101
|
Valentini A, Cardillo C, Della Morte D, Tesauro M. The Role of Perivascular Adipose Tissue in the Pathogenesis of Endothelial Dysfunction in Cardiovascular Diseases and Type 2 Diabetes Mellitus. Biomedicines 2023; 11:3006. [PMID: 38002006 PMCID: PMC10669084 DOI: 10.3390/biomedicines11113006] [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: 09/25/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
Cardiovascular diseases (CVDs) and type 2 diabetes mellitus (T2DM) are two of the four major chronic non-communicable diseases (NCDs) representing the leading cause of death worldwide. Several studies demonstrate that endothelial dysfunction (ED) plays a central role in the pathogenesis of these chronic diseases. Although it is well known that systemic chronic inflammation and oxidative stress are primarily involved in the development of ED, recent studies have shown that perivascular adipose tissue (PVAT) is implicated in its pathogenesis, also contributing to the progression of atherosclerosis and to insulin resistance (IR). In this review, we describe the relationship between PVAT and ED, and we also analyse the role of PVAT in the pathogenesis of CVDs and T2DM, further assessing its potential therapeutic target with the aim of restoring normal ED and reducing global cardiovascular risk.
Collapse
Affiliation(s)
- Alessia Valentini
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (A.V.); (D.D.M.)
| | - Carmine Cardillo
- Department of Aging, Policlinico A. Gemelli IRCCS, 00168 Roma, Italy;
- Department of Translational Medicine and Surgery, Catholic University, 00168 Rome, Italy
| | - David Della Morte
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (A.V.); (D.D.M.)
| | - Manfredi Tesauro
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (A.V.); (D.D.M.)
| |
Collapse
|
102
|
Mohammed AQ, Abdu FA, Liu L, Yin G, Mareai RM, Mohammed AA, Xu Y, Che W. Coronary microvascular dysfunction and myocardial infarction with non-obstructive coronary arteries: Where do we stand? Eur J Intern Med 2023; 117:8-20. [PMID: 37482469 DOI: 10.1016/j.ejim.2023.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/15/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
In the past decade, scientific and clinical research has provided a translational perspective on myocardial infarction (MI) with non-obstructive coronary arteries (MINOCA). MINOCA is characterized by clinical documentation of an acute MI but angiography shows no significant coronary artery obstruction (stenosis <50%). The prevalence of MINOCA is estimated to range from approximately 6 to 10% among MI patients, and those with this condition have a poor prognosis, experiencing high rates of mortality, rehospitalization, and socioeconomic burden. MINOCA represents a major unmet need in cardiovascular medicine, with uncertain clinical management. It is a complex condition that can be caused by various factors, including atherosclerosis, plaque rupture, coronary vasospasm, and microvascular dysfunction. Effective management of MINOCA depends on identifying the underlying mechanism of the infarction, thus a systematic diagnostic approach is recommended. Contemporary data shows that a significant number of patients exhibit structural and functional abnormalities in coronary microcirculation, which is referred to as coronary microvascular dysfunction (CMD). CMD plays a crucial role in patients with signs and symptoms of myocardial ischemia and non-obstructive coronary artery stenosis, including MINOCA. Furthermore, conducting a thorough evaluation of coronary function can have significant prognostic and therapeutic implications, since personalized patient management strategies based on this assessment have been shown to improve symptoms and prognosis. Therefore, an accurate and timely diagnosis of CMD is essential for effective patient management, which can be achieved through various invasive and non-invasive methods. This review will discuss the pathophysiological understanding, current diagnostic techniques, and management strategies of patients with MINOCA and CMD.
Collapse
Affiliation(s)
- Abdul-Quddus Mohammed
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fuad A Abdu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guoqing Yin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Redhwan M Mareai
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ayman A Mohammed
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Department of Cardiology, Shanghai Tenth People's Hospital Chongming Branch, Shanghai, China.
| |
Collapse
|
103
|
Abagnale L, Candia C, Motta A, Galloway B, Ambrosino P, Molino A, Maniscalco M. Flow-mediated dilation as a marker of endothelial dysfunction in pulmonary diseases: A narrative review. Respir Med Res 2023; 84:101049. [PMID: 37826872 DOI: 10.1016/j.resmer.2023.101049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/12/2023] [Accepted: 09/04/2023] [Indexed: 10/14/2023]
Abstract
The endothelium is an active and crucial component of vessels and produces several key regulatory factors for the homeostasis of the entire organism. Endothelial function can be investigated invasively or non-invasively, both in the coronary and peripheral circulation. A widely accepted method for the assessment of endothelial function is measurement of flow-mediated dilation (FMD), which evaluates the vascular response to changes in blood flow. In this current review, we describe FMD applications in the clinical setting of different respiratory diseases: acute SARS-COV2 infection, pulmonary embolism; post-acute SARS-COV2 infection, Chronic Obstructive Pulmonary Disease, Obstructive Sleep Apneas Syndrome, Pulmonary Hypertension, Interstitial Lung Diseases. Emerging evidence shows that FMD might be an effective tool to assess the cardiovascular risk in patients suffering from the undermentioned respiratory diseases as well as an independent predictive factor of disease severity and/or recovery.
Collapse
Affiliation(s)
- Lucia Abagnale
- Department of Medicine and Surgery, Federico II University, Naples, Italy
| | - Claudio Candia
- Department of Medicine and Surgery, Federico II University, Naples, Italy
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli, Naples, Italy
| | - Brurya Galloway
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, Telese Terme, Italy
| | - Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, Directorate of Telese Terme Institute, Telese Terme, Italy
| | - Antonio Molino
- Department of Medicine and Surgery, Federico II University, Naples, Italy
| | - Mauro Maniscalco
- Department of Medicine and Surgery, Federico II University, Naples, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, Telese Terme, Italy.
| |
Collapse
|
104
|
Durukan E, Jensen CFS, Skaarup KG, Østergren PB, Sønksen J, Biering-Sørensen T, Fode M. Erectile Dysfunction Is Associated with Left Ventricular Diastolic Dysfunction: A Systematic Review and Meta-analysis. Eur Urol Focus 2023; 9:903-912. [PMID: 37355365 DOI: 10.1016/j.euf.2023.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/21/2023] [Accepted: 06/09/2023] [Indexed: 06/26/2023]
Abstract
CONTEXT Erectile dysfunction (ED) is associated with an increased risk of cardiovascular morbidity and mortality. OBJECTIVE To systematically review and analyze the cardiac structure and function in men with ED assessed with echocardiography. EVIDENCE ACQUISITION We performed a systematic review and meta-analysis according to the guideline of the Preferred Reporting Items for Systematic Reviews and Meta-analyses. We searched PubMed and the Cochrane Library on June 2, 2022, and included studies evaluating cardiac structure and function using echocardiography in men with ED compared with controls without ED. The Newcastle-Ottawa Quality Assessment Scale was used for assessing the quality of studies. We analyzed the mean differences in left ventricular ejection fraction (LVEF), the ratio of early transmitral filling velocity to early diastolic mitral annular velocity (E/e'), ratio of the early to late diastolic transmitral flow velocity (E/A), isovolumic relaxation time (IVRT), and left ventricular mass index (LVMi) in a random-effect model computed using means and standard deviations. The review was preregistered with PROSPERO (CRD42022337183). We received no funding. EVIDENCE SYNTHESIS We included ten studies with 763 men diagnosed with ED (mean age: 55.6 yr) and 358 control men (mean age: 54.4 yr). E/e' was significantly worse in men with ED than in controls (mean absolute difference = 1.17, 95% confidence interval or CI [0.68, 1.65], p < 0.005). No significant differences were observed in LVEF, E/A, IVRT, or LVMi (-0.06, 95% CI [-1.06, 0.95], p = 0.91; -0.06, 95% CI [-0.24, 0.13], p = 0.55; 11.76, 95% CI [-0.88, 24.39], p = 0.07; and 4.37, 95% CI [-2.91, 11.65], respectively). The studies exhibited heterogeneity regarding study populations, reported echocardiography data, and variations in adjustments for confounding factors. CONCLUSIONS Left ventricle diastolic dysfunction, as assessed by E/e', was more frequent in men with ED than in matched controls without ED. The results imply that echocardiography may be useful in the cardiovascular evaluation of men with ED to help identify myocardial impairment. PATIENT SUMMARY This study reviewed for the first time previous research on cardiac structure and function in men with erectile dysfunction (ED), as assessed by echocardiography. We found that men with ED, compared with men without ED, had a higher ratio of early transmitral filling velocity to early diastolic mitral annular velocity , indicating a potentially higher rate of impaired diastolic function-a potential early indicator of heart disease. Identification of early signs of heart problems in men with ED may help initiate necessary lifestyle modifications or preventative therapies before the development of heart disease. However, more research is required to determine the clinical utility of using echocardiography as a risk assessment method.
Collapse
Affiliation(s)
- Emil Durukan
- Department of Urology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Copenhagen, Denmark.
| | | | | | - Peter Busch Østergren
- Department of Urology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jens Sønksen
- Department of Urology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Tor Biering-Sørensen
- Department of Cardiology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Fode
- Department of Urology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
105
|
Nasoni MG, Crinelli R, Iuliano L, Luchetti F. When nitrosative stress hits the endoplasmic reticulum: Possible implications in oxLDL/oxysterols-induced endothelial dysfunction. Free Radic Biol Med 2023; 208:178-185. [PMID: 37544487 DOI: 10.1016/j.freeradbiomed.2023.08.008] [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: 06/12/2023] [Revised: 07/14/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Oxidized LDL (oxLDL) and oxysterols are known to play a crucial role in endothelial dysfunction (ED) by inducing endoplasmic reticulum stress (ERS), inflammation, and apoptosis. However, the precise molecular mechanisms underlying these pathophysiological processes remain incompletely understood. Emerging evidence strongly implicates excessive nitric oxide (NO) production in the progression of various pathological conditions. The accumulation of reactive nitrogen species (RNS) leading to nitrosative stress (NSS) and aberrant protein S-nitrosylation contribute to NO toxicity. Studies have highlighted the involvement of NSS and S-nitrosylation in perturbing ER signaling through the modification of ER sensors and resident isomerases in neurons. This review focuses on the existing evidence that strongly associates NO with ERS and the possible implications in the context of ED induced by oxLDL and oxysterols. The potential effects of perturbed NO synthesis on signaling effectors linking NSS with ERS in endothelial cells are discussed to provide a conceptual framework for further investigations and the development of novel therapeutic strategies targeting ED.
Collapse
Affiliation(s)
- M G Nasoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - R Crinelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - L Iuliano
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Latina, Italy.
| | - F Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
106
|
Zhang X, Zheng Y, Wang Z, Gan J, Yu B, Lu B, Jiang X. Melatonin as a therapeutic agent for alleviating endothelial dysfunction in cardiovascular diseases: Emphasis on oxidative stress. Biomed Pharmacother 2023; 167:115475. [PMID: 37722190 DOI: 10.1016/j.biopha.2023.115475] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/20/2023] Open
Abstract
The vascular endothelium is vital in maintaining cardiovascular health by regulating vascular permeability and tone, preventing thrombosis, and controlling vascular inflammation. However, when oxidative stress triggers endothelial dysfunction, it can lead to chronic cardiovascular diseases (CVDs). This happens due to oxidative stress-induced mitochondrial dysfunction, inflammatory responses, and reduced levels of nitric oxide. These factors cause damage to endothelial cells, leading to the acceleration of CVD progression. Melatonin, a natural antioxidant, has been shown to inhibit oxidative stress and stabilize endothelial function, providing cardiovascular protection. The clinical application of melatonin in the prevention and treatment of CVDs has received widespread attention. In this review, based on bibliometric studies, we first discussed the relationship between oxidative stress-induced endothelial dysfunction and CVDs, then summarized the role of melatonin in the treatment of atherosclerosis, hypertension, myocardial ischemia-reperfusion injury, and other CVDs. Finally, the potential clinical use of melatonin in the treatment of these diseases is discussed.
Collapse
Affiliation(s)
- Xiaolu Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Yujia Zheng
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Ziyu Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Jiali Gan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Bin Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Bin Lu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Xijuan Jiang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
107
|
Hamrangsekachaee M, Wen K, Yazdani N, Willits RK, Bencherif SA, Ebong EE. Endothelial glycocalyx sensitivity to chemical and mechanical sub-endothelial substrate properties. Front Bioeng Biotechnol 2023; 11:1250348. [PMID: 38026846 PMCID: PMC10643223 DOI: 10.3389/fbioe.2023.1250348] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Glycocalyx (GCX) is a carbohydrate-rich structure that coats the surface of endothelial cells (ECs) and lines the blood vessel lumen. Mechanical perturbations in the vascular environment, such as blood vessel stiffness, can be transduced and sent to ECs through mechanosensors such as GCX. Adverse stiffness alters GCX-mediated mechanotransduction and leads to EC dysfunction and eventually atherosclerotic cardiovascular diseases. To understand GCX-regulated mechanotransduction events, an in vitro model emulating in vivo vessel conditions is needed. To this end, we investigated the impact of matrix chemical and mechanical properties on GCX expression via fabricating a tunable non-swelling matrix based on the collagen-derived polypeptide, gelatin. To study the effect of matrix composition, we conducted a comparative analysis of GCX expression using different concentrations (60-25,000 μg/mL) of gelatin and gelatin methacrylate (GelMA) in comparison to fibronectin (60 μg/mL), a standard coating material for GCX-related studies. Using immunocytochemistry analysis, we showed for the first time that different substrate compositions and concentrations altered the overall GCX expression on human umbilical vein ECs (HUVECs). Subsequently, GelMA hydrogels were fabricated with stiffnesses of 2.5 and 5 kPa, representing healthy vessel tissues, and 10 kPa, corresponding to diseased vessel tissues. Immunocytochemistry analysis showed that on hydrogels with different levels of stiffness, the GCX expression in HUVECs remained unchanged, while its major polysaccharide components exhibited dysregulation in distinct patterns. For example, there was a significant decrease in heparan sulfate expression on pathological substrates (10 kPa), while sialic acid expression increased with increased matrix stiffness. This study suggests the specific mechanisms through which GCX may influence ECs in modulating barrier function, immune cell adhesion, and mechanotransduction function under distinct chemical and mechanical conditions of both healthy and diseased substrates.
Collapse
Affiliation(s)
| | - Ke Wen
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
| | - Narges Yazdani
- Bioengineering Department, Northeastern University, Boston, MA, United States
| | - Rebecca K. Willits
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
| | - Sidi A. Bencherif
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
- Laboratoire de BioMécanique et BioIngénierie (BMBI), UMR CNRS, Sorbonne Universités, Université de Technologie of Compiègne (UTC), Compiègne, France
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| | - Eno E. Ebong
- Chemical Engineering Department, Northeastern University, Boston, MA, United States
- Bioengineering Department, Northeastern University, Boston, MA, United States
- Neuroscience Department, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
108
|
Allbritton-King JD, García-Cardeña G. Endothelial cell dysfunction in cardiac disease: driver or consequence? Front Cell Dev Biol 2023; 11:1278166. [PMID: 37965580 PMCID: PMC10642230 DOI: 10.3389/fcell.2023.1278166] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
The vascular endothelium is a multifunctional cellular system which directly influences blood components and cells within the vessel wall in a given tissue. Importantly, this cellular interface undergoes critical phenotypic changes in response to various biochemical and hemodynamic stimuli, driving several developmental and pathophysiological processes. Multiple studies have indicated a central role of the endothelium in the initiation, progression, and clinical outcomes of cardiac disease. In this review we synthesize the current understanding of endothelial function and dysfunction as mediators of the cardiomyocyte phenotype in the setting of distinct cardiac pathologies; outline existing in vivo and in vitro models where key features of endothelial cell dysfunction can be recapitulated; and discuss future directions for development of endothelium-targeted therapeutics for cardiac diseases with limited existing treatment options.
Collapse
Affiliation(s)
- Jules D. Allbritton-King
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Guillermo García-Cardeña
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
109
|
Le HH, Hagen MW, Louey S, Tavori H, Thornburg KL, Giraud GD, Hinds MT, Barnes AP. Development of a novel Guinea Pig model producing transgenerational endothelial transcriptional changes driven by maternal food restriction and a second metabolic insult of high fat diet. Front Physiol 2023; 14:1266444. [PMID: 37942229 PMCID: PMC10628814 DOI: 10.3389/fphys.2023.1266444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/26/2023] [Indexed: 11/10/2023] Open
Abstract
Developmental programming of chronic adverse cardiovascular health outcomes has been studied both using numerous human populations and an array of animal models. However, the mechanisms that produce transgenerational effects have been difficult to study due to a lack of developmentally relevant models. As such, how increased disease risk is carried to the second generation has been poorly studied. We hypothesized that the endothelium which mediates many acute and chronic vascular inflammatory responses is a key player in these effects, and epidemiological studies implicate transgenerational nutritional effects on endothelial health. To study the mutigenerational effects of maternal undernutrition on offspring endothelial health, we developed a model of transgenerational nutritional stress in guinea pigs, a translationally relevant precocial species with a relatively short lifespan. First- and second-generation offspring were subjected to a high fat diet in adolescence to exacerbate negative cardiovascular health. To assess transcriptional changes, we performed bulk RNA-sequencing in carotid artery endothelial cells, with groups stratified as prenatal control or food restricted, and postnatal control or high fat diet. We detected statistically significant gene alterations for each dietary permutation, some of which were unique to treatments and other transcriptional signatures shared by multiple or all conditions. These findings highlight a core group of genes altered by high fat diet that is shared by all cohorts and a divergence of transgenerational effects between the prenatal ad libitum and dietary restriction groups. This study establishes the groundwork for this model to be used to better understand the interplay of prenatal stress and genetic reprogramming.
Collapse
Affiliation(s)
- Hillary H. Le
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| | - Matthew W. Hagen
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| | - Samantha Louey
- Center for Developmental Health, Portland, OR, United States
- Knight Cardiovascular Institute, Portland, OR, United States
| | - Hagai Tavori
- Knight Cardiovascular Institute, Portland, OR, United States
| | - Kent L. Thornburg
- Center for Developmental Health, Portland, OR, United States
- Knight Cardiovascular Institute, Portland, OR, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
| | - George D. Giraud
- Center for Developmental Health, Portland, OR, United States
- Knight Cardiovascular Institute, Portland, OR, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
- VA Portland Health Care System, Portland, OR, United States
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
- Center for Developmental Health, Portland, OR, United States
| | | |
Collapse
|
110
|
Ermolinskiy P, Gurfinkel Y, Sovetnikov E, Lugovtsov A, Priezzhev A. Correlation between the Capillary Blood Flow Characteristics and Endothelium Function in Healthy Volunteers and Patients Suffering from Coronary Heart Disease and Atrial Fibrillation: A Pilot Study. Life (Basel) 2023; 13:2043. [PMID: 37895425 PMCID: PMC10608205 DOI: 10.3390/life13102043] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/26/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Coronary heart disease (CHD) and atrial fibrillation (AF) pose significant health risks and require accurate diagnostic tools to assess the severity and progression of the diseases. Traditional diagnostic methods have limitations in providing detailed information about blood flow characteristics, particularly in the microcirculation. This study's objective was to examine and compare the microcirculation in both healthy volunteers and patient groups with CHD and AF. Furthermore, this study aimed to identify a relationship between blood microcirculation parameters and endothelial function. Digital capillaroscopy was employed to assess the microcirculation parameters, for example, such as capillary blood flow velocity, the size of red blood cell aggregates, and the number of aggregates per min and per running mm. The results indicate significant alterations in blood flow characteristics among patients with CHD and AF compared to healthy volunteers. For example, capillary blood flow velocity is statistically significantly decreased in the case of CHD and AF compared to the healthy volunteers (p < 0.001). Additionally, the correlation between the measured parameters is different for the studied groups of patients and healthy volunteers. These findings highlight the potential of digital capillaroscopy as a non-invasive tool for evaluating blood flow abnormalities (red blood cell aggregates and decreased capillary blood flow velocity) in cardiovascular diseases, aiding in early diagnosis and disease management.
Collapse
Affiliation(s)
- Petr Ermolinskiy
- Department of Physics, Lomonosov Moscow State University, 1-2 Leninskie Gory, Moscow 119991, Russia; (P.E.); (A.L.)
| | - Yury Gurfinkel
- Medical Research and Education Center, Lomonosov Moscow State University, 27-10 Lomonosovsky pr-t, Moscow 119991, Russia; (Y.G.); (E.S.)
| | - Egor Sovetnikov
- Medical Research and Education Center, Lomonosov Moscow State University, 27-10 Lomonosovsky pr-t, Moscow 119991, Russia; (Y.G.); (E.S.)
| | - Andrei Lugovtsov
- Department of Physics, Lomonosov Moscow State University, 1-2 Leninskie Gory, Moscow 119991, Russia; (P.E.); (A.L.)
| | - Alexander Priezzhev
- Department of Physics, Lomonosov Moscow State University, 1-2 Leninskie Gory, Moscow 119991, Russia; (P.E.); (A.L.)
| |
Collapse
|
111
|
Jin L, Wu L, Chen J, Zhang M, Sun J, Shen C, Du L, She X, Li Z. Uncoupling of the center-to-periphery arterial stiffness gradient and pulse pressure amplification in viral pneumonia infection. BMC Infect Dis 2023; 23:657. [PMID: 37798630 PMCID: PMC10552441 DOI: 10.1186/s12879-023-08650-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023] Open
Abstract
OBJECTIVES Arterial stiffness is a common manifestation of viral pneumonia infections, including COVID-19. Nevertheless, the relationship between the center-to-periphery arterial stiffness gradient and pulse pressure amplification (PPA) in infectious diseases remains unclear. This study aimed to investigate this relationship utilizing arterial pressure volume index (API) and arterial velocity pulse index (AVI) ratio. METHODS API/AVI and PPA were measured in 219 participants with COVID-19 and 374 normal participants. Multiple linear regression was used to assess the association of API/AVI and PPA, and restricted cubic spline was used to investigate the non-linear relationship between API/AVI and PPA. Receiver operating characteristic curve (ROC) analysis was used to evaluate the effects of API/AVI in identifying COVID-19 infection and severe stage. RESULTS There was a significant J-shaped relationship between API/AVI and PPA in COVID-19 group, while a M-shaped relationship was observed in normal group. API/AVI decreased rapidly as PPA decreased until API/AVI decreased slowly at PPA of 1.07, and then API/AVI decreased slowly again at PPA of 0.78. ROC results showed that API/AVI demonstrated excellent accuracy in identifying COVID-19 infection (AUC = 0.781) and a high specificity (84.88%) in identifying severe stage. CONCLUSIONS There was a J-shaped association between the API/AVI and PPA in viral infected patients, while a M-shaped relationship in the normal participants. API/AVI is better for identifying infected and uninfected patients, with a high specificity in identifying those in severe stages of the disease. The attenuation or reversal of API/AVI may be associated with the loss of PPA coupling.
Collapse
Affiliation(s)
- Lin Jin
- Department of Ultrasound, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200052, China
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Lingheng Wu
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
- Department of Ultrasound, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China
| | - Jianxiong Chen
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
- Department of Ultrasound, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China
| | - Mengjiao Zhang
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Jiali Sun
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Cuiqin Shen
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Lianfang Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xiaoyin She
- Department of Emergency and Critical Care, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Zhaojun Li
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China.
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
112
|
Shechter M, Natanzon SS, Lerman A, Cohn H, Prasad M, Goitein O, Goldkorn R, Naroditsky M, Koren-Morag N, Matetzky S. Endothelial function predicts 5-year adverse outcome in patients hospitalized in an emergency department chest pain unit. J Cardiovasc Med (Hagerstown) 2023; 24:729-736. [PMID: 37222628 DOI: 10.2459/jcm.0000000000001502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
BACKGROUND Although endothelial function is a marker for cardiovascular risk, endothelial dysfunction assessment is not routinely used in daily clinical practice. A growing challenge has emerged in identifying patients prone to cardiovascular events. We aim to investigate whether abnormal endothelial function may be associated with adverse 5-year outcomes in patients presenting to a chest pain unit (CPU). METHODS Following endothelial function testing using EndoPAT 2000 in 300 consecutive patients without a history of coronary artery disease, patients underwent coronary computerized tomographic angiography (CCTA) or single-photon emission computed tomography according to availability. RESULTS Mean 10-year Framingham risk score (FRS) was 6.6 ± 5.9%; mean 10-year atherosclerotic cardiovascular disease (ASCVD) risk was 7.1 ± 7.2%; median reactive hyperemia index (RHI) as a measure of an endothelial function 2.0 and mean was 2.0 ± 0.4. During a 5-year follow-up, the 30 patients who developed major adverse cardiovascular events (MACE), including all-cause mortality, nonfatal myocardial infarction, hospitalization for heart failure or angina pectoris, stroke, coronary artery bypass grafting, and percutaneous coronary interventions, had higher 10-year FRS (9.6 ± 7.8 vs. 6.3 ± 5.6%; P = 0.032), higher 10-year ASCVD risk (10.4 ± 9.2 vs. 6.7 ± 6.9%; P = 0.042), lower baseline RHI (1.6 ± 0.5 vs. 2.1 ± 0.4; P < 0.001) and a greater degree of coronary atherosclerotic lesions (53 vs. 3%, P < 0.001) on CCTA compared with patients without MACE. Multivariate analysis demonstrated that RHI below the median was an independent predictor of 5-year MACE (odds ratio 5.567, 95% confidence interval 1.955-15.853; P = 0.001). CONCLUSION Our findings suggest that noninvasive endothelial function testing may contribute to clinical efficacy in triaging patients in the CPU and in predicting 5-year MACE. CLINICAL TRIALSGOV IDENTIFIER NCT01618123.
Collapse
Affiliation(s)
- Michael Shechter
- Leviev Heart and Vascular Center
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Shalom Natanzon
- Leviev Heart and Vascular Center
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo Clinic and College of Medicine, Rochester, USA
| | - Herold Cohn
- Leviev Heart and Vascular Center
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Megha Prasad
- Division of Cardiovascular Diseases, Mayo Clinic and College of Medicine, Rochester, USA
| | - Orly Goitein
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Diagnostic Imaging, Chaim Sheba Medical Center, Tel Hashomer, Tel Hashomer, Israel
| | - Ronen Goldkorn
- Leviev Heart and Vascular Center
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Naroditsky
- Leviev Heart and Vascular Center
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nira Koren-Morag
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shlomi Matetzky
- Leviev Heart and Vascular Center
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
113
|
Atawia RT, Batori R, Jordan CR, Kennard S, Antonova G, Bruder-Nascimento T, Mehta V, Saeed MI, Patel VS, Fukai T, Ushio-Fukai M, Huo Y, Fulton DJR, de Chantemèle EJB. Type 1 Diabetes Impairs Endothelium-Dependent Relaxation Via Increasing Endothelial Cell Glycolysis Through Advanced Glycation End Products, PFKFB3, and Nox1-Mediated Mechanisms. Hypertension 2023; 80:2059-2071. [PMID: 37729634 PMCID: PMC10514399 DOI: 10.1161/hypertensionaha.123.21341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 08/02/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Type 1 diabetes (T1D) is a major cause of endothelial dysfunction. Although cellular bioenergetics has been identified as a new regulator of vascular function, whether glycolysis, the primary bioenergetic pathway in endothelial cells (EC), regulates vascular tone and contributes to impaired endothelium-dependent relaxation (EDR) in T1D remains unknown. METHODS Experiments were conducted in Akita mice with intact or selective deficiency in EC PFKFB3 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3), the main regulator of glycolysis. Seahorse analyzer and myography were employed to measure glycolysis and mitochondrial respiration, and EDR, respectively, in aortic explants. EC PFKFB3 (Ad-PFKFB3) and glycolysis (Ad-GlycoHi) were increased in situ via adenoviral transduction. RESULTS T1D increased EC glycolysis and elevated EC expression of PFKFB3 and NADPH oxidase Nox1 (NADPH oxidase homolog 1). Functionally, pharmacological and genetic inhibition of PFKFB3 restored EDR in T1D, while in situ aorta EC transduction with Ad-PFKFB3 or Ad-GlycoHi reproduced the impaired EDR associated with T1D. Nox1 inhibition restored EDR in aortic rings from Akita mice, as well as in Ad-PFKFB3-transduced aorta EC and lactate-treated wild-type aortas. T1D increased the expression of the advanced glycation end product precursor methylglyoxal in the aortas. Exposure of the aortas to methylglyoxal impaired EDR, which was prevented by PFKFB3 inhibition. T1D and exposure to methylglyoxal increased EC expression of HIF1α (hypoxia-inducible factor 1α), whose inhibition blunted methylglyoxal-mediated EC PFKFB3 upregulation. CONCLUSIONS EC bioenergetics, namely glycolysis, is a new regulator of vasomotion and excess glycolysis, a novel mechanism of endothelial dysfunction in T1D. We introduce excess methylglyoxal, HIF1α, and PFKFB3 as major effectors in T1D-mediated increased EC glycolysis.
Collapse
Affiliation(s)
- Reem T. Atawia
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abasia, Cairo, Egypt
| | - Robert Batori
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Coleton R. Jordan
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Simone Kennard
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Galina Antonova
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | | | - Vinay Mehta
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Muhammad I. Saeed
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Vijay S Patel
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - David JR Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | | |
Collapse
|
114
|
Gao S, Chen H. Therapeutic potential of apelin and Elabela in cardiovascular disease. Biomed Pharmacother 2023; 166:115268. [PMID: 37562237 DOI: 10.1016/j.biopha.2023.115268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/12/2023] Open
Abstract
Apelin and Elabela (Ela) are peptides encoded by APLN and APELA, respectively, which act on their receptor APJ and play crucial roles in the body. Recent research has shown that they not only have important effects on the endocrine system, but also promote vascular development and maintain the homeostasis of myocardial cells. From a molecular biology perspective, we explored the roles of Ela and apelin in the cardiovascular system and summarized the mechanisms of apelin-APJ signaling in the progression of myocardial infarction, ischemia-reperfusion injury, atherosclerosis, pulmonary arterial hypertension, preeclampsia, and congenital heart disease. Evidences indicated that apelin and Ela play important roles in cardiovascular diseases, and there are many studies focused on developing apelin, Ela, and their analogues for clinical treatments. However, the literature on the therapeutic potential of apelin, Ela and their analogues and other APJ agonists in the cardiovascular system is still limited. This review summarized the regulatory pathways of apelin/ELA-APJ axis in cardiovascular function and cardiovascular-related diseases, and the therapeutic effects of their analogues in cardiovascular diseases were also included.
Collapse
Affiliation(s)
- Shenghan Gao
- Department of Histology and embryology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Hongping Chen
- Department of Histology and embryology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
115
|
Patterson PD, Friedman JC, Ding S, Miller RS, Martin-Gill C, Hostler D, Platt TE. Acute Effect of Night Shift Work on Endothelial Function with and without Naps: A Scoping Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6864. [PMID: 37835134 PMCID: PMC10572584 DOI: 10.3390/ijerph20196864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/23/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023]
Abstract
We examined the breadth and depth of the current evidence investigating napping/sleeping during night shift work and its impact on non-invasive measures of endothelial function. We used a scoping review study design and searched five databases: Ovid Medline, EMBASE, Ovid APA PsycInfo, Web of Science Core Collection, and EBSCO CINAHL. We limited our search to English language and publications from January 1980 to September 2022. Our reporting adhered to the PRISMA-ScR guidance for scoping reviews. Our search strategy yielded 1949 records (titles and abstracts) after deduplication, of which 36 were retained for full-text review. Five articles were retained, describing three observational and two experimental research studies with a total sample of 110 individuals, which examined the non-invasive indicators of endothelial function in relation to the exposure to night shift work. While there is some evidence of an effect of night shift work on the non-invasive indicators of endothelial function, this evidence is incomplete, limited to a small samples of shift workers, and is mostly restricted to one measurement technique for assessing endothelial function with diverse protocols. In addition, there is no identifiable research investigating the potential benefits of napping during night shift work on non-invasive measures of endothelial function.
Collapse
Affiliation(s)
- Paul D. Patterson
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Community Health Services and Rehabilitation Sciences, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jacob C. Friedman
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Community Health Services and Rehabilitation Sciences, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Samuel Ding
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Community Health Services and Rehabilitation Sciences, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Rebekah S. Miller
- Health Sciences Library System, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Christian Martin-Gill
- Department of Emergency Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David Hostler
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA;
| | - Thomas E. Platt
- Department of Community Health Services and Rehabilitation Sciences, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
116
|
Awad AM, Hansen K, Del Rio D, Flores D, Barghash RF, Kakkola L, Julkunen I, Awad K. Insights into COVID-19: Perspectives on Drug Remedies and Host Cell Responses. Biomolecules 2023; 13:1452. [PMID: 37892134 PMCID: PMC10604481 DOI: 10.3390/biom13101452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
In light of the COVID-19 global pandemic caused by SARS-CoV-2, ongoing research has centered on minimizing viral spread either by stopping viral entry or inhibiting viral replication. Repurposing antiviral drugs, typically nucleoside analogs, has proven successful at inhibiting virus replication. This review summarizes current information regarding coronavirus classification and characterization and presents the broad clinical consequences of SARS-CoV-2 activation of the angiotensin-converting enzyme 2 (ACE2) receptor expressed in different human cell types. It provides publicly available knowledge on the chemical nature of proposed therapeutics and their target biomolecules to assist in the identification of potentially new drugs for the treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ahmed M. Awad
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Kamryn Hansen
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Diana Del Rio
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Derek Flores
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Reham F. Barghash
- Institute of Chemical Industries Research, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Laura Kakkola
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
- Clinical Microbiology, Turku University Hospital, 20521 Turku, Finland
| | - Kareem Awad
- Institute of Biomedicine, Faculty of Medicine, University of Turku, 20014 Turku, Finland
- Department of Therapeutic Chemistry, Institute of Pharmaceutical and Drug Industries Research, National Research Center, Dokki, Cairo 12622, Egypt
| |
Collapse
|
117
|
Ait-Aissa K, Guo X, Klemmensen M, Leng LN, Koval OM, Grumbach IM. Short-term statin treatment reduces, and long-term statin treatment abolishes chronic vascular injury by radiation therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.558723. [PMID: 37790532 PMCID: PMC10542122 DOI: 10.1101/2023.09.20.558723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Background The incidental use of statins during radiation therapy has been associated with a reduced long-term risk of developing atherosclerotic cardiovascular disease. Objectives Determine if irradiation causes chronic vascular injury and whether short-term administration of statins during and after irradiation is sufficient to prevent chronic injury compared to long-term administration. Methods C57Bl/6 mice were pretreated with pravastatin for 72 hours and then exposed to 12 Gy x-ray head-and-neck irradiation. Subsequently, they received pravastatin either for one additional day or for one year. Carotid arteries were tested for vascular reactivity and altered gene expression one year after irradiation. Results Treatment with pravastatin for 24 hours reduced the loss of endothelium-dependent vasorelaxation and protected against enhanced vasoconstriction after IR. It reduced the expression of some markers associated with inflammation and oxidative stress and modulated that of subunits of the voltage and Ca2+ activated K+ (BK) channel in the carotid artery one year after irradiation. Treatment with pravastatin for one year completely reversed the changes caused by irradiation. Conclusions In mice, short-term administration of pravastatin is sufficient to reduce chronic vascular injury after irradiation. Long-term administration eliminates the effects of irradiation. These findings suggest that a prospective treatment strategy involving statins could be effective in patients undergoing radiation therapy. The optimal duration of treatment in humans has yet to be determined.
Collapse
Affiliation(s)
- Karima Ait-Aissa
- Abboud Cardiovascular Research Center, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Xutong Guo
- Abboud Cardiovascular Research Center, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Madelyn Klemmensen
- Abboud Cardiovascular Research Center, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Linette N. Leng
- Abboud Cardiovascular Research Center, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Olha M. Koval
- Abboud Cardiovascular Research Center, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Isabella M. Grumbach
- Abboud Cardiovascular Research Center, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA
- Iowa City VA Healthcare System, Iowa City, IA
| |
Collapse
|
118
|
Polecka A, Olszewska N, Danielski Ł, Olszewska E. Association between Obstructive Sleep Apnea and Heart Failure in Adults-A Systematic Review. J Clin Med 2023; 12:6139. [PMID: 37834783 PMCID: PMC10573908 DOI: 10.3390/jcm12196139] [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: 08/23/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Heart failure (HF) patients commonly experience obstructive sleep apnea (OSA), which may worsen their condition. We reviewed a diverse range of studies to investigate the prevalence of OSA in HF patients, the effects of positive airway pressure (PAP) treatment, and the potential impact of sodium-glucose cotransporter-2 inhibitors (SGLT2i) and sacubitril/valsartan on OSA outcomes. METHODS We analyzed case-control, observational studies, and randomized controlled trials. Prevalence rates, PAP treatment, and HF pharmacotherapy were assessed. RESULTS Numerous studies revealed a high prevalence of OSA in HF patients, particularly with preserved ejection fraction. PAP treatment consistently improved an apnea-hypopnea index, left ventricular ejection fraction, oxygen saturation, and overall quality of life. Emerging evidence suggests that SGLT2i and sacubitril/valsartan might influence OSA outcomes through weight loss, improved metabolic profiles, and potential direct effects on upper airway muscles. CONCLUSIONS The complex interplay between OSA and HF necessitates a multifaceted approach. PAP treatment has shown promising results in improving OSA symptoms and HF parameters. Additionally, recent investigations into the effects of HF pharmacotherapy on OSA suggest their potential as adjunctive therapy. This review provides insights for clinicians and researchers, highlighting the importance of addressing OSA and HF in patient management strategies.
Collapse
Affiliation(s)
- Agnieszka Polecka
- Doctoral School of the Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Natalia Olszewska
- Student Research Group, Department of Otolaryngology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Łukasz Danielski
- Student Research Group, Department of Otolaryngology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Ewa Olszewska
- Sleep Apnea Surgery Center, Department of Otolaryngology, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
119
|
Miao R, Yu R, Zhou H, Liu L, Peng T, Wang J. Novel use of structural equation modelling to examine diet and metabolic traits associated with microvascular endothelial dysfunction in middle-aged Chinese males: a cross-sectional study. BMJ Open 2023; 13:e073357. [PMID: 37709331 PMCID: PMC10921913 DOI: 10.1136/bmjopen-2023-073357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND The present study aims to use structural equation modelling (SEM) with multiple regression pathways to examine direct and indirect links from diet and metabolic traits to microvascular endothelial dysfunction (ED) among middle-aged Chinese males. METHODS The study was conducted in middle-aged Chinese males, who underwent a health check-up between 2018 and 2019. Data on lifestyle behaviour factors (physical activity, diet pattern, sleep quality and diet data underwent factor analysis in advance) and metabolic risk factors referring to metabolic traits were introduced into the SEM to examine inter-relationship among these factors and their association with ED, as evaluated by the reactive hyperaemia index (RHI). RESULTS Both exploratory factor analysis and confirmatory factor analysis identified two major dietary patterns: 'prudent pattern' and 'western pattern'. The univariate test suggested that only triglycerides (TGs) and prudent dietary pattern were directly associated with RHI. Furthermore, prudent dietary pattern had an indirect association with RHI via TG (prudent diet→TG: β=-0.15, p<0.05; TG→RHI: β=-0.17, p<0.001). As to confirming the hypothesised association between variables apart, physical activity frequency was correlated to the decrease in TG (β=-0.29, p<0.001), but had no direct correlation to RHI. CONCLUSION The network of direct and indirect associations among diet pattern and cardiometabolic risk factors with RHI measured ED among middle-aged males. The most significant modifiable factors identified were TG and prudent diet pattern, which needs to be targeted as preventive strategies for early microvascular impairment.
Collapse
Affiliation(s)
- Rujia Miao
- Health Management Center, Central South University Third Xiangya Hospital, Changsha, China
| | - Renhe Yu
- Department of Epidemiology and Health Statistics, Central South University, Changsha, China
| | - Hui Zhou
- Health Management Center, Central South University Third Xiangya Hospital, Changsha, China
| | - Lei Liu
- Health Management Center, Central South University Third Xiangya Hospital, Changsha, China
| | - Ting Peng
- Health Management Center, Central South University Third Xiangya Hospital, Changsha, China
| | - Jiangang Wang
- Health Management Center, Central South University Third Xiangya Hospital, Changsha, China
| |
Collapse
|
120
|
Fiedorczuk P, Olszewska E, Polecka A, Walasek M, Mroczko B, Kulczyńska-Przybik A. Investigating the Role of Serum and Plasma IL-6, IL-8, IL-10, TNF-alpha, CRP, and S100B Concentrations in Obstructive Sleep Apnea Diagnosis. Int J Mol Sci 2023; 24:13875. [PMID: 37762178 PMCID: PMC10530258 DOI: 10.3390/ijms241813875] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a prevalent and underdiagnosed condition associated with cardiovascular diseases, depression, accidents, and stroke. There is an increasing need for alternative diagnostic tools beyond overnight sleep studies that measure the Apnea/Hypopnea Index (AHI). In this single-center, case-control study, we evaluated serum and plasma concentrations of IL-6, IL-8, IL-10, TNF-α, CRP, and S100B in 80 subjects, including 52 OSA patients (27 moderate [15 ≤ AHI ˂ 30], 25 severe [AHI ≥ 30]) and 28 non-OSA controls (AHI 0-5). Participants with OSA showed approximately 2 times higher median concentrations of CRP in plasma, and IL-6 in serum, as well as 1.3 to 1.7 times higher concentrations of TNF-α and IL-8 in plasma compared with the control group. Receiver Operator Characteristic (ROC) curve analysis was performed to evaluate the predictive capabilities of these serum and plasma biomarkers in distinguishing between the OSA and control groups, revealing varying sensitivity and specificity. In summary, in this study, serum and plasma biomarkers CRP, S100B, IL-6, TNF-α, and IL-8 have been shown to be elevated in patients with OSA, correlated positively with disease severity, age, and BMI. These results support the potential role of these biomarkers in diagnosing OSA, supplementing traditional methods such as overnight sleep studies.
Collapse
Affiliation(s)
- Piotr Fiedorczuk
- Department of Otolaryngology, Medical University of Bialystok, 15-089 Bialystok, Poland; (P.F.); (M.W.)
| | - Ewa Olszewska
- Department of Otolaryngology, Medical University of Bialystok, 15-089 Bialystok, Poland; (P.F.); (M.W.)
| | - Agnieszka Polecka
- Doctoral School of the Medical, University of Bialystok, 15-089 Bialystok, Poland;
| | - Marzena Walasek
- Department of Otolaryngology, Medical University of Bialystok, 15-089 Bialystok, Poland; (P.F.); (M.W.)
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok 15-089 Bialystok, Poland; (B.M.); (A.K.-P.)
| | - Agnieszka Kulczyńska-Przybik
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok 15-089 Bialystok, Poland; (B.M.); (A.K.-P.)
| |
Collapse
|
121
|
Liu C, Lei S, Cai T, Cheng Y, Bai J, Fu W, Huang M. Inducible nitric oxide synthase activity mediates TNF-α-induced endothelial cell dysfunction. Am J Physiol Cell Physiol 2023; 325:C780-C795. [PMID: 37575057 DOI: 10.1152/ajpcell.00153.2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 08/15/2023]
Abstract
Inducible nitric oxide synthase (iNOS) and vascular endothelial dysfunction have been implicated in the development and progression of atherosclerosis. This study aimed to elucidate the role of iNOS in vascular endothelial dysfunction. Ultrahigh performance liquid chromatography-quadrupole time-of-flight mass spectrometry combined with multivariate data analysis was used to characterize the metabolic changes in human umbilical vein endothelial cells (HUVECs) in response to different treatment conditions. In addition, molecular biology techniques were employed to explain the molecular mechanisms underlying the role of iNOS in vascular endothelial dysfunction. Tumor necrosis factor-α (TNF-α) enhances the expression of iNOS, TXNIP, and the level of reactive oxygen species (ROS) facilitates the entry of nuclear factor-κB (NF-κB) into the nucleus and promotes injury in HUVECs. iNOS deficiency reversed the TNF-α-mediated pathological changes in HUVECs. Moreover, TNF-α increased the expression of tumor necrosis factor receptor-2 (TNFR-2) and the levels of p-IκBα and IL-6 proteins and CD31, ICAM-1, and VCAM-1 protein expression, which was significantly reduced in HUVECs with iNOS deficiency. In addition, treating HUVECs in the absence or presence of TNF-α or iNOS, respectively, enabled the identification of putative endogenous biomarkers associated with endothelial dysfunction. These biomarkers were involved in critical metabolic pathways, including glycosylphosphatidylinositol-anchor biosynthesis, amino acid metabolism, sphingolipid metabolism, and fatty acid metabolism. iNOS deficiency during vascular endothelial dysfunction may affect the expression of TNFR-2, vascular adhesion factors, and the level of ROS via cellular metabolic changes, thereby attenuating vascular endothelial dysfunction.NEW & NOTEWORTHY Inducible nitric oxide synthase (iNOS) deficiency during vascular endothelial dysfunction may affect the expression of tumor necrosis factor receptor-2 and vascular adhesion factors via cellular metabolic changes, thereby attenuating vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Chen Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Sujuan Lei
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Tianying Cai
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yonglang Cheng
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Junjie Bai
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Meizhou Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
122
|
Knowles KA, Stewart KJ, Ouyang P, Magliato K, Whitt MD, Silber HA. A Novel Noninvasive Device and Method for Assessing Endothelial Function Is Associated With Measures of Obesity and Fitness. J Cardiopulm Rehabil Prev 2023; 43:390-391. [PMID: 37311026 DOI: 10.1097/hcr.0000000000000805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Affiliation(s)
- Kellen A Knowles
- Division of Cardiology, Department of Medicine (Drs Knowles, Stewart, Ouyang, and Silber), Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Cardiothoracic Surgery, Providence Saint John's Health Center, Santa Monica, California (Dr Magliato); and Department of Biomedical Engineering, California Polytechnic State University, San Luis Obispo, California (Dr Whitt)
| | | | | | | | | | | |
Collapse
|
123
|
Zhao J, Xie Y, Meng Z, Liu C, Wu Y, Zhao F, Ma X, Christopher TA, Lopez BJ, Wang Y. COVID-19 and cardiovascular complications: updates of emergency medicine. EMERGENCY AND CRITICAL CARE MEDICINE 2023; 3:104-114. [PMID: 38314258 PMCID: PMC10836842 DOI: 10.1097/ec9.0000000000000095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and SARS-CoV-2 variants, has become a global pandemic resulting in significant morbidity and mortality. Severe cases of COVID-19 are characterized by hypoxemia, hyper-inflammation, cytokine storm in lung. Clinical studies have reported an association between COVID-19 and cardiovascular disease (CVD). Patients with CVD tend to develop severe symptoms and mortality if contracted COVID-19 with further elevations of cardiac injury biomarkers. Furthermore, COVID-19 itself can induce and promoted CVD development, including myocarditis, arrhythmia, acute coronary syndrome, cardiogenic shock, and venous thromboembolism. Although the direct etiology of SARS-CoV-2 induced cardiac injury remains unknown and under-investigated, it is suspected that it is related to myocarditis, cytokine-mediated injury, microvascular injury, and stress-related cardiomyopathy. Despite vaccinations having provided the most effective approach to reducing mortality overall, an adapted treatment paradigm and regular monitoring of cardiac injury biomarkers is critical for improving outcomes in vulnerable populations at risk for severe COVID-19. In this review, we focus on the latest progress in clinic and research on the cardiovascular complications of COVID-19 and provide a perspective of treating cardiac complications deriving from COVID-19 in Emergency Medicine.
Collapse
Affiliation(s)
- Jianli Zhao
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, USA
| | - Yaoli Xie
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhijun Meng
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | - Caihong Liu
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yalin Wu
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, USA
| | - Fujie Zhao
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, USA
| | - Xinliang Ma
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Bernard J. Lopez
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yajing Wang
- Emergency Medicine Department, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL, USA
| |
Collapse
|
124
|
Lazaro CM, Victorio JA, Davel AP, Oliveira HCF. CETP expression ameliorates endothelial function in female mice through estrogen receptor-α and endothelial nitric oxide synthase pathway. Am J Physiol Heart Circ Physiol 2023; 325:H592-H600. [PMID: 37539470 DOI: 10.1152/ajpheart.00365.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/05/2023]
Abstract
Endothelial dysfunction is an early manifestation of atherosclerosis. The cholesteryl ester transfer protein (CETP) has been considered proatherogenic by reducing plasma HDL levels. However, CETP may exhibit cell- or tissue-specific effects. We have previously reported that male mice expressing the human CETP gene show impaired endothelium-mediated vascular relaxation associated with oxidative stress. Although sexual dimorphisms on the metabolic role of CETP have been proposed, possible sex differences in the vascular effects of CETP were not previously studied. Thus, here we investigated the endothelial function of female CETP transgenic mice as compared with nontransgenic controls (NTg). Aortas from CETP females presented preserved endothelium-dependent relaxation to acetylcholine and an endothelium-dependent reduction of phenylephrine-induced contraction. eNOS phosphorylation (Ser1177) and calcium-induced NO levels were enhanced, whereas reactive oxygen species (ROS) production and NOX2 and SOD2 expression were reduced in the CETP female aortas. Furthermore, CETP females exhibited increased aortic relaxation to 17β-estradiol (E2) and upregulation of heat shock protein 90 (HSP90) and caveolin-1, proteins that stabilize estrogen receptor (ER) in the caveolae. Indeed, CETP females showed an increased E2-induced relaxation in a manner sensitive to estrogen receptor-α (ERα) and HSP90 inhibitors methylpiperidinopyrazole (MPP) and geldanamycin, respectively. MPP also impaired the relaxation response to acetylcholine in CETP but not in NTg females. Altogether, the study indicates that CETP expression ameliorates the anticontractile endothelial effect and relaxation to E2 in females. This was associated with less ROS production, and increased eNOS-NO and E2-ERα pathways. These results highlight the need for considering the sex-specific effects of CETP on cardiovascular risk.NEW & NOTEWORTHY Here we demonstrated that CETP expression has a sex-specific impact on the endothelium function. Contrary to what was described for males, CETP-expressing females present preserved endothelium-dependent relaxation to acetylcholine and improved relaxation response to 17β-estradiol. This was associated with less ROS production, increased eNOS-derived NO, and increased expression of proteins that stabilize estrogen receptor-α (ERα), thus increasing E2-ERα signaling sensitivity. These results highlight the need for considering the sex-specific effects of CETP on cardiovascular risk.
Collapse
Affiliation(s)
- Carolina M Lazaro
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Jamaira A Victorio
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Ana Paula Davel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Helena C F Oliveira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| |
Collapse
|
125
|
Izquierdo-Gómez R, Esteban-Cornejo I, Cabanas-Sánchez V, Marcos A, Gómez-Martínez S, Castro-Piñero J, Veiga ÓL. Are obesity and physical fitness associated with cardiovascular disease risk in adolescents with Down syndrome? The longitudinal UP&DOWN study. JOURNAL OF INTELLECTUAL & DEVELOPMENTAL DISABILITY 2023; 48:247-259. [PMID: 39815918 DOI: 10.3109/13668250.2023.2188876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 02/21/2023] [Indexed: 01/18/2025]
Abstract
INTRODUCTION The aims of this study were to examine cross-sectional, and longitudinal association of different measures of obesity and physical fitness with cardiovascular disease (CVD) risk factors in adolescents with Down syndrome. METHODS Longitudinal analysis included 90 adolescents with Down syndrome. Obesity and physical fitness were measured following the ALPHA battery. Blood pressure and blood samplings (glucose, total cholesterol, high-density lipoprotein cholesterol, and serum lipid triglycerides levels (mg/dl) were determinated. RESULTS In cross-sectional analyses, obesity was positively associated with single CVD risk factors and CVD risk factor index (all p < .05). In the prospective analyses, obesity was not associated with single CVD risk factors, but components of physical fitness at baseline impact in some single CVD risk factors (all p < .05). CONCLUSION Obesity might be more strongly related to current cardiovascular-health, while components of physical fitness might be more related to future cardiovascular-health in adolescents with Down syndrome.
Collapse
Affiliation(s)
- Rocio Izquierdo-Gómez
- GALENO Research Group, Department of Physical Education, Faculty of Education Sciences, University of Cádiz, Cádiz, Spain
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Cádiz, Spain
| | - Irene Esteban-Cornejo
- PROFITH "PROmoting FITness and Health Through Physical Activity" Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical Education and Sports, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | | | - Ascensión Marcos
- Department of Nutrition and Metabolism (DMN), Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), Madrid, Spain
| | - Sonia Gómez-Martínez
- Department of Nutrition and Metabolism (DMN), Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), Madrid, Spain
| | - José Castro-Piñero
- GALENO Research Group, Department of Physical Education, Faculty of Education Sciences, University of Cádiz, Cádiz, Spain
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Cádiz, Spain
| | - Óscar L Veiga
- Department of Physical Education, Sport and Human Movement, Autonomous University of Madrid, Madrid, Spain
| |
Collapse
|
126
|
Gram-Kampmann EM, Olesen TB, Hansen CD, Hugger MB, Jensen JM, Handberg A, Beck-Nielsen H, Krag A, Olsen MH, Højlund K. A six-month low-carbohydrate diet high in fat does not adversely affect endothelial function or markers of low-grade inflammation in patients with type 2 diabetes: an open-label randomized controlled trial. Cardiovasc Diabetol 2023; 22:212. [PMID: 37592243 PMCID: PMC10436534 DOI: 10.1186/s12933-023-01956-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND While a low-carbohydrate diet (LCD) reduces HbA1c in patients with type 2 diabetes (T2D), the associated high intake of fat may adversely affect cardiovascular risk factors. To address this, we examined the effect of a non-calorie-restricted LCD high in fat on endothelial function and markers of low-grade inflammation in T2D over 6 months. METHODS In an open-label randomized controlled trial, 71 patients with T2D were randomized 2:1 to either a LCD (< 20 E% carbohydrates, 50-60 E% fat) or a control diet (50-60 E% carbohydrates, 20-30 E% fat) for six months. Flow-mediated vasodilation (FMD) and nitroglycerine-induced vasodilation (NID) were assessed by ultrasound in the brachial artery together with plasma interleukin-6 (IL-6) and serum high-sensitivity C-reactive protein (hsCRP) in the participants at baseline (n = 70) and after six months (n = 64). RESULTS The FMD and NID were unaltered in both groups after six months, and there were no between-group differences in change of either FMD (p = 0.34) or NID (p = 0.53) in response to the interventions. The circulating hsCRP and IL-6 levels decreased only in response to LCD (both p < 0.05). However, comparing changes over time with the control diet, the LCD did not reduce either IL-6 (p = 0.25) or hsCRP (p = 0.07) levels. The lack of changes in FMD and NID in response to LCD persisted after adjustment for cardiovascular risk factors. CONCLUSION A LCD high in fat for six months does not adversely affect endothelial function or selected markers of low-grade inflammation, which suggests that this nutritional approach does not increase the risk of cardiovascular disease. Trial registration ClinicalTrials.gov (NCT03068078).
Collapse
Affiliation(s)
- Eva M Gram-Kampmann
- Steno Diabetes Center Odense, Odense University Hospital, Kløvervænget 10, Entrance 112, 5000, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
| | - Thomas B Olesen
- Steno Diabetes Center Odense, Odense University Hospital, Kløvervænget 10, Entrance 112, 5000, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Camilla D Hansen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Mie B Hugger
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Jane M Jensen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Ålborg, Denmark
- Department of Clinical Medicine, Aalborg University, Ålborg, Denmark
| | - Henning Beck-Nielsen
- Steno Diabetes Center Odense, Odense University Hospital, Kløvervænget 10, Entrance 112, 5000, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Aleksander Krag
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Michael H Olsen
- Department of Internal Medicine 1, Holbæk Hospital, and Steno Diabetes Center Zealand, Holbæk, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Kløvervænget 10, Entrance 112, 5000, Odense, Denmark.
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
127
|
Santoro L, Zaccone V, Falsetti L, Ruggieri V, Danese M, Miro C, Di Giorgio A, Nesci A, D’Alessandro A, Moroncini G, Santoliquido A. Role of Endothelium in Cardiovascular Sequelae of Long COVID. Biomedicines 2023; 11:2239. [PMID: 37626735 PMCID: PMC10452509 DOI: 10.3390/biomedicines11082239] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
The global action against coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2 infection, shed light on endothelial dysfunction. Although SARS-CoV-2 primarily affects the pulmonary system, multiple studies have documented pan-vascular involvement in COVID-19. The virus is able to penetrate the endothelial barrier, damaging it directly or indirectly and causing endotheliitis and multi-organ injury. Several mechanisms cooperate to development of endothelial dysfunction, including endothelial cell injury and pyroptosis, hyperinflammation and cytokine storm syndrome, oxidative stress and reduced nitric oxide bioavailability, glycocalyx disruption, hypercoagulability, and thrombosis. After acute-phase infection, some patients reported signs and symptoms of a systemic disorder known as long COVID, in which a broad range of cardiovascular (CV) disorders emerged. To date, the exact pathophysiology of long COVID remains unclear: in addition to the persistence of acute-phase infection mechanisms, specific pathways of CV damage have been postulated, such as persistent viral reservoirs in the heart or an autoimmune response to cardiac antigens through molecular mimicry. The aim of this review is to provide an overview of the main molecular patterns of enduring endothelial activation following SARS-CoV-2 infection and to offer the latest summary of CV complications in long COVID.
Collapse
Affiliation(s)
- Luca Santoro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Vincenzo Zaccone
- Department of Emergency Medicine, Internal and Sub-Intensive Medicine, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy
| | - Lorenzo Falsetti
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Vittorio Ruggieri
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Martina Danese
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Chiara Miro
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (V.R.); (M.D.); (C.M.)
| | - Angela Di Giorgio
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Antonio Nesci
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Alessia D’Alessandro
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
| | - Gianluca Moroncini
- Clinica Medica, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.F.); (G.M.)
| | - Angelo Santoliquido
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (L.S.); (A.D.G.); (A.N.); (A.D.); (A.S.)
- Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
128
|
Perticone M, Maio R, Gigliotti S, Shehaj E, Toscani AF, Capomolla A, Fabiani G, Sciacqua A, Perticone F. Mutual Effect Modification between Insulin Resistance and Endothelial Dysfunction in Predicting Incident Heart Failure in Hypertensives. Biomedicines 2023; 11:2188. [PMID: 37626686 PMCID: PMC10452906 DOI: 10.3390/biomedicines11082188] [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: 07/13/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Insulin resistance and endothelial dysfunction are associated with heart failure (HF). Our objective was to investigate whether endothelial dysfunction and insulin resistance are independent predictors of incident HF and if a possible interaction exists between them. We enrolled 705 white never-treated hypertensives. Endothelium-dependent vasodilation was investigated by intra-arterial infusion of acetylcholine. During the follow-up [median: 117 months (range: 31-211)], we documented 223 new cases of HF (3.3 events/100 patient-years). We stratified the study population into progressors and non-progressors; progressors showed an older age and a higher prevalence of females, as well as higher mean values of baseline glucose, insulin, homeostasis model assessment (HOMA), creatinine, and high-sensitivity C-reactive protein (hs-CRP), whereas the estimated glomerular filtration rate (e-GFR) and endothelium-dependent vasodilation were lower. In the multiple Cox regression analysis, serum hs-CRP (HR = 1.362, (95% CI = 1.208-1.536), HOMA (HR = 1.293, 95% CI = 1.142-1.465), maximal acetylcholine (Ach)-stimulated forearm blood flow (FBF) (100% increment, HR = 0.807, 95% CI = 0.697-0.934), and e-GFR (10 mL/min/1.73 m2 increment, HR = 0.552, 95% CI = 0.483-0.603) maintained an independent association with incident HF. HOMA and endothelial dysfunction interact between them in a competitive manner (HR = 6.548, 95% CI = 4.034-10.629), also showing a mutual effect modification. Our findings demonstrate that both endothelial dysfunction and HOMA are independent and strong predictors of incident HF in hypertensives, these two risk factors interact between them with a competitive mechanism.
Collapse
Affiliation(s)
- Maria Perticone
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (A.F.T.); (A.S.); (F.P.)
| | - Raffaele Maio
- Geriatrics Unit, Azienda Ospedaliero-Universitaria Renato Dulbecco, 88100 Catanzaro, Italy;
| | - Simona Gigliotti
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Ermal Shehaj
- Cardiology and CICU Unit, Giovanni Paolo II Hospital, 88046 Lamezia Terme, Italy;
| | - Alfredo Francesco Toscani
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (A.F.T.); (A.S.); (F.P.)
| | | | - Ginevra Fabiani
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy;
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (A.F.T.); (A.S.); (F.P.)
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (A.F.T.); (A.S.); (F.P.)
| |
Collapse
|
129
|
Qiao S, Wang X, Li H, Zhang C, Wang A, Zhang S. Atherosclerosis-associated endothelial dysfunction is promoted by miR-199a-5p/SIRT1 axis regulated by circHIF1ɑ. Nutr Metab Cardiovasc Dis 2023; 33:1619-1631. [PMID: 37336718 DOI: 10.1016/j.numecd.2023.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 04/29/2023] [Accepted: 05/03/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis (AS) is a chronic inflammatory disease that damages the arterial wall as a result of hyperlipidemia and causes endothelial cell dysfunction, which increases the risk of atherothrombotic events. Multiple pathological conditions have shown ectopic miR-199a-5p levels to cause endothelial injury, but its role in the AS competitive endogenous RNA (CeRNA) network is still unknown. METHODS AND RESULTS The high-fat diet (HFD) apoE-/- mouse model was constructed in vivo, and ECs were cultured under ox-LDL treatment to induce EC injury in vitro. Immunohistochemistry and immunofluorescence staining were used to assess the effect of miR-199a-5p on the macrophage, SMC, collagen content, and endothelial coverage in the artery wall of mouse model. miR-199a-5p level was validated to be overexpression in the aorta tissue of HFD apoE-/- mice and in the ox-LDL-treated ECs, and even in the plasma EVs of the patients with cerebral AS. Silencing of miR-199a-5p significantly attenuated atherosclerotic progress in HFD apoE-/- mice, and the gain/loss-of-function assay indicated that miR-199a-5p overexpression aggravated ox-LDL-induced disabilities of endothelial proliferation, motility, and neovascularization based on cell counting kit-8 assay, transwell assay and matrigel assay. Mechanistically, miR-199a-5p prevented EC activation by activating the FOXO signaling pathway by targeting SIRT1. Additionally, circular RNA (circRNA) circHIF1ɑ was identified as having a low expression in the ox-LDL-treated EC and mediated SIRT1 expression via sponging miR-199a-5p to rescue ox-LDL-induced EC injury. CONCLUSIONS Our study demonstrated the vital role of miR-199a-5p/SIRT1 axis regulated by circHIF1ɑ in AS pathogenesis and provided novel effective targets for AS treatment.
Collapse
Affiliation(s)
- Shan Qiao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Jinan, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China; Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Laibo Biotechnology Co., Ltd, China
| | - Xing Wang
- Department of Neurology, Tianyou Affiliated Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Haiyun Li
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Canling Zhang
- Nursing Department, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Jinan, China
| | - Aihua Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Jinan, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Shanchao Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Jinan, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China; School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
130
|
Mace EH, Kimlinger MJ, Billings FT, Lopez MG. Targeting Soluble Guanylyl Cyclase during Ischemia and Reperfusion. Cells 2023; 12:1903. [PMID: 37508567 PMCID: PMC10378692 DOI: 10.3390/cells12141903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Ischemia and reperfusion (IR) damage organs and contribute to many disease states. Few effective treatments exist that attenuate IR injury. The augmentation of nitric oxide (NO) signaling remains a promising therapeutic target for IR injury. NO binds to soluble guanylyl cyclase (sGC) to regulate vasodilation, maintain endothelial barrier integrity, and modulate inflammation through the production of cyclic-GMP in vascular smooth muscle. Pharmacologic sGC stimulators and activators have recently been developed. In preclinical studies, sGC stimulators, which augment the reduced form of sGC, and activators, which activate the oxidized non-NO binding form of sGC, increase vasodilation and decrease cardiac, cerebral, renal, pulmonary, and hepatic injury following IR. These effects may be a result of the improved regulation of perfusion and decreased oxidative injury during IR. sGC stimulators are now used clinically to treat some chronic conditions such as heart failure and pulmonary hypertension. Clinical trials of sGC activators have been terminated secondary to adverse side effects including hypotension. Additional clinical studies to investigate the effects of sGC stimulation and activation during acute conditions, such as IR, are warranted.
Collapse
Affiliation(s)
- Eric H Mace
- Department of Surgery, Vanderbilt University Medical Center, Medical Center North, Suite CCC-4312, 1161 21st Avenue South, Nashville, TN 37232-2730, USA
| | - Melissa J Kimlinger
- Vanderbilt University School of Medicine, 428 Eskind Family Biomedical Library and Learning Center, Nashville, TN 37240-0002, USA
| | - Frederic T Billings
- Department of Anesthesiology, Division of Critical Care Medicine, Vanderbilt University Medical Center, Medical Arts Building, Suite 422, 1211 21st Avenue South, Nashville, TN 37212-1750, USA
| | - Marcos G Lopez
- Department of Anesthesiology, Division of Critical Care Medicine, Vanderbilt University Medical Center, Medical Arts Building, Suite 422, 1211 21st Avenue South, Nashville, TN 37212-1750, USA
| |
Collapse
|
131
|
van Genugten EAJ, van Lith TJ, van den Heuvel FMA, van Steenis JL, Ten Heggeler RM, Brink M, Rodwell L, Meijer FJA, Lobeek D, Hagmolen Of Ten Have W, van de Veerdonk FL, Netea MG, Prokop M, Nijveldt R, Tuladhar AM, Aarntzen EHJG. Gallium-68 labelled RGD PET/CT imaging of endothelial activation in COVID-19 patients. Sci Rep 2023; 13:11507. [PMID: 37460572 DOI: 10.1038/s41598-023-37390-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 06/21/2023] [Indexed: 07/20/2023] Open
Abstract
In coronavirus disease 2019 (COVID-19), endothelial cells play a central role and an inadequate response is associated with vascular complications. PET imaging with gallium-68 labelled RGD-peptide (68Ga-RGD) targets αvβ3 integrin expression which allows quantification of endothelial activation. In this single-center, prospective observational study, we included ten hospitalized patients with COVID-19 between October 2020 and January 2021. Patients underwent 68Ga-RGD PET/CT followed by iodine mapping of lung parenchyma. CT-based segmentation of lung parenchyma, carotid arteries and myocardium was used to quantify tracer uptake by calculating standardized uptake values (SUV). Five non-COVID-19 patients were used as reference. The study population was 68.5 (IQR 52.0-74.5) years old, with median oxygen need of 3 l/min (IQR 0.9-4.0). 68Ga-RGD uptake quantified as SUV ± SD was increased in lungs (0.99 ± 0.32 vs. 0.45 ± 0.18, p < 0.01) and myocardium (3.44 ± 1.59 vs. 0.65 ± 0.22, p < 0.01) of COVID-19 patients compared to reference but not in the carotid arteries. Iodine maps showed local variations in parenchymal perfusion but no correlation with SUV. In conclusion, using 68Ga-RGD PET/CT in COVID-19 patients admitted with respiratory symptoms, we demonstrated increased endothelial activation in the lung parenchyma and myocardium. Our findings indicate the involvement of increased and localized endothelial cell activation in the cardiopulmonary system in COVID-19 patients.Trail registration: NCT04596943.
Collapse
Affiliation(s)
- Evelien A J van Genugten
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
| | - Theresa J van Lith
- Department of Neurology, Donders Center for Medical Neurosciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Josee L van Steenis
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
- Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Romy M Ten Heggeler
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
- Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Monique Brink
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
| | - Laura Rodwell
- Department of Health Evidence, Section Biostatistics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frederick J A Meijer
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
| | - Daphne Lobeek
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
| | | | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Respiratory Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Mathias Prokop
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
| | - Robin Nijveldt
- Department of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anil M Tuladhar
- Department of Neurology, Donders Center for Medical Neurosciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands.
| |
Collapse
|
132
|
Luck JC, Blaha C, Cauffman A, Gao Z, Arnold AC, Cui J, Sinoway LI. Autonomic and vascular function testing in collegiate athletes following SARS-CoV-2 infection: an exploratory study. Front Physiol 2023; 14:1225814. [PMID: 37528892 PMCID: PMC10389084 DOI: 10.3389/fphys.2023.1225814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/03/2023] [Indexed: 08/03/2023] Open
Abstract
Introduction: Recent studies suggest that SARS-CoV-2 infection alters autonomic and vascular function in young, otherwise healthy, adults. However, whether these alterations exist in young competitive athletes remains unknown. This study aimed to assess the effects of COVID-19 on cardiac autonomic control and vascular function in collegiate athletes who tested positive for COVID-19, acknowledging the limitations imposed by the early stages of the pandemic. Methods: Sixteen collegiate athletes from various sports underwent a battery of commonly used autonomic and vascular function tests (23 ± 9, range: 12-44 days post-infection). Additionally, data from 26 healthy control participants were included. Results: In response to the Valsalva maneuver, nine athletes had a reduced early phase II blood pressure response and/or reduced Valsalva ratio. A depressed respiratory sinus arrhythmia amplitude was observed in three athletes. Three athletes became presyncopal during standing and did not complete the 10-min orthostatic challenge. Brachial artery flow-mediated dilation, when allometrically scaled to account for differences in baseline diameter, was not different between athletes and controls (10.0% ± 3.5% vs. 7.1% ± 2.4%, p = 0.058). Additionally, no differences were observed between groups when FMD responses were normalized by shear rate (athletes: 0.055% ± 0.026%/s-1, controls: 0.068% ± 0.049%/s-1, p = 0.40). Discussion: Few atypical and borderline responses to autonomic function tests were observed in athletes following an acute SARS-CoV-2 infection. The most meaningful autonomic abnormality being the failure of three athletes to complete a 10-min orthostatic challenge. These findings suggest that some athletes may develop mild alterations in autonomic function in the weeks after developing COVID-19, while vascular function is not significantly impaired.
Collapse
Affiliation(s)
- J. Carter Luck
- Milton S. Hershey Medical Center, Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Cheryl Blaha
- Milton S. Hershey Medical Center, Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Aimee Cauffman
- Milton S. Hershey Medical Center, Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Zhaohui Gao
- Milton S. Hershey Medical Center, Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Amy C. Arnold
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Jian Cui
- Milton S. Hershey Medical Center, Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Lawrence I. Sinoway
- Milton S. Hershey Medical Center, Penn State Heart and Vascular Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
133
|
Sun L, Li X, Luo H, Guo H, Zhang J, Chen Z, Lin F, Zhao G. EZH2 can be used as a therapeutic agent for inhibiting endothelial dysfunction. Biochem Pharmacol 2023; 213:115594. [PMID: 37207700 DOI: 10.1016/j.bcp.2023.115594] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
Enhancer of zeste homolog 2 (EZH2) is a catalytic subunit of polycomb repressor complex 2 and plays important roles in endothelial cell homeostasis. EZH2 functionally methylates lysine 27 of histone H3 and represses gene expression through chromatin compaction. EZH2 mediates the effects of environmental stimuli by regulating endothelial functions, such as angiogenesis, endothelial barrier integrity, inflammatory signaling, and endothelial mesenchymal transition. Numerous studies have been conducted to determine the significance of EZH2 in endothelial function. The aim of this review is to provide a concise summary of the roles EZH2 plays in endothelial function and elucidate its therapeutic potential in cardiovascular diseases.
Collapse
Affiliation(s)
- Li Sun
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Xuefang Li
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Hui Luo
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Huige Guo
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Jie Zhang
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Zhigang Chen
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China
| | - Fei Lin
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China.
| | - Guoan Zhao
- Cardiovascular Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China; Key Laboratory of Cardiovascular Injury and Repair Medicine of Henan, Weihui, China.
| |
Collapse
|
134
|
Song JY, Huang JY, Hsu YC, Lo MT, Lin C, Shen TC, Liao MT, Lu KC. Coronavirus disease 2019 and cardiovascular disease. Tzu Chi Med J 2023; 35:213-220. [PMID: 37545802 PMCID: PMC10399840 DOI: 10.4103/tcmj.tcmj_219_22] [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: 08/13/2022] [Revised: 10/28/2022] [Accepted: 05/17/2023] [Indexed: 08/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus behind the coronavirus disease 2019 (COVID-19) pandemic, is a type of RNA virus that is nonsegmented. Cardiovascular diseases (CVDs) increase the mortality risk of patients. In this review article, we overview the existing evidence regarding the potential mechanisms of myocardial damage in coronavirus disease 2019 (COVID-19) patients. Having a comprehensive knowledge of the cardiovascular damage caused by SARS-CoV-2 and its underlying mechanisms is essential for providing prompt and efficient treatment, ultimately leading to a reduction in mortality rates. Severe COVID-19 causes acute respiratory distress syndrome and shock in patients. In addition, awareness regarding COVID-19 cardiovascular manifestations has increased, including the adverse impact on prognosis with cardiovascular involvement. Angiotensin-converting enzyme 2 receptor may play a role in acute myocardial injury caused by SARS-CoV-2 infection. COVID-19 patients experiencing heart failure may have their condition exacerbated by various contributing factors and mechanisms. Increased oxygen demand, myocarditis, stress cardiomyopathy, elevated pulmonary pressures, and venous thrombosis are potential health issues. The combination of these factors may lead to COVID-19-related cardiogenic shock, resulting in acute systolic heart failure. Extracorporeal membrane oxygenation (ECMO) and left ventricular assist devices (LVADs) are treatment options when inotropic support fails for effective circulatory support. To ensure effective COVID-19-related cardiovascular disease (CVD) surveillance, it is crucial to closely monitor the future host adaptation, viral evolution, and transmissibility of SARS-CoV-2, given the virus's pandemic potential.
Collapse
Affiliation(s)
- Jenn-Yeu Song
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Jian-You Huang
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Anesthesiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Men-Tzung Lo
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Chen Lin
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Ta-Chung Shen
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu, Taiwan
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| |
Collapse
|
135
|
Zhou M, Yao Y, Ma S, Zou M, Chen Y, Cai S, Zhao F, Wu H, Xiao F, Abudushalamu G, Fan X, Wu G. Dual-targeted and dual-sensitive self-assembled protein nanocarrier delivering hVEGI-192 for triple-negative breast cancer. Int J Biol Macromol 2023:125475. [PMID: 37353129 DOI: 10.1016/j.ijbiomac.2023.125475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 06/25/2023]
Abstract
Breast cancer is a highly prevalent malignancy worldwide among women with an increasing incidence in recent years. Triple-negative breast cancer (TNBC), a specific type of breast cancer, occurs primarily in young women and exhibits large tumor size, high clinical stage, and extremely poor prognosis with a high rate of lymph node, liver, and lung metastases. TNBC is insensitive to endocrine therapy and trastuzumab treatment, and there is an urgent need for effective therapeutics and treatment guidelines. However, investigations into antiangiogenic agents for the treatment of TNBC are ongoing. In this study, we successfully engineered a self-assembled protein nanocarrier TfRBP9-hVEGI-192-ELP fusion protein (TVEFP) to deliver the therapeutic protein, human vascular endothelial growth inhibitor (hVEGI-192). This was found to be effective in inhibiting tumor angiogenesis in vivo. The protein nanocarrier effectively inhibited the progression of TNBC in vivo and showed the behavior of self-assembly, thermoresponsiveness, enzyme stimulation-responsiveness, tumor-targeting, biocompatibility, and biodegradability. Near-infrared imaging studies showed that fluorescent dye-stained TVEFP effectively aggregated at the tumor site. The TVEFP nanocarrier significantly expands the application of the therapeutic protein hVEGI-192 and improves the imaging and biotherapeutic effects in TNBC, chiefly based on anti-angiogenesis effects.
Collapse
Affiliation(s)
- Meiling Zhou
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yuming Yao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Shuo Ma
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Mingyuan Zou
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yaya Chen
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Shijie Cai
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Fengfeng Zhao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Huina Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Feng Xiao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - GuliNazhaer Abudushalamu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiaobo Fan
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Guoqiu Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China; Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Laboratory Medcine, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
136
|
Gao J, Hou T. Cardiovascular disease treatment using traditional Chinese medicine:Mitochondria as the Achilles' heel. Biomed Pharmacother 2023; 164:114999. [PMID: 37311280 DOI: 10.1016/j.biopha.2023.114999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/30/2023] [Accepted: 06/07/2023] [Indexed: 06/15/2023] Open
Abstract
Cardiovascular disease (CVD), involving the pathological alteration of the heart or blood vessels, is one of the main causes of disability and death worldwide, with an estimated 18.6 million deaths per year. CVDs are caused by a variety of risk factors, including inflammation, hyperglycemia, hyperlipidemia, and increased oxidative stress. Mitochondria, the hub of ATP production and the main generator of reactive oxygen species (ROS), are linked to multiple cellular signaling pathways that regulate the progression of CVD and therefore are recognized as an essential target for CVD management. Initial treatment of CVD generally focuses on diet and lifestyle interventions; proper drugs or surgery can prolong or save the patient's life. Traditional Chinese medicine (TCM), a holistic medical care system with an over 2500-year history, has been proven to be efficient in curing CVD and other illnesses, with a strengthening effect on the body. However, the mechanisms underlying TCM alleviation of CVD remain elusive. Recent studies have recognized that TCM can alleviate cardiovascular disease by manipulating the quality and function of mitochondria. This review systematically summarizes the association of mitochondria with cardiovascular risk factors, and the relationships between mitochondrial dysfunction and CVD progression. We will investigate the research progress of managing cardiovascular disease by TCM and cover widely used TCMs that target mitochondria for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Jie Gao
- Chengdu Integrated TCM and Western Medicine Hospital and Chengdu University of Traditional Chinese Medicine, Chengdu 610041 China
| | - Tianshu Hou
- Chengdu Integrated TCM and Western Medicine Hospital and Chengdu University of Traditional Chinese Medicine, Chengdu 610041 China.
| |
Collapse
|
137
|
Wang Y, Chen B, Ciaccio EJ, Jneid H, Virani SS, Lavie CJ, Lebovits J, Green PHR, Krittanawong C. Celiac Disease and the Risk of Cardiovascular Diseases. Int J Mol Sci 2023; 24:9974. [PMID: 37373122 DOI: 10.3390/ijms24129974] [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: 05/16/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Celiac disease (CD) is a chronic autoimmune disorder that affects the small intestine in genetically predisposed individuals. Previous studies have investigated the potential link between CD and cardiovascular disease (CVD); however, the findings have been inconsistent. We aimed to provide an updated review of the literature on the association between CD and CVD. PubMed was searched from inception to January 2023 using keywords including CD, cardiovascular disease, coronary artery disease, cardiac arrhythmia, heart failure, cardiomyopathy, and myocarditis. We summarized the results of the studies, including meta-analyses and original investigations, and presented them according to the different forms of CVD. Meta-analyses published in 2015 provided mixed results regarding the relationship between CD and CVD. However, subsequent original investigations have shed new light on this association. Recent studies indicate that individuals with CD are at a higher risk of developing overall CVD, including an increased risk of myocardial infarction and atrial fibrillation. However, the link between CD and stroke is less established. Further research is needed to determine the link between CD and other cardiac arrhythmias, such as ventricular arrhythmia. Moreover, the relationship between CD and cardiomyopathy or heart failure, as well as myopericarditis, remains ambiguous. CD patients have a lower prevalence of traditional cardiac risk factors, such as smoking, hypertension, hyperlipidemia, and obesity. Therefore, it is important to discover strategies to identify patients at risk and reduce the risk of CVD in CD populations. Lastly, it is unclear whether adherence to a gluten-free diet can diminish or increase the risk of CVD among individuals with CD, necessitating further research in this area. To fully comprehend the correlation between CD and CVD and to determine the optimal prevention strategies for CVD in individuals with CD, additional research is necessary.
Collapse
Affiliation(s)
- Yichen Wang
- Mercy Internal Medicine Service, Trinity Health of New England, Springfield, MA 01104, USA
| | - Bing Chen
- Department of Gastroenterology and Nutrition, Geisinger Medical Center, Danville, PA 17821, USA
| | - Edward J Ciaccio
- Department of Medicine, Celiac Disease Center, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Hani Jneid
- Division of Cardiology, University of Texas Medical Branch, Houston, TX 77030, USA
| | - Salim S Virani
- Section of Cardiology and Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Office of the Vice Provost (Research), The Aga Khan University, Karachi 74800, Pakistan
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School, University of Queensland School of Medicine, New Orleans, LA 70121, USA
| | - Jessica Lebovits
- Department of Medicine, Celiac Disease Center, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Peter H R Green
- Department of Medicine, Celiac Disease Center, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | |
Collapse
|
138
|
Wang E, Feng B, Chakrabarti S. MicroRNA 9 Is a Regulator of Endothelial to Mesenchymal Transition in Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2023; 64:13. [PMID: 37279396 PMCID: PMC10249683 DOI: 10.1167/iovs.64.7.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/23/2023] [Indexed: 06/08/2023] Open
Abstract
Purpose Diabetic retinopathy (DR) is a significant cause of blindness. Most research around DR focus on late-stage developments rather than early changes such as early endothelial dysfunction. Endothelial-to-mesenchymal transition (EndMT), an epigenetically regulated process whereby endothelial cells lose endothelial characteristics and adopt mesenchymal-like phenotypes, contributes to early endothelial changes in DR. The epigenetic regulator microRNA 9 (miR-9) is suppressed in the eyes during DR. MiR-9 plays a role in various diseases and regulates EndMT-related processes in other organs. We investigated the role miR-9 plays in glucose-induced EndMT in DR. Methods We examined the effects of glucose on miR-9 and EndMT using human retinal endothelial cells (HRECs). We then used HRECs and an endothelial-specific miR-9 transgenic mouse line to investigate the effect of miR-9 on glucose-induced EndMT. Finally, we used HRECs to probe the mechanisms through which miR-9 may regulate EndMT. Results We found that miR-9 inhibition was both necessary and sufficient for glucose-induced EndMT. Overexpression of miR-9 prevented glucose-induced EndMT, whereas suppressing miR-9 caused glucose-like EndMT changes. We also found that preventing EndMT with miR-9 overexpression improved retinal vascular leakage in DR. Finally, we showed that miR-9 regulates EndMT at an early stage by regulating EndMT-inducing signals such as proinflammatory and TGF-β pathways. Conclusions We have shown that miR-9 is an important regulator of EndMT in DR, potentially making it a good target for RNA-based therapy in early DR.
Collapse
Affiliation(s)
- Eric Wang
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Biao Feng
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| |
Collapse
|
139
|
Okoh AK, Young A, Garcia M, Sullivan S, Almuwaqqat Z, Hu Y, Liu C, Moazzami K, Uphoff I, Lima BB, Ko YA, Elon L, Jajeh N, Rout P, Gupta S, Shah AJ, Bremner JD, Lewis T, Quyyumi A, Vaccarino V. Racial Differences in Mental Stress-Induced Transient Endothelial Dysfunction and Its Association With Cardiovascular Outcomes. Psychosom Med 2023; 85:431-439. [PMID: 37053106 PMCID: PMC10239336 DOI: 10.1097/psy.0000000000001201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
OBJECTIVE This study aimed to investigate differences in transient endothelial dysfunction (TED) with mental stress in Black and non-Black individuals with coronary heart disease (CHD), and their potential impact on cardiovascular outcomes. METHODS We examined 812 patients with stable CHD between June 2011 and March 2016 and followed through February 2020 at a university-affiliated hospital network. Flow-mediated vasodilation (FMD) was assessed before and 30 minutes after mental stress. TED was defined as a lower poststress FMD than prestress FMD. We compared prestress FMD, post-stress FMD, and TED between Black and non-Black participants. In both groups, we examined the association of TED with an adjudicated composite end point of cardiovascular death or nonfatal myocardial infarction (first and recurring events) after adjusting for demographic, clinical, and socioeconomic factors. RESULTS Prestress FMD was lower in Black than non-Black participants (3.7 [2.8] versus 4.9 [3.8], p < .001) and significantly declined with mental stress in both groups. TED occurred more often in Black (76%) than non-Black patients (67%; multivariable-adjusted odds ratio = 1.6, 95% confidence interval = 1.5-1.7). Over a median (interquartile range) follow-up period of 75 (65-82) months, 142 (18%) patients experienced either cardiovascular death or nonfatal myocardial infarction. Black participants had a 41.9% higher risk of the study outcome than non-Black participants (95% confidence interval = 1.01-1.95). TED with mental stress explained 69% of this excess risk. CONCLUSIONS Among CHD patients, Black individuals are more likely than non-Black individuals to develop endothelial dysfunction with mental stress, which in turn explains a substantial portion of their excess risk of adverse events.
Collapse
Affiliation(s)
- Alexis K Okoh
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - An Young
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| | - Mariana Garcia
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| | - Samaah Sullivan
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Zakaria Almuwaqqat
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| | - Yingtian Hu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Chang Liu
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Kasra Moazzami
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| | - Irina Uphoff
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| | - Bruno B. Lima
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Lisa Elon
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Nour Jajeh
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Pratik Rout
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Shishir Gupta
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Amit J Shah
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
- Atlanta VA Medical Center, Decatur, GA
| | - J. Douglas Bremner
- Atlanta VA Medical Center, Decatur, GA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Tene Lewis
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Arshed Quyyumi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| | - Viola Vaccarino
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| |
Collapse
|
140
|
Kirsten N, Ohmes J, Mikkelsen MD, Nguyen TT, Blümel M, Wang F, Tasdemir D, Seekamp A, Meyer AS, Fuchs S. Impact of Enzymatically Extracted High Molecular Weight Fucoidan on Lipopolysaccharide-Induced Endothelial Activation and Leukocyte Adhesion. Mar Drugs 2023; 21:339. [PMID: 37367664 DOI: 10.3390/md21060339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/24/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
The endothelial cell lining creates an interface between circulating blood and adjoining tissue and forms one of the most critical barriers and targets for therapeutical intervention. Recent studies suggest that fucoidans, sulfated and fucose-rich polysaccharides from brown seaweed, show multiple promising biological effects, including anti-inflammatory properties. However, their biological activity is determined by chemical characteristics such as molecular weight, sulfation degree, and molecular structure, which vary depending on the source, species, and harvesting and isolation method. In this study, we investigated the impact of high molecular weight (HMW) fucoidan extract on endothelial cell activation and interaction with primary monocytes (MNCs) in lipopolysaccharide (LPS)-induced inflammation. Gentle enzyme-assisted extraction combined with fractionation by ion exchange chromatography resulted in well-defined and pure fucoidan fractions. FE_F3, with a molecular weight ranging from 110 to 800 kDa and a sulfate content of 39%, was chosen for further investigation of its anti-inflammatory potential. We observed that along with higher purity of fucoidan fractions, the inflammatory response in endothelial mono- and co-cultures with MNCs was reduced in a dose-dependent manner when testing two different concentrations. This was demonstrated by a decrease in IL-6 and ICAM-1 on gene and protein levels and a reduced gene expression of TLR-4, GSK3β and NF-kB. Expression of selectins and, consequently, the adhesion of monocytes to the endothelial monolayer was reduced after fucoidan treatment. These data indicate that the anti-inflammatory effect of fucoidans increases with their purity and suggest that fucoidans might be useful in limiting the inflammatory response of endothelial cells in cases of LPS-induced bacterial infection.
Collapse
Affiliation(s)
- Nora Kirsten
- Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Julia Ohmes
- Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Maria Dalgaard Mikkelsen
- Protein Chemistry and Enzyme Technology Section, DTU Bioengineering, Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Thuan Thi Nguyen
- Protein Chemistry and Enzyme Technology Section, DTU Bioengineering, Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Martina Blümel
- GEOMAR Centre for Marine Biotechnology (GEOMAR-Biotech), Research Unit Marine Natural Products Chemistry, GEOMAR Helmholtz Centre for Ocean Research Kiel, 24106 Kiel, Germany
| | - Fanlu Wang
- Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Deniz Tasdemir
- GEOMAR Centre for Marine Biotechnology (GEOMAR-Biotech), Research Unit Marine Natural Products Chemistry, GEOMAR Helmholtz Centre for Ocean Research Kiel, 24106 Kiel, Germany
- Faculty of Mathematics and Natural Science, Kiel University, 24118 Kiel, Germany
| | - Andreas Seekamp
- Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Anne S Meyer
- Protein Chemistry and Enzyme Technology Section, DTU Bioengineering, Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Sabine Fuchs
- Experimental Trauma Surgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| |
Collapse
|
141
|
Marchenko V, Zelinskaya I, Toropova Y, Podyacheva E, Martynov M, Mukhametdinova D, Lioznov D, Zhilinskaya IN. Influenza A(H1N1)pdm09 Virus Aggravates Pathology of Blood Vessels in Wistar Rats with Premorbid Acute Cardiomyopathy. Viruses 2023; 15:v15051114. [PMID: 37243200 DOI: 10.3390/v15051114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
Influenza virus can infect the vascular endothelium and cause endothelial dysfunction. Persons at higher risk for severe influenza are patients with acute and chronic cardiovascular disorders; however, the mechanism of influenza-induced cardiovascular system alteration remains not fully understood. The aim of the study was to assess the functional activity of mesenteric blood vessels of Wistar rats with premorbid acute cardiomyopathy infected with Influenza A(H1N1)pdm09 virus. For this, we determined (1) the vasomotor activity of mesenteric blood vessels of Wistar rats using wire myography, (2) the level of expression of three endothelial factors: endothelial nitric oxide synthase (eNOS), plasminogen activator inhibitor-1 (PAI-1), and tissue plasminogen activator (tPA) in the endothelium of mesenteric blood vessels using immunohistochemistry, and (3) the concentration of PAI-1 and tPA in the blood plasma using ELISA. Acute cardiomyopathy in animals was induced by doxorubicin (DOX) following infection with rat-adapted Influenza A(H1N1)pdm09 virus. The functional activity of mesenteric blood vessels was analyzed at 24 and 96 h post infection (hpi). Thus, the maximal response of mesenteric arteries to both vasoconstrictor and vasodilator at 24 and 96 hpi was significantly decreased compared with control. Expression of eNOS in the mesenteric vascular endothelium was modulated at 24 and 96 hpi. PAI-1 expression increased 3.47-fold at 96 hpi, while the concentration of PAI-1 in the blood plasma increased 6.43-fold at 24 hpi compared with control. The tPA concentration in plasma was also modulated at 24 hpi and 96 hpi. The obtained data indicate that influenza A(H1N1)pdm09 virus aggravates the course of premorbid acute cardiomyopathy in Wistar rats, causing pronounced dysregulation of endothelial factor expression and vasomotor activity impairment of mesenteric arteries.
Collapse
Affiliation(s)
- Vladimir Marchenko
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376 St. Petersburg, Russia
- Smorodintsev Research Institute of Influenza, Department of Medical Microbiology, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia
| | - Irina Zelinskaya
- Almazov National Medical Research Centre, Russian Ministry of Health, 197341 St. Petersburg, Russia
| | - Yana Toropova
- Almazov National Medical Research Centre, Russian Ministry of Health, 197341 St. Petersburg, Russia
| | - Ekaterina Podyacheva
- Almazov National Medical Research Centre, Russian Ministry of Health, 197341 St. Petersburg, Russia
| | - Mikhail Martynov
- Almazov National Medical Research Centre, Russian Ministry of Health, 197341 St. Petersburg, Russia
| | - Daria Mukhametdinova
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376 St. Petersburg, Russia
- Almazov National Medical Research Centre, Russian Ministry of Health, 197341 St. Petersburg, Russia
| | - Dmitry Lioznov
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376 St. Petersburg, Russia
| | - Irina N Zhilinskaya
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376 St. Petersburg, Russia
- Smorodintsev Research Institute of Influenza, Department of Medical Microbiology, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia
| |
Collapse
|
142
|
Lopes KG, de Souza MDGC, Bouskela E, Kraemer-Aguiar LG. Microvascular Function, Inflammatory Status, and Oxidative Stress in Post-Bariatric Patients with Weight Regain. Nutrients 2023; 15:2135. [PMID: 37432255 DOI: 10.3390/nu15092135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 07/12/2023] Open
Abstract
Weight loss after bariatric surgery in obesity improves vascular function and metabolic/inflammatory profiles and reduces cardiovascular mortality but there are limited data on the effects of weight regain on vascular health. We compared the metabolic/inflammatory profiles, oxidative status, and vascular function of post-bariatric patients with a high ratio of weight regain (RWR) vs. non-surgical controls. Thirty-two post-bariatric patients [Roux-en-Y gastric bypass; aged = 44 ± 8 years, BMI = 40.1 ± 7.7 kg/m2, and RWR = 58.7 ± 24.3%] and thirty controls that were BMI-, age-, and gender-matched entered the study. We collected clinical data, metabolic/inflammatory/oxidative stress circulating biomarkers, and endothelial/microvascular reactivity through Venous occlusion plethysmography and Laser speckle contrast imaging. The bariatric group exhibited lower neck circumference, fasting glucose, and triglycerides than the non-surgical group, while HDL-cholesterol was higher in the bariatric group (p < 0.001). There was no significant difference between groups for endothelial/microvascular reactivities (p ≥ 0.06). Resistin, leptin, endothelin-1, soluble forms of intercellular cell adhesion molecule-1 and vascular cell adhesion molecule-1, tumor necrosis factor-α, and thiobarbituric acid reactive substances did not differ significantly between groups (p ≥ 0.09) either. The adiponectin level was higher in the bariatric compared to the non-surgical group, while interleukin-6 was lower in the bariatric group (p < 0.001). Despite the fact that endothelial/microvascular functions were not significantly different between groups, post-bariatric patients present partially preserved metabolic/inflammatory benefits even with high RWR.
Collapse
Affiliation(s)
- Karynne Grutter Lopes
- Obesity Unit, Centro de Pesquisas Clínicas Multiusuário (CePeM), Hospital Universitário Pedro Ernesto (HUPE), State University of Rio de Janeiro, Rio de Janeiro 20550-013, RJ, Brazil
- Postgraduate Program in Clinical and Experimental Physiopathology (Fisclinex), Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro 20550-013, RJ, Brazil
- Laboratory for Clinical and Experimental Research on Vascular Biology (Biovasc), Rio de Janeiro State University, Rio de Janeiro 20550-013, RJ, Brazil
| | - Maria das Graças Coelho de Souza
- Obesity Unit, Centro de Pesquisas Clínicas Multiusuário (CePeM), Hospital Universitário Pedro Ernesto (HUPE), State University of Rio de Janeiro, Rio de Janeiro 20550-013, RJ, Brazil
- Postgraduate Program in Clinical and Experimental Physiopathology (Fisclinex), Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro 20550-013, RJ, Brazil
- Laboratory for Clinical and Experimental Research on Vascular Biology (Biovasc), Rio de Janeiro State University, Rio de Janeiro 20550-013, RJ, Brazil
| | - Eliete Bouskela
- Obesity Unit, Centro de Pesquisas Clínicas Multiusuário (CePeM), Hospital Universitário Pedro Ernesto (HUPE), State University of Rio de Janeiro, Rio de Janeiro 20550-013, RJ, Brazil
- Postgraduate Program in Clinical and Experimental Physiopathology (Fisclinex), Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro 20550-013, RJ, Brazil
- Laboratory for Clinical and Experimental Research on Vascular Biology (Biovasc), Rio de Janeiro State University, Rio de Janeiro 20550-013, RJ, Brazil
| | - Luiz Guilherme Kraemer-Aguiar
- Obesity Unit, Centro de Pesquisas Clínicas Multiusuário (CePeM), Hospital Universitário Pedro Ernesto (HUPE), State University of Rio de Janeiro, Rio de Janeiro 20550-013, RJ, Brazil
- Postgraduate Program in Clinical and Experimental Physiopathology (Fisclinex), Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro 20550-013, RJ, Brazil
- Laboratory for Clinical and Experimental Research on Vascular Biology (Biovasc), Rio de Janeiro State University, Rio de Janeiro 20550-013, RJ, Brazil
- Endocrinology, Department of Internal Medicine, Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro 20550-013, RJ, Brazil
| |
Collapse
|
143
|
Rehman M, Chaudhary R, Rajput S, Agarwal V, Kaushik AS, Srivastava S, Srivastava S, Singh R, Aziz I, Singh S, Mishra V. Butein Ameliorates Chronic Stress Induced Atherosclerosis via Targeting Anti-inflammatory, Anti-fibrotic and BDNF Pathways. Physiol Behav 2023; 267:114207. [PMID: 37100219 DOI: 10.1016/j.physbeh.2023.114207] [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: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/23/2023] [Indexed: 04/28/2023]
Abstract
Chronic stress is a major risk factor for various diseases, including cardiovascular diseases (CVDs). Chronic stress enhances the release of pro-inflammatory cytokines like IL-1β, IL-6, and TNF-α, making individuals susceptible to atherosclerosis which is dominant cause for CVDs. In present study, we validated a mouse model of chronic unpredictable stress (CUS), and assessed the characteristic features of atherosclerosis in thoracic aortas of CUS mice. The CUS procedure consisted of exposing groups of mice to random stressors daily for 10-weeks. The stress response was verified by presence of depressive-like behaviors and increased serum corticosterone in mice which was determined by battery of behavioural tests (SPT, EPMT, NSFT) and ELISA, respectively. Atherosclerosis parameters in CUS mice were evaluated by lipid indices estimation followed by histological assessment of plaque deposition and fibrosis in thoracic aorta. Further, we assessed the efficacy of a polyphenol, i.e. Butein in conferring protection against chronic stress-induced atherosclerosis and the possible mechanism of action. Butein (20mg/kg x 28 days, alternatively, i.p.) was administered to CUS mice after 6-weeks of CUS exposure till the end of the protocol. Butein treatment decreased peripheral IL-1β and enhanced peripheral as well as central BDNF levels. Histological assessment revealed decreased macrophage expression and reduced fibrosis in thoracic aorta of Butein treated mice. Further, treatment with Butein lowered lipid indices in CUS mice. Our findings thus, suggest that 10-weeks of CUS induce characteristic features of atherosclerosis in mice and Butein can offer protection in CUS-induced atherosclerosis through multiple mechanisms including anti-inflammatory, antifibrotic and anti-adipogenic actions.
Collapse
Affiliation(s)
- Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Sonu Rajput
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Rohit Singh
- Centre for Animal Disease Research and Diagnosis, ICAR-Indian Veterinary Research Institute, Izzatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Irfan Aziz
- Integral University, Kursi road, Lucknow, Uttar Pradesh 226026, India
| | - Sanjay Singh
- Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025, India.
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India.
| |
Collapse
|
144
|
Petrova M, Gavino A, Li Y, McLachlan CS. Comparison of Parameters for Assessment of Carotid Stiffness and Their Association with Carotid Atherosclerosis in Rural Australian Adults: A Pilot Study. J Clin Med 2023; 12:jcm12082935. [PMID: 37109272 PMCID: PMC10144104 DOI: 10.3390/jcm12082935] [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: 03/07/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Carotid stiffness has been associated with the development and progression of carotid artery disease and is an independent factor for stroke and dementia. There has also been a lack of comparison of different ultrasound-derived carotid stiffness parameters and their association with carotid atherosclerosis. This pilot study aimed to investigate the associations between carotid stiffness parameters (derived via ultrasound echo tracking) and the presence of carotid plaques in Australian rural adults. In cross-sectional analyses, we assessed forty-six subjects (68 ± 9 years; mean ± SD) who underwent carotid ultrasound examinations. Carotid stiffness was assessed by a noninvasive echo-tracking method, measuring and comparing multiple carotid stiffness parameters, including stroke change in diameter (ΔD), stroke change in lumen area (ΔA), β- stiffness index, pulse wave velocity beta (PWV-β), compliance coefficient (CC), distensibility coefficient (DC), Young's elastic modulus (YEM), Peterson elastic modulus (Ep), and strain. Carotid atherosclerosis was assessed bilaterally by the presence of plaques in the common and internal carotid arteries, while carotid stiffness was assessed at the right common carotid artery. β-stiffness index, PWV-β, and Ep were significantly higher (p = 0.006, p = 0.004, p = 0.02, respectively), whilst ΔD, CC, DC, and strain were lower among subjects with carotid plaques (p = 0.036, p = 0.032, p = 0.01, p = 0.02, respectively) comparing to subjects without carotid plaques. YEM and ΔA did not significantly differ among the groups. Carotid plaques were associated with age, history of stroke, coronary artery disease, and previous coronary interventions. These results suggest that unilateral carotid stiffness is associated with the presence of carotid plaques.
Collapse
Affiliation(s)
- Marjana Petrova
- Centre for Healthy Futures, Torrens University Australia, Sydney, NSW 2010, Australia
| | - Alex Gavino
- Centre for Healthy Futures, Torrens University Australia, Sydney, NSW 2010, Australia
| | - Yujie Li
- Centre for Healthy Futures, Torrens University Australia, Sydney, NSW 2010, Australia
| | - Craig S McLachlan
- Centre for Healthy Futures, Torrens University Australia, Sydney, NSW 2010, Australia
| |
Collapse
|
145
|
Ricci TA, Boonpattrawong N, Laher I, Devlin AM. Maternal nutrition and effects on offspring vascular function. Pflugers Arch 2023:10.1007/s00424-023-02807-x. [PMID: 37041303 DOI: 10.1007/s00424-023-02807-x] [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: 02/14/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/13/2023]
Abstract
Maternal nutrition during pregnancy may have profound effects on the developing fetus and impact risk for cardiovascular disease later in life. Here, we provide a narrative review on the impact of maternal diet during pregnancy on offspring vascular function. We review studies reporting effects of maternal micronutrient (folic acid, iron) intakes, high-fat diets, dietary energy restriction, and low protein intake on offspring endothelial function. We discuss the differences in study design and outcomes and potential underlying mechanisms contributing to the vascular phenotypes observed in the offspring. We further highlight key gaps in the literature and identify targets for future investigations.
Collapse
Affiliation(s)
- Taylor A Ricci
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Nicha Boonpattrawong
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology, and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Angela M Devlin
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada.
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.
| |
Collapse
|
146
|
Kennedy KG, Islam AH, Karthikeyan S, Metcalfe AWS, McCrindle BW, MacIntosh BJ, Black S, Goldstein BI. Differential association of endothelial function with brain structure in youth with versus without bipolar disorder. J Psychosom Res 2023; 167:111180. [PMID: 36764023 DOI: 10.1016/j.jpsychores.2023.111180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/22/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Mood symptoms and disorders are associated with impaired endothelial function, a marker of early atherosclerosis. Given the increased vascular burden and neurostructural differences among individuals with mood disorders, we investigated the endothelial function and brain structure interface in relation to youth bipolar disorder (BD). METHODS This cross-sectional case-controlled study included 115 youth, ages 13-20 years (n = 66 BD; n = 49 controls [CG]). Cortical thickness and volume for regions of interest (ROI; insular cortex, ventrolateral prefrontal cortex [vlPFC], temporal lobe) were acquired from FreeSurfer processed T1-weighted MRI images. Endothelial function was assessed using pulse amplitude tonometry, yielding a reactive hyperemia index (RHI). ROI and vertex-wise analyses controlling for age, sex, obesity, and intracranial volume investigated for RHI-neurostructural associations, and RHI-by-diagnosis interactions. RESULTS In ROI analyses, higher RHI (i.e., better endothelial function) was associated with lower thickness in the insular cortex (β = -0.19, pFDR = 0.03), vlPFC (β = -0.30, pFDR = 0.003), and temporal lobe (β = -0.22, pFDR = 0.01); and lower temporal lobe volume (β = -0.16, pFDR = 0.01) in the overall sample. In vertex-wise analyses, higher RHI was associated with lower cortical thickness and volume in the insular cortex, prefrontal cortex (e.g., vlPFC), and temporal lobe. Additionally, higher RHI was associated with lower vlPFC and temporal lobe volume to a greater extent in youth with BD vs. CG. CONCLUSIONS Better endothelial function was associated with lower regional brain thickness and volume, contrasting the hypothesized associations. Additionally, we found evidence that this pattern was exaggerated in youth with BD. Future studies examining the direction of the observed associations and underlying mechanisms are warranted.
Collapse
Affiliation(s)
- Kody G Kennedy
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada.
| | - Alvi H Islam
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada; Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Sudhir Karthikeyan
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada.
| | - Arron W S Metcalfe
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada
| | - Brian W McCrindle
- Faculty of Medicine, University of Toronto, Toronto, Canada; Hospital for Sick Children, Toronto, Canada; Labatt Family Heart Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada.
| | - Bradley J MacIntosh
- Hurvitz Brain Sciences, Sunnybrook Research Institute, Toronto, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Computational Radiology & Artificial Intelligence (CRAI) Unit, Dept of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.
| | - Sandra Black
- Faculty of Medicine, University of Toronto, Toronto, Canada; Hurvitz Brain Sciences, Sunnybrook Research Institute, Toronto, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Canada.
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada; Faculty of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
147
|
Tang Y, Jin L, Qi W, Gao Y, Xie Y, Xie X, Lv J, Jiang Z, Jiang H, Fan C, Yan J. N-acetyl-L-cysteine attenuated the toxicity of ZIF-8 on EA.hy926 endothelial cells by wnt/β-catenin pathway. Toxicol In Vitro 2023; 88:105553. [PMID: 36634885 DOI: 10.1016/j.tiv.2023.105553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/07/2023] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
As kinds of porous crystalline compounds, zeolitic imidazolate frameworks (ZIFs) have been developed quickly and attracted considerable attention for use in nano drug delivery systems, which raised concerns about cardiovascular disorders. At the present, the cytotoxic mechanism of ZIFs in cardiovascular disorders was still unclear. Our experiment explored the toxicity of ZIF-8, a typical kind of ZIFs, on human EA.hy926 vascular endothelial cells. The cell viability, ROS formation, apoptosis level, inflammatory response level, wound healing ability and atherosclerosis-related indicators of EA.hy926 endothelial cells were analyzed after ZIF-8 treatment. Meanwhile, we evaluated the ability of antioxidant N-Acetyl-L-cysteine (NAC) to attenuate the toxicity of ZIF-8 on EA.hy926 endothelial cells. As results, NAC attenuated ROS formation, cell apoptosis, LDH formation and endothelial dysfunction caused by ZIF-8. As the Wnt/β-catenin pathway was involved in endothelial cell dysfunction, we also studied the expression level of β-catenin and LEF1 in ZIF-8 and/or NAC treated EA.hy926 cells. As expected, ZIF-8 increased the protein expressions of β-catenin and LEF1in the IC50 group, which was significantly inhibited by co-treatment with NAC. Taken together, this study could help improve our understanding about the mechanism of ZIF-8-induced endothelial cells injury and NAC had therapeutic potential in preventing ZIF-8-associated endothelial dysfunction by wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yaxin Tang
- School of Life Science, Shaoxing University, Shaoxing, Zhejiang, China
| | - Lifang Jin
- School of Life Science, Shaoxing University, Shaoxing, Zhejiang, China; Shaoxing Academy of Biomedicine of Zhejiang Sci-Tech University, Shaoxing, Zhejiang, China
| | - Wenwen Qi
- Xiangzhou District People's Hospital, Xiangyang, Hubei, China
| | - Yue Gao
- School of Life Science, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yixia Xie
- School of Life Science, Shaoxing University, Shaoxing, Zhejiang, China
| | - Xueying Xie
- School of Life Science, Shaoxing University, Shaoxing, Zhejiang, China
| | - Jianan Lv
- School of Life Science, Shaoxing University, Shaoxing, Zhejiang, China
| | - Zhikai Jiang
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - He Jiang
- The First Clinical Medical School of Zhejiang Chinese Medical University, Hangzhou, China
| | - Caixia Fan
- School of Life Science, Shaoxing University, Shaoxing, Zhejiang, China.
| | - Junyan Yan
- School of Life Science, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
148
|
Vorn R, Yoo HY. Food Restriction Augmented Alpha1-Adrenergic Mediated Contraction in Mesenteric Arteries. Biol Res Nurs 2023; 25:198-209. [PMID: 36203228 DOI: 10.1177/10998004221132247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Food restriction (FR) enhances sensitivity to cardiopulmonary reflexes and α1-adrenoreceptors in females in the presence of hypotension. However, the effect of FR on cardiopulmonary and vascular function in males is not well-understood. This study examines the effects of FR on cardiopulmonary, isolated arterial function, and potential underlying mechanisms. Male Sprague-Dawley (SD) rats were randomly divided into 3 groups and monitored for 5 weeks: (1) control (n = 30), (2) 20% food reduction (FR20, n = 30), and (3) 40% food reduction (FR40, n = 30). Non-invasive blood pressure was measured twice a week. Pulmonary arterial pressure (PAP) was measured using isolated/perfused lungs. The isolated vascular reactivity was assessed using double-wire myographs. FR rats exhibited a lower mean arterial pressure and heart rate; however, only the FR40 group exhibited statistically significant differences. We observed that FR enhanced sensitivity (EC50) to vasoconstriction induced by the α1-adrenoreceptor phenylephrine (PhE) but not to serotonin, U46619, or high K+ in the mesenteric arteries. PhE-mediated vasoconstriction in the mesenteric arteries was eliminated in the presence of the eNOS inhibitor (L-NAME). In addition, incubation with NOX2/4 inhibitors (apocynin, GKT137831, and VAS2870) and the reactive oxygen species (ROS) scavenger inhibitor (Tiron) eliminated the differences in PhE-mediated vasoconstriction, but the cyclooxygenase inhibitor (indomethacin) in the mesenteric arteries did not. Augmentation of α1-adrenergic-mediated contraction via the inhibition of the eNOS-NO pathway increased the activation of ROS through NOX2/4 in response to FR. Reduced eNOS-NO signaling may be a pathophysiological counterbalance to prevent hypovolemic shock in response to FR.
Collapse
Affiliation(s)
- Rany Vorn
- Department of Nursing, 26729Chung-Ang University, Seoul, Korea
- School of Nursing, 1466Johns Hopkins University, Baltimore, MD, USA
| | - Hae Young Yoo
- Department of Nursing, 26729Chung-Ang University, Seoul, Korea
| |
Collapse
|
149
|
Qin K, Bai S, Chen W, Li J, Guo VY. Association of comorbid depression and obesity with cardiometabolic multimorbidity among middle-aged and older Chinese adults: A cohort study. Arch Gerontol Geriatr 2023; 107:104912. [PMID: 36565606 DOI: 10.1016/j.archger.2022.104912] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/24/2022] [Accepted: 12/18/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE To evaluate the association of comorbid depression and obesity with the risk of incident cardiometabolic multimorbidity among middle-aged and older Chinese adults. METHODS This cohort study extracted data from the 2011 and 2015 waves of the China Health and Retirement Longitudinal Study (CHARLS). Depression was confirmed by the 10-item Center for Epidemiologic Studies Depression Scale (CESD-10) with a cut-off score ≥10. Obesity was defined as a body mass index ≥28 kg/m2. Participants were categorized into four groups based on depression and obesity status at baseline, i.e., with neither condition, depression only, obesity only, and with both conditions. Cardiometabolic multimorbidity was defined as the coexistence of two or more of heart diseases, stroke, and diabetes mellitus. Logistic regression models were established to estimate the associations. RESULTS A total of 9,308 participants without cardiometabolic multimorbidity at baseline were included (mean [SD] age, 58.8 [9.0] years; 4,449 [47.8%] were males). During four-year of follow-up, 349 (3.8%) participants developed cardiometabolic multimorbidity. Compared to participants without depression or obesity, comorbid depression and obesity was associated with greater risk of cardiometabolic multimorbidity (adjusted OR: 4.79, 95% CI: 3.09-7.43) than that in participants with depression alone (adjusted OR: 1.84, 95% CI: 1.37-2.46) or obesity alone (adjusted OR: 2.26, 95% CI: 1.48-3.45). The findings were consistent in different gender and age groups. CONCLUSIONS Comorbid depression and obesity was associated with excessive risk of cardiometabolic multimorbidity. Intervention targeting at individuals with both depression and obesity might have substantial benefit in minimizing the risk of cardiometabolic multimorbidity.
Collapse
Affiliation(s)
- Kang Qin
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Shigen Bai
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weiqing Chen
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jinghua Li
- Department of Biostatistics, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Vivian Yawei Guo
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
150
|
Milusev A, Despont A, Shaw J, Rieben R, Sorvillo N. Inflammatory stimuli induce shedding of heparan sulfate from arterial but not venous porcine endothelial cells leading to differential proinflammatory and procoagulant responses. Sci Rep 2023; 13:4483. [PMID: 36934164 PMCID: PMC10024017 DOI: 10.1038/s41598-023-31396-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/10/2023] [Indexed: 03/20/2023] Open
Abstract
Endothelial dysfunction is an early event of vascular injury defined by a proinflammatory and procoagulant endothelial cell (EC) phenotype. Although endothelial glycocalyx disruption is associated with vascular damage, how various inflammatory stimuli affect the glycocalyx and whether arterial and venous cells respond differently is unknown. Using a 3D round-channel microfluidic system we investigated the endothelial glycocalyx, particularly heparan sulfate (HS), on porcine arterial and venous ECs. Heparan sulfate (HS)/glycocalyx expression was observed already under static conditions on venous ECs while it was flow-dependent on arterial cells. Furthermore, analysis of HS/glycocalyx response after stimulation with inflammatory cues revealed that venous, but not arterial ECs, are resistant to HS shedding. This finding was observed also on isolated porcine vessels. Persistence of HS on venous ECs prevented complement deposition and clot formation after stimulation with tumor necrosis factor α or lipopolysaccharide, whereas after xenogeneic activation no glycocalyx-mediated protection was observed. Contrarily, HS shedding on arterial cells, even without an inflammatory insult, was sufficient to induce a proinflammatory and procoagulant phenotype. Our data indicate that the dimorphic response of arterial and venous ECs is partially due to distinct HS/glycocalyx dynamics suggesting that arterial and venous thrombo-inflammatory disorders require targeted therapies.
Collapse
Affiliation(s)
- Anastasia Milusev
- Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 24, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Alain Despont
- Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 24, 3008, Bern, Switzerland
| | - Jane Shaw
- Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 24, 3008, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 24, 3008, Bern, Switzerland
| | - Nicoletta Sorvillo
- Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 24, 3008, Bern, Switzerland.
| |
Collapse
|