151
|
Ziogos E, Kwapong YA, Weiss RG, Schär M, Brown TT, Bagchi S, Soleimanifard A, Harb T, Piggott DA, Gerstenblith G, Leucker TM, Hays AG. Coronary artery endothelial function and aging in people with HIV and HIV-negative individuals. Am J Physiol Heart Circ Physiol 2023; 325:H1099-H1107. [PMID: 37682238 PMCID: PMC10907030 DOI: 10.1152/ajpheart.00143.2023] [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: 03/13/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/09/2023]
Abstract
Coronary artery disease (CAD) is a common comorbidity in people with human immunodeficiency virus (HIV) (PWH) and impaired coronary endothelial function (CEF) plays a central role in the pathogenesis of CAD. Age-related impaired CEF among PWH, however, is not well characterized. We investigated the association between CEF and age in males and females with and without HIV using 3-T magnetic resonance imaging (MRI). We measured the changes in coronary cross-sectional area (CSA) and coronary blood flow during isometric handgrip exercise (IHE), an established endothelial-dependent stressor with smaller increases in CSA and coronary blood flow indicative of impaired CEF. We included 106 PWH and 82 individuals without HIV. Differences in demographic and clinical characteristics between PWH and individuals without HIV were explored using Pearson's χ2 test for categorical variables and Welch's t test for continuous variables. Linear regression models were used to examine the association between CEF and age. CEF was significantly lower in PWH as compared with individuals without HIV. Coronary endothelial dysfunction was also present at younger ages in PWH than in the individuals without HIV and there were significant differences in CEF between the PWH and individuals without HIV across age groups. Among the individuals without HIV, the percent changes in CSA were inversely related to age in unadjusted and adjusted models. There was no significant association between CEF and age in PWH. To the best of our knowledge, this is the first study to examine the relationship between age and CEF in PWH, and our results suggest that factors other than age significantly impair CEF in PWH across the life span.NEW & NOTEWORTHY This is the first study to examine the relationship between age and coronary endothelial function (CEF) in people with human immunodeficiency virus (HIV) (PWH). CEF was assessed using magnetic resonance imaging (MRI) in people with and without HIV. Although age and CEF were significantly inversely related in individuals without HIV, there was no association between age and CEF in PWH.
Collapse
Affiliation(s)
- Efthymios Ziogos
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Yaa A Kwapong
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Robert G Weiss
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Michael Schär
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Todd T Brown
- Division of Endocrinology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Shashwatee Bagchi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Division of Infectious Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Alborz Soleimanifard
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Tarek Harb
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Damani A Piggott
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Gary Gerstenblith
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Thorsten M Leucker
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Allison G Hays
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
152
|
Pu Y, Cheng CK, Zhang H, Luo JY, Wang L, Tomlinson B, Huang Y. Molecular mechanisms and therapeutic perspectives of peroxisome proliferator-activated receptor α agonists in cardiovascular health and disease. Med Res Rev 2023; 43:2086-2114. [PMID: 37119045 DOI: 10.1002/med.21970] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 03/10/2023] [Accepted: 04/12/2023] [Indexed: 04/30/2023]
Abstract
The prevalence of cardiovascular disease (CVD) has been rising due to sedentary lifestyles and unhealthy dietary patterns. Peroxisome proliferator-activated receptor α (PPARα) is a nuclear receptor regulating multiple biological processes, such as lipid metabolism and inflammatory response critical to cardiovascular homeostasis. Healthy endothelial cells (ECs) lining the lumen of blood vessels maintains vascular homeostasis, where endothelial dysfunction associated with increased oxidative stress and inflammation triggers the pathogenesis of CVD. PPARα activation decreases endothelial inflammation and senescence, contributing to improved vascular function and reduced risk of atherosclerosis. Phenotypic switch and inflammation of vascular smooth muscle cells (VSMCs) exacerbate vascular dysfunction and atherogenesis, in which PPARα activation improves VSMC homeostasis. Different immune cells participate in the progression of vascular inflammation and atherosclerosis. PPARα in immune cells plays a critical role in immunological events, such as monocyte/macrophage adhesion and infiltration, macrophage polarization, dendritic cell (DC) embedment, T cell activation, and B cell differentiation. Cardiomyocyte dysfunction, a major risk factor for heart failure, can also be alleviated by PPARα activation through maintaining cardiac mitochondrial stability and inhibiting cardiac lipid accumulation, oxidative stress, inflammation, and fibrosis. This review discusses the current understanding and future perspectives on the role of PPARα in the regulation of the cardiovascular system as well as the clinical application of PPARα ligands.
Collapse
Affiliation(s)
- Yujie Pu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Hongsong Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jiang-Yun Luo
- Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Brian Tomlinson
- Faculty of Medicine, Macau University of Science & Technology, Macau, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
153
|
Karmazyn M, Gan XT. Probiotics as potential treatments to reduce myocardial remodelling and heart failure via the gut-heart axis: State-of-the-art review. Mol Cell Biochem 2023; 478:2539-2551. [PMID: 36892791 DOI: 10.1007/s11010-023-04683-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/17/2023] [Indexed: 03/10/2023]
Abstract
Probiotics are considered to represent important modulators of gastrointestinal health through increased colonization of beneficial bacteria thus altering the gut microflora. Although these beneficial effects of probiotics are now widely recognized, emerging evidence suggests that alterations in the gut microflora also affect numerous other organ systems including the heart through a process generally referred to as the gut-heart axis. Moreover, cardiac dysfunction such as that seen in heart failure can produce an imbalance in the gut flora, known as dysbiosis, thereby further contributing to cardiac remodelling and dysfunction. The latter occurs by the production of gut-derived pro-inflammatory and pro-remodelling factors which exacerbate cardiac pathology. One of the key contributors to gut-dependent cardiac pathology is trimethylamine N-oxide (TMAO), a choline and carnitine metabolic by-product first synthesized as trimethylamine which is then converted into TMAO by a hepatic flavin-containing monooxygenase. The production of TMAO is particularly evident with regular western diets containing high amounts of both choline and carnitine. Dietary probiotics have been shown to reduce myocardial remodelling and heart failure in animal models although the precise mechanisms for these effects are not completely understood. A large number of probiotics have been shown to possess a reduced capacity to synthesize gut-derived trimethylamine and therefore TMAO thereby suggesting that inhibition of TMAO is a factor mediating the beneficial cardiac effects of probiotics. However, other potential mechanisms may also be important contributing factors. Here, we discuss the potential benefit of probiotics as effective therapeutic tools for attenuating myocardial remodelling and heart failure.
Collapse
Affiliation(s)
- Morris Karmazyn
- Department of Pharmacology and Physiology, University of Western Ontario, London, ON, N6G 2X6, Canada.
| | - Xiaohong Tracey Gan
- Department of Pharmacology and Physiology, University of Western Ontario, London, ON, N6G 2X6, Canada
| |
Collapse
|
154
|
Li Z, Giarto J, Zhang J, Kulkarni N, Mahalingam E, Klipstine W, Turng LS. Anti-thrombotic poly(AAm-co-NaAMPS)-xanthan hydrogel-expanded polytetrafluoroethylene (ePTFE) vascular grafts with enhanced endothelialization and hemocompatibility properties. BIOMATERIALS ADVANCES 2023; 154:213625. [PMID: 37722163 PMCID: PMC10841274 DOI: 10.1016/j.bioadv.2023.213625] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/23/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death among all non-communicable diseases globally. Although expanded polytetrafluoroethylene (ePTFE) has been widely used for larger-diameter vascular graft transplantation, the persistent thrombus formation and intimal hyperplasia of small-diameter vascular grafts (SDVGs) made of ePTFE to treat severe CVDs remain the biggest challenges due to lack of biocompatibility and endothelium. In this study, bi-layered poly(acrylamide-co-2-Acrylamido-2-methyl-1-propanesulfonic acid sodium)-xanthan hydrogel-ePTFE (poly(AAm-co-NaAMPS)-xanthan hydrogel-ePTFE) vascular grafts capable of promoting endothelialization and prohibiting thrombosis were synthesized and fabricated. While the external ePTFE layer of the vascular grafts provided the mechanical stability, the inner hydrogel layer offered much-needed cytocompatibility, hemocompatibility, and endothelialization functions. The interface morphology between the inner hydrogel layer and the outer ePTFE layer was observed by scanning electron microscope (SEM), which revealed that the hydrogel was well attached to the porous ePTFE through mechanical interlocking. Among all the hydrogel compositions tested with cell culture using human umbilical vein endothelial cells (HUVECs), the hydrogel with the molar ratio of 40:60 (NaAMPS/AAm) composition (i.e., Hydrogel 40:60) exhibited the best endothelialization function, as it produced the largest endothelialization area that was three times more than of that of plain ePTFE on day 14, maintained the highest average cell viability, and had the best cell morphology. Hydrogel 40:60 also showed excellent hemocompatibility, prolonged activated partial thromboplastin time (aPTT), and good mechanical properties. Overall, bi-layered poly(AAm-co-NaAMPS)-xanthan hydrogel-ePTFE vascular grafts with the Hydrogel 40:60 composition could potentially solve the critical challenge of thrombus formation in vascular graft transplantation applications.
Collapse
Affiliation(s)
- Zhutong Li
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Joshua Giarto
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jue Zhang
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Neha Kulkarni
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Esha Mahalingam
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; College of Letters and Science, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Will Klipstine
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Lih-Sheng Turng
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Mechanical Engineering, Chang Gung University, Tao-Yuan 33302, Taiwan.
| |
Collapse
|
155
|
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
|
156
|
Kologrivova IV, Suslova TE, Koshelskaya OA, Kravchenko ES, Kharitonova OA, Romanova EA, Vyrostkova AI, Boshchenko AA. Intermediate Monocytes and Circulating Endothelial Cells: Interplay with Severity of Atherosclerosis in Patients with Coronary Artery Disease and Type 2 Diabetes Mellitus. Biomedicines 2023; 11:2911. [PMID: 38001912 PMCID: PMC10669450 DOI: 10.3390/biomedicines11112911] [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: 09/21/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
The aim was to investigate the association of monocyte heterogeneity and presence of circulating endothelial cells with the severity of coronary atherosclerosis in patients with coronary artery disease (CAD) and type 2 diabetes mellitus (T2DM). We recruited 62 patients with CAD, including 22 patients with DM2. The severity of atherosclerosis was evaluated using Gensini Score. Numbers of classical (CD14++CD16-), intermediate (CD14++CD16+), and non-classical (CD14+CD16++) monocyte subsets; circulating endothelial progenitor cells; and the presence of circulating endothelial cells were evaluated. Counts and frequencies of intermediate monocytes, but not glycaemia parameters, were associated with the severity of atherosclerosis in diabetic CAD patients (rs = 0.689; p = 0.001 and rs = 0.632; p = 0.002, respectively). Frequency of Tie2+ cells was lower in classical than in non-classical monocytes in CAD patients (p = 0.007), while in patients with association of CAD and T2DM, differences between Tie2+ monocytes subsets disappeared (p = 0.080). Circulating endothelial cells were determined in 100% of CAD+T2DM patients, and counts of CD14++CD16+ monocytes and concentration of TGF-β predicted the presence of circulating endothelial cells (sensitivity 92.3%; specificity 90.9%; AUC = 0.930). Thus, intermediate monocytes represent one of the key determinants of the appearance of circulating endothelial cells in all the patients with CAD, but are associated with the severity of atherosclerosis only in patients with association of CAD and T2DM.
Collapse
Affiliation(s)
- Irina V. Kologrivova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Tatiana E. Suslova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Olga A. Koshelskaya
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Elena S. Kravchenko
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Olga A. Kharitonova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Ekaterina A. Romanova
- Department of Biomedicine, Siberian State Medical University, 2 Moskovskii trakt, Tomsk 634050, Russia; (E.A.R.); (A.I.V.)
| | - Alexandra I. Vyrostkova
- Department of Biomedicine, Siberian State Medical University, 2 Moskovskii trakt, Tomsk 634050, Russia; (E.A.R.); (A.I.V.)
| | - Alla A. Boshchenko
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| |
Collapse
|
157
|
Zhang S, Yang Y, Lv X, Liu W, Zhu S, Wang Y, Xu H. Unraveling the Intricate Roles of Exosomes in Cardiovascular Diseases: A Comprehensive Review of Physiological Significance and Pathological Implications. Int J Mol Sci 2023; 24:15677. [PMID: 37958661 PMCID: PMC10650316 DOI: 10.3390/ijms242115677] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Exosomes, as potent intercellular communication tools, have garnered significant attention due to their unique cargo-carrying capabilities, which enable them to influence diverse physiological and pathological functions. Extensive research has illuminated the biogenesis, secretion, and functions of exosomes. These vesicles are secreted by cells in different states, exerting either protective or harmful biological functions. Emerging evidence highlights their role in cardiovascular disease (CVD) by mediating comprehensive interactions among diverse cell types. This review delves into the significant impacts of exosomes on CVD under stress and disease conditions, including coronary artery disease (CAD), myocardial infarction, heart failure, and other cardiomyopathies. Focusing on the cellular signaling and mechanisms, we explore how exosomes mediate multifaceted interactions, particularly contributing to endothelial dysfunction, oxidative stress, and apoptosis in CVD pathogenesis. Additionally, exosomes show great promise as biomarkers, reflecting differential expressions of NcRNAs (miRNAs, lncRNAs, and circRNAs), and as therapeutic carriers for targeted CVD treatment. However, the specific regulatory mechanisms governing exosomes in CVD remain incomplete, necessitating further exploration of their characteristics and roles in various CVD-related contexts. This comprehensive review aims to provide novel insights into the biological implications of exosomes in CVD and offer innovative perspectives on the diagnosis and treatment of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China; (S.Z.); (Y.Y.); (X.L.); (W.L.); (S.Z.)
| | - Hongfei Xu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China; (S.Z.); (Y.Y.); (X.L.); (W.L.); (S.Z.)
| |
Collapse
|
158
|
Postma RJ, Broekhoven AG, Verspaget HW, de Boer H, Hankemeier T, Coenraad MJ, van Duinen V, van Zonneveld AJ. Novel Morphological Profiling Assay Connects ex Vivo Endothelial Cell Responses to Disease Severity in Liver Cirrhosis. GASTRO HEP ADVANCES 2023; 3:238-249. [PMID: 39129954 PMCID: PMC11307659 DOI: 10.1016/j.gastha.2023.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/16/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims Endothelial cell (EC) dysfunction in response to circulating plasma factors is a known causal factor in many systemic diseases. However, no appropriate assay is available to investigate this causality ex vivo. In liver cirrhosis, systemic inflammation is identified as central mechanism in progression from compensated to decompensated cirrhosis (DC), but the role of ECs therein is unknown. We aimed to develop a novel ex vivo assay for assessing EC responses to patient-derived plasma (PDP) and assess the potential of this assay in a cohort of liver cirrhosis patients. Methods Image-based morphological profiling was utilized to assess the impact of PDP on cultured ECs. Endothelial cell (EC) monolayers were exposed to 25% stabilized PDP (20 compensated cirrhoses, 20 DCs, and 20 healthy controls (HCs). Single-cell morphological profiles were extracted by automated image-analysis following staining of multiple cellular components and high-content imaging. Patient profiles were created by dimension reduction and cell-to-patient data aggregation, followed by multivariate-analysis to stratify patients and identify discriminating features. Results Patient-derived plasma (PDP) exposure induced profound changes in EC morphology, displaying clear differences between controls and DC patients. Compensated cirrhosis patients showed overlap with healthy controls and DC patients. Supervised analysis showed Child-Pugh (CP) class could be predicted from EC morphology. Most importantly, CP-C patients displayed distinct EC phenotypes, in which mitochondrial changes were most discriminative. Conclusion Morphological profiling presents a viable tool to assess the endothelium ex vivo. We demonstrated that the EC phenotype corresponds with disease severity in liver cirrhosis. Moreover, our results suggest the presence of mitochondrial dysfunction in ECs of CP-C patient.
Collapse
Affiliation(s)
- Rudmer J. Postma
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Annelotte G.C. Broekhoven
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hein W. Verspaget
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hetty de Boer
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas Hankemeier
- Department of Analytical BioSciences, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Minneke J. Coenraad
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vincent van Duinen
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- MIMETAS B.V., Oegstgeest, The Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
159
|
Hasan M, Al-Thani H, El-Menyar A, Zeidan A, Al-Thani A, Yalcin HC. Disturbed hemodynamics and oxidative stress interaction in endothelial dysfunction and AAA progression: Focus on Nrf2 pathway. Int J Cardiol 2023; 389:131238. [PMID: 37536420 DOI: 10.1016/j.ijcard.2023.131238] [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: 02/03/2023] [Revised: 04/30/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Hemodynamic shear stress is one of the major factors that are involved in the pathogenesis of many cardiovascular diseases including atherosclerosis and abdominal aortic aneurysm (AAA), through its modulatory effect on the endothelial cell's redox homeostasis and mechanosensitive gene expression. Among important mechanisms, oxidative stress, endoplasmic reticulum stress activation, and the subsequent endothelial dysfunction are attributed to disturbed blood flow and low shear stress in the vascular curvature and bifurcations which are considered atheroprone regions and aneurysm occurrence spots. Many pathways were shown to be involved in AAA progression. Of particular interest from recent findings is, the (Nrf2)/Keap-1 pathway, where Nrf2 is a transcription factor that has antioxidant properties and is strongly associated with several CVDs, yet, the exact mechanism by which Nrf2 alleviates CVDs still to be elucidated. Nrf2 expression is closely affected by shear stress and was shown to participate in AAA. In the current review paper, we discussed the link between disturbed hemodynamics and its effect on Nrf2 as a mechanosensitive gene and its role in the development of endothelial dysfunction which is linked to the progression of AAA.
Collapse
Affiliation(s)
- Maram Hasan
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Hassan Al-Thani
- Department of Surgery, Trauma and Vascular Surgery, Hamad General Hospital, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Ayman El-Menyar
- Department of Surgery, Trauma and Vascular Surgery, Hamad General Hospital, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar; Clinical Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Asad Zeidan
- Department of Basic Sciences, College of Medicine, QU health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Asmaa Al-Thani
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar; Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Huseyin C Yalcin
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
160
|
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
|
161
|
Naderi-Meshkin H, Cornelius VA, Eleftheriadou M, Potel KN, Setyaningsih WAW, Margariti A. Vascular organoids: unveiling advantages, applications, challenges, and disease modelling strategies. Stem Cell Res Ther 2023; 14:292. [PMID: 37817281 PMCID: PMC10566155 DOI: 10.1186/s13287-023-03521-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023] Open
Abstract
Understanding mechanisms and manifestations of cardiovascular risk factors, including diabetes, on vascular cells such as endothelial cells, pericytes, and vascular smooth muscle cells, remains elusive partly due to the lack of appropriate disease models. Therefore, here we explore different aspects for the development of advanced 3D in vitro disease models that recapitulate human blood vessel complications using patient-derived induced pluripotent stem cells, which retain the epigenetic, transcriptomic, and metabolic memory of their patient-of-origin. In this review, we highlight the superiority of 3D blood vessel organoids over conventional 2D cell culture systems for vascular research. We outline the key benefits of vascular organoids in both health and disease contexts and discuss the current challenges associated with organoid technology, providing potential solutions. Furthermore, we discuss the diverse applications of vascular organoids and emphasize the importance of incorporating all relevant cellular components in a 3D model to accurately recapitulate vascular pathophysiology. As a specific example, we present a comprehensive overview of diabetic vasculopathy, demonstrating how the interplay of different vascular cell types is critical for the successful modelling of complex disease processes in vitro. Finally, we propose a strategy for creating an organ-specific diabetic vasculopathy model, serving as a valuable template for modelling other types of vascular complications in cardiovascular diseases by incorporating disease-specific stressors and organotypic modifications.
Collapse
Affiliation(s)
- Hojjat Naderi-Meshkin
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Victoria A Cornelius
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Magdalini Eleftheriadou
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Koray Niels Potel
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Wiwit Ananda Wahyu Setyaningsih
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
- Department of Anatomy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Sleman, D.I. Yogyakarta, 55281, Indonesia
| | - Andriana Margariti
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
162
|
Favre J, Roy C, Guihot AL, Drouin A, Laprise M, Gillis MA, Robson SC, Thorin E, Sévigny J, Henrion D, Kauffenstein G. NTPDase1/CD39 Ectonucleotidase Is Necessary for Normal Arterial Diameter Adaptation to Flow. Int J Mol Sci 2023; 24:15038. [PMID: 37894719 PMCID: PMC10606763 DOI: 10.3390/ijms242015038] [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: 08/28/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
NTPDase1/CD39, the major vascular ectonucleotidase, exerts thrombo-immunoregulatory function by controlling endothelial P2 receptor activation. Despite the well-described release of ATP from endothelial cells, few data are available regarding the potential role of CD39 as a regulator of arterial diameter. We thus investigated the contribution of CD39 in short-term diameter adaptation and long-term arterial remodeling in response to flow using Entpd1-/- male mice. Compared to wild-type littermates, endothelial-dependent relaxation was modified in Entpd1-/- mice. Specifically, the vasorelaxation in response to ATP was potentiated in both conductance (aorta) and small resistance (mesenteric and coronary) arteries. By contrast, the relaxing responses to acetylcholine were supra-normalized in thoracic aortas while decreased in resistance arteries from Entpd1-/- mice. Acute flow-mediated dilation, measured via pressure myography, was dramatically diminished and outward remodeling induced by in vivo chronic increased shear stress was altered in the mesenteric resistance arteries isolated from Entpd1-/- mice compared to wild-types. Finally, changes in vascular reactivity in Entpd1-/- mice were also evidenced by a decrease in the coronary output measured in isolated perfused hearts compared to the wild-type mice. Our results highlight a key regulatory role for purinergic signaling and CD39 in endothelium-dependent short- and long-term arterial diameter adaptation to increased flow.
Collapse
Affiliation(s)
- Julie Favre
- MITOVASC Institute, CARFI Facility, CNRS UMR 6015, INSERM U1083, Angers University, 49045 Angers, France; (J.F.); (D.H.)
| | - Charlotte Roy
- MITOVASC Institute, CARFI Facility, CNRS UMR 6015, INSERM U1083, Angers University, 49045 Angers, France; (J.F.); (D.H.)
| | - Anne-Laure Guihot
- MITOVASC Institute, CARFI Facility, CNRS UMR 6015, INSERM U1083, Angers University, 49045 Angers, France; (J.F.); (D.H.)
| | - Annick Drouin
- Montreal Heart Institute, Department of Surgery, Université de Montréal, Montreal, QC H1T 1C8, Canada
| | - Manon Laprise
- Animal Physiology Service, Institut de Recherches Cliniques de Montreal (IRCM), Montreal, QC H2W 1R7, Canada;
| | - Marc-Antoine Gillis
- Montreal Heart Institute, Department of Surgery, Université de Montréal, Montreal, QC H1T 1C8, Canada
| | - Simon C. Robson
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Eric Thorin
- Montreal Heart Institute, Department of Surgery, Université de Montréal, Montreal, QC H1T 1C8, Canada
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC G1V 4G2, Canada
- Département de Microbiologie-Infectiologie et D’immunologie, Faculté de Médecine, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - Daniel Henrion
- MITOVASC Institute, CARFI Facility, CNRS UMR 6015, INSERM U1083, Angers University, 49045 Angers, France; (J.F.); (D.H.)
| | - Gilles Kauffenstein
- MITOVASC Institute, CARFI Facility, CNRS UMR 6015, INSERM U1083, Angers University, 49045 Angers, France; (J.F.); (D.H.)
- INSERM UMR 1260—Regenerative Nanomedicine, CRBS, Strasbourg University, 67000 Strasbourg, France
| |
Collapse
|
163
|
Shahdadian S, Wang X, Liu H. Directed physiological networks in the human prefrontal cortex at rest and post transcranial photobiomodulation. RESEARCH SQUARE 2023:rs.3.rs-3393702. [PMID: 37886539 PMCID: PMC10602070 DOI: 10.21203/rs.3.rs-3393702/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Cerebral infra-slow oscillation (ISO) is a source of vasomotion in endogenic (E; 0.005-0.02 Hz), neurogenic (N; 0.02-0.04 Hz), and myogenic (M; 0.04-0.2 Hz) frequency bands. In this study, we quantified changes in prefrontal concentrations of oxygenated hemoglobin ( Δ [ H b O ] ) and redox-state cytochrome c oxidase ( Δ [ C C O ] ) as hemodynamic and metabolic activity metrics, and electroencephalogram (EEG) powers as electrophysiological activity, using concurrent measurements of 2-channel broadband near-infrared spectroscopy and EEG on the forehead of 22 healthy participants at rest. After preprocessing, the multi-modality signals were analyzed using generalized partial directed coherence to construct unilateral neurophysiological networks among the three neurophysiological metrics (with simplified symbols of HbO, CCO, and EEG) in each E/N/M frequency band. The links in these networks represent neurovascular, neurometabolic, and metabolicvascular coupling (NVC, NMC, and MVC). The results illustrate that the demand for oxygen by neuronal activity and metabolism (EEG and CCO) drives the hemodynamic supply (HbO) in all E/N/M bands in the resting prefrontal cortex. Furthermore, to investigate the effect of transcranial photobiomodulation (tPBM), we performed a sham-controlled study by delivering an 800-nm laser beam to the left and right prefrontal cortex of the same participants. After performing the same data processing and statistical analysis, we obtained novel and important findings: tPBM delivered on either side of the prefrontal cortex triggered the alteration or reversal of directed network couplings among the three neurophysiological entities (i.e., HbO, CCO, and EEG frequency-specific powers) in the physiological network in the E and N bands, demonstrating that during the post-tPBM period, both metabolism and hemodynamic supply drive electrophysiological activity in directed network coupling of the PFC. Overall, this study revealed that tPBM facilitates significant modulation of the directionality of neurophysiological networks in electrophysiological, metabolic, and hemodynamic activities.
Collapse
|
164
|
Martin ZT, Al-Daas IO, Cardenas N, Kolade JO, Merlau ER, Vu JK, Brown KK, Brothers RM. Peripheral and Cerebral Vasodilation in Black and White Women: Examining the Impact of Psychosocial Stress Exposure Versus Internalization and Coping. Hypertension 2023; 80:2122-2134. [PMID: 37534492 PMCID: PMC10530116 DOI: 10.1161/hypertensionaha.123.21230] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Black women have among the highest rates of cardiovascular and cerebrovascular disease prevalence and mortality in part due to blunted vascular function. Psychosocial stress likely also contributes but its relationship to vascular function remains incompletely understood. Recent studies suggest that stress internalization and coping strategies are more important than stress exposures alone. We hypothesized that Black women would have blunted peripheral and cerebral vasodilation and that, among Black women, this would be inversely related with psychosocial stress internalization/coping but not stress exposures. METHODS Healthy Black (n=21; 20±2 years) and White (n=16; 25±7 years) women underwent testing for forearm reactive hyperemia, brachial artery flow-mediated dilation (FMD), and cerebrovascular reactivity. Psychosocial stress exposures (adverse childhood experiences; past week discrimination) and internalization/coping techniques (John Henryism Active Coping Scale; Giscombe Superwoman Schema Questionnaire) were assessed. RESULTS Reactive hyperemia and cerebrovascular reactivity were not different between groups (P>0.05), whereas FMD was lower in Black women (P=0.007). Neither adverse childhood experiences nor past week discrimination were associated with FMD in either group (P>0.05 for all). John Henryism Active Coping Scale scores were negatively associated with FMD in Black women (P=0.014) but positively associated with FMD in White women (P=0.042). Superwoman Schema-Succeed was negatively associated (P=0.044) and Superwoman Schema-Vulnerable tended to be negatively associated (P=0.057) with FMD in Black women. CONCLUSIONS These findings indicate that blunted FMD in Black women may be due more to stress internalization and maladaptive coping than stress exposures alone.
Collapse
Affiliation(s)
- Zachary T Martin
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX
| | - Iman O Al-Daas
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX
| | - Natalia Cardenas
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX
| | - John O Kolade
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX
| | - Emily R Merlau
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX
| | - Joshua K Vu
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX
| | - Kyrah K Brown
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX
| | - R Matthew Brothers
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX
| |
Collapse
|
165
|
Shimabukuro M. L-Arginine, Nitric Oxide, and Endothelial Dysfunction Underlying Atherosclerotic Cardiovascular Disease (ASCVD). J Atheroscler Thromb 2023; 30:1311-1312. [PMID: 37245961 PMCID: PMC10564653 DOI: 10.5551/jat.ed235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/30/2023] Open
Affiliation(s)
- Michio Shimabukuro
- Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
166
|
Geethika M, Singh N, Kumar S, Kumar SKN, Mugesh G. A Redox Modulatory SOD Mimetic Nanozyme Prevents the Formation of Cytotoxic Peroxynitrite and Improves Nitric Oxide Bioavailability in Human Endothelial Cells. Adv Healthc Mater 2023; 12:e2300621. [PMID: 37524524 DOI: 10.1002/adhm.202300621] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/22/2023] [Indexed: 08/02/2023]
Abstract
The endothelium-derived signalling molecule nitric oxide (NO) in addition to controlling multifarious servo-regulatory functions, suppresses key processes in vascular lesion formation and prevents atherogenesis and other vascular abnormalities. The conversion of NO into cytotoxic and powerful oxidant peroxynitrite (ONOO- ) in a superoxide (O2 .- )-rich environment has emerged as a major reason for reduced NO levels in vascular walls, leading to endothelial dysfunction and cardiovascular complications. So, designing superoxide dismutase (SOD) mimetics that can selectively catalyze the dismutation of O2 .- in the presence of NO, considering their rapid reaction is challenging and is of therapeutic relevance. Herein, the authors report that SOD mimetic cerium vanadate (CeVO4 ) nanozymes effectively regulate the bioavailability of both NO and O2 .- , the two vital constitutive molecules of vascular endothelium, even in the absence of cellular SOD enzyme. The nanozymes optimally modulate the O2 .- level in endothelial cells under oxidative stress conditions and improve endogenously generated NO levels by preventing the formation of ONOO- . Furthermore, nanoparticles exhibit size- and morphology-dependent uptake into the cells and internalize via the clathrin-mediated endocytosis pathway. Intravenous administration of CeVO4 nanoparticles in mice caused no definite organ toxicity and unaltered haematological and biochemical parameters, indicating their biosafety and potential use in biological applications.
Collapse
Affiliation(s)
- Motika Geethika
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Namrata Singh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Sagar Kumar
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | | | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
167
|
Cheng S, Ni J, Deng W, Wang P. Diagnostic Ability of Perivascular Fat Attenuation Index in Predicting Atherosclerotic Plaque Formation Proximal to Myocardial Bridging of the Left Anterior Descending Artery within 3 Years. Acad Radiol 2023; 30:2234-2242. [PMID: 37474348 DOI: 10.1016/j.acra.2023.06.019] [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: 05/13/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/22/2023]
Abstract
RATIONALE AND OBJECTIVES This study was designed to investigate the association between the perivascular fat attenuation index (FAI) and atherosclerotic plaque formation proximal to myocardial bridging (MB) of the left anterior descending artery (LAD) within 3 years. MATERIALS AND METHODS LAD-MB patients who underwent coronary computed tomography angiography at least twice between January 2016 and December 2022 were retrospectively included in this study. In total, 99 LAD-MB patients were included in the study. Based on the formation of atherosclerotic plaques proximal to LAD-MB during follow-up, the patients were classified into two groups: LAD-MB with plaque formation and LAD-MB without plaque formation within 3 years. The anatomical features, clinical factors, and proximal perivascular FAI of LAD-MB were measured and recorded. The association of the previously mentioned factors with the presence of atherosclerotic plaque proximal to LAD-MB was determined. RESULTS The results showed that MB length, MB stenosis, and the perivascular FAI were significant predictors of the formation of atherosclerotic plaques proximal to LAD-MB. The area under the curve of the combined predictive model incorporating MB length, MB stenosis, and the perivascular FAI was 0.901(95% confidence interval: 0.824-0.952), with higher diagnostic performance than any other single parameter (all P < 0.05). Moreover, dynamic changes in the perivascular FAI of the vascular segments proximal to LAD-MB were higher in high-risk plaques than in non-high-risk plaques (P = 0.003). CONCLUSION The combined use of the perivascular FAI, MB length, and MB stenosis may enable prediction of the probability of atherosclerotic plaque formation proximal to LAD-MB within 3 years. Dynamic changes in perivascular FAI were associated with the vulnerability of plaques proximal to LAD-MB.
Collapse
Affiliation(s)
- Shuihua Cheng
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China (S.C., J.N., P.W.)
| | - Jiong Ni
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China (S.C., J.N., P.W.)
| | - Weiwei Deng
- CT Clinical Science, Philips Healthcare, Shanghai, China (W.D.)
| | - Peijun Wang
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China (S.C., J.N., P.W.).
| |
Collapse
|
168
|
Wang J, Tan Y, Dai Y, Hu K, Tan X, Jiang S, Li G, Zhang X, Kang L, Wang X, Xu B. Intranasal Delivery of Endothelial Cell-Derived Extracellular Vesicles with Supramolecular Gel Attenuates Myocardial Ischemia-Reperfusion Injury. Int J Nanomedicine 2023; 18:5495-5510. [PMID: 37791323 PMCID: PMC10544033 DOI: 10.2147/ijn.s420301] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
PURPOSE Myocardial ischemia-reperfusion injury after myocardial infarction has always been a difficult problem in clinical practice. Endothelial cells and their secreted extracellular vesicles are closely related to inflammation, thrombosis formation, and other processes after injury. Meanwhile, low-molecular-weight gelators have shown great potential for nasal administration. This study aims to explore the therapeutic effects and significance of endothelial cell-derived extracellular vesicles combined with a hydrogel for nasal administration on myocardial ischemia-reperfusion injury. METHODS We chose a gel system composed of a derivative of glutamine amide and benzaldehyde as the extracellular vesicle delivery vehicle. This hydrogel was combined with extracellular vesicles extracted from mouse aortic endothelial cells and administered multiple times intranasally in a mouse model of ischemia-reperfusion injury to the heart. The delivery efficiency of the extracellular vesicle-hydrogel combination was evaluated by flow cytometry and immunofluorescence. Echocardiography, TTC Evan's Blue and Masson's staining were used to assess mouse cardiac function, infarct area, and cardiac fibrosis level. Flow cytometry, ELISA, and immunofluorescence staining were used to investigate changes in mouse inflammatory cells, cytokines, and vascular neogenesis. RESULTS The vesicles combined with the hydrogel have good absorption in the nasal cavity. The hydrogel combined with vesicles reduces the levels of pro-inflammatory Ly6C (high) monocytes/macrophages and neutrophils. It can also reduce the formation of microcirculation thrombi in the infarcted area, improve endothelial barrier function, and increase microvascular density in the injured area. As a result, the heart function of mice is improved and the infarct area is reduced. CONCLUSION We first demonstrated that the combination of extracellular vesicles and hydrogel has a better absorption efficiency in the nasal cavity, which can improve myocardial ischemia-reperfusion injury by inhibiting inflammatory reactions and protecting endothelial function. Nasal administration of vesicles combined with hydrogel is a potential therapeutic direction.
Collapse
Affiliation(s)
- Junzhuo Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Ying Tan
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Yang Dai
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Geriatrics, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Ke Hu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xi Tan
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Shaoli Jiang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, People’s Republic of China
| | - Guannan Li
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Xinlin Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Xiaojian Wang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, People’s Republic of China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
169
|
Zheng PF, Rong JJ, Zheng ZF, Liu ZY, Pan HW, Liu P. Investigating the causal effect of Dickkopf-1 on coronary artery disease and ischemic stroke: a Mendelian randomization study. Aging (Albany NY) 2023; 15:9797-9808. [PMID: 37742224 PMCID: PMC10564415 DOI: 10.18632/aging.205050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023]
Abstract
Epidemiological investigations have indicated a correlation between elevated plasma levels of Dickkopf-related protein 1 (DKK1) and the presence of atherosclerosis. However, the exact causal relationship of DKK1 with the development of coronary artery disease (CAD) and ischemic stroke (IS) remains unclear. To address this gap, our study aimed to explore their causal association using a two-sample Mendelian randomization (MR) approach. We obtained summary statistics from genome-wide association studies (GWAS) meta-analyses conducted by Folkersen et al. and Nikpay et al., which included data from 21,758 individuals for DKK1 and 42,096 cases of CAD. Additionally, we obtained data from the FinnGen biobank analysis round 5, which included 10,551 cases of IS. Eight MR methods were employed to estimate causal effects and detect directional pleiotropy. Our findings demonstrated that genetic liability to DKK1 was associated with increased risks of CAD (odds ratio [OR]: 1.087; 95% confidence interval [CI]: 1.024-1.154; P = 0.006) and IS (OR: 1.096; 95% CI: 1.004-1.195; P = 0.039). These results establish a causal link between genetic liability to DKK1 and elevated risks of CAD and IS. Consequently, DKK1 may represent a promising therapeutic target for the prevention and treatment of CAD and IS.
Collapse
Affiliation(s)
- Peng-Fei Zheng
- Cardiology Department, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, Furong, Changsha 410000, Hunan, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
| | - Jing-Jing Rong
- Cardiology Department, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, Furong, Changsha 410000, Hunan, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
| | - Zhao-Fen Zheng
- Cardiology Department, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, Furong, Changsha 410000, Hunan, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
| | - Zheng-Yu Liu
- Cardiology Department, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, Furong, Changsha 410000, Hunan, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
| | - Hong-Wei Pan
- Cardiology Department, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, Furong, Changsha 410000, Hunan, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
| | - Peng Liu
- Department of Cardiology, The Central Hospital of ShaoYang, Shaoyang 422000, Hunan, China
| |
Collapse
|
170
|
Lu H, Jiang X, He L, Ji X, Li X, Liu S, Sun Y, Qin X, Xiong X, Philipsen S, Xi B, Zhang M, Yang J, Zhang C, Zhang Y, Zhang W. Endothelial Sp1/Sp3 are essential to the effect of captopril on blood pressure in male mice. Nat Commun 2023; 14:5891. [PMID: 37735515 PMCID: PMC10514286 DOI: 10.1038/s41467-023-41567-1] [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: 04/01/2022] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
Endothelial dysfunction represents a major cardiovascular risk factor for hypertension. Sp1 and Sp3 belong to the specificity protein and Krüppel-like transcription factor families. They are ubiquitously expressed and closely associated with cardiovascular development. We investigate the role of Sp1 and Sp3 in endothelial cells in vivo and evaluate whether captopril, an angiotensin-converting enzyme inhibitor (ACEI), targets Sp1/Sp3 to exert its effects. Inducible endothelial-specific Sp1/Sp3 knockout mice are generated to elucidate their role in endothelial cells. Tamoxifen-induced deletion of endothelial Sp1 and Sp3 in male mice decreases the serum nitrite/nitrate level, impairs endothelium-dependent vasodilation, and causes hypertension and cardiac remodeling. The beneficial actions of captopril are abolished by endothelial-specific deletion of Sp1/Sp3, indicating that they may be targets for ACEIs. Captopril increases Sp1/Sp3 protein levels by recruiting histone deacetylase 1, which elevates deacetylation and suppressed degradation of Sp1/Sp3. Sp1/Sp3 represents innovative therapeutic target for captopril to prevent cardiovascular diseases.
Collapse
Affiliation(s)
- Hanlin Lu
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiuxin Jiang
- Department of Bariatric and Metabolic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lifan He
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xuyang Ji
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinyun Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shaozhuang Liu
- Department of Bariatric and Metabolic Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuanyuan Sun
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoteng Qin
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiwen Xiong
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Bo Xi
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jianmin Yang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yun Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wencheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
171
|
Zhao H, Wu D, Gyamfi MA, Wang P, Luecht C, Pfefferkorn AM, Ashraf MI, Kamhieh-Milz J, Witowski J, Dragun D, Budde K, Schindler R, Zickler D, Moll G, Catar R. Expanded Hemodialysis ameliorates uremia-induced impairment of vasculoprotective KLF2 and concomitant proinflammatory priming of endothelial cells through an ERK/AP1/cFOS-dependent mechanism. Front Immunol 2023; 14:1209464. [PMID: 37795100 PMCID: PMC10546407 DOI: 10.3389/fimmu.2023.1209464] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/31/2023] [Indexed: 10/06/2023] Open
Abstract
Aims Expanded hemodialysis (HDx) therapy with improved molecular cut-off dialyzers exerts beneficial effects on lowering uremia-associated chronic systemic microinflammation, a driver of endothelial dysfunction and cardiovascular disease (CVD) in hemodialysis (HD) patients with end-stage renal disease (ESRD). However, studies on the underlying molecular mechanisms are still at an early stage. Here, we identify the (endothelial) transcription factor Krüppel-like factor 2 (KLF2) and its associated molecular signalling pathways as key targets and regulators of uremia-induced endothelial micro-inflammation in the HD/ESRD setting, which is crucial for vascular homeostasis and controlling detrimental vascular inflammation. Methods and results First, we found that human microvascular endothelial cells (HMECs) and other typical endothelial and kidney model cell lines (e.g. HUVECs, HREC, and HEK) exposed to uremic serum from patients treated with two different hemodialysis regimens in the Permeability Enhancement to Reduce Chronic Inflammation II (PERCI-II) crossover clinical trial - comparing High-Flux (HF) and Medium Cut-Off (MCO) membranes - exhibited strongly reduced expression of vasculoprotective KLF2 with HF dialyzers, while dialysis with MCO dialyzers led to the maintenance and restoration of physiological KLF2 levels in HMECs. Mechanistic follow-up revealed that the strong downmodulation of KLF2 in HMECs exposed to uremic serum was mediated by a dominant engagement of detrimental ERK instead of beneficial AKT signalling, with subsequent AP1-/c-FOS binding in the KLF2 promoter region, followed by the detrimental triggering of pleiotropic inflammatory mediators, while the introduction of a KLF2 overexpression plasmid could restore physiological KLF2 levels and downmodulate the detrimental vascular inflammation in a mechanistic rescue approach. Conclusion Uremia downmodulates vasculoprotective KLF2 in endothelium, leading to detrimental vascular inflammation, while MCO dialysis with the novel improved HDx therapy approach can maintain physiological levels of vasculoprotective KLF2.
Collapse
Affiliation(s)
- Hongfan Zhao
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Dashan Wu
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Michael Adu Gyamfi
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Pinchao Wang
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Christian Luecht
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | | | | | - Julian Kamhieh-Milz
- Institute of Transfusion Medicine, at Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Janusz Witowski
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Duska Dragun
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Ralf Schindler
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Daniel Zickler
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT) and Berlin-Brandenburg School for Regenerative Therapies (BSRT), at Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, at Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
172
|
Qiu Y, Xu S, Chen X, Wu X, Zhou Z, Zhang J, Tu Q, Dong B, Liu Z, He J, Zhang X, Liu S, Su C, Huang H, Xia W, Tao J. NAD + exhaustion by CD38 upregulation contributes to blood pressure elevation and vascular damage in hypertension. Signal Transduct Target Ther 2023; 8:353. [PMID: 37718359 PMCID: PMC10505611 DOI: 10.1038/s41392-023-01577-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 06/16/2023] [Accepted: 07/23/2023] [Indexed: 09/19/2023] Open
Abstract
Hypertension is characterized by endothelial dysfunction and arterial stiffness, which contribute to the pathogenesis of atherosclerotic cardiovascular diseases. Nicotinamide adenine dinucleotide (NAD+) is an indispensable cofactor in all living cells that is involved in fundamental biological processes. However, in hypertensive patients, alterations in NAD+ levels and their relation with blood pressure (BP) elevation and vascular damage have not yet been studied. Here we reported that hypertensive patients exhibited lower NAD+ levels, as detected by high-performance liquid chromatography-mass spectrometry (HPLC-MS), in both peripheral blood mononuclear cells (PBMCs) and aortas, which was parallel to vascular dysfunction. NAD+ boosting therapy with nicotinamide mononucleotide (NMN) supplement reduced BP and ameliorated vascular dysfunction in hypertensive patients (NCT04903210) and AngII-induced hypertensive mice. Upregulation of CD38 in endothelial cells led to endothelial NAD+ exhaustion by reducing NMN bioavailability. Pro-inflammatory macrophages infiltration and increase in IL-1β generation derived from pro-inflammatory macrophages resulted in higher CD38 expression by activating JAK1-STAT1 signaling pathway. CD38 KO, CD38 inhibitors treatment, or adeno-associated virus (AAV)-mediated endothelial CD38 knockdown lowered BP and improved vascular dysfunction in AngII-induced hypertensive mice. The present study demonstrated for the first time that endothelial CD38 activation and subsequently accelerated NAD+ degradation due to enhanced macrophage-derived IL-1β production was responsible for BP elevation and vascular damage in hypertension. NAD+ boosting therapy can be used as a novel therapeutic strategy for the management of hypertensive patients.
Collapse
Affiliation(s)
- Yumin Qiu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Shiyue Xu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Xi Chen
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Xing Wu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Zhe Zhou
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Jianning Zhang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Qiang Tu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Bing Dong
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Zhefu Liu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Jiang He
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Xiaoyu Zhang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Shuangshuang Liu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Chen Su
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, 518033, Shenzhen, China.
| | - Wenhao Xia
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China.
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China.
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China.
- Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, 530022, Nanning, China.
| | - Jun Tao
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China.
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, 510080, Guangzhou, China.
- Key Laboratory on Assisted Circulation, Ministry of Health, 510080, Guangzhou, China.
| |
Collapse
|
173
|
Shields KL, Jarrett CL, Bisconti AV, Park SH, Craig JC, Broxterman RM, Richardson RS. Preserved endothelium-independent vascular function with aging in men and women: evidence from the peripheral and cerebral vasculature. J Appl Physiol (1985) 2023; 135:559-571. [PMID: 37391885 PMCID: PMC10538978 DOI: 10.1152/japplphysiol.00571.2022] [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: 09/27/2022] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023] Open
Abstract
In the peripheral and cerebral vasculature, the impact of aging and sex on the endothelial-independent functional capacity of vascular smooth muscle cells (VSMCs) is not well understood, nor is it known whether such VSMC functions in these vascular beds reflect one another. Therefore, endothelium-independent dilation, at both the conduit (Δ diameter) and microvascular (Δ vascular conductance, VC) level, elicited by sublingual nitroglycerin (NTG, 0.8 mg of Nitrostat), compared with sham-delivery (control), was assessed using Doppler ultrasound in the popliteal (PA) and middle cerebral (MCA) artery of 20 young [23 ± 4 yr, 10 males (YM)/10 females (YF)] and 21 old [69 ± 5 yr, 11 males (OM)/10 females (OF)] relatively healthy adults. In the PA, compared with zero, NTG significantly increased diameter in all groups (YM: 0.29 ± 0.13, YF: 0.35 ± 0.26, OM: 0.30 ± 0.18, OF: 0.31 ± 0.14 mm), while control did not. The increase in VC only achieved significance in the OF (0.22 ± 0.31 mL/min/mmHg). In the MCA, compared with zero, NTG significantly increased diameter and VC in all groups (YM: 0.89 ± 0.30, 1.06 ± 1.28; YF: 0.97 ± 0.31, 1.84 ± 1.07; OM: 0.90 ± 0.42, 0.72 ± 0.99; OF: 0.74 ± 0.32, 1.19 ± 1.18, mm and mL/min/mmHg, respectively), while control did not. There were no age or sex differences or age-by-sex interactions for both the NTG-induced PA and MCA dilation and VC. In addition, PA and MCA dilation and VC responses to NTG were not related when grouped by age, sex, or as all subjects (r = 0.04-0.44, P > 0.05). Thus, peripheral and cerebral endothelial-independent VSMC function appears to be unaffected by age or sex, and variations in such VSMC function in one of these vascular beds are not reflected in the other.NEW & NOTEWORTHY To confidently explain peripheral and cerebral vascular dysfunction, it is essential to have a clear understanding of the endothelial-independent function of VSMCs across age and sex. By assessing endothelium-independent dilation using sublingual nitroglycerin, endothelial-independent VSMC function in the periphery (popliteal artery), and in the cerebral circulation (middle cerebral artery), was not different due to age or sex. In addition, endothelial-independent VSMC function in one of these vascular beds is not reflected in the other.
Collapse
Affiliation(s)
- Katherine L Shields
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Catherine L Jarrett
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Angela V Bisconti
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Soung Hun Park
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Jesse C Craig
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Ryan M Broxterman
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
- Center on Aging, University of Utah, Salt Lake City, Utah, United States
| | - Russell S Richardson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Medical Center, Salt Lake City, Utah, United States
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
- Center on Aging, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
174
|
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
|
175
|
Wojtasińska A, Frąk W, Lisińska W, Sapeda N, Młynarska E, Rysz J, Franczyk B. Novel Insights into the Molecular Mechanisms of Atherosclerosis. Int J Mol Sci 2023; 24:13434. [PMID: 37686238 PMCID: PMC10487483 DOI: 10.3390/ijms241713434] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Atherosclerosis is one of the most fatal diseases in the world. The associated thickening of the arterial wall and its background and consequences make it a very composite disease entity with many mechanisms that lead to its creation. It is an active process, and scientists from various branches are engaged in research, including molecular biologists, cardiologists, and immunologists. This review summarizes the available information on the pathophysiological implications of atherosclerosis, focusing on endothelium dysfunction, inflammatory factors, aging, and uric acid, vitamin D, and miRNA expression as recent evidence of interactions of the molecular and cellular elements. Analyzing new discoveries for the underlying causes of this condition assists the general research to improve understanding of the mechanism of pathophysiology and thus prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Armanda Wojtasińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Weronika Frąk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Wiktoria Lisińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Natalia Sapeda
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (W.L.)
| |
Collapse
|
176
|
Xue J, Zhang Z, Sun Y, Jin D, Guo L, Li X, Zhao D, Feng X, Qi W, Zhu H. Research Progress and Molecular Mechanisms of Endothelial Cells Inflammation in Vascular-Related Diseases. J Inflamm Res 2023; 16:3593-3617. [PMID: 37641702 PMCID: PMC10460614 DOI: 10.2147/jir.s418166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Endothelial cells (ECs) are widely distributed inside the vascular network, forming a vital barrier between the bloodstream and the walls of blood vessels. These versatile cells serve myriad functions, including the regulation of vascular tension and the management of hemostasis and thrombosis. Inflammation constitutes a cascade of biological responses incited by biological, chemical, or physical stimuli. While inflammation is inherently a protective mechanism, dysregulated inflammation can precipitate a host of vascular pathologies. ECs play a critical role in the genesis and progression of vascular inflammation, which has been implicated in the etiology of numerous vascular disorders, such as atherosclerosis, cardiovascular diseases, respiratory diseases, diabetes mellitus, and sepsis. Upon activation, ECs secrete potent inflammatory mediators that elicit both innate and adaptive immune reactions, culminating in inflammation. To date, no comprehensive and nuanced account of the research progress concerning ECs and inflammation in vascular-related maladies exists. Consequently, this review endeavors to synthesize the contributions of ECs to inflammatory processes, delineate the molecular signaling pathways involved in regulation, and categorize and consolidate the various models and treatment strategies for vascular-related diseases. It is our aspiration that this review furnishes cogent experimental evidence supporting the established link between endothelial inflammation and vascular-related pathologies, offers a theoretical foundation for clinical investigations, and imparts valuable insights for the development of therapeutic agents targeting these diseases.
Collapse
Affiliation(s)
- Jiaojiao Xue
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Ziwei Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yuting Sun
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Di Jin
- Department of Nephrology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Liming Guo
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiaochun Feng
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Haoyu Zhu
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| |
Collapse
|
177
|
Hong KY, Ghafari A, Mei Y, Williams JS, Attia D, Forsyth J, Wang K, Wyeld T, Sun C, Glogauer M, King TJ. Oral inflammatory load predicts vascular function in a young adult population: a pilot study. FRONTIERS IN ORAL HEALTH 2023; 4:1233881. [PMID: 37670806 PMCID: PMC10476491 DOI: 10.3389/froh.2023.1233881] [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: 06/02/2023] [Accepted: 07/13/2023] [Indexed: 09/07/2023] Open
Abstract
Background The periodontium is a highly vascularized area of the mouth, and periodontitis initiates negative functional and structural changes in the vasculature. However, mild oral inflammation, including levels experienced by many apparently healthy individuals, has an unclear impact on cardiovascular function. The purpose of this pilot study is to investigate the effects of objectively measured whole mouth oral inflammatory load (OIL) on vascular function in apparently healthy individuals. Methods In this cross-sectional and correlational analysis, we recruited 28 young (18-30 years) and systemically healthy participants (16 male, 12 female). Using oral neutrophil counts, a validated measure for OIL, we collected participant's mouth rinse samples and quantified OIL. Blood pressure, arterial stiffness (pulse-wave velocity) and endothelial function (brachial artery flow-mediated dilation) were also measured. Results Only oral neutrophil count significantly predicted flow-mediated dilation % (p = 0.04; R2 = 0.16, β = - 1.05) and those with OIL levels associated with >2.5 × 105 neutrophil counts (n = 8) had a lower flow-mediated dilation % (6.0 ± 2.3%) than those with counts associated with gingival health with less than 2.5 × 105 neutrophil counts (10.0 ± 5.2%, p = 0.05). There were no significant predictors for arterial stiffness. Conclusion We found that OIL was a predictor of reduced flow-mediated dilation. An impairment in flow-mediated dilation is an indicator of future possible risk of cardiovascular disease-one of the leading causes of death in North America. Therefore, this study provides evidence for the importance of oral health and that OIL may impact endothelial function.
Collapse
Affiliation(s)
- Ker-Yung Hong
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Avin Ghafari
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Yixue Mei
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Jennifer S. Williams
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Dina Attia
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Jourdyn Forsyth
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Kevin Wang
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Trevor Wyeld
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
| | - Chunxiang Sun
- Faculty of Dentistry, University of Toronto, and Dental Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, and Dental Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Trevor J. King
- Department of Kinesiology, Faculty of Science, McMaster University, Hamilton, ON, Canada
- Department of Health and Physical Education, Faculty of Health, Community and Education, Mount Royal University, Calgary, AB, Canada
| |
Collapse
|
178
|
Das D, Shruthi NR, Banerjee A, Jothimani G, Duttaroy AK, Pathak S. Endothelial dysfunction, platelet hyperactivity, hypertension, and the metabolic syndrome: molecular insights and combating strategies. Front Nutr 2023; 10:1221438. [PMID: 37614749 PMCID: PMC10442661 DOI: 10.3389/fnut.2023.1221438] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023] Open
Abstract
Metabolic syndrome (MetS) is a multifaceted condition that increases the possibility of developing atherosclerotic cardiovascular disease. MetS includes obesity, hypertension, dyslipidemia, hyperglycemia, endothelial dysfunction, and platelet hyperactivity. There is a concerning rise in the occurrence and frequency of MetS globally. The rising incidence and severity of MetS need a proactive, multipronged strategy for identifying and treating those affected. For many MetS patients, achieving recommended goals for healthy fat intake, blood pressure control, and blood glucose management may require a combination of medicine therapy, lifestyles, nutraceuticals, and others. However, it is essential to note that lifestyle modification should be the first-line therapy for MetS. In addition, MetS requires pharmacological, nutraceutical, or other interventions. This review aimed to bring together the etiology, molecular mechanisms, and dietary strategies to combat hypertension, endothelial dysfunction, and platelet dysfunction in individuals with MetS.
Collapse
Affiliation(s)
- Diptimayee Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Nagainallur Ravichandran Shruthi
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Ganesan Jothimani
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Asim K. Duttaroy
- Faculty of Medicine, Department of Nutrition, Institute of Medical Sciences, University of Oslo, Oslo, Norway
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| |
Collapse
|
179
|
Masaki N, Adachi T, Tomiyama H, Kohro T, Suzuki T, Ishizu T, Ueda S, Yamazaki T, Furumoto T, Kario K, Inoue T, Koba S, Takemoto Y, Hano T, Sata M, Ishibashi Y, Node K, Maemura K, Ohya Y, Furukawa T, Ito H, Higashi Y, Yamashina A, Takase B. Reduced reactive hyperemia of the brachial artery in diabetic patients assessed by repeated measurements: The FMD-J B study. Physiol Rep 2023; 11:e15786. [PMID: 37607768 PMCID: PMC10444575 DOI: 10.14814/phy2.15786] [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: 07/05/2023] [Accepted: 07/23/2023] [Indexed: 08/24/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a major cause of microvascular dysfunction. However, its effect on blood flow patterns during ischemic demand has not been adequately elucidated. In this study, we investigated the hypothesis that microvascular dysfunction in patients with T2DM manifests as brachial reactive hyperemia (BRH), defined as the ratio of peak blood flow velocities in a brachial artery before and after forearm cuff occlusion. The study enrolled 943 subjects (men, n = 152 [T2DM] and n = 371 [non-T2DM]; women, n = 107 [T2DM] and n = 313 [non-T2DM], respectively) with no history of cardiovascular disease. Semiautomatic measurements were obtained three times at 1.5-year intervals to confirm the reproducibility of factors involved in BRH for each sex. An age-adjusted mixed model demonstrated attenuated BRH in the presence of T2DM in both men (p = 0.022) and women (p = 0.031) throughout the study period. Post hoc analysis showed that the estimated BRH was significantly attenuated in patients with T2DM regardless of sex, except at baseline in women. In multivariate regression analysis, T2DM was a negative predictor of BRH at every measurement in men. For women, BRH was more strongly associated with alcohol consumption. Repeated measurements analysis revealed that T2DM was associated with attenuated postocclusion reactive hyperemia.
Collapse
Affiliation(s)
- Nobuyuki Masaki
- Department of Intensive Care MedicineNational Defense Medical CollegeTokorozawaJapan
| | - Takeshi Adachi
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | | | - Takahide Kohro
- Department of Hospital Planning and Management, Medical InformaticsJichi Medical University School of MedicineTochigiJapan
| | - Toru Suzuki
- Cardiovascular MedicineUniversity of LeicesterLeicesterUK
| | - Tomoko Ishizu
- Cardiovascular DivisionInstitute of Clinical Medicine, University of TsukubaIbarakiJapan
| | - Shinichiro Ueda
- Department of Clinical Pharmacology and TherapeuticsUniversity of the Ryukyu School of MedicineOkinawaJapan
| | - Tsutomu Yamazaki
- Department of Clinical Epidemiology and Systems, Faculty of MedicineThe University of TokyoTokyoJapan
| | - Tomoo Furumoto
- Department of Cardiovascular MedicineHokkaido University Graduate School of MedicineSapporoJapan
| | - Kazuomi Kario
- Division of Cardiovascular MedicineJichi Medical University School of MedicineTochigiJapan
| | - Teruo Inoue
- Dokkyo Medical University; Nasu Red Cross HospitalTochigiJapan
| | - Shinji Koba
- Department of Medicine, Division of CardiologyShowa University School of MedicineTokyoJapan
| | - Yasuhiko Takemoto
- Department of Internal Medicine and CardiologyOsaka City University Graduate School of MedicineOsakaJapan
| | - Takuzo Hano
- Department of Medical Education and Population‐based Medicine, Postgraduate School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Masataka Sata
- Department of Cardiovascular MedicineInstitute of Health Biosciences, The University of Tokushima Graduate SchoolTokushimaJapan
| | - Yutaka Ishibashi
- Department of General MedicineShimane University Faculty of MedicineShimaneJapan
| | - Koichi Node
- Department of Cardiovascular MedicineSaga UniversitySagaJapan
| | - Koji Maemura
- Department of Cardiovascular Medicine, Course of Medical and Dental Sciences, Graduate School of Biomedical SciencesNagasaki UniversityNagasakiJapan
| | - Yusuke Ohya
- The Third Department of Internal MedicineUniversity of the RyukyusOkinawaJapan
| | - Taiji Furukawa
- Department of Internal MedicineTeikyo University School of MedicineTokyoJapan
| | - Hiroshi Ito
- Department of Cardiovascular MedicineOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Yukihito Higashi
- Department of Regenerative MedicineResearch Institute for Radiation Biology and Medicine, Hiroshima UniversityHiroshimaJapan
| | | | - Bonpei Takase
- Department of Intensive Care MedicineNational Defense Medical CollegeTokorozawaJapan
| |
Collapse
|
180
|
Simeunovic A, Brunborg C, Heier M, Seljeflot I, Dahl-Jørgensen K, Margeirsdottir HD. Sustained low-grade inflammation in young participants with childhood onset type 1 diabetes: The Norwegian atherosclerosis and childhood diabetes (ACD) study. Atherosclerosis 2023; 379:117151. [PMID: 37349194 DOI: 10.1016/j.atherosclerosis.2023.05.020] [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: 11/15/2022] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND AND AIMS Persons with type 1 diabetes (T1D) have increased mortality from cardiovascular disease. Early inflammation is important in the development of atherosclerosis. We aimed to evaluate the extent of inflammation and difference in mean over a five-year period in young persons with T1D compared to healthy controls. METHODS The Norwegian Atherosclerosis and Childhood Diabetes (ACD) study is a prospective population-based cohort study on atherosclerosis development in childhood-onset T1D compared to healthy controls, with follow-ups every fifth year. The original study cohort consisted of 314 children with T1D on intensive insulin treatment and 120 healthy controls of similar age. Circulating levels of VCAM-1, TNA-α, P-selectin, E-selectin, CRP, IL-6, IL-18, MCP-1, MMP-9 and TIMP-1 were measured by ELISAs at baseline and at the five-year follow-up. RESULTS The group with T1D had mean age 13.7 (SD = 2.8) years, disease duration 5.6 (SD = 3.4) years and HbA1c 68 (SD = 13.1) mmol/mol at baseline. Levels of almost all inflammatory markers were significantly increased in the group with T1D compared to controls, and significant mean-difference between the two groups over the five-year period was observed in four markers: IL-18, P-selectin, E-selectin and TIMP-1. CONCLUSIONS The early low-grade inflammation present in young individuals with T1D five years after diagnosis is sustained at ten-year disease duration, with moderate changes for most markers of inflammation over time. The evolving inflammatory profile indicates an accelerated chain of events in the progression of early atheromatosis in T1D.
Collapse
Affiliation(s)
- Aida Simeunovic
- Department of Paediatric and Adolescent Medicine, Akershus University Hospital, Lørenskog, Norway; Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway.
| | - Cathrine Brunborg
- Centre for Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital, Oslo, Norway
| | - Martin Heier
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway
| | - Ingebjørg Seljeflot
- University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway; Center for Clinical Heart Research and Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Knut Dahl-Jørgensen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway
| | - Hanna Dis Margeirsdottir
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; Oslo Diabetes Research Centre, Oslo, Norway
| |
Collapse
|
181
|
Liu W, Huang J, He S, Du R, Shi W, Wang Y, Du D, Du Y, Liu Q, Wang Y, Wang G, Yin T. Senescent endothelial cells' response to the degradation of bioresorbable scaffold induces intimal dysfunction accelerating in-stent restenosis. Acta Biomater 2023; 166:266-277. [PMID: 37211308 DOI: 10.1016/j.actbio.2023.05.028] [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: 01/17/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Atherosclerotic cardiovascular disease is a typical age-related disease accompanied by stiffening arteries. We aimed to elucidate the influence of aged arteries on in-stent restenosis (ISR) after the implantation of bioresorbable scaffolds (BRS). Histology and optical coherence tomography showed increased lumen loss and ISR in the aged abdominal aorta of Sprague-Dawley rats, with apparent scaffold degradation and deformation, which induce lower wall shear stress (WSS). This was also the case at the distal end of BRS, where the scaffolds degraded faster, and significant lumen loss was followed by a lower WSS. In addition, early thrombosis, inflammation, and delayed re-endothelialization were presented in the aged arteries. Degradation of BRS causes more senescent cells in the aged vasculature, increasing endothelial cell dysfunction and the risk of ISR. Thus, profoundly understanding the mechanism between BRS and senescent cells may give a meaningful guide for the age-related scaffold design. STATEMENT OF SIGNIFICANCE: The degradation of bioresorbable scaffolds aggravates senescent endothelial cells and a much lower wall shear stress areas in the aged vasculature, lead to intimal dysfunction and increasing in-stent restenosis risk. Early thrombosis and inflammation, as well as delayed re-endothelialization, are presented in the aged vasculature after bioresorbable scaffolds implantation. Age stratification during the clinical evaluation and senolytics in the design of new bioresorbable scaffolds should be considered, especially for old patients.
Collapse
Affiliation(s)
- Wanling Liu
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Modern Life Science Experiment Teaching Center, Bioengineering College of Chongqing University, Chongqing 400030, PR China
| | - Junyang Huang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Modern Life Science Experiment Teaching Center, Bioengineering College of Chongqing University, Chongqing 400030, PR China
| | - Shicheng He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ruolin Du
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Modern Life Science Experiment Teaching Center, Bioengineering College of Chongqing University, Chongqing 400030, PR China
| | - Wen Shi
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Modern Life Science Experiment Teaching Center, Bioengineering College of Chongqing University, Chongqing 400030, PR China
| | - Yang Wang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Modern Life Science Experiment Teaching Center, Bioengineering College of Chongqing University, Chongqing 400030, PR China
| | - Dingyuan Du
- Department of Traumatology, and Department of Cardiothoracic Surgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, China
| | - Yan Du
- Ultrasonography Department, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, China
| | - Qing Liu
- Beijing Advanced Medical Technologies Inc., Beijing 102609, China
| | - Yazhou Wang
- School of Medicine, Chongqing University, Chongqing 400044, PR China.
| | - Guixue Wang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Modern Life Science Experiment Teaching Center, Bioengineering College of Chongqing University, Chongqing 400030, PR China.
| | - Tieying Yin
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Modern Life Science Experiment Teaching Center, Bioengineering College of Chongqing University, Chongqing 400030, PR China.
| |
Collapse
|
182
|
Radwan B, Prabhakaran A, Rocchetti S, Matuszyk E, Keyes TE, Baranska M. Uptake and anti-inflammatory effects of liposomal astaxanthin on endothelial cells tracked by Raman and fluorescence imaging. Mikrochim Acta 2023; 190:332. [PMID: 37500736 PMCID: PMC10374751 DOI: 10.1007/s00604-023-05888-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023]
Abstract
Astaxanthin (AXT) is a lipophilic antioxidant and anti-inflammatory natural pigment whose cellular uptake and bioavailability could be improved via liposomal encapsulation. Endothelial cells (EC) line the lumen of all blood vessels and are tasked with multiple roles toward maintaining cardiovascular homeostasis. Endothelial dysfunction is linked to the development of many diseases and is closely interconnected with oxidative stress and vascular inflammation. The uptake of free and liposomal AXT into EC was investigated using Raman and fluorescence microscopies. AXT was either encapsulated in neutral or cationic liposomes. Enhanced uptake and anti-inflammatory effects of liposomal AXT were observed. The anti-inflammatory effects of liposomal AXT were especially prominent in reducing EC lipid unsaturation, lowering numbers of lipid droplets (LDs), and decreasing intercellular adhesion molecule 1 (ICAM-1) overexpression, which is considered a well-known marker for endothelial inflammation. These findings highlight the benefits of AXT liposomal encapsulation on EC and the applicability of Raman imaging to investigate such effects.
Collapse
Affiliation(s)
- Basseem Radwan
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Krakow, Poland
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387, Krakow, Poland
| | - Amrutha Prabhakaran
- School of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland
| | - Stefano Rocchetti
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Krakow, Poland
| | - Ewelina Matuszyk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Krakow, Poland
| | - Tia E Keyes
- School of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland
| | - Malgorzata Baranska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348, Krakow, Poland.
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387, Krakow, Poland.
| |
Collapse
|
183
|
Navodnik MP, Janež A, Žuran I. The Effect of Additional Treatment with Empagliflozin or Semaglutide on Endothelial Function and Arterial Stiffness in Subjects with Type 1 Diabetes Mellitus-ENDIS Study. Pharmaceutics 2023; 15:1945. [PMID: 37514131 PMCID: PMC10385568 DOI: 10.3390/pharmaceutics15071945] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
We investigated the effect of additional treatment with newer antidiabetic drugs on endothelium function and arterial stiffness in subjects with type 1 diabetes mellitus (T1DM) without cardiovascular diseases. A total of 89 participants, all users of CGMS (continuous monitoring glucose system), were randomized into three comparable groups, receiving empagliflozin (E; n = 30), receiving semaglutide (S; n = 30), and a control group (C; n = 29). At baseline and 12 weeks post treatment, we measured FMD (brachial artery flow-mediated dilation) and FBF (forearm blood flow as reactive hyperemia assessed with strain gauge plethysmography) as parameters of endothelial function, as well as pulse wave velocity (PWV) and peripheral resistance as parameters of arterial stiffness. Improvement in FMD was significant in both intervention groups compared to controls (E group 2.0-fold, p = 0.000 and S group 1.9-fold, p = 0.000), with no changes between those two groups (p = 0.745). During the evaluation of FBF, there were statistically insignificant improvements in both therapeutic groups compared to controls (E group 1.39-fold, p = 0.074 and S group 1.22-fold, p = 0.701). In arterial stiffness parameters, improvements were seen only in the semaglutide group, with a decline in peripheral resistance by 5.1% (p = 0.046). We can conclude that, for arterial stiffness, semaglutide seems better, but both drugs positively impact endothelial function and, thus, could also have a protective role in T1DM.
Collapse
Affiliation(s)
- Maja Preložnik Navodnik
- Department of Angiology, Endocrinology and Rheumatology, General Hospital Celje, Oblakova ul. 5, 3000 Celje, Slovenia
| | - Andrej Janež
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia
| | - Ivan Žuran
- Department of Angiology, Endocrinology and Rheumatology, General Hospital Celje, Oblakova ul. 5, 3000 Celje, Slovenia
| |
Collapse
|
184
|
Inoue Y, Shue F, Bu G, Kanekiyo T. Pathophysiology and probable etiology of cerebral small vessel disease in vascular dementia and Alzheimer's disease. Mol Neurodegener 2023; 18:46. [PMID: 37434208 PMCID: PMC10334598 DOI: 10.1186/s13024-023-00640-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is commonly caused by vascular injuries in cerebral large and small vessels and is a key driver of age-related cognitive decline. Severe VCID includes post-stroke dementia, subcortical ischemic vascular dementia, multi-infarct dementia, and mixed dementia. While VCID is acknowledged as the second most common form of dementia after Alzheimer's disease (AD) accounting for 20% of dementia cases, VCID and AD frequently coexist. In VCID, cerebral small vessel disease (cSVD) often affects arterioles, capillaries, and venules, where arteriolosclerosis and cerebral amyloid angiopathy (CAA) are major pathologies. White matter hyperintensities, recent small subcortical infarcts, lacunes of presumed vascular origin, enlarged perivascular space, microbleeds, and brain atrophy are neuroimaging hallmarks of cSVD. The current primary approach to cSVD treatment is to control vascular risk factors such as hypertension, dyslipidemia, diabetes, and smoking. However, causal therapeutic strategies have not been established partly due to the heterogeneous pathogenesis of cSVD. In this review, we summarize the pathophysiology of cSVD and discuss the probable etiological pathways by focusing on hypoperfusion/hypoxia, blood-brain barriers (BBB) dysregulation, brain fluid drainage disturbances, and vascular inflammation to define potential diagnostic and therapeutic targets for cSVD.
Collapse
Affiliation(s)
- Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Guojun Bu
- SciNeuro Pharmaceuticals, Rockville, MD 20850 USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| |
Collapse
|
185
|
Zhang K, Chen R, Cai Z, Hou L, Li X, Xu X, Sun Y, Lu X, Jiang Q. The effect of ozone short-term exposure on flow-mediated dilation: Using data before and after COVID-19 lockdown in Shanghai. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 881:163485. [PMID: 37068686 PMCID: PMC10105378 DOI: 10.1016/j.scitotenv.2023.163485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/02/2023] [Accepted: 04/09/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Short-term ambient ozone exposure has been shown to have an adverse impact on endothelial function, contributing to major cardiovascular diseases and premature death. However, only limited studies have focused on the impact of short-term ozone exposure on Flow-mediated Dilation (FMD), and their results have been inconsistent. The current study aims to explore the relationship between short-term ambient ozone exposure and FMD. In addition, the study aims to investigate how lockdown measures for COVID-19 may influence ozone concentration in the atmosphere. METHODS Participants were recruited from a hospital in Shanghai from December 2020 to August 2022. Individuals' ozone exposure was determined using residential addresses. A distributed lag nonlinear model was adopted to assess the exposure-response relationship between short-term ozone exposure and FMD. A comparison was made between ambient ozone concentration and FMD data collected before and after Shanghai's lockdown in 2022. RESULTS When ozone concentration was between 150 and 200 μg/m3, there was a significant reduction in FMD with a 2-day lag. Elderly individuals (age ≥ 65), females, non-drinkers, and non-smokers were found to be more susceptible to high concentrations of ozone exposure. The lockdown did elevate ambient ozone concentration compared to the same period previously. INTERPRETATION This study proposes that an ambient ozone concentration of 150-200 μg/m3 is harmful to endothelial function, and that a reduction in human activity during lockdown increased the concentration, which in turn reduced FMD. However, the underlying mechanism requires further research.
Collapse
Affiliation(s)
- Kai Zhang
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Rukun Chen
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China; Faculty of Medicine, University of Southampton, United Kingdom of Great Britain and Northern Ireland
| | - Zhenzhen Cai
- Shanghai Fourth People's Hospital, School of Medicine, Tongji University, China
| | - Lei Hou
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xiaoguang Li
- Shanghai Fourth People's Hospital, School of Medicine, Tongji University, China
| | - Xin Xu
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yishuai Sun
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xiaotong Lu
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Qixia Jiang
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China.
| |
Collapse
|
186
|
Martin ZT, Akins JD, Merlau ER, Kolade JO, Al-Daas IO, Cardenas N, Vu JK, Brown KK, Brothers RM. The acute effect of whole-body heat therapy on peripheral and cerebral vascular reactivity in Black and White females. Microvasc Res 2023; 148:104536. [PMID: 37024072 PMCID: PMC10908357 DOI: 10.1016/j.mvr.2023.104536] [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: 01/04/2023] [Revised: 03/17/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Among females in the U.S., Black females suffer the most from cardiovascular disease and stroke. While the reasons for this disparity are multifactorial, vascular dysfunction likely contributes. Chronic whole-body heat therapy (WBHT) improves vascular function, but few studies have examined its acute effect on peripheral or cerebral vascular function, which may help elucidate chronic adaptative mechanisms. Furthermore, no studies have investigated this effect in Black females. We hypothesized that Black females would have lower peripheral and cerebral vascular function relative to White females and that one session of WBHT would mitigate these differences. Eighteen young, healthy Black (n = 9; 21 ± 3 yr; BMI: 24.7 ± 4.5 kg/m2) and White (n = 9; 27 ± 3 yr; BMI: 24.8 ± 4.1 kg/m2) females underwent one 60 min session of WBHT (49 °C water via a tube-lined suit). Pre- and 45 min post-testing measures included post-occlusive forearm reactive hyperemia (peripheral microvascular function, RH), brachial artery flow-mediated dilation (peripheral macrovascular function, FMD), and cerebrovascular reactivity (CVR) to hypercapnia. Prior to WBHT, there were no differences in RH, FMD, or CVR (p > 0.05 for all). WBHT improved peak RH in both groups (main effect of WBHT: 79.6 ± 20.1 cm/s to 95.9 ± 30.0 cm/s; p = 0.004, g = 0.787) but not Δ blood velocity (p > 0.05 for both groups). WBHT improved FMD in both groups (6.2 ± 3.4 % to 8.8 ± 3.7 %; p = 0.016, g = 0.618) but had no effect on CVR in either group (p = 0.077). These data indicate that one session of WBHT acutely improves peripheral micro- and macrovascular but not cerebral vascular function in Black and White females.
Collapse
Affiliation(s)
- Zachary T Martin
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX, USA
| | - John D Akins
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX, USA
| | - Emily R Merlau
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX, USA
| | - John O Kolade
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX, USA
| | - Iman O Al-Daas
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX, USA
| | - Natalia Cardenas
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX, USA
| | - Joshua K Vu
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX, USA
| | - Kyrah K Brown
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX, USA
| | - R Matthew Brothers
- Department of Kinesiology, The University of Texas at Arlington, Arlington, TX, USA.
| |
Collapse
|
187
|
Wabel E, Orr A, Flood ED, Thompson JM, Xie H, Demireva EY, Abolibdeh B, Honke Hulbert D, Mullick AE, Garver H, Fink GD, Kung TA, Watts SW. Chemerin is resident to vascular tunicas and contributes to vascular tone. Am J Physiol Heart Circ Physiol 2023; 325:H172-H186. [PMID: 37294893 PMCID: PMC11467446 DOI: 10.1152/ajpheart.00239.2023] [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: 04/25/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/11/2023]
Abstract
The adipokine chemerin may support blood pressure, evidenced by a fall in mean arterial pressure after whole body antisense oligonucleotide (ASO)-mediated knockdown of chemerin protein in rat models of normal and elevated blood pressure. Although the liver is the greatest contributor of circulating chemerin, liver-specific ASOs that abolished hepatic-derived chemerin did not change blood pressure. Thus, other sites must produce the chemerin that supports blood pressure. We hypothesize that the vasculature is a source of chemerin independent of the liver that supports arterial tone. RNAScope, PCR, Western blot analyses, ASOs, isometric contractility, and radiotelemetry were used in the Dahl salt-sensitive (SS) rat (male and female) on a normal diet. Retinoic acid receptor responder 2 (Rarres2) mRNA was detected in the smooth muscle, adventitia, and perivascular adipose tissue of the thoracic aorta. Chemerin protein was detected immunohistochemically in the endothelium, smooth muscle cells, adventitia, and perivascular adipose tissue. Chemerin colocalized with the vascular smooth muscle marker α-actin and the adipocyte marker perilipin. Importantly, chemerin protein in the thoracic aorta was not reduced when liver-derived chemerin was abolished by a liver-specific ASO against chemerin. Chemerin protein was similarly absent in arteries from a newly created global chemerin knockout in Dahl SS rats. Inhibition of the receptor Chemerin1 by the receptor antagonist CCX832 resulted in the loss of vascular tone that supports potential contributions of chemerin by both perivascular adipose tissue and the media. These data suggest that vessel-derived chemerin may support vascular tone locally through constitutive activation of Chemerin1. This posits chemerin as a potential therapeutic target in blood pressure regulation.NEW & NOTEWORTHY Vascular tunicas synthesizing chemerin is a new finding. Vascular chemerin is independent of hepatic-derived chemerin. Vasculature from both males and females have resident chemerin. Chemerin1 receptor activity supports vascular tone.
Collapse
Affiliation(s)
- Emma Wabel
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Alexis Orr
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Emma D Flood
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Janice M Thompson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Huirong Xie
- Transgenic and Genome Editing Facility, Research Technology Support Facility, and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, United States
| | - Elena Y Demireva
- Transgenic and Genome Editing Facility, Research Technology Support Facility, and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, United States
| | - Bana Abolibdeh
- Transgenic and Genome Editing Facility, Research Technology Support Facility, and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, United States
| | - Darcy Honke Hulbert
- Cardiovascular Division, Campus Animal Resources, Michigan State University, East Lansing, Michigan, United States
| | - Adam E Mullick
- Ionis Pharmaceuticals, Carlsbad, California, United States
| | - Hannah Garver
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Gregory D Fink
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Theodore A Kung
- Section of Plastic and Reconstructive Surgery, Department of Surgery, Michigan Medicine, Ann Arbor, Michigan, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| |
Collapse
|
188
|
Baba MS, Laway BA, Misgar RA, Wani AI, Bashir MI, Bhat IA, Haq MG, Shah ZA. Metabolic Abnormalities, Inflammatory Markers and Endothelial Dysfunction in Hyperprolactinemia due to Prolactinoma before and after Normalization of Serum Prolactin: A Prospective Case Control Study. Indian J Endocrinol Metab 2023; 27:357-364. [PMID: 37867992 PMCID: PMC10586551 DOI: 10.4103/ijem.ijem_201_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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 10/20/2022] [Accepted: 01/06/2023] [Indexed: 10/24/2023] Open
Abstract
Background Hyperprolactinemia is associated with obesity, dyslipidemia, insulin resistance, and low-grade inflammation which may promote endothelial dysfunction (EnD). Limited work has been done on EnD in prolactinomas and we, therefore, studied serum markers of inflammation and EnD in patients with prolactinomas before and after treatment with dopamine agonists. Methodology Fifty-six treatment naïve patients with prolactinomas and fifty-three (apparently healthy age and sex-matched) controls were enrolled in the study and subjected to clinical assessment and laboratory investigations including blood glucose, total cholesterol, triglycerides, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, urea, creatinine, uric acid, erythrocyte sedimentation rate (ESR), highly sensitive C-reactive protein (hsCRP) and markers of EnD i.e., intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). Patients were treated with a dopamine agonist (cabergoline) and parameters (like ESR, hsCRP, ICAM-1, and VCAM-1) were measured at 12 weeks. Results The majority of the patients (84%) were female, more than half (52%) had metabolic syndrome and over a third (36%) were obese. Blood glucose fasting, HbA1c, lipid fractions, ESR, hsCRP, ICAM-1, and VCAM-1 were significantly higher in patients than in controls. Median ICAM-1 was 1331.95 ng/ml (IQR 803.43-1825.99) in patients vs 753.04 ng/ml (IQR 402.04-871.55) in controls, P < 0.001 and median VCAM-1in patients was 971.35 ng/ml (IQR 695.03-1285.23) as against 634.56 ng/ml (IQR 177.49-946.50) in controls, p0.001. Serum ICAM-1 and VCAM-1 correlated positively with hsCRP. On multivariate regression analysis, serum hsCRP was the only significant predictor of change in ICAM-1 and VCAM-1. Normalization of serum PRL with CAB resulted in a significant decrease in metabolic parameters, ESR, hsCRP, ICAM-1, and VCAM-1. Conclusion Hyperprolactinemia because of prolactinoma is associated with EnD secondary to systemic inflammation and metabolic abnormalities which improve after treatment with DA.
Collapse
Affiliation(s)
- Mohammad Salem Baba
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Bashir Ahmad Laway
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Raiz Ahmad Misgar
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Arshad Iqbal Wani
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Mir Iftikhar Bashir
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Imtiyaz Ahmad Bhat
- Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Malik Gawharul Haq
- Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Zafar Amin Shah
- Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
189
|
Perks J, Zaccardi F, Rayt H, Sayers R, Brady EM, Davies MJ, Rowlands AV, Edwardson CL, Hall A, Yates T, Henson J. Device-measured physical activity behaviours, and physical function, in people with type 2 diabetes mellitus and peripheral artery disease: A cross-sectional study. Exp Gerontol 2023; 178:112207. [PMID: 37196824 DOI: 10.1016/j.exger.2023.112207] [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: 03/28/2023] [Revised: 05/11/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
AIM To quantify differences in device-measured physical activity (PA) behaviours, and physical function (PF), in people with type 2 diabetes mellitus (T2DM) with and without peripheral artery disease (PAD). MATERIALS AND METHODS Participants from the Chronotype of Patients with T2DM and Effect on Glycaemic Control cross-sectional study wore accelerometers on their non-dominant wrist for up to 8-days to quantify: volume and intensity distribution of PA, time spent inactive, time in light PA, moderate-to-vigorous PA in at least 1-minute bouts (MVPA1min), and the average intensity achieved during the most active continuous 2, 5, 10, 30, and 60-minute periods of the 24-h day. PF was assessed using the short physical performance battery (SPPB), the Duke Activity Status Index (DASI), sit-to-stand repetitions in 60 s (STS-60); hand-grip strength was also assessed. Differences between subjects with and without PAD were estimated using regressions adjusted for possible confounders. RESULTS 736 participants with T2DM (without diabetic foot ulcers) were included in the analysis, 689 had no PAD. People with T2DM and PAD undertake less PA (MVPA1min: -9.2 min [95 % CI: -15.3 to -3.0; p = 0.004]) (light intensity PA: -18.7 min [-36.4 to -1.0; p = 0.039]), spend more time inactive (49.2 min [12.1 to 86.2; p = 0.009]), and have reduced PF (SPPB score: -1.6 [-2.5 to -0.8; p = 0.001]) (DASI score: -14.8 [-19.8 to -9.8; p = 0.001]) (STS-60 repetitions: -7.1 [-10.5 to -3.8; p = 0.001]) compared to people without; some differences in PA were attenuated by confounders. Reduced intensity of activity for the most active continuous 2-30 min in the 24-h day, and reduced PF, persisted after accounting for confounders. There were no significant differences in hand-grip strength. CONCLUSIONS Findings from this cross-sectional study suggest that, the presence of PAD in T2DM may have been associated with lower PA levels and PF.
Collapse
Affiliation(s)
- Jemma Perks
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK.
| | - Francesco Zaccardi
- Diabetes Research Centre, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Harjeet Rayt
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Robert Sayers
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Emer M Brady
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Melanie J Davies
- Diabetes Research Centre, University Hospitals of Leicester NHS Trust, Leicester, UK; NIHR Leicester Biomedical Research Centre and Diabetes Research Centre, College of Life Sciences, University of Leicester, Leicester, UK
| | - Alex V Rowlands
- Diabetes Research Centre, University Hospitals of Leicester NHS Trust, Leicester, UK; NIHR Leicester Biomedical Research Centre and Diabetes Research Centre, College of Life Sciences, University of Leicester, Leicester, UK
| | - Charlotte L Edwardson
- Diabetes Research Centre, University Hospitals of Leicester NHS Trust, Leicester, UK; NIHR Leicester Biomedical Research Centre and Diabetes Research Centre, College of Life Sciences, University of Leicester, Leicester, UK
| | - Andrew Hall
- The Hanning Sleep Laboratory, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Thomas Yates
- Diabetes Research Centre, University Hospitals of Leicester NHS Trust, Leicester, UK; NIHR Leicester Biomedical Research Centre and Diabetes Research Centre, College of Life Sciences, University of Leicester, Leicester, UK
| | - Joseph Henson
- Diabetes Research Centre, University Hospitals of Leicester NHS Trust, Leicester, UK; NIHR Leicester Biomedical Research Centre and Diabetes Research Centre, College of Life Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
190
|
Ristovska EC, Genadieva-Dimitrova M, Todorovska B, Milivojevic V, Rankovic I, Samardziski I, Bojadzioska M. The Role of Endothelial Dysfunction in the Pathogenesis of Pregnancy-Related Pathological Conditions: A Review. Pril (Makedon Akad Nauk Umet Odd Med Nauki) 2023; 44:113-137. [PMID: 37453122 DOI: 10.2478/prilozi-2023-0032] [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] [Indexed: 07/18/2023]
Abstract
In the recent decades, endothelial dysfunction (ED) has been recognized as a significant contributing factor in the pathogenesis of many pathological conditions. In interaction with atherosclerosis, hypercholesterolemia, and hypertension, ED plays a crucial role in the pathogenesis of coronary artery disease, chronic renal disease, and microvascular complications in diabetes mellitus. Although ED plays a significant role in the pathogenesis of several pregnancy-related disorders such as preeclampsia, HELLP syndrome, fetal growth restriction, and gestational diabetes mellitus, the exact pathogenetic mechanisms are still a matter of debate. The increased prevalence of these entities in patients with preexisting vascular diseases highlights the essential pathological role of the preexisting ED in these patients. The abnormal uteroplacental circulation and the release of soluble factors from the ischemic placenta into the maternal bloodstream are the main causes of the maternal ED underlying the characteristic preeclamptic phenotype. Besides the increased risk for maternal and fetal poor outcomes, the preexisting ED also increases the risk of development of future cardiovascular diseases in these patients. This study aimed to look deeper into the role of ED in the pathogenesis of several pregnancy-related hypertensive and liver diseases. Hopefully, it could contribute to improvement of the awareness, knowledge, and management of these conditions and also to the reduction of the adverse outcomes and additional long-term cardiovascular complications.
Collapse
Affiliation(s)
- Elena Curakova Ristovska
- 1University Clinic for Gastroenterohepatology, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Skopje, RN Macedonia
| | - Magdalena Genadieva-Dimitrova
- 1University Clinic for Gastroenterohepatology, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Skopje, RN Macedonia
| | - Beti Todorovska
- 1University Clinic for Gastroenterohepatology, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Skopje, RN Macedonia
| | - Vladimir Milivojevic
- 2Section for Internal Medicine, Medcompass Alliance, School of Medicine, Belgrade University, Belgrade, Serbia
| | - Ivan Rankovic
- 3Section for Internal Medicine, Medcompass Alliance, Belgrade, Serbia
| | - Igor Samardziski
- 4University Clinic for Gynecology and Obstetrics, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Skopje, RN Macedonia
| | - Maja Bojadzioska
- 5University Clinic for Rheumatology, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Skopje, RN Macedonia
| |
Collapse
|
191
|
Lee WE, Genetzakis E, Figtree GA. Novel Strategies in the Early Detection and Treatment of Endothelial Cell-Specific Mitochondrial Dysfunction in Coronary Artery Disease. Antioxidants (Basel) 2023; 12:1359. [PMID: 37507899 PMCID: PMC10376062 DOI: 10.3390/antiox12071359] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Although elevated cholesterol and other recognised cardiovascular risk factors are important in the development of coronary artery disease (CAD) and heart attack, the susceptibility of humans to this fatal process is distinct from other animals. Mitochondrial dysfunction of cells in the arterial wall, particularly the endothelium, has been strongly implicated in the pathogenesis of CAD. In this manuscript, we review the established evidence and mechanisms in detail and explore the potential opportunities arising from analysing mitochondrial function in patient-derived cells such as endothelial colony-forming cells easily cultured from venous blood. We discuss how emerging technology and knowledge may allow us to measure mitochondrial dysfunction as a potential biomarker for diagnosis and risk management. We also discuss the "pros and cons" of animal models of atherosclerosis, and how patient-derived cell models may provide opportunities to develop novel therapies relevant for humans. Finally, we review several targets that potentially alleviate mitochondrial dysfunction working both via direct and indirect mechanisms and evaluate the effect of several classes of compounds in the cardiovascular context.
Collapse
Affiliation(s)
- Weiqian E. Lee
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Elijah Genetzakis
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Gemma A. Figtree
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW 2065, Australia
| |
Collapse
|
192
|
Dubsky M, Veleba J, Sojakova D, Marhefkova N, Fejfarova V, Jude EB. Endothelial Dysfunction in Diabetes Mellitus: New Insights. Int J Mol Sci 2023; 24:10705. [PMID: 37445881 DOI: 10.3390/ijms241310705] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Endothelial dysfunction (ED) is an important marker of future atherosclerosis and cardiovascular disease, especially in people with diabetes. This article summarizes the evidence on endothelial dysfunction in people with diabetes and adds different perspectives that can affect the presence and severity of ED and its consequences. We highlight that data on ED in type 1 diabetes are lacking and discuss the relationship between ED and arterial stiffness. Several interesting studies have been published showing that ED modulates microRNA, microvesicles, lipid levels, and the endoplasmatic reticulum. A better understanding of ED could provide important insights into the microvascular complications of diabetes, their treatment, and even their prevention.
Collapse
Affiliation(s)
- Michal Dubsky
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic
- First Faculty of Medicine, Charles University, 14021 Prague, Czech Republic
| | - Jiri Veleba
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic
| | - Dominika Sojakova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic
- First Faculty of Medicine, Charles University, 14021 Prague, Czech Republic
| | - Natalia Marhefkova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic
- First Faculty of Medicine, Charles University, 14021 Prague, Czech Republic
| | - Vladimira Fejfarova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic
| | - Edward B Jude
- Diabetes Center, Tameside and Glossop Integrated Care NHS Foundation Trust, Ashton-under-Lyne OL6 9RW, UK
- Department of Endocrinology and Gastroenterology, University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
193
|
Li Z, Liu J, Ballard K, Liang C, Wang C. Low-dose albumin-coated gold nanorods induce intercellular gaps on vascular endothelium by causing the contraction of cytoskeletal actin. J Colloid Interface Sci 2023; 649:844-854. [PMID: 37390532 DOI: 10.1016/j.jcis.2023.06.154] [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: 03/17/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023]
Abstract
Cytotoxicity of nanoparticles, typically evaluated by biochemical-based assays, often overlook the cellular biophysical properties such as cell morphology and cytoskeletal actin, which could serve as more sensitive indicators for cytotoxicity. Here, we demonstrate that low-dose albumin-coated gold nanorods (HSA@AuNRs), although being considered noncytotoxic in multiple biochemical assays, can induce intercellular gaps and enhance the paracellular permeability between human aortic endothelial cells (HAECs). The formation of intercellular gaps can be attributed to the changed cell morphology and cytoskeletal actin structures, as validated at the monolayer and single cell levels using fluorescence staining, atomic force microscopy, and super-resolution imaging. Molecular mechanistic study shows the caveolae-mediated endocytosis of HSA@AuNRs induces the calcium influx and activates actomyosin contraction in HAECs. Considering the important roles of endothelial integrity/dysfunction in various physiological/pathological conditions, this work suggests a potential adverse effect of albumin-coated gold nanorods on the cardiovascular system. On the other hand, this work also offers a feasible way to modulate the endothelial permeability, thus promoting drug and nanoparticle delivery across the endothelium.
Collapse
Affiliation(s)
- Zhengqiang Li
- Nanoscience and Biomedical Engineering, South Dakota School of Mines and Technology, 501 E St Joseph Street, Rapid City, SD 57701, USA; BioSystems Networks & Translational Research (BioSNTR), 501 E St Joseph Street, Rapid City, SD 57701, USA
| | - Jinyuan Liu
- Nanoscience and Biomedical Engineering, South Dakota School of Mines and Technology, 501 E St Joseph Street, Rapid City, SD 57701, USA; BioSystems Networks & Translational Research (BioSNTR), 501 E St Joseph Street, Rapid City, SD 57701, USA
| | - Katherine Ballard
- Nanoscience and Biomedical Engineering, South Dakota School of Mines and Technology, 501 E St Joseph Street, Rapid City, SD 57701, USA; BioSystems Networks & Translational Research (BioSNTR), 501 E St Joseph Street, Rapid City, SD 57701, USA
| | - Chao Liang
- Department of Anesthesiology, Zhongshan Hospital (Xiamen) Fudan University, Xiamen 361015, China; Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Congzhou Wang
- Nanoscience and Biomedical Engineering, South Dakota School of Mines and Technology, 501 E St Joseph Street, Rapid City, SD 57701, USA; BioSystems Networks & Translational Research (BioSNTR), 501 E St Joseph Street, Rapid City, SD 57701, USA.
| |
Collapse
|
194
|
Fico BG, Maharaj A, Pena GS, Huang CJ. The Effects of Obesity on the Inflammatory, Cardiovascular, and Neurobiological Responses to Exercise in Older Adults. BIOLOGY 2023; 12:865. [PMID: 37372149 DOI: 10.3390/biology12060865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023]
Abstract
Obesity with advancing age leads to increased health complications that are involved in various complex physiological processes. For example, inflammation is a critical cardiovascular disease risk factor that plays a role in the stages of atherosclerosis in both aging and obesity. Obesity can also induce profound changes to the neural circuitry that regulates food intake and energy homeostasis with advancing age. Here we discuss how obesity in older adults impacts inflammatory, cardiovascular, and neurobiological functions with an emphasis on how exercise mediates each topic. Although obesity is a reversible disorder through lifestyle changes, it is important to note that early interventions are crucial to prevent pathological changes seen in the aging obese population. Lifestyle modifications such as physical activity (including aerobic and resistance training) should be considered as a main intervention to minimize the synergistic effect of obesity on age-related conditions, such as cerebrovascular disease.
Collapse
Affiliation(s)
- Brandon G Fico
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Arun Maharaj
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Gabriel S Pena
- Department of Kinesiology, University of Maryland, College Park, MD 20742, USA
| | - Chun-Jung Huang
- Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
195
|
Yang Z, Liu Y, Li Z, Feng S, Lin S, Ge Z, Fan Y, Wang Y, Wang X, Mao J. Coronary microvascular dysfunction and cardiovascular disease: Pathogenesis, associations and treatment strategies. Biomed Pharmacother 2023; 164:115011. [PMID: 37321056 DOI: 10.1016/j.biopha.2023.115011] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023] Open
Abstract
Coronary microvascular dysfunction (CMD) is a high-risk factor for a variety of cardiovascular events. Due to its complex aetiology and concealability, knowledge of the pathophysiological mechanism of CMD is still limited at present, which greatly restricts its clinical diagnosis and treatment. Studies have shown that CMD is closely related to a variety of cardiovascular diseases, can aggravate the occurrence and development of cardiovascular diseases, and is closely related to a poor prognosis in patients with cardiovascular diseases. Improving coronary microvascular remodelling and increasing myocardial perfusion might be promising strategies for the treatment of cardiovascular diseases. In this paper, the pathogenesis and functional assessment of CMD are reviewed first, along with the relationship of CMD with cardiovascular diseases. Then, the latest strategies for the treatment of CMD and cardiovascular diseases are summarized. Finally, urgent scientific problems in CMD and cardiovascular diseases are highlighted and future research directions are proposed to provide prospective insights for the prevention and treatment of CMD and cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Yangxi Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Zhenzhen Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Shaoling Feng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Shanshan Lin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Zhao Ge
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Yujian Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Yi Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
196
|
Faconti L, Farukh B, McNally RJ, Brett S, Chowienczyk PJ. Impaired β 2 -adrenergic endothelium-dependent vasodilation in patients previously hospitalized with coronavirus disease 2019. J Hypertens 2023; 41:951-957. [PMID: 37016904 DOI: 10.1097/hjh.0000000000003420] [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] [Indexed: 04/06/2023]
Abstract
AIM The pulse wave response to salbutamol (PWRS) - change in augmentation index (AIx) - provides a means to assess endothelial vasodilator function in vivo . Endothelial dysfunction plays a relevant role in the pathogenesis of hypertension and cardiovascular disease and appears to underlie many of the complications of coronavirus disease 2019 (COVID-19). However, to what degree this persists after recovery is unknown. METHODS Individuals previously hospitalized with COVID-19, those recovered from mild symptoms and seronegative controls with well known risk factors for endothelial dysfunction were studied. To assess the involvement of nitric oxide-cyclic guanosine monophosphate pathway (NO-cGMP) on PWRS, sildenafil was also administrated in a subsample. RESULTS One hundred and one participants (60 men) aged 47.8 ± 14.1 (mean ± SD) years of whom 33 were previously hospitalized with COVID-19 were recruited. Salbutamol had minimal effect on haemodynamics including blood pressure and heart rate. It reduced AIx in controls ( n = 34) and those recovered from mild symptoms of COVID-19 ( n = 34) but produced an increase in AIx in those previously hospitalized: mean change [95% confidence interval] -2.85 [-5.52, -0.188] %, -2.32 [-5.17,0.54] %, and 3.03 [0.06, 6.00] % for controls, those recovered from mild symptoms and those previously hospitalized, respectively ( P = 0.001). In a sub-sample ( n = 22), sildenafil enhanced PWRS (change in AIx 0.05 [-2.15,2.24] vs. -3.96 [-7.01. -2.18], P = 0.006) with no significant difference between hospitalized ( n = 12) and nonhospitalized participants ( n = 10). CONCLUSIONS In patients previously hospitalized with COVID-19, there is long-lasting impairment of endothelial function as measured by the salbutamol-induced stimulation of the NO-cGMP pathway that may contribute to cardiovascular complications.
Collapse
Affiliation(s)
- Luca Faconti
- King's College London, Department of Clinical Pharmacology, St Thomas' Hospital, London, UK
| | | | | | | | | |
Collapse
|
197
|
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
|
198
|
Brosolo G, Da Porto A, Marcante S, Picci A, Capilupi F, Capilupi P, Bertin N, Vivarelli C, Bulfone L, Vacca A, Catena C, Sechi LA. Omega-3 Fatty Acids in Arterial Hypertension: Is There Any Good News? Int J Mol Sci 2023; 24:9520. [PMID: 37298468 PMCID: PMC10253816 DOI: 10.3390/ijms24119520] [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: 05/12/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (ω-3 PUFAs), including alpha-linolenic acid (ALA) and its derivatives eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are "essential" fatty acids mainly obtained from diet sources comprising plant oils, marine blue fish, and commercially available fish oil supplements. Many epidemiological and retrospective studies suggested that ω-3 PUFA consumption decreases the risk of cardiovascular disease, but results of early intervention trials have not consistently confirmed this effect. In recent years, some large-scale randomized controlled trials have shed new light on the potential role of ω-3 PUFAs, particularly high-dose EPA-only formulations, in cardiovascular prevention, making them an attractive tool for the treatment of "residual" cardiovascular risk. ω-3 PUFAs' beneficial effects on cardiovascular outcomes go far beyond the reduction in triglyceride levels and are thought to be mediated by their broadly documented "pleiotropic" actions, most of which are directed to vascular protection. A considerable number of clinical studies and meta-analyses suggest the beneficial effects of ω-3 PUFAs in the regulation of blood pressure in hypertensive and normotensive subjects. These effects occur mostly through regulation of the vascular tone that could be mediated by both endothelium-dependent and independent mechanisms. In this narrative review, we summarize the results of both experimental and clinical studies that evaluated the effect of ω-3 PUFAs on blood pressure, highlighting the mechanisms of their action on the vascular system and their possible impact on hypertension, hypertension-related vascular damage, and, ultimately, cardiovascular outcomes.
Collapse
Affiliation(s)
- Gabriele Brosolo
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Andrea Da Porto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Stefano Marcante
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Alessandro Picci
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Filippo Capilupi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Patrizio Capilupi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Nicole Bertin
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Cinzia Vivarelli
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
| | - Luca Bulfone
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Antonio Vacca
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Cristiana Catena
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
| | - Leonardo A. Sechi
- Department of Medicine, University of Udine, 33100 Udine, Italy; (A.D.P.); (S.M.); (A.P.); (F.C.); (P.C.); (N.B.); (C.V.); (L.B.); (A.V.); (C.C.)
- European Hypertension Excellence Center, Clinica Medica, University of Udine, 33100 Udine, Italy
- Diabetes and Metabolism Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
- Thrombosis and Hemostasis Unit, Clinica Medica, University of Udine, 33100 Udine, Italy
| |
Collapse
|
199
|
Dorresteijn MJ, Dekker D, Zwaag J, Heemskerk S, Roelofs HM, Smits P, van der Hoeven JG, Wagener FA, Pickkers P. Atazanavir-induced unconjugated hyperbilirubinemia prevents vascular hyporeactivity during experimental human endotoxemia. Front Immunol 2023; 14:1176775. [PMID: 37261364 PMCID: PMC10228648 DOI: 10.3389/fimmu.2023.1176775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/24/2023] [Indexed: 06/02/2023] Open
Abstract
Objective Inflammation-induced free radical release is important in the pathogenesis of several diseases, including atherosclerosis and sepsis. Heme oxygenase (HO) breaks down heme into carbon monoxide, iron, and biliverdin. Biliverdin IXα is directly converted to bilirubin by biliverdin reductase. Unconjugated bilirubin is a powerful antioxidant, and elevated levels have beneficial effects in preclinical models and human cardiovascular disease. However, its impact during acute inflammation in humans is unknown. In the present study, we investigated the impact of atazanavir-induced (unconjugated) hyperbilirubinemia on antioxidant capacity, inflammation, and vascular dysfunction in human experimental endotoxemia. Approach and results Following double-blinded four-day treatment with atazanavir 2dd300 mg (or placebo), twenty healthy male volunteers received 2 ng/kg Escherichia coli lipopolysaccharide intravenously. Blood was drawn to determine the bilirubin levels, antioxidant capacity, and cytokine response. It was demonstrated that following atazanavir treatment, total bilirubin concentrations increased to maximum values of 4.67 (95%CI 3.91-5.59) compared to 0.82 (95%CI 0.64-1.07) mg/dL in the control group (p<0.01). Furthermore, the anti-oxidant capacity, as measured by the ferric-reducing ability of plasma (FRAP), was significantly increased with 36% in hyperbilirubinemia subjects (p<0.0001), and FRAP concentrations correlated strongly to bilirubin concentrations (R2 = 0.77, p<0.001). Hyperbilirubinemia attenuated the release of interleukin-10 from 377 (95%CI 233-609) to 219 (95%CI 152-318) pg/mL (p=0.01), whereas the release of pro-inflammatory cytokines remained unaltered. In vitro, in the absence of hyperbilirubinemia, atazanavir did not influence lipopolysaccharide-induced cytokine release in a whole blood assay. Vascular function was assessed using forearm venous occlusion plethysmography after intra-arterial infusion of acetylcholine and nitroglycerin. Hyperbilirubinemia completely prevented the LPS-associated blunted vascular response to acetylcholine and nitroglycerin. Conclusions Atazanavir-induced hyperbilirubinemia increases antioxidant capacity, attenuates interleukin-10 release, and prevents vascular hyporesponsiveness during human systemic inflammation elicited by experimental endotoxemia. Clinical trial registration http://clinicaltrials.gov, identifier NCT00916448.
Collapse
Affiliation(s)
- Mirrin J. Dorresteijn
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Pharmacology and Toxicology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Douwe Dekker
- Department of Pharmacology and Toxicology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jelle Zwaag
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Suzanne Heemskerk
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Pharmacology and Toxicology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hennie M.J. Roelofs
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Paul Smits
- Department of Pharmacology and Toxicology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johannes G. van der Hoeven
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frank A.D.T.G. Wagener
- Dentistry-Orthodontics and Craniofacial Biology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
200
|
Mousa S, Saif A, Fathy M, Mansour M, Abd Elhamid AM, Atef A, Galal M, Saad S, Aboulsoud S. Assessment of early vascular changes in adult females with polycystic ovary syndrome: correlation with insulin resistance. Gynecol Endocrinol 2023; 39:2210226. [PMID: 37182540 DOI: 10.1080/09513590.2023.2210226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the commonest endocrine disorder in females in the reproductive period with estimated prevalence of 5% to 18% [1]. It contributes to the mortality and morbidity in patients with PCOS due to the increased risk of different metabolic and cardiovascular (CV) complications [2]. Despite the presence of obesity in 40-60% of cases [3], the disease may occur in non-obese women. The occurrence of metabolic disorders in non-obese PCOS patients, suggests that the syndrome itself may play a role in the development of metabolic and CV co-morbidities [4]. The identification of early stages of atherosclerosis in patients with PCOS might be useful in the development of new strategies to control modifiable CV risk factors [5]. Assessment of vascular endothelial function (ED) as an initial reversible step in atherosclerosis development, may serve as an integral index for CV risk factor burden [6]. In addition, carotid intima media thickness (CIMT) is a helpful marker for atherosclerosis and for the identification of increased risk of CV disease [7]. Our study assessed the early vascular changes in Egyptian women with PCOS both physically and functionally by looking at the CIMT using high resolution Doppler ultrasound and by measuring ED using brachial artery flow-mediated vasodilatation (FMD). Our results indicate that patients with PCOS have significant ED and premature atherosclerosis which is, to a great extent, independent of obesity and IR. This suggests that PCOS patients are at increased risk for premature CVD and may benefit from early detection and management.
Collapse
Affiliation(s)
- Shrook Mousa
- Faculty of Medicine, Internal Medicine-Endocrinology Unit, Cairo University, Cairo, Egypt
| | - Aasem Saif
- Faculty of Medicine, Internal Medicine-Endocrinology Unit, Cairo University, Cairo, Egypt
| | - Marwa Fathy
- Faculty of Medicine, Internal Medicine-Endocrinology Unit, Cairo University, Cairo, Egypt
| | - Mona Mansour
- Faculty of Medicine, Internal Medicine-Endocrinology Unit, Cairo University, Cairo, Egypt
| | | | - Adel Atef
- Faculty of Medicine, Gynaecology and Obstetrics Department, Cairo University, Cairo, Egypt
| | - Mai Galal
- Faculty of Medicine, Internal Medicine-Vascular Unit, Cairo University, Cairo, Egypt
| | - Samar Saad
- Faculty of Medicine, Cairo University, Clinical Pathology, Cairo, Egypt
| | - Samar Aboulsoud
- Faculty of Medicine, Internal Medicine Department, Cairo University, Cairo, Egypt
| |
Collapse
|