measured by ELISA. Relative expression of circulating miRNA-146a-5p and miRNA-155-5p were determined using RT-qPCR. Compared to controls, circulating miRNA-155-5p, VCAM-1 and PTX-3 concentrations were increased, and vessel relaxation was impaired in the CIA (all p<0.05), but not in the CIA+etanercept (all p<0.05) groups. The CIA group had greater miRNA-146a-5p expression compared to the CIA+etanercept group (p = 0.005). Independent of blood pressure, miRNA-146a-5p expression was associated with increased PTX-3 concentrations (p = 0.03), while miRNA-155-5p expression was associated with impaired vessel relaxation (p = 0.01). In conclusion, blocking circulating TNF-α impacted systemic inflammation-induced increased expression of miRNA-146a-5p and miRNA-155-5p, which were associated with endothelial inflammation and impaired endothelial dependent vasorelaxation, respectively.
Collapse
Affiliation(s)
- Sulè Gunter
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- * E-mail:
| | - Frederic S. Michel
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Serena S. Fourie
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mikayra Singh
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Regina le Roux
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ashmeetha Manilall
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Lebogang P. Mokotedi
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Aletta M. E. Millen
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
232
|
Ahmed S, Akotat O, Sajeesh V, Alabi M, Datta S. A Case of Low-Flow Priapism as a Complication of COVID-19 Infection. Cureus 2022; 14:e22613. [PMID: 35371780 PMCID: PMC8958113 DOI: 10.7759/cureus.22613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2022] [Indexed: 11/21/2022] Open
Abstract
On 11 March 2020, the World Health Organisation (WHO) declared the severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) (COVID-19) a pandemic. With a global incidence of over 414 million cases, as of 16 February 2022, it presents a significant burden on healthcare. COVID-19 is primarily considered a respiratory illness; however, a wide range of presentations have been reported including a tendency for thrombotic complications. We report a case of a 58-year-old man who presented with dyspnoea, pyrexia and dry cough. Upon admission, he was noted to be in a severe type 1 respiratory failure with bilateral pulmonary infiltrates suggestive of COVID-19 infection. Rapid transfer to intensive therapy unit (ITU) ensued with intubation and ventilation. The patient was noted to have developed priapism one day following admission with subsequent aspiration by the Urology team, achieving detumescence. Priapism is a state of persistent penile erection that continues for four hours beyond sexual stimulation. Our case highlights the role of thrombosis, dysregulation of the clotting cascade and acute disseminated intravascular coagulation (DIC) as shared pathologies in priapism and COVID-19 infection. We put forth an example of one of the extra-pulmonary manifestations of the COVID-19 secondary to the pro-thrombotic state associated with the COVID-19 infection.
Collapse
|
233
|
Zampiccoli E, Barthelmes J, Kreysing L, Nägele MP, Nebunu D, Haider T, Eckardstein A, Gerber B, Schwotzer R, Ruschitzka F, Sudano I, Flammer AJ. Eyes on amyloidosis: microvascular retinal dysfunction in cardiac amyloidosis. ESC Heart Fail 2022; 9:1186-1194. [PMID: 35060356 PMCID: PMC8934987 DOI: 10.1002/ehf2.13792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/08/2021] [Accepted: 12/14/2021] [Indexed: 11/10/2022] Open
Abstract
Aims Methods and results Conclusions
Collapse
Affiliation(s)
- Emanuel Zampiccoli
- Department of Cardiology University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Jens Barthelmes
- Department of Cardiology University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Leonie Kreysing
- Department of Cardiology University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Matthias P. Nägele
- Department of Cardiology University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Delia Nebunu
- Department of Cardiology University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Thomas Haider
- Department of Cardiology University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Arnold Eckardstein
- Department of Clinical Chemistry University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Bernhard Gerber
- Clinic of Haematology Oncology Institute of Southern Switzerland Piazza Indipendenza 2 Bellinzona 6500 Switzerland
| | - Rahel Schwotzer
- Department of Medical Oncology and Haematology University Hospital Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Frank Ruschitzka
- Department of Cardiology University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Isabella Sudano
- Department of Cardiology University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| | - Andreas J. Flammer
- Department of Cardiology University Hospital Zurich, University of Zurich Raemistrasse 100 Zurich 8091 Switzerland
| |
Collapse
|
234
|
Tarnawski AS, Ahluwalia A. Endothelial cells and blood vessels are major targets for COVID-19-induced tissue injury and spreading to various organs. World J Gastroenterol 2022; 28:275-289. [PMID: 35110950 PMCID: PMC8771611 DOI: 10.3748/wjg.v28.i3.275] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/02/2021] [Accepted: 01/11/2022] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) infected so far over 250 million people and caused the death of over 5 million worldwide. Aging, diabetes, and cardiovascular diseases, conditions with preexisting impaired endothelial functions predispose to COVID-19. While respiratory epithelium is the main route of virus entry, the endothelial cells (ECs) lining pulmonary blood vessels are also an integral part of lung injury in COVID-19 patients. COVID-19 not only affects the lungs and respiratory system but also gastrointestinal (GI) tract, liver, pancreas, kidneys, heart, brain, and skin. Blood vessels are likely conduits for the virus dissemination to these distant organs. Importantly, ECs are also critical for vascular regeneration during injury/lesions healing and restoration of vascular network. The World Journal of Gastroenterology has published in last two years over 67 outstanding papers on COVID-19 infection with a focus on the GI tract, liver, pancreas, etc., however, the role of the endothelial and vascular components as major targets for COVID-19-induced tissue injury, spreading to various organs, and injury healing have not been sufficiently emphasized. In the present article, we focus on these subjects and on current treatments including the most recent oral drugs molnupiravir and paxlovid that show a dramatic, significant efficacy in controlling severe COVID-19 infection.
Collapse
Affiliation(s)
- Andrzej S Tarnawski
- Gastroenterology Research Department, University of California Irvine and the Veterans Administration Long Beach Healthcare System, Long Beach, CA 90822, United States
| | - Amrita Ahluwalia
- Research Service, Veterans Administration Long Beach Healthcare System, Long Beach, CA 90822, United States
| |
Collapse
|
235
|
Kiyooka T, Ohanyan V, Yin L, Pung YF, Chen YR, Chen CL, Kang PT, Hardwick JP, Yun J, Janota D, Peng J, Kolz C, Guarini G, Wilson G, Shokolenko I, Stevens DA, Chilian WM. Mitochondrial DNA integrity and function are critical for endothelium-dependent vasodilation in rats with metabolic syndrome. Basic Res Cardiol 2022; 117:3. [PMID: 35039940 PMCID: PMC9030679 DOI: 10.1007/s00395-021-00908-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 01/31/2023]
Abstract
Endothelial dysfunction in diabetes is generally attributed to oxidative stress, but this view is challenged by observations showing antioxidants do not eliminate diabetic vasculopathy. As an alternative to oxidative stress-induced dysfunction, we interrogated if impaired mitochondrial function in endothelial cells is central to endothelial dysfunction in the metabolic syndrome. We observed reduced coronary arteriolar vasodilation to the endothelium-dependent dilator, acetylcholine (Ach), in Zucker Obese Fatty rats (ZOF, 34 ± 15% [mean ± standard deviation] 10-3 M) compared to Zucker Lean rats (ZLN, 98 ± 11%). This reduction in dilation occurred concomitantly with mitochondrial DNA (mtDNA) strand lesions and reduced mitochondrial complex activities in the endothelium of ZOF versus ZLN. To demonstrate endothelial dysfunction is linked to impaired mitochondrial function, administration of a cell-permeable, mitochondria-directed endonuclease (mt-tat-EndoIII), to repair oxidatively modified DNA in ZOF, restored mitochondrial function and vasodilation to Ach (94 ± 13%). Conversely, administration of a cell-permeable, mitochondria-directed exonuclease (mt-tat-ExoIII) produced mtDNA strand breaks in ZLN, reduced mitochondrial complex activities and vasodilation to Ach in ZLN (42 ± 16%). To demonstrate that mitochondrial function is central to endothelium-dependent vasodilation, we introduced (via electroporation) liver mitochondria (from ZLN) into the endothelium of a mesenteric vessel from ZOF and restored endothelium-dependent dilation to vasoactive intestinal peptide (VIP at 10-5 M, 4 ± 3% vasodilation before mitochondrial transfer and 48 ± 36% after transfer). Finally, to demonstrate mitochondrial function is key to endothelium-dependent dilation, we administered oligomycin (mitochondrial ATP synthase inhibitor) and observed a reduction in endothelium-dependent dilation. We conclude that mitochondrial function is critical for endothelium-dependent vasodilation.
Collapse
Affiliation(s)
- Takahiko Kiyooka
- Division of Cardiology, Tokai University Oiso Hospital, Oiso, Kanagawa, Japan
| | - Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Yuh Fen Pung
- Division of Biomedical Sciences, University of Nottingham, Malaysia Campus, Selangor, Malaysia
| | - Yeong-Renn Chen
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Chwen-Lih Chen
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Patrick T Kang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - James P Hardwick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - June Yun
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Danielle Janota
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Joanna Peng
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Christopher Kolz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Giacinta Guarini
- Cardiovascular Unit, Spedali Riuniti Santa Maria Maddalena, Volterra, Italy
| | - Glenn Wilson
- Department of Biomedical Science, University of South Alabama, Mobile, USA
| | - Inna Shokolenko
- Department of Biomedical Science, University of South Alabama, Mobile, USA
| | - Donte A Stevens
- Division of Biological Sciences, University of California-San Diego, San Diego, USA
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA.
| |
Collapse
|
236
|
Markey O, Vasilopoulou D, Kliem KE, Fagan CC, Grandison AS, Sutton R, Humphries DJ, Todd S, Jackson KG, Givens DI, Lovegrove JA. Effect of fat-reformulated dairy food consumption on postprandial flow-mediated dilatation and cardiometabolic risk biomarkers compared with conventional dairy: a randomized controlled trial. Am J Clin Nutr 2022; 115:679-693. [PMID: 35020795 PMCID: PMC8895219 DOI: 10.1093/ajcn/nqab428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/20/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Longer-term consumption of SFA-reduced, MUFA-enriched dairy products has been reported to improve fasting flow-mediated dilatation (FMD). Yet, their impact on endothelial function in the postprandial state warrants investigation. OBJECTIVES The aim was to compare the impact of a fatty acid (FA) modified with a conventional (control) dairy diet on the postprandial %FMD (primary outcome) and systemic cardiometabolic responses to representative meals, and retrospectively explore whether treatment effects differ by apolipoprotein E (APOE) or endothelial NO synthase (eNOS) Glu298Asp gene polymorphisms. METHODS In a crossover-design randomized controlled study, 52 adults with moderate cardiovascular disease risk consumed dairy products [38% of total energy intake (%TE) from fat: FA-modified (target: 16%TE SFAs; 14%TE MUFAs) or control (19%TE SFAs; 11%TE MUFAs)] for 12 wk, separated by an 8-wk washout. Blood sampling and FMD measurements (0-480 min) were performed pre- and postintervention after sequential mixed meals that were representative of the assigned dairy diets (0 min, ∼50 g fat; 330 min, ∼30 g fat). RESULTS Relative to preintervention (∆), the FA-modified dairy diet and meals (treatment) attenuated the increase in the incremental AUC (iAUC), but not AUC, for the %FMD response observed with the conventional treatment (-135 ± 69% vs. +199 ± 82% × min; P = 0.005). The ∆ iAUC, but not AUC, for the apoB response decreased after the FA-modified treatment yet increased after the conventional treatment (-4 ± 3 vs. +3 ± 3 mg/mL × min; P = 0.004). The ∆ iAUC decreased for plasma total SFAs (P = 0.003) and trans 18:1 (P < 0.0001) and increased for cis-MUFAs (P < 0.0001) following the conventional relative to the FA-modified treatment. No treatment × APOE or eNOS genotype interactions were evident for any outcome. CONCLUSIONS This study provides novel insights into the longer-term effects of FA-modified dairy food consumption on postprandial cardiometabolic responses.
Collapse
Affiliation(s)
- Oonagh Markey
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, United Kingdom
| | - Dafni Vasilopoulou
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, United Kingdom
| | - Kirsty E Kliem
- Animal, Dairy, and Food Chain Sciences, University of Reading, Reading, United Kingdom,Institute for Food, Nutrition, and Health, University of Reading, Reading, United Kingdom
| | - Colette C Fagan
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, United Kingdom,Institute for Food, Nutrition, and Health, University of Reading, Reading, United Kingdom
| | - Alistair S Grandison
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, United Kingdom
| | - Rachel Sutton
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, United Kingdom
| | - David J Humphries
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, United Kingdom,Institute for Food, Nutrition, and Health, University of Reading, Reading, United Kingdom
| | - Susan Todd
- Department of Mathematics and Statistics, University of Reading, Reading, United Kingdom
| | - Kim G Jackson
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, United Kingdom,Institute for Food, Nutrition, and Health, University of Reading, Reading, United Kingdom
| | - David I Givens
- Institute for Food, Nutrition, and Health, University of Reading, Reading, United Kingdom
| | | |
Collapse
|
237
|
Nageeb RS, Azmy AM, Tantawy HF, Nageeb GS, Omran AA. Subclinical thyroid dysfunction and autoantibodies in acute ischemic and hemorrhagic stroke patients: relation to long term stroke outcome. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2022. [DOI: 10.1186/s41983-021-00439-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Data regarding the relation between both subclinical thyroid dysfunction, thyroid autoantibodies and clinical outcomes in stroke patients are limited. This study aimed to evaluate subclinical thyroid dysfunction and thyroid autoantibodies production in acute stroke patients and their relation to long term stroke outcome. We recruited 138 patients who were subjected to thorough general, neurological examination and brain imaging. Blood samples were collected for measurement of levels of serum thyroid function [free tri-iodothyronine (FT3), free thyroxin (FT4), thyroid stimulating hormone (TSH)], thyroid autoantibodies within 48 h after hospital admission. FT4 and TSH after 1 year were done. The stroke severity was assessed at admission by the National Institutes of Health Stroke Scale (NIHSS). The stroke outcome was assessed at 3 months and after 1 year by the modified Rankin Scale (mRS). We divided the patients into two groups according to thyroid autoantibodies (positive and negative groups).
Results
Subclinical hyperthyroidism was found in 23% of patients, and subclinical hypothyroidism in 10% of patients. Euthyroidism was detected in 67% of patients. 34% patients had positive thyroid autoantibody. Positive thyroid autoantibodies were commonly found in those with subclinical hyperthyroidism (28%), followed by subclinical hypothyroidism (21%) and euthyroidism (14%). 73% and 59% of stroke patients had poor outcomes (mRS was > 2) at 3 months and 1 year respectively with no significant difference between ischemic and hemorrhagic stroke patients. In the positive group final TSH level, NIHSS score at admission, and disability at 1 year were significantly higher compared with the negative group. Poor outcome was significantly associated with higher NIHSS score at admission, positive thyroid autoantibodies, subclinical hyperthyroidism, and atrial fibrillation.
Conclusions
Subclinical thyroid dysfunction could be found in stroke patients with positive thyroid autoantibodies. Subclinical hyperthyroidism and thyroid autoantibodies were associated with a poor outcome at 1 year in first-ever acute stroke patients especially in those presented with atrial fibrillation and higher NIHSS score at admission.
Collapse
|
238
|
Brown PA. Differential and targeted vesiculation: pathologic cellular responses to elevated arterial pressure. Mol Cell Biochem 2022; 477:1023-1040. [PMID: 34989921 DOI: 10.1007/s11010-021-04351-7] [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: 10/11/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022]
Abstract
Extracellular vesicles are small membrane-enclosed particles released during cell activation or injury. They have been investigated for several decades and found to be secreted in various diseases. Their pathogenic role is further supported by the presence of several important molecules among their cargo, including proteins, lipids, and nucleic acids. Many studies have reported enhanced and targeted extracellular vesicle biogenesis in diseases that involve chronic or transient elevation of arterial pressure resulting in endothelial dysfunction, within either the general circulatory system or specific local vascular beds. In addition, several associated pathologic processes have been studied and reported. However, the role of elevated pressure as a common pathogenic trigger across vascular domains and disease chronicity has not been previously described. This review will therefore summarize our current knowledge of the differential and targeted biogenesis of extracellular vesicles in major diseases that are characterized by elevated arterial pressure leading to endothelial dysfunction and propose a unified theory of pressure-induced extracellular vesicle-mediated pathogenesis.
Collapse
Affiliation(s)
- Paul A Brown
- Department of Basic Medical Sciences, Faculty of Medical Sciences Teaching and Research Complex, The University of the West Indies, Mona, Kingston 7, Jamaica.
| |
Collapse
|
239
|
da Silva JF, Bolsoni JA, da Costa RM, Alves JV, Bressan AFM, Silva LEV, Costa TJ, Oliveira AER, Manzato CP, Aguiar CA, Fazan R, Cunha FQ, Nakaya HI, Carneiro FS, Tostes RC. Aryl-hydrocarbon receptor (AhR) activation contributes to high-fat diet-induced vascular dysfunction. Br J Pharmacol 2022; 179:2938-2952. [PMID: 34978070 DOI: 10.1111/bph.15789] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Metabolic and vascular dysfunction are common features of obesity. Aryl hydrocarbons receptors (AhR) regulate lipid metabolism and vascular homeostasis, but whether vascular AhR are activated in obesity or if AhR have protective or harmful effects on vascular function in obesity are not known. Thus, our study addressed whether AhR activation contributes to obesity-associated vascular dysfunction and the mechanisms involved in the AhR effects. EXPERIMENTAL APPROACH Male AhRKO (AhR knockout) and WT (wild type) mice were fed either a control or a HF (high-fat) diet for ten weeks. Metabolic and inflammatory parameters were measured in serum and adipose tissue. Vascular reactivity (isometric force) was evaluated using a myography. eNOS and AhR protein expression was determined by Western blot; Cyp1A1 and eNOS gene expression by RT-PCR. Nitric oxide (NO) production was quantified by DAF fluorescence. KEY RESULTS HF diet increased serum total, HDL, and LDL cholesterol, as well as vascular AhR protein expression and proinflammatory cytokines in the adipose tissue. HF diet decreased endothelium-dependent vasodilation. AhR deletion protected mice from HF diet-induced dyslipidemia, weight gain, and inflammatory processes. HF diet-induced endothelial dysfunction was attenuated in AhRKO mice. Vessels from AhRKO mice exhibited a greater NO reserve. In cultured endothelial cells, lysophosphatidylcholine (LPC, a major component of LDL and oxLDL) reduced eNOS gene expression and NO production. Antagonism of AhR abrogated LPC effects on endothelial cells and LPC-induced decreased endothelium-dependent vasodilation. CONCLUSION AND IMPLICATIONS AhR deletion attenuates HF diet-induced dyslipidemia and vascular dysfunction by improving eNOS/NO signalling. Targeting AhR may prevent obesity-associated vascular dysfunction.
Collapse
Affiliation(s)
- Josiane Fernandes da Silva
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Juliana A Bolsoni
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Rafael M da Costa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Academic Unit on Health Sciences, Jataí Federal University, Jataí, Brazil
| | - Juliano V Alves
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Alecsander F M Bressan
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Luiz Eduardo V Silva
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Antonio E R Oliveira
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, Sao Paulo, Brazil
| | - Carla P Manzato
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Carlos A Aguiar
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Rubens Fazan
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, Sao Paulo, Brazil.,Israelita Albert Einstein Hospital, Sao Paulo, Brazil
| | - Fernando S Carneiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
240
|
Kurian C, Mayer S, Kaur G, Sahni R, Feldstein E, Samaan M, Viswanathan D, Sami T, Ali S, Al-Shammari H, Bloomfield J, Bravo M, Nuoman R, Gulko E, Gandhi C, Al-Mufti F. Bihemispheric ischemic strokes in patients with COVID-19. Brain Circ 2022; 8:10-16. [PMID: 35372732 PMCID: PMC8973449 DOI: 10.4103/bc.bc_65_21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/29/2021] [Accepted: 02/06/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND: There is emerging evidence that COVID-19 can trigger thrombosis because of a hypercoagulable state, including large-vessel occlusion ischemic strokes. Bihemispheric ischemic stroke is uncommon and is thought to indicate an embolic source. Here, we examine the findings and outcomes of patients with bihemispheric stroke in the setting of COVID-19. METHODS: We performed a retrospective cohort study at a quaternary academic medical center between March 1, 2020, and April 30, 2020. We identified all patients with laboratory-confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection who presented with simultaneous bihemispheric ischemic strokes. RESULTS: Of 637 COVID-19 admissions during the 2-month period, 13 had a diagnosis of acute ischemic stroke, including 5 who developed bihemispheric cerebral infarction. Three of those 5 (60%) were female, median age was 54 (range 41–67), and all five were being managed for severe COVID-19-related pneumonia complicated by acute kidney injury and liver failure before the diagnosis of cerebral infarction was established. Five presented with elevated ferritin, lactate dehydrogenase, and interleukin-6 (IL-6) levels, and four had lymphopenia and elevated D-dimer levels. All patients underwent neuroimaging with computed tomography for persistent depressed mentation, with or without a focal neurologic deficit, demonstrating multifocal ischemic strokes with bihemispheric involvement. Outcome was poor in all patients: two were discharged to a rehabilitation facility with moderate-to-severe disability and three (60%) patients died. CONCLUSIONS: Stroke is implicated in SARS-CoV-2 infection. Although causality cannot be established, we present the imaging and clinical findings of patients with COVID-19 and simultaneous bihemispheric ischemic strokes. Multifocal ischemic strokes with bihemispheric involvement should be considered in COVID-19 patients with severe infection and poor neurologic status and may be associated with poor outcomes.
Collapse
|
241
|
Watany MM, Abdou S, Elkolaly R, Elgharbawy N, Hodeib H. Evaluation of admission levels of P, E and L selectins as predictors for thrombosis in hospitalized COVID-19 patients. Clin Exp Med 2022; 22:567-575. [PMID: 35061142 PMCID: PMC8778493 DOI: 10.1007/s10238-021-00787-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/15/2021] [Indexed: 01/20/2023]
Abstract
Thromboembolic complications are the most reported cause of death in coronavirus disease-2019 (COVID-19). Hypercoagulability, platelets activation and endotheliopathy are well-recognized features in COVID-19 patients. The aim of this work was to evaluate circulating soluble selectins P, E and L at the time of hospital admission as predictors for upcoming thrombosis. This retrospective study included 103 hospitalized COVID-19 patients and 50 healthy volunteer controls. COVID-19 patients were categorized into two groups; group 1 who developed thrombosis during hospitalization and group 2 who did not. Soluble selectins were quantitated using ELISA technique. Higher levels of sP-selectin, sE-selectin and sL-selectin were detected in COVID-19 patients compared to controls. Furthermore, significantly higher levels were found in group 1 compared to group 2. Their means were [5.86 ± 1.72 ng/mL vs. 2.51 ± 0.81 ng/mL]; [50 ± 8.57 ng/mL vs. 23.96 ± 6.31 ng/mL] and [4.66 ± 0.83 ng/mL vs. 2.95 ± 0.66 ng/mL] for sP-selectin, sE-selectin and sL-selectin respectively. The elevated selectins correlated with the currently used laboratory biomarkers of disease severity. After adjustment of other factors, sP-selectin, sE-selectin and sL-selectin were independent predictors for thrombosis. At sP-selectin ≥ 3.2 ng/mL, sE-selectin ≥ 32.5 ng/mL and sL-selectin ≥ 3.6 ng/mL thrombosis could be predicted with 97.1%, 97.6% and 96.5% sensitivity. A panel of the three selectins provided 100% clinical sensitivity. Admission levels of circulating soluble selectins P, E and L can predict thrombosis in COVID-19 patients and could be used to identify patients who need prophylactic anticoagulants. E-selectin showed a superior clinical performance, as thrombo-inflammation biomarker, to the most commonly studied P-selectin.
Collapse
Affiliation(s)
- Mona M Watany
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Medical Campus, El Giesh st., Tanta, El-Gharbia Governorate, Tanta, 31527, Egypt.
| | - Saied Abdou
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Medical Campus, El Giesh st., Tanta, El-Gharbia Governorate, Tanta, 31527, Egypt
| | - Reham Elkolaly
- Chest department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Nashwa Elgharbawy
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hossam Hodeib
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Medical Campus, El Giesh st., Tanta, El-Gharbia Governorate, Tanta, 31527, Egypt
| |
Collapse
|
242
|
Bertucci T, Kakarla S, Kim D, Dai G. Differentiating Human Pluripotent Stem Cells to Vascular Endothelial Cells for Regenerative Medicine, Tissue Engineering, and Disease Modeling. Methods Mol Biol 2022; 2375:1-12. [PMID: 34591294 DOI: 10.1007/978-1-0716-1708-3_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Vasculature plays a vital role in human biology as blood vessels transport nutrients and oxygen throughout the body. Endothelial cells (ECs), specifically, are key as they maintain barrier functions between the circulating blood and the surrounding tissues. ECs derived from human pluripotent stem cells (hPSCs) are utilized to study vascular development and disease mechanisms within in vitro models. Additionally, ECs derived from induced pluripotent stem cells (iPSCs) hold great promise for advancing personalized medicine, cell therapies, and tissue-engineered constructs by creating patient-specific cell populations. Here, we describe a xeno-free, serum-free differentiation protocol for deriving ECs from hPSCs. In brief, mesoderm progenitor cells are derived via WNT pathway activation. Following this, EC maturation is achieved with exogenous vascular endothelial growth factor A (VEGFA) and basic fibroblast growth factor 2 (bFGF2). We have characterized these cells as expressing mature EC markers and have illustrated their functionality in vitro.
Collapse
Affiliation(s)
| | - Shravani Kakarla
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Diana Kim
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
243
|
Gluvic ZM, Zafirovic SS, Obradovic MM, Sudar-Milovanovic EM, Rizzo M, Isenovic ER. Hypothyroidism and Risk of Cardiovascular Disease. Curr Pharm Des 2022; 28:2065-2072. [PMID: 35726428 DOI: 10.2174/1381612828666220620160516] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/12/2022] [Indexed: 11/22/2022]
Abstract
Thyroid hormones (TH) have a significant impact on cellular oxidative metabolism. Besides that, they maintain vascular homeostasis by positive effects on endothelial and vascular smooth muscle cells. Subclinical (SCH) and clinical (CH) hypothyroidism influences target organs by changing their morphology and function and impaired blood and oxygen supply induced by accelerated atherosclerosis. The increased risk of acceleration and extension of atherosclerosis in patients with SCH and CH could be explained by dyslipidemia, diastolic hypertension, increased arterial stiffness, endothelial dysfunction, and altered blood coagulation. Instability of atherosclerotic plaque in hypothyroidism could cause excessive activity of the elements of innate immunity, which are characterized by the significant presence of macrophages in atherosclerotic plaques, increased nuclear factor kappa B (NFkB) expression, and elevated levels of tumor necrosis factor α (TNF-α) and matrix metalloproteinase (MMP) 9, with reduced interstitial collagen; all of them together creates inflammation milieu, resulting in plaque rupture. Optimal substitution by levothyroxine (LT4) restores biochemical euthyroidism. In postmenopausal women and elderly patients with hypothyroidism and associated vascular comorbidity, excessive LT4 substitution could lead to atrial rhythm disorders and osteoporosis. Therefore, it is of interest to maintain thyroid-stimulating hormone (TSH) levels in the reference range, thus eliminating the deleterious effects of lower or higher TSH levels on the cardiovascular system. This review summarizes the recent literature on subclinical and clinical hypothyroidism and atherosclerotic cardiovascular disease and discusses the effects of LT4 replacement therapy on restoring biochemical euthyroidism and atherosclerosis processes.
Collapse
Affiliation(s)
- Zoran M Gluvic
- Department of Endocrinology and Diabetes, Clinic for Internal Medicine, Zemun Clinical Hospital, School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Sonja S Zafirovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Milan M Obradovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Emina M Sudar-Milovanovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Manfredi Rizzo
- Promise Department, School of Medicine, University of Palermo, Palermo, Italy
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
244
|
Kreslová M, Sýkorová A, Bittenglová R, Schwarz J, Pomahačová R, Jehlička P, Kobr J, Trefil L, Sýkora J. Age-Related Progression of Microvascular Dysfunction in Cystic Fibrosis: New Detection Ways and Clinical Outcomes. Physiol Res 2021. [DOI: 10.33549//physiolres.934743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
There are concerns about altered vascular functions that could play an important role in the pathogenesis and influence the severity of chronic disease, however, increased cardiovascular risk in paediatric cystic fibrosis (CF) has not been yet fully understood. Aim was to analyse vascular disease risk and investigate changes over times in CF and controls. We prospectively enrolled 22 CF subjects (a median age of 16.07 years), and 22 healthy demographically matched controls (a median age of 17.28 years) and determined endothelial function. We utilised a combined diagnostic approach by measuring the plethysmographic Reactive Hyperemia Index (RHI) as the post-to preocclusive endothelium-dependent changes of vascular tone, and biomarkers that are known to be related to endothelial dysfunction (ED): asymmetric dimethyl arginine (ADMA), high-sensitive CRP (hsCRP), VCAM-1 and E-selectin. RHI values were significantly lower in CF young adults (p<0.005). HsCRP (p<0.005), E-selectin (p<0.001) and VCAM-1 (p<0.001) were significantly increased in CF patients since childhood. The findings have provided a detailed account of the ongoing process of microvascular dysfunction with gradual progression with the age of CF patients, making them further at risk of advanced vascular disease. Elevations of biomarkers in CF children with not yet demonstrated RHI changes but with significantly reduced RHI in adulthood and lipid profile changes indicate the possible occurrence of ED with CF-related specific risk factors over time and will enable us to provide the best possible support.
Collapse
Affiliation(s)
- M. Kreslová
- Department of Pediatrics, Charles University in Prague, Faculty of Medicine in Pilsen, Faculty Hospital, Pilsen, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Willems LH, Nagy M, Ten Cate H, Spronk HMH, Groh LA, Leentjens J, Janssen NAF, Netea MG, Thijssen DHJ, Hannink G, van Petersen AS, Warlé MC. Sustained inflammation, coagulation activation and elevated endothelin-1 levels without macrovascular dysfunction at 3 months after COVID-19. Thromb Res 2021; 209:106-114. [PMID: 34922160 PMCID: PMC8642246 DOI: 10.1016/j.thromres.2021.11.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/14/2021] [Accepted: 11/24/2021] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Endothelial damage and thrombosis caused by COVID-19 may imperil cardiovascular health. More than a year since the WHO declared COVID-19 pandemic, information on its effects beyond the acute phase is lacking. We investigate endothelial dysfunction, coagulation and inflammation, 3 months post-COVID-19. MATERIALS AND METHODS A cohort study was conducted including 203 patients with prior COVID-19. Macrovascular dysfunction was assessed by measuring the carotid artery diameter in response to hand immersion in ice-water. A historic cohort of 312 subjects served as controls. Propensity score matching corrected for baseline differences. Plasma concentrations of endothelin-1 were measured in patients post-COVID-19, during the acute phase, and in matched controls. Coagulation enzyme:inhibitor complexes and inflammatory cytokines were studied. RESULTS AND CONCLUSIONS The prevalence of macrovascular dysfunction did not differ between the COVID-19 (18.6%) and the historic cohort (22.5%, RD -4%, 95%CI: -15-7, p = 0.49). Endothelin-1 levels were significantly higher in acute COVID-19 (1.67 ± 0.64 pg/mL) as compared to controls (1.24 ± 0.37, p < 0.001), and further elevated 3 months post-COVID-19 (2.74 ± 1.81, p < 0.001). Thrombin:antithrombin(AT) was high in 48.3%. Markers of contact activation were increased in 16-30%. FVIIa:AT (35%) and Von Willebrand Factor:antigen (80.8%) were elevated. Inflammatory cytokine levels were high in a majority: interleukin(IL)-18 (73.9%), IL-6 (47.7%), and IL-1ra (48.9%). At 3 months after acute COVID-19 there was no indication of macrovascular dysfunction; there was evidence, however, of sustained endothelial cell involvement, coagulation activity and inflammation. Our data highlight the importance of further studies on SARS-CoV-2 related vascular inflammation and thrombosis, as well as longer follow-up in recovered patients.
Collapse
Affiliation(s)
- L H Willems
- Department of Surgery, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - M Nagy
- Departments of Internal medicine and Biochemistry, MUMC and CARIM School for Cardiovascular diseases, Maastricht, the Netherlands
| | - H Ten Cate
- Departments of Internal medicine and Biochemistry, MUMC and CARIM School for Cardiovascular diseases, Maastricht, the Netherlands; Center for Thrombosis and Haemostasis, Gutenberg University Medical Center, Mainz, Germany
| | - H M H Spronk
- Departments of Internal medicine and Biochemistry, MUMC and CARIM School for Cardiovascular diseases, Maastricht, the Netherlands
| | - L A Groh
- Department of Surgery, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - J Leentjens
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - N A F Janssen
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - M G Netea
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - D H J Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands/Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - G Hannink
- Department of Operating Rooms, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - A S van Petersen
- Department of Surgery, Bernhoven Hospital, Uden, the Netherlands
| | - M C Warlé
- Department of Surgery, Radboud University Medical Centre, Nijmegen, the Netherlands.
| |
Collapse
|
246
|
Zhang J, Rojas S, Singh S, Musich PR, Gutierrez M, Yao Z, Thewke D, Jiang Y. Wnt2 Contributes to the Development of Atherosclerosis. Front Cardiovasc Med 2021; 8:751720. [PMID: 34901211 PMCID: PMC8652052 DOI: 10.3389/fcvm.2021.751720] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/21/2021] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis, is a chronic inflammatory disease, characterized by the narrowing of the arteries resulting from the formation of intimal plaques in the wall of arteries. Yet the molecular mechanisms responsible for maintaining the development and progression of atherosclerotic lesions have not been fully defined. In this study, we show that TGF-β activates the endothelial-to-mesenchymal transition (EndMT) in cultured human aortic endothelial cells (HAECs) and this transition is dependent on the key executor of the Wnt signaling pathway in vitro. This study presents the first evidence describing the mechanistic details of the TGF-β-induced EndMT signaling pathway in HAECs by documenting the cellular transition to the mesenchymal phenotype including the expression of mesenchymal markers α-SMA and PDGFRα, and the loss of endothelial markers including VE-cadherin and CD31. Furthermore, a short hairpin RNA (shRNA) screening revealed that Wnt2 signaling is required for TGF-β-mediated EndMT of HAECs. Also, we found that LDLR−/− mice fed on a high-fat western-type diet (21% fat, 0.2% cholesterol) expressed high levels of Wnt2 protein in atherosclerotic lesions, confirming that this signaling pathway is involved in atherosclerosis in vivo. These findings suggest that Wnt2 may contribute to atherosclerotic plaque development and this study will render Wnt2 as a potential target for therapeutic intervention aiming at controlling atherosclerosis.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Samuel Rojas
- Department of Biological Sciences, College of Arts and Sciences, East Tennessee State University, Johnson City, TN, United States
| | - Sanjay Singh
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Phillip R Musich
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Matthew Gutierrez
- Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, TN, United States
| | - Zhiqiang Yao
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Douglas Thewke
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Yong Jiang
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
247
|
Stanic B, Kokai D, Tesic B, Fa S, Samardzija Nenadov D, Pogrmic-Majkic K, Andric N. Integration of data from the in vitro long-term exposure study on human endothelial cells and the in silico analysis: A case of dibutyl phthalate-induced vascular dysfunction. Toxicol Lett 2021; 356:64-74. [PMID: 34902519 DOI: 10.1016/j.toxlet.2021.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/22/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022]
Abstract
General population is exposed to dibutyl phthalate (DBP) through continuous use of various consumer products. DBP exhibits its effects mainly on the endocrine and reproductive system but it can also affect the function of the vasculature; however, the underlying mechanisms behind DBP-induced vascular dysfunction are not fully understood. To infer pathways, molecular functions, biological processes, and human diseases associated with DBP exposure, we integrated the toxicogenomic data obtained from the 4-week-long exposure of human vascular endothelial cells (ECs) to three environmentally relevant concentrations of DBP with the in silico analysis. Nine genes were affected by DBP exposure: six of the integrin family, VCAM1, ICAM1, and MMP2. As shown by the in silico analysis, changes in DBP-affected genes could affect extracellular matrix and binding of molecules and cells to ECs, thereby altering cell adhesion and migration. Several pathways, molecular functions, and biological processes were further identified to provide insight into the DBP-vascular disease relationships and the potential mechanism of action. The top three human disease categories associated with DBP exposure and vascular dysfunction include cardiovascular, cerebrovascular, and immune system diseases. Integration of experimental and in silico approaches may offer better understanding of the potential human health risks associated with DBP exposure.
Collapse
Affiliation(s)
- Bojana Stanic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | - Dunja Kokai
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | - Biljana Tesic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | - Svetlana Fa
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | | | | | - Nebojsa Andric
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia.
| |
Collapse
|
248
|
COVID-19 and Venous Thromboembolism: From Pathological Mechanisms to Clinical Management. J Pers Med 2021; 11:jpm11121328. [PMID: 34945800 PMCID: PMC8706381 DOI: 10.3390/jpm11121328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/02/2021] [Accepted: 12/04/2021] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), which is becoming a global pandemic, is caused by SARS-CoV-2 infection. In COVID-19, thrombotic events occur frequently, mainly venous thromboembolism (VTE), which is closely related to disease severity and clinical prognosis. Compared with historical controls, the occurrence of VTE in hospitalized and critical COVID-19 patients is incredibly high. However, the pathophysiology of thrombosis and the best strategies for thrombosis prevention in COVID-19 remain unclear, thus needing further exploration. Virchow’s triad elements have been proposed as important risk factors for thrombotic diseases. Therefore, the three factors outlined by Virchow can also be applied to the formation of venous thrombosis in the COVID-19 setting. A thorough understanding of the complex interactions in these processes is important in the search for effective treatments for COVID-19. In this work, we focus on the pathological mechanisms of VTE in COVID-19 from the aspects of endothelial dysfunction, hypercoagulability, abnormal blood flow. We also discuss the treatment of VTE as well as the ongoing clinical trials of heparin anticoagulant therapy. In addition, according to the pathophysiological mechanism of COVID-19-associated thrombosis, we extended the range of antithrombotic drugs including antiplatelet drugs, antifibrinolytic drugs, and anti-inflammatory drugs, hoping to find effective drug therapy and improve the prognosis of VTE in COVID-19 patients.
Collapse
|
249
|
Khismatullin RR, Ponomareva AA, Nagaswami C, Ivaeva RA, Montone KT, Weisel JW, Litvinov RI. Pathology of lung-specific thrombosis and inflammation in COVID-19. J Thromb Haemost 2021; 19:3062-3072. [PMID: 34538029 PMCID: PMC8646730 DOI: 10.1111/jth.15532] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/29/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Infection by SARS-CoV-2 produces significant pulmonary pathology including endothelial damage with resultant thrombotic events. While pathologic features were described, there are limited data on the relationship of these changes to the inflammatory response and the production of thromboses. OBJECTIVE To investigate pathology of COVID-19-related immunothrombosis. PATIENTS/METHODS Tissue samples from lung, kidney, brain and heart that were collected from 45 patients who died of COVID-19. Histopathological examination was performed after H&E and Picro-Mallory staining in combination with (immuno)fluorescence to visualize neutrophil extracellular traps. Ultrastructural alterations in lungs were studied with scanning and transmission electron microscopy. RESULTS Inflammatory changes and thrombosis were substantially more pronounced in the lung than in the kidney, heart, and brain. The most common pathologic finding was diffuse alveolar damage. In addition, most lung samples showed thrombi in vessels. The cause of death in single cases was massive pulmonary embolism. Ultrastructural examination revealed neutrophils attached to endothelium, perhaps as a step towards transendothelial migration. In addition, platelets were identified in the midst of fibrin as individual procoagulant balloon-like cells. Ultrastructural examination demonstrated numerous virion-like particles. CONCLUSIONS Studying (ultra)structural features of the autopsy lung samples from patients with COVID-19 has provided evidence for a pathogenic link between inflammation and thrombosis. The major features in the lungs of COVID-19 patients comprised primary inflammatory thrombosis associated with diffuse alveolar damage. The lungs had pronounced circulatory changes with inflammation-dependent intravascular blood clotting, whereas heart, brain, and kidneys had predominantly degenerative changes that were distinct from the lung pathology.
Collapse
Affiliation(s)
- Rafael R Khismatullin
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Anastasia A Ponomareva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
- Kazan Institute of Biochemistry and Biophysics, FRC KSC of RAS, Kazan, Russian Federation
| | - Chandrasekaran Nagaswami
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rozalina A Ivaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Kathleen T Montone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
250
|
Zaninotto M, Mion MM, Marchioro L, Padoan A, Plebani M. Endothelial dysfunction and Mid-Regional proAdrenomedullin: What role in SARS-CoV-2 infected Patients? Clin Chim Acta 2021; 523:185-190. [PMID: 34555411 PMCID: PMC8452353 DOI: 10.1016/j.cca.2021.09.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Endothelial dysfunction, a major complication of SARS-CoV-2 infectionplaying a key-role in multi-organ damage, carries high risk of mortality. AIM To investigate the potential role of Mid-Regional pro-Adrenomedullin (MR-proADM) in detecting endothelial damage with a view to stratifying the risk of adverse events (length of stay, death, admission in Intensive Care Unit) and/or disease resolution. MATERIALS AND METHODS In 135 consecutive patients with SARS-CoV-2 infection, MR-proADM was measured in EDTA-K2 plasma samples using B.R.A.H.M.S. KRYPTOR® COMPACT Plus method (Thermo Fisher Scientific, Hennigsdorf, Germany) RESULTS: Patients were subdivided into three groups based on their MR-proADM value (nmol/L): 1 (n = 20, MR-proADM ≤ 0.55); 2 (n = 82, 0.55 < MR-proADM ≤ 1.50); 3 (n = 33, MR-proADM > 1.50). The higher the MR-proADM value, the greater the patients' age, the more frequent the occurrence of pneumonia, the requiring of more aggressive treatment, the longer the hospitalization and the more frequent a fatal event. Significant differences were found between the three groups for MR-proADM, White-blood cell count, Neutrophil count, D-dimer, C-reactive Protein, Procalcitonin and hs-Troponin I. At logistic regression,it was found that MR-proADM and Log10D-dimer were the most significant predictors of adverse events. CONCLUSION The findings made in the present study highlight the relevance of MR-proADM values in providing clinically useful information, particularly for stratifying COVID-19 patients according to the risk of a more severe form of disease and to the development of adverse events.
Collapse
Affiliation(s)
- Martina Zaninotto
- Department of Laboratory Medicine, University-Hospital, Padua, Italy.
| | - Monica Maria Mion
- Department of Laboratory Medicine, University-Hospital, Padua, Italy
| | - Lucio Marchioro
- Department of Laboratory Medicine, University-Hospital, Padua, Italy
| | - Andrea Padoan
- Department of Laboratory Medicine, University-Hospital, Padua, Italy
| | - Mario Plebani
- Department of Laboratory Medicine, University-Hospital, Padua, Italy; Department of Medicine-DIMED, University of Padua, Italy
| |
Collapse
|