151
|
Diaz Sanchez L, Sanchez-Aranguren L, Wang K, Spickett CM, Griffiths HR, Dias IHK. TNF-α-Mediated Endothelial Cell Apoptosis Is Rescued by Hydrogen Sulfide. Antioxidants (Basel) 2023; 12:antiox12030734. [PMID: 36978982 PMCID: PMC10045727 DOI: 10.3390/antiox12030734] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Endothelial dysfunction is implicated in the development and aggravation of cardiovascular complications. Among the endothelium-released vasoactive factors, hydrogen sulfide (H2S) has been investigated for its beneficial effects on the vasculature through anti-inflammatory and redox-modulating regulatory mechanisms. Reduced H2S bioavailability is reported in chronic diseases such as cardiovascular disease, diabetes, atherosclerosis and preeclampsia, suggesting the value of investigating mechanisms, by which H2S acts as a vasoprotective gasotransmitter. We explored whether the protective effects of H2S were linked to the mitochondrial health of endothelial cells and the mechanisms by which H2S rescues apoptosis. Here, we demonstrate that endothelial dysfunction induced by TNF-α increased endothelial oxidative stress and induced apoptosis via mitochondrial cytochrome c release and caspase activation over 24 h. TNF-α also affected mitochondrial morphology and altered the mitochondrial network. Post-treatment with the slow-releasing H2S donor, GYY4137, alleviated oxidising redox state, decreased pro-caspase 3 activity, and prevented endothelial apoptosis caused by TNF-α alone. In addition, exogenous GYY4137 enhanced S-sulfhydration of pro-caspase 3 and improved mitochondrial health in TNF-α exposed cells. These data provide new insights into molecular mechanisms for cytoprotective effects of H2S via the mitochondrial-driven pathway.
Collapse
Affiliation(s)
- Lorena Diaz Sanchez
- Aston Medical School, College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Lissette Sanchez-Aranguren
- Aston Medical School, College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Keqing Wang
- Aston Medical School, College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Corinne M. Spickett
- School of Biosciences, College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Helen R. Griffiths
- Swansea Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | - Irundika H. K. Dias
- Aston Medical School, College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
- Correspondence: ; Tel.: +0121-204-4678
| |
Collapse
|
152
|
Willems L, Vermeulen J, Wiegerinck A, Fekkes S, Reijnen M, Warlé M, De Korte C, Thijssen D. Construct Validity and Reproducibility of Handheld Ultrasound Devices in Carotid Artery Diameter Measurement. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:866-874. [PMID: 36567191 DOI: 10.1016/j.ultrasmedbio.2022.11.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 06/17/2023]
Abstract
The construct validity and reproducibility of three commonly used handheld ultrasound (US) devices in measuring carotid arterial diameter was evaluated: Telemed MicrUs EXT-1H (Telemed, Vilnius, Lithuania), Butterfly iQ (Butterfly Network, Inc., Guilford, CT, USA) and Philips Lumify (Philips Healthcare, Best, The Netherlands). An in vitro setup was built to evaluate construct validity, compared with high-end US, and intra-observer variability of handheld US devices. Handheld devices showed a mean difference of 0.023 ± 0.030 cm, 0.012 ± 0.037 cm and 0.009 ± 0.046 cm for, respectively, Telemed, Butterfly and Lumify in comparison with high-end US devices. Intraclass agreement with the high-end system as well as intra-observer variability for handheld US devices was classified as excellent, with all values greater than 0.95. Subsequently, inter-observer variability of handheld US devices was investigated in an in vivo setup with 20 healthy volunteers. Inter-observer variability was classified as excellent for Telemed (0.901), good for Lumify (0.827) and moderate for Butterfly (0.684) with a difference of, respectively, 0.005 ± 0.031 cm, 0.020 ± 0.050 cm and -0.003 ± 0.033 cm. In conclusion, handheld US devices demonstrated an excellent construct validity and intra-observer variability. Additionally, excellent-to-good inter-observer variability for Telemed and Lumify was observed, and Butterfly demonstrated a moderate inter-observer agreement. These results indicate that handheld US devices are effective for measuring carotid arterial diameter.
Collapse
Affiliation(s)
- Loes Willems
- Department of Surgery, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Jenske Vermeulen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Surgery, Rijnstate Hospital, Arnhem, The Netherlands
| | - Anouk Wiegerinck
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Stein Fekkes
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Michel Reijnen
- Department of Surgery, Rijnstate Hospital, Arnhem, The Netherlands; Multi-Modality Medical Imaging Group, TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Michiel Warlé
- Department of Surgery, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Chris De Korte
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Dick Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
153
|
Koueik J, Wesley UV, Dempsey RJ. Pathophysiology, cellular and molecular mechanisms of large and small vessel diseases. Neurochem Int 2023; 164:105499. [PMID: 36746322 DOI: 10.1016/j.neuint.2023.105499] [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: 12/08/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/07/2023]
Abstract
Cerebrovascular disease (CVD) is the second most common cause of cognitive impairment and dementia in aged population. CVD presents in a myriad number of clinical ways based on the functional location of pathology. While primary clinical emphasis has been placed on motor, speech and visual deficits, vascular cognitive decline is a vastly under recognized and devastating condition afflicting millions of Americans. CVD, a disease of the blood vessels that supply blood to brain involves an integration between small and large vessels. Cerebral large vessel diseases (LVD) are associated with atherosclerosis, artery-to-artery embolism, intracardiac embolism and a large vessel stroke leading to substantial functional disability. Cerebral small vessel disease (SVD) is critically involved in stroke, brain hemorrhages, cognitive decline and functional loss in elderly patients. An evolving understanding of cellular and molecular mechanisms emphasizes that inflammatory vascular changes contribute to systemic pathologic conditions of the central nervous systems (CNS), with specific clinical presentations including, cognitive decline. Advances in an understanding of pathophysiology of disease processes and therapeutic interventions may help improve outcomes. This review will focus on large and small vessels diseases and their relationship to vascular cognitive decline, atherosclerosis, stroke, and inflammatory neurodegeneration. We will also emphasize the molecular and cellular mechanisms, as well as genetic and epigenetic factors associated with LVD and SVD.
Collapse
Affiliation(s)
- Joyce Koueik
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA
| | - Umadevi V Wesley
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA
| | - Robert J Dempsey
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA.
| |
Collapse
|
154
|
Zamani M, Rezaiian F, Saadati S, Naseri K, Ashtary-Larky D, Yousefi M, Golalipour E, Clark CCT, Rastgoo S, Asbaghi O. The effects of folic acid supplementation on endothelial function in adults: a systematic review and dose-response meta-analysis of randomized controlled trials. Nutr J 2023; 22:12. [PMID: 36829207 PMCID: PMC9951414 DOI: 10.1186/s12937-023-00843-y] [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: 11/09/2022] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Endothelial dysfunction serves as an early marker for the risk of cardiovascular disease (CVD); therefore, it is an attractive site of therapeutic interventions to reduce the risk of CVD. This study was conducted to investigate the effect of folic acid supplementation on endothelial function markers in randomized controlled trials (RCTs). METHODS PubMed, ISI web of science, and Scopus databases were searched up to July 2022 for detecting eligible studies. A random-effects model was used for meta-analysis, and linear Meta-regression and non-linear dose-response analysis were performed to assess whether the effect of folic acid supplementation was affected by the dose and duration of intervention. Cochrane tools were also used to assess the risk of bias in the included studies. RESULTS Twenty-one studies, including 2025 participants (1010 cases and 1015 controls), were included in the present meta-analysis. Folic acid supplementation significantly affected the percentage of flow-mediated dilation (FMD%) (WMD: 2.59%; 95% CI: 1.51, 3.67; P < 0.001) and flow-mediated dilation (FMD) (WMD: 24.38 μm; 95% CI: 3.08, 45.68; P = 0.025), but not end-diastolic diameter (EDD) (WMD: 0.21 mm; 95% CI: - 0.09, 0.52; P = 0.176), and intercellular adhesion molecule (ICAM) (WMD: 0.18 ng/ml; 95% CI: - 10.02, 13.81; P = 0.755). CONCLUSIONS These findings suggest that folic acid supplementation may improve endothelial function by increasing FMD and FMD% levels. TRIAL REGISTRATION PROSPERO registration cod: CRD42021289744.
Collapse
Affiliation(s)
- Mohammad Zamani
- grid.411705.60000 0001 0166 0922Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Rezaiian
- grid.411600.2National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeede Saadati
- grid.1002.30000 0004 1936 7857Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Kaveh Naseri
- grid.411600.2Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Damoon Ashtary-Larky
- grid.411230.50000 0000 9296 6873Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohsen Yousefi
- grid.411600.2Faculty of Medicine, Shahid Beheshti University of Medical sciences, Tehran, Iran
| | - Elnaz Golalipour
- grid.411600.2Faculty of Medicine, Shahid Beheshti University of Medical sciences, Tehran, Iran
| | - Cain C. T. Clark
- grid.8096.70000000106754565Centre for Intelligent Healthcare, Coventry University, Coventry, CV1 5FB UK
| | - Samira Rastgoo
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Cellular and Molecular Nutrition, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Omid Asbaghi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
155
|
Li JX, Tian R, Lu N. Quercetin Attenuates Vascular Endothelial Dysfunction in Atherosclerotic Mice by Inhibiting Myeloperoxidase and NADPH Oxidase Function. Chem Res Toxicol 2023; 36:260-269. [PMID: 36719041 DOI: 10.1021/acs.chemrestox.2c00334] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Myeloperoxidase (MPO) exhibits a unique property to use H2O2 to oxidize chloride and lead to the generation of a strong oxidant, hypochlorous acid (HOCl), which plays important roles in atherosclerosis. A lot of evidence indicates that quercetin, a natural polyphenol derived from human diet, effectively contributes to cardiovascular health. Herein, we found that dietary quercetin significantly inhibited vascular endothelial dysfunction and atherosclerosis in apolipoprotein E-deficient (ApoE-/-) mice. Mechanistic studies revealed that dietary quercetin effectively suppressed the MPO level and activity in the vessels of ApoE-/- animals, and p47phox expression and NADPH oxidase activity were simultaneously attenuated after quercetin treatment. In vascular endothelial cells, NADPH oxidase was demonstrated to be the major source of H2O2 formation. Moreover, quercetin effectively attenuated MPO/H2O2-mediated HOCl production and toxicity to human vascular endothelial cells, and this compound was not toxic. The inhibitory effect on MPO activity was likely attributed to that quercetin significantly inhibited NADPH oxidase-derived H2O2 formation in human endothelial cells and could act as an effective mediator for MPO intermediates, subsequently preventing HOCl production by the MPO/H2O2 system. Collectively, it was suggested that quercetin effectively suppressed endothelial dysfunction in atherosclerotic vasculature through the reduction of MPO/NADPH oxidase-mediated HOCl production.
Collapse
Affiliation(s)
- Jia-Xin Li
- College of Chemistry and Chemical Engineering, Jiangxi Key Laboratory of Green Chemistry, Jiangxi Normal University, Ziyang Road 99, Nanchang, Jiangxi 330022, China
| | - Rong Tian
- College of Chemistry and Chemical Engineering, Jiangxi Key Laboratory of Green Chemistry, Jiangxi Normal University, Ziyang Road 99, Nanchang, Jiangxi 330022, China
| | - Naihao Lu
- College of Chemistry and Chemical Engineering, Jiangxi Key Laboratory of Green Chemistry, Jiangxi Normal University, Ziyang Road 99, Nanchang, Jiangxi 330022, China
| |
Collapse
|
156
|
Vascular Function, Systemic Inflammation, and Coagulation Activation 18 Months after COVID-19 Infection: An Observational Cohort Study. J Clin Med 2023; 12:jcm12041413. [PMID: 36835948 PMCID: PMC9965558 DOI: 10.3390/jcm12041413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
INTRODUCTION Among its effect on virtually all other organs, COVID-19 affects the cardiovascular system, potentially jeopardizing the cardiovascular health of millions. Previous research has shown no indication of macrovascular dysfunction as reflected by carotid artery reactivity, but has shown sustained microvascular dysfunction, systemic inflammation, and coagulation activation at 3 months after acute COVID-19. The long-term effects of COVID-19 on vascular function remain unknown. MATERIALS AND METHODS This cohort study involved 167 patients who participated in the COVAS trial. At 3 months and 18 months after acute COVID-19, macrovascular dysfunction was evaluated by measuring the carotid artery diameter in response to cold pressor testing. Additionally, plasma endothelin-1, von Willebrand factor, Interleukin(IL)-1ra, IL-6, IL-18, and coagulation factor complexes were measured using ELISA techniques. RESULTS The prevalence of macrovascular dysfunction did not differ between 3 months (14.5%) and 18 months (11.7%) after COVID-19 infection (p = 0.585). However, there was a significant decrease in absolute carotid artery diameter change, 3.5% ± 4.7 vs. 2.7% ± 2.5, p-0.001, respectively. Additionally, levels of vWF:Ag were persistently high in 80% of COVID-19 survivors, reflecting endothelial cell damage and possibly attenuated endothelial function. Furthermore, while levels of the inflammatory cytokines interleukin(IL)-1RA and IL-18 were normalized and evidence of contact pathway activation was no longer present, the concentrations of IL-6 and thrombin:antithrombin complexes were further increased at 18 months versus 3 months (2.5 pg/mL ± 2.6 vs. 4.0 pg/mL ± 4.6, p = 0.006 and 4.9 μg/L ± 4.4 vs. 18.2 μg/L ± 11.4, p < 0.001, respectively). DISCUSSION This study shows that 18 months after COVID-19 infection, the incidence of macrovascular dysfunction as defined by a constrictive response during carotid artery reactivity testing is not increased. Nonetheless, plasma biomarkers indicate sustained endothelial cell activation (vWF), systemic inflammation (IL-6), and extrinsic/common pathway coagulation activation (FVII:AT, TAT) 18 months after COVID-19 infection.
Collapse
|
157
|
Jiang X, Gao H, Cao Y, Chen S, Huang F, Feng Y, Zhang Y, Feng S. SiNPs induce ferroptosis in HUVECs through p38 inhibiting NrF2 pathway. Front Public Health 2023; 11:1024130. [PMID: 36844840 PMCID: PMC9945284 DOI: 10.3389/fpubh.2023.1024130] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 01/16/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Despite of growing evidence linking silica nanoparticles (SiNPs), one of the global-top-three-produced and -used nanoparticle (NP), to human health risks, there remain many knowledge gaps over the adverse effects of SiNPs exposure on cardiovascular system and the underlying molecular mechanisms. Methods In this study, the ferroptotic effects of SiNPs (20 nm; 0, 25, 50, and 100 μg/mL) on human umbilical vein endothelial cells (HUVECs) and the possible molecular mechanism were studied with the corresponding biochemical and molecular biology assays. Results and discussion The results showed that at the tested concentrations, SiNPs could decrease HUVEC viability, but the deferoxamine mesylate (an iron ion chelator) might rescue this reduction of cell viability. Also, increased levels of intracellular reactive oxygen species and enhanced mRNA expression of lipid oxidation enzymes (ACSL4 and LPCAT3) with increase in lipid peroxidation (malondialdehyde), but decreased ratios of intracellular GSH/total-GSH and mitochondrial membrane potential as well as reduced enzymatic activities of anti-oxidative enzymes (CAT, SOD, and GSH-PX), were found in the SiNPs-treated HUVECs. Meanwhile, increase in p38 protein phosphorylation and decrease in NrF2 protein phosphorylation with reduced mRNA expressions of downstream anti-oxidative enzyme genes (CAT, SOD1, GSH-PX, and GPX4) was identified in the SiNPs-exposed HUVECs. These data indicated that SiNPs exposure might induce ferroptosis in HUVECs via p38 inhibiting NrF2 pathway. Ferroptosis of HUVECs will become a useful biomarker for assessing the cardiovascular health risks of environmental contaminants.
Collapse
Affiliation(s)
- Xiaojun Jiang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, China,The School of Public Health, University of South China, Hengyang, China
| | - Huiqian Gao
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, China
| | - Yunchang Cao
- The Department of Molecular Biology, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| | - Shuting Chen
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, China,The School of Public Health, University of South China, Hengyang, China
| | - Fangfang Huang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, China
| | - Yashi Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, China
| | - Yuqi Zhang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, China
| | - Shaolong Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, China,The School of Public Health, University of South China, Hengyang, China,The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, China,*Correspondence: Shaolong Feng ✉
| |
Collapse
|
158
|
Fan Z, Feng Y, Zang L, Guo Y, Zhong XY. Association of circulating MtDNA with CVD in hemodialysis patients and in vitro effect of exogenous MtDNA on cardiac microvascular inflammation. BMC Cardiovasc Disord 2023; 23:74. [PMID: 36755219 PMCID: PMC9906832 DOI: 10.1186/s12872-023-03104-2] [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: 09/12/2022] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) patients sustain a fairly high prevalence of cardiovascular disease (CVD). Microvascular inflammation is an early manifestation of CVD, and the released mitochondrial DNA (MtDNA) has been proposed to be a crucial integrator of inflammatory signals. Herein, the aim of this study was to determine the relationship between CVD, microvessel, and circulating MtDNA in the settings of uremia. METHODS Forty-two maintenance hemodialysis (MHD) patients and 36 health controls were enrolled in this study. Plasma cell-free MtDNA was detected by TaqMan-based qPCR assay. CVD risk markers including high-sensitive C-reactive protein (Hs-CRP), monocyte chemoattractant protein-1 (MCP-1), fibrinogen, and erythrocyte sedimentation rate (ESR) were measured by standard assays. Ten-year CVD risk was calculated from the Framingham risk score (FRS) model. In vitro study, human cardiac microvascular endothelial cells (HCMECs) were incubated with normal or uremic serum, with or without exogenous MtDNA. Intracellular toll-like receptor 9 (TLR9), adhesion molecule 1 (ICAM-1), MCP-1 and tumor necrosis factor-α (TNF-α) and cytosolic MtDNA were detected by qPCR. RESULTS Plasma MtDNA in MHD patients was significantly higher than healthy controls (4.74 vs. 2.41 × 105 copies/mL; p = 0.000). Subsequently, the MHD patients were classified into two groups based on the MtDNA median (4.34 × 105 copies/mL). In stratified analyses, the levels of Hs-CRP (5.02 vs. 3.73 mg/L; p = 0.042) and MCP-l (99.97 vs. 64.72 pg/mL; p = 0.008) and FRS (21.80 vs. 16.52; p = 0.016) in the high plasma MtDNA group were higher than those in the low plasma MtDNA group. In vitro study, we found that exogenous MtDNA aggravated uremic serum-induced microvascular inflammation (ICAM-1 and TNF-α) in HCMECs (all p < 0.05). Besides, the addition of MtDNA to the medium resulted in a further increase in cytosolic MtDNA and TLR9 levels in uremic serum-treated cells (all p < 0.05). In patients with MHD, MtDNA levels in plasma were significantly reduced after a single routine hemodialysis (pre 4.47 vs. post 3.45 × 105 copies/mL; p = 0.001) or hemodiafiltration (pre 4.85 vs. post 4.09 × 105 copies/mL; p = 0.001). These two approaches seem similar in terms of MtDNA clearance rate (21.26% vs. 11.94%; p = 0.172). CONCLUSIONS Overall, the present study suggests that MtDNA released into the circulation under the uremic toxin environment may adversely affect the cardiovascular system by exacerbating microvascular inflammation, and that reducing circulating MtDNA might be a future therapeutic strategy for the prevention of MHD-related CVD.
Collapse
Affiliation(s)
- Zhen Fan
- Department of Geriatrics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan China
| | - Ya Feng
- grid.414880.1Department of Nephrology, Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan China
| | - Li Zang
- grid.414880.1Department of Nephrology, Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan China
| | - Yi Guo
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Section 2, Yihuan Road, Qingyang District, Chengdu, 610072, Sichuan, China.
| | - Xiao-yi Zhong
- grid.417298.10000 0004 1762 4928Department of Nephrology, Xinqiao Hospital, Army Medical University (Third Military Medical University), No. 83 Xinqiao Zhengjie, Shapingba District, Chongqing, 400037 China
| |
Collapse
|
159
|
Hluchanova A, Kollar B, Klobucnikova K, Hardonova M, Poddany M, Zitnanova I, Dvorakova M, Konarikova K, Tedla M, Urik M, Klail P, Skopek P, Turcani P, Siarnik P. Lipoprotein Subfractions Associated with Endothelial Function in Previously Healthy Subjects with Newly Diagnosed Sleep Apnea-A Pilot Study. Life (Basel) 2023; 13:441. [PMID: 36836798 PMCID: PMC9962671 DOI: 10.3390/life13020441] [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: 12/28/2022] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) activates several pathophysiological mechanisms which can lead to the development of vascular diseases. Endothelial dysfunction (ED) is an initial step in the development of atherosclerosis. The association between ED and OSA has been described in several studies, even in previously healthy subjects. High-density lipoproteins (HDL) were generally considered to be atheroprotective, and low-density lipoprotein (LDL) to be an atherogenic component of lipoproteins. However, recent findings suggest a pro-atherogenic role of small HDL subfractions (8-10) and LDL subfractions (3-7). This study aimed to evaluate the relationship between endothelial function and lipid subfractions in previously healthy OSA subjects. MATERIAL AND METHODS We prospectively enrolled 205 subjects with sleep monitoring. Plasma levels of triacylglycerols, total cholesterol, LDL, HDL, and their subfractions were assessed. Endothelial function was determined using peripheral arterial tonometry, and reperfusion hyperemia index (RHI) was assessed. RESULTS Plasma levels of small and intermediate HDL subfractions have statistically significant pro-atherogenic correlations with endothelial function (p = 0.015 and p = 0.019). In other lipoprotein levels, no other significant correlation was found with RHI. In stepwise multiple linear regression analysis, small HDL (beta = -0.507, p = 0.032) was the only significant contributor in the model predicting RHI. CONCLUSIONS In our studied sample, a pro-atherogenic role of small HDL subfractions in previously healthy subjects with moderate-to-severe OSA was proven.
Collapse
Affiliation(s)
- Alzbeta Hluchanova
- Department of Neurology, University Hospital Bratislava, 85107 Bratislava, Slovakia
| | - Branislav Kollar
- 1st Department of Neurology, Faculty of Medicine, Comenius University, University Hospital, 81499 Bratislava, Slovakia
| | - Katarina Klobucnikova
- 1st Department of Neurology, Faculty of Medicine, Comenius University, University Hospital, 81499 Bratislava, Slovakia
| | - Miroslava Hardonova
- 1st Department of Neurology, Faculty of Medicine, Comenius University, University Hospital, 81499 Bratislava, Slovakia
| | - Michal Poddany
- Department of Neurology, General Hospital, 03123 Liptovsky Mikulas, Slovakia
| | - Ingrid Zitnanova
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 81499 Bratislava, Slovakia
| | - Monika Dvorakova
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 81499 Bratislava, Slovakia
| | - Katarina Konarikova
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 81499 Bratislava, Slovakia
| | - Miroslav Tedla
- Department of ENT and HNS, Faculty of Medicine, Comenius University, University Hospital, 81499 Bratislava, Slovakia
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SQ, UK
| | - Milan Urik
- Department of Pediatric Otorhinolaryngology, University Hospital Brno, 61300 Brno, Czech Republic
- Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Pavel Klail
- Department of Otorhinolaryngology, University Hospital in Pilsen, Faculty of Medicine, Charles University, 11000 Pilsen, Czech Republic
| | - Petr Skopek
- Department of Otorhinolaryngology, University Hospital in Pilsen, Faculty of Medicine, Charles University, 11000 Pilsen, Czech Republic
| | - Peter Turcani
- 1st Department of Neurology, Faculty of Medicine, Comenius University, University Hospital, 81499 Bratislava, Slovakia
| | - Pavel Siarnik
- 1st Department of Neurology, Faculty of Medicine, Comenius University, University Hospital, 81499 Bratislava, Slovakia
| |
Collapse
|
160
|
Cirillo M, Argento FR, Attanasio M, Becatti M, Ladisa I, Fiorillo C, Coccia ME, Fatini C. Atherosclerosis and Endometriosis: The Role of Diet and Oxidative Stress in a Gender-Specific Disorder. Biomedicines 2023; 11:biomedicines11020450. [PMID: 36830986 PMCID: PMC9953736 DOI: 10.3390/biomedicines11020450] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Background: Accelerated atherosclerosis in patients with endometriosis has been hypothesised, and lifestyle improvement might control cardiovascular risk. We explored cardiometabolic markers and oxidative stress and evaluated the effects of the Mediterranean Diet (MD) in modulating these markers. Methods: In this prospective study, we included 35 women with endometriosis. At baseline (T0) and after 3 (T1) and 6 (T2) months from the start of the diet, we investigated cardiometabolic parameters, lifestyle and oxidative stress. Results: After a 3-month intervention with MD, we observed a significant reduction in total cholesterol (p = 0.01) and LDL-c (p = 0.003). We observed at T1 an increase in B12 and E vitamins, folate and zinc. After 6 months, zinc (p = 0.04) and folate (p = 0.08) increased in comparison to T0. A reduction in homocysteine from T0 to T1 (p = 0.01) was found. After 3 months, an increase in Rapid Assessment of Physical Activity tool 1 (RAPA) (p < 0.001) and RAPA 2 was observed (p = 0.009). We observed high levels of oxidative stress markers at baseline. After 6 months of MD, a significant improvement in lymphocyte Reactive Oxygen Species (ROS) (p < 0.001) and total antioxidant capacity was observed (p = 0.02). Conclusions: The improvement of lifestyle, and in particular the Mediterranean dietary intervention, allowed the improvement of the metabolic and oxidative profile and overall health-related quality of life.
Collapse
Affiliation(s)
- Michela Cirillo
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Centre for Assisted Reproductive Technology, Division of Obstetrics and Gynaecology, Careggi University Hospital, 50134 Florence, Italy
| | - Flavia Rita Argento
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Monica Attanasio
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Matteo Becatti
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Irene Ladisa
- Centre for Assisted Reproductive Technology, Division of Obstetrics and Gynaecology, Careggi University Hospital, 50134 Florence, Italy
| | - Claudia Fiorillo
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Maria Elisabetta Coccia
- Centre for Assisted Reproductive Technology, Division of Obstetrics and Gynaecology, Careggi University Hospital, 50134 Florence, Italy
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, 50134 Florence, Italy
| | - Cinzia Fatini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Centre for Assisted Reproductive Technology, Division of Obstetrics and Gynaecology, Careggi University Hospital, 50134 Florence, Italy
- Correspondence:
| |
Collapse
|
161
|
Association between the Oxidative Balance Score and Incident Chronic Kidney Disease in Adults. Antioxidants (Basel) 2023; 12:antiox12020335. [PMID: 36829895 PMCID: PMC9952833 DOI: 10.3390/antiox12020335] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress is a novel risk factor for chronic kidney disease (CKD). The oxidative balance score (OBS) was developed to represent the overall oxidative balance based on dietary and lifestyle pro-oxidant and antioxidant components. The aim of this study is to verify the relationship between the OBS and the incidence of CKD. Data from 5795 participants without CKD at the baseline survey of the Korean Genome and Epidemiology Study were analyzed. Participants were classified into sex-specific OBS tertiles. During the mean follow-up period of 13.6 years, 286 men and 382 women newly developed CKD. The Cox proportional hazard spline curve revealed an inverse dose-response association between the OBS and incident CKD in both men and women. Multiple Cox proportional hazard regression analysis revealed that the adjusted hazard ratios (95% confidence intervals) for sex-specific highest (T3) and middle (T2) OBS tertile groups were 0.80 (0.59-1.08) and 0.70 (0.51-0.95), respectively, in men and 0.76 (0.59-0.98) and 0.73 (0.55-0.96), respectively, in women, with the sex-specific lowest OBS tertile group (T1) as the reference. These results suggest that a healthy diet and lifestyle that increases the OBS may help prevent CKD in both men and women.
Collapse
|
162
|
Fiedorczuk P, Olszewska E, Rogalska J, Brzóska MM. Osteoprotegerin, Chitinase 3-like Protein 1, and Cardiotrophin-1 as Potential Biomarkers of Obstructive Sleep Apnea in Adults-A Case-Control Study. Int J Mol Sci 2023; 24:ijms24032607. [PMID: 36768925 PMCID: PMC9916736 DOI: 10.3390/ijms24032607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/15/2023] [Accepted: 01/28/2023] [Indexed: 02/03/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a prevalent, underdiagnosed disease and is considered a risk factor for cardiovascular diseases, depression, accidents, and stroke. Recent clinical practice guidelines for OSA expressed the need for a new clinical tool that establishes the Apnea-Hypopnea Index (AHI) to determine the disease burden. The serum and plasma concentrations of Osteoprotegerin (OPG), Chitinase 3-like protein 1 (YKL-40), and Cardiotrophin-1 (CT-1) in 80 subjects-52 OSA patients, 27 moderate (15 ≤ AHI ˂ 30) and 25 severe (AHI ≥ 30), and 28 non-OSA controls (AHI 0-5)-were determined. Moreover, the Total Oxidative Status (TOS), Total Antioxidative Status (TAS), and Oxidative Stress Index (OSI) were assessed in the serum and plasma to evaluate whether the severity of OSA and the concentrations of OPG, YKL-40, and CT-1 correlate with the oxidative/reductive status. The serum and plasma concentrations of YKL-40 and CT-1 were higher in the OSA group, whereas the serum and plasma concentrations of OPG were lower compared to the control group. The concentrations of OPG, YKL-40, and CT-1 in the serum and plasma correlated with AHI; however, a better correlation of the concentrations was obtained for the above-mentioned proteins in the plasma. The concentrations of YKL-40 and CT-1 in the serum and OPG in the plasma show better diagnostic capabilities for moderate and severe OSA than the concentrations of YKL-40 and CT-1 in the plasma and the concentrations of OPG in the serum.
Collapse
Affiliation(s)
- Piotr Fiedorczuk
- Department of Otolaryngology, Medical University of Bialystok, 15-089 Bialystok, Poland
- Correspondence: ; Tel.: +48-663-751-516
| | - Ewa Olszewska
- Department of Otolaryngology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Joanna Rogalska
- Department of Toxicology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | | |
Collapse
|
163
|
Zhou F, Zhu X, Liu Y, Sun Y, Zhang Y, Cheng D, Wang W. Coronary atherosclerosis and chemotherapy: From bench to bedside. Front Cardiovasc Med 2023; 10:1118002. [PMID: 36742069 PMCID: PMC9892653 DOI: 10.3389/fcvm.2023.1118002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular disease, particularly coronary artery disease, is the leading cause of death in humans worldwide. Coronary heart disease caused by chemotherapy affects the prognosis and survival of patients with tumors. The most effective chemotherapeutic drugs for cancer include proteasome inhibitors, tyrosine kinase inhibitors, immune checkpoint inhibitors, 5-fluorouracil, and anthracyclines. Animal models and clinical trials have consistently shown that chemotherapy is closely associated with coronary events and can cause serious adverse cardiovascular events. Adverse cardiovascular events after chemotherapy can affect the clinical outcome, treatment, and prognosis of patients with tumors. In recent years, with the development of new chemotherapeutic drugs, new discoveries have been made about the effects of drugs used for chemotherapy on cardiovascular disease and its related mechanisms, such as inflammation. This review article summarizes the effects of chemotherapeutic drugs on coronary artery disease and its related mechanisms to guide efforts in reducing cardiovascular adverse events during tumor chemotherapy, preventing the development of coronary heart disease, and designing new prevention and treatment strategies for cardiotoxicity caused by clinical tumor chemotherapy.
Collapse
Affiliation(s)
- Fanghui Zhou
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Zhu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yao Liu
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Sun
- Department of Blood and Endocrinology, The 962nd Hospital of the PLA Joint Logistic Support Force, Harbin, Heilongjiang, China
| | - Ying Zhang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | | | - Wei Wang
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,*Correspondence: Wei Wang,
| |
Collapse
|
164
|
Bkaily G, Jacques D. Morphological and Functional Remodeling of Vascular Endothelium in Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24031998. [PMID: 36768314 PMCID: PMC9916505 DOI: 10.3390/ijms24031998] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/21/2023] Open
Abstract
The vascular endothelium plays a vital role during embryogenesis and aging and is a cell monolayer that lines the blood vessels. The immune system recognizes the endothelium as its own. Therefore, an abnormality of the endothelium exposes the tissues to the immune system and provokes inflammation and vascular diseases such as atherosclerosis. Its secretory role allows it to release vasoconstrictors and vasorelaxants as well as cardio-modulatory factors that maintain the proper functioning of the circulatory system. The sealing of the monolayer provided by adhesion molecules plays an important role in cardiovascular physiology and pathology.
Collapse
|
165
|
Xiao CY, Ma YH, Ou YN, Zhao B, Hu HY, Wang ZT, Tan L. Association between Kidney Function and the Burden of Cerebral Small Vessel Disease: An Updated Meta-Analysis and Systematic Review. Cerebrovasc Dis 2023; 52:376-386. [PMID: 36599326 DOI: 10.1159/000527069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/31/2022] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Due to anatomical and functional similarities in microvascular beds, the brain and kidney share distinctive susceptibilities to vascular injury and common risk factors of small vessel disease. The aim of this updated meta-analysis is to explore the association between kidney function and the burden of cerebral small vessel disease (CSVD). METHODS PubMed, EMBASE, and Cochrane Library were systematically searched for observational studies that explored the association between the indicators of kidney function and CSVD neuroimaging markers. The highest-adjusted risk estimates and their 95% confidence intervals (CIs) were aggregated using random-effect models. RESULTS Twelve longitudinal studies and 51 cross-sectional studies with 57,030 subjects met the inclusion criteria of systematic review, of which 52 were included in quantitative synthesis. According to the pooled results, we found that low estimated glomerular filtration rate (eGFR <60 mL/min/1.73 m2) was associated with cerebral microbleeds (odds ratio (OR) = 1.55, 95% CI = 1.26-1.90), white matter hyperintensities (OR = 1.40, 95% CI = 1.05-1.86), and lacunar infarctions (OR = 1.50, 95% CI = 1.18-1.92), but not with severe perivascular spaces (OR = 1.20, 95% CI = 0.77-1.88). Likewise, patients with proteinuria (OR = 1.75, 95% CI = 1.47-2.09) or elevated serum cystatin C (OR = 1.51, 95% CI = 1.25-1.83) also had an increased risk of CSVD. CONCLUSION The association between kidney function and CSVD has been comprehensively updated through this study, that kidney insufficiency manifested as low eGFR, proteinuria, and elevated serum cystatin C was independently associated with CSVD burden.
Collapse
Affiliation(s)
- Chu-Yun Xiao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Bing Zhao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
166
|
Prognostic value of reactive hyperemia index using peripheral artery tonometry in patients with heart failure. Sci Rep 2023; 13:125. [PMID: 36599885 PMCID: PMC9812967 DOI: 10.1038/s41598-023-27454-1] [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: 09/05/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Given the high prevalence and poor prognosis of heart failure (HF), finding prognostic factors for patients with HF is crucial. This study investigated the prognostic value of reactive hyperemia index (RHI), a measure of endothelial function, in HF. A total of 90 HF patients (mean age, 63.7 ± 13.2 years; female, 25.6%) with a history of hospitalization for HF treatment were prospectively enrolled. RHI was measured using digital arterial tonometry in a stable condition. Clinical events, including all-cause death and HF admission, were assessed. During the median follow-up of 3.66 years (interquartile range, 0.91-4.94 years), 26 clinical events (28.9%) occurred. Although there were no significant differences in risk factors and laboratory findings according to the occurrence of clinical events, the RHI value was significantly lower in patients with clinical events than in those without (1.21 ± 0.34 vs. 1.68 ± 0.48; P < 0.001). Kaplan-Meier survival analysis showed that a lower RHI value (< 1.48) was associated with a significantly higher incidence rate of clinical events (log-rank P < 0.001). In multivariable cox regression analysis, a low RHI value (< 1.48) was associated with an increased risk of clinical events (hazard ratio, 14.09; 95% confidence interval, 3.61-54.99; P < 0.001) even after controlling for potential confounders. Our study showed that reduced RHI was associated with an increased risk of adverse clinical outcomes in HF. This suggests that endothelial dysfunction may be an important prognostic marker in patients with HF.
Collapse
|
167
|
Sivaraj N, Boppana S, Ravula N, Simhachalam Kutikuppala LV, Raja Hamsa CH, Mahajan S, Chaitanya PS. Prevalence of resistant hypertension and its relation with plasma lipoprotein-associated phospholipase A2. INTERNATIONAL JOURNAL OF NONCOMMUNICABLE DISEASES 2023. [DOI: 10.4103/jncd.jncd_80_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
|
168
|
Feng S, Huang F, Zhang Y, Feng Y, Zhang Y, Cao Y, Wang X. The pathophysiological and molecular mechanisms of atmospheric PM 2.5 affecting cardiovascular health: A review. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114444. [PMID: 38321663 DOI: 10.1016/j.ecoenv.2022.114444] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 02/08/2024]
Abstract
BACKGROUND Exposure to ambient fine particulate matter (PM2.5, with aerodynamic diameter less than 2.5 µm) is a leading environmental risk factor for global cardiovascular health concern. OBJECTIVE To provide a roadmap for those new to this field, we reviewed the new insights into the pathophysiological and cellular/molecular mechanisms of PM2.5 responsible for cardiovascular health. MAIN FINDINGS PM2.5 is able to disrupt multiple physiological barriers integrity and translocate into the systemic circulation and get access to a range of secondary target organs. An ever-growing body of epidemiological and controlled exposure studies has evidenced a causal relationship between PM2.5 exposure and cardiovascular morbidity and mortality. A variety of cellular and molecular biology mechanisms responsible for the detrimental cardiovascular outcomes attributable to PM2.5 exposure have been described, including metabolic activation, oxidative stress, genotoxicity, inflammation, dysregulation of Ca2+ signaling, disturbance of autophagy, and induction of apoptosis, by which PM2.5 exposure impacts the functions and fates of multiple target cells in cardiovascular system or related organs and further alters a series of pathophysiological processes, such as cardiac autonomic nervous system imbalance, increasing blood pressure, metabolic disorder, accelerated atherosclerosis and plaque vulnerability, platelet aggregation and thrombosis, and disruption in cardiac structure and function, ultimately leading to cardiovascular events and death. Therein, oxidative stress and inflammation were suggested to play pivotal roles in those pathophysiological processes. CONCLUSION Those biology mechanisms have deepen insights into the etiology, course, prevention and treatment of this public health concern, although the underlying mechanisms have not yet been entirely clarified.
Collapse
Affiliation(s)
- Shaolong Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China; Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China; The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China.
| | - Fangfang Huang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yuqi Zhang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yashi Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Ying Zhang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yunchang Cao
- The Department of Molecular Biology, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, China
| | - Xinming Wang
- Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China; The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China
| |
Collapse
|
169
|
Nayeem MA, Geldenhuys WJ, Hanif A. Role of cytochrome P450-epoxygenase and soluble epoxide hydrolase in the regulation of vascular response. ADVANCES IN PHARMACOLOGY 2023; 97:37-131. [DOI: 10.1016/bs.apha.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
170
|
Bei YR, Zhang SC, Song Y, Tang ML, Zhang KL, Jiang M, He RC, Wu SG, Liu XH, Wu LM, Dai XY, Hu YW. EPSTI1 promotes monocyte adhesion to endothelial cells in vitro via upregulating VCAM-1 and ICAM-1 expression. Acta Pharmacol Sin 2023; 44:71-80. [PMID: 35778487 DOI: 10.1038/s41401-022-00923-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/21/2022] [Indexed: 01/18/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease of arterial wall, and circulating monocyte adhesion to endothelial cells is a crucial step in the pathogenesis of atherosclerosis. Epithelial-stromal interaction 1 (EPSTI1) is a novel gene, which is dramatically induced by epithelial-stromal interaction in human breast cancer. EPSTI1 expression is not only restricted to the breast but also in other normal tissues. In this study we investigated the role of EPSTI1 in monocyte-endothelial cell adhesion and its expression pattern in atherosclerotic plaques. We showed that EPSTI1 was dramatically upregulated in human and mouse atherosclerotic plaques when compared with normal arteries. In addition, the expression of EPSTI1 in endothelial cells of human and mouse atherosclerotic plaques is significantly higher than that of the normal arteries. Furthermore, we demonstrated that EPSTI1 promoted human monocytic THP-1 cell adhesion to human umbilical vein endothelial cells (HUVECs) via upregulating VCAM-1 and ICAM-1 expression in HUVECs. Treatment with LPS (100, 500, 1000 ng/mL) induced EPSTI1 expression in HUVECs at both mRNA and protein levels in a dose- and time-dependent manner. Knockdown of EPSTI1 significantly inhibited LPS-induced monocyte-endothelial cell adhesion via downregulation of VCAM-1 and ICAM-1. Moreover, we revealed that LPS induced EPSTI1 expression through p65 nuclear translocation. Thus, we conclude that EPSTI1 promotes THP-1 cell adhesion to endothelial cells by upregulating VCAM-1 and ICAM-1 expression, implying its potential role in the development of atherosclerosis.
Collapse
Affiliation(s)
- Yan-Rou Bei
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shun-Chi Zhang
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, 510620, China
| | - Yu Song
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Mao-Lin Tang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Ke-Lan Zhang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Min Jiang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Run-Chao He
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou, 510620, China
| | - Shao-Guo Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, 510620, China
| | - Xue-Hui Liu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, 510620, China
| | - Li-Mei Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou Medical University, Guangzhou, 510620, China
| | - Xiao-Yan Dai
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou, 510620, China.
| |
Collapse
|
171
|
Chook CYB, Cheung YM, Ma KY, Leung FP, Zhu H, Niu QJ, Wong WT, Chen ZY. Physiological concentration of protocatechuic acid directly protects vascular endothelial function against inflammation in diabetes through Akt/eNOS pathway. Front Nutr 2023; 10:1060226. [PMID: 37025617 PMCID: PMC10070727 DOI: 10.3389/fnut.2023.1060226] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
Background Cardiovascular diseases (CVDs) have been the major cause of mortality in type 2 diabetes. However, new approaches are still warranted since current diabetic medications, which focus mainly on glycemic control, do not effectively lower cardiovascular mortality rate in diabetic patients. Protocatechuic acid (PCA) is a phenolic acid widely distributed in garlic, onion, cauliflower and other plant-based foods. Given the anti-oxidative effects of PCA in vitro, we hypothesized that PCA would also have direct beneficial effects on endothelial function in addition to the systemic effects on vascular health demonstrated by previous studies. Methods and results Since IL-1β is the major pathological contributor to endothelial dysfunction in diabetes, the anti-inflammatory effects of PCA specific on endothelial cells were further verified by the use of IL-1β-induced inflammation model. Direct incubation of db/db mouse aortas with physiological concentration of PCA significantly ameliorated endothelium-dependent relaxation impairment, as well as reactive oxygen species overproduction mediated by diabetes. In addition to the well-studied anti-oxidative activity, PCA demonstrated strong anti-inflammatory effects by suppressing the pro-inflammatory cytokines MCP1, VCAM1 and ICAM1, as well as increasing the phosphorylation of eNOS and Akt in the inflammatory endothelial cell model induced by the key player in diabetic endothelial dysfunction IL-1β. Upon blocking of Akt phosphorylation, p-eNOS/eNOS remained low and the inhibition of pro-inflammatory cytokines by PCA ceased. Conclusion PCA exerts protection on vascular endothelial function against inflammation through Akt/eNOS pathway, suggesting daily acquisition of PCA may be encouraged for diabetic patients.
Collapse
Affiliation(s)
- Chui Yiu Bamboo Chook
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yiu Ming Cheung
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ka Ying Ma
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fung Ping Leung
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hanyue Zhu
- School of Food Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Qingshan Jason Niu
- Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong, China
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Wing Tak Wong,
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Zhen-Yu Chen,
| |
Collapse
|
172
|
Bradley C, Berry C. Retinal microvascular function: a tractable biomarker of cardiovascular risk? Cardiovasc Res 2022; 118:3169-3170. [PMID: 36310539 DOI: 10.1093/cvr/cvac170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 01/25/2023] Open
Affiliation(s)
- Conor Bradley
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
- NHS Golden Jubilee, Agamemnon Street, Clydebank, G81 4DY, United Kingdom
| | - Colin Berry
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow G12 8TA, United Kingdom
- NHS Golden Jubilee, Agamemnon Street, Clydebank, G81 4DY, United Kingdom
| |
Collapse
|
173
|
Faisal M, Alzahraa Al-Hattab F, Mohammed Al-boinin A, Mahmoud Al-Qudah B, Waheed MA, Danjuma M. The effects of thrombocytopenia, type 2 diabetes mellitus, and endothelial dysfunction on clinical outcomes in patients with COVID-19. Qatar Med J 2022; 2023:3. [PMID: 36588776 PMCID: PMC9800283 DOI: 10.5339/qmj.2023.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/30/2022] [Indexed: 12/29/2022] Open
Abstract
Diabetes mellitus is a well-recognized contributor to increased COVID-19 severity. Endothelial dysfunction has been implicated in the pathogenesis of COVID-19, while thrombocytopenia has been identified as a potential risk factor for severe COVID-19. In this study, we evaluated the combined effect of thrombocytopenia and other markers of endothelial dysfunction on disease outcomes in patients with type 2 diabetes and active COVID-19 infection. Our aim was to risk stratify patients with COVID-19 and type 2 diabetes mellitus, which can help identify patients with high-risk features who will benefit the most from hospital admission and a high level of care. This cross-sectional study was performed after reviewing secondary data of 932 patients with COVID-19 and type 2 diabetes mellitus in the outpatient and inpatient settings across Qatar between March 1, 2020 and May 7, 2020. Univariate and multivariate analyses, with adjustment for low platelet counts, were performed for the following variables: age, hemoglobin, white blood cells (WBC), lymphocytes, monocytes, eosinophils, alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, ferritin, D-dimer, and interleukin 6. Increasing age was associated with an increased risk for death and/or intensive care unit admission in diabetic patients with COVID-19 who have low platelet counts. These findings support the evidence found in the literature and give special attention to COVID-19 patients with low platelet counts and diabetes mellites. These results can guide physicians in making clinical decisions regarding hospital admission and escalation of care during follow-up in this population of patients.
Collapse
Affiliation(s)
- Mohanad Faisal
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957,E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Fatima Alzahraa Al-Hattab
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Aisha Mohammed Al-boinin
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Bara Mahmoud Al-Qudah
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Muhammad Aamir Waheed
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| | - Mohammed Danjuma
- Hamad Medical Corporation, Doha, Qatar. E-mail: ORCID: E-mail: https://orcid.org/0000-0002-2086-2957
| |
Collapse
|
174
|
Shcheblykin DV, Bolgov AA, Pokrovskii MV, Stepenko JV, Tsuverkalova JM, Shcheblykina OV, Golubinskaya PA, Korokina LV. Endothelial dysfunction: developmental mechanisms and therapeutic strategies. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.80376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction: Every year the importance of the normal functioning of the endothelial layer of the vascular wall in maintaining the health of the body becomes more and more obvious.
The physiological role of the endothelium: The endothelium is a metabolically active organ actively involved in the regulation of hemostasis, modulation of inflammation, maintenance of hemovascular homeostasis, regulation of angiogenesis, vascular tone, and permeability.
Risk factors for the development of endothelial dysfunction: Currently, insufficient bioavailability of nitric oxide is considered the most significant risk factor for endothelial dysfunction.
Mechanisms of development of endothelial dysfunction: The genesis of endothelial dysfunction is a multifactorial process. Among various complex mechanisms, this review examines oxidative stress, inflammation, hyperglycemia, vitamin D deficiency, dyslipidemia, excess visceral fat, hyperhomocysteinemia, hyperuricemia, as well as primary genetic defect of endotheliocytes, as the most common causes in the population underlying the development of endothelial dysfunction.
Markers of endothelial dysfunction in various diseases: This article discusses the main biomarkers of endothelial dysfunction currently used, as well as promising biomarkers in the future for laboratory diagnosis of this pathology.
Therapeutic strategies: Therapeutic approaches to the endothelium in order to prevent or reduce a degree of damage to the vascular wall are briefly described.
Conclusion: Endothelial dysfunction is a typical pathological process involved in the pathogenesis of many diseases. Thus, pharmacological agents with endothelioprotective properties can provide more therapeutic benefits than a drug without such an effect.
Collapse
|
175
|
Potential Diagnostic and Monitoring Biomarkers of Obstructive Sleep Apnea-Umbrella Review of Meta-Analyses. J Clin Med 2022; 12:jcm12010060. [PMID: 36614858 PMCID: PMC9821668 DOI: 10.3390/jcm12010060] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a prevalent, underdiagnosed disease that imposes a significant impact on the health and wellbeing of patients and a financial burden on individuals, their families, and society. Development of new methods of testing other than an overnight sleep study, such as measurement of serum or plasma biomarkers, may provide an easier diagnostic process to identify patients with OSA and allow earlier initiation of treatment, which might prevent serious comorbidities. We conducted a systematic review and quality assessment of available meta-analyses regarding potential diagnostic and monitoring biomarkers of obstructive sleep apnea. A total of 14 sets of candidate biomarkers displayed differences in levels or concentrations in OSA patients compared to non-OSA controls, and decreased after OSA treatment: CRP, IL-6, TNF-α, Il-8, HCY, ICAM-1, VCAM-1, VEGF, TC, LDLc, HDLc, TG, leptin, MDA, ALT, AST, IGF-1, adiponectin, and cortisol. This review summarizes the evidence for OSA-associated potential biomarkers and demonstrates that the quality of available studies, as measured by AMSTAR2, is often low and associated with a high risk of bias.
Collapse
|
176
|
Kozlov S, Okhota S, Avtaeva Y, Melnikov I, Matroze E, Gabbasov Z. Von Willebrand factor in diagnostics and treatment of cardiovascular disease: Recent advances and prospects. Front Cardiovasc Med 2022; 9:1038030. [PMID: 36531725 PMCID: PMC9755348 DOI: 10.3389/fcvm.2022.1038030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/21/2022] [Indexed: 10/10/2023] Open
Abstract
Von Willebrand factor (VWF) is a large multimeric glycoprotein involved in hemostasis. It is essential for platelet adhesion to the subendothelium of the damaged endothelial layer at high shear rates. Such shear rates occur in small-diameter arteries, especially at stenotic sites. Moreover, VWF carries coagulation factor VIII and protects it from proteolysis in the bloodstream. Deficiency or dysfunction of VWF predisposes to bleeding. In contrast, an increase in the concentration of high molecular weight multimers (HMWM) of VWF is closely associated with arterial thrombotic events. Severe aortic stenosis (AS) or hypertrophic obstructive cardiomyopathy (HOCM) can deplete HMWM of VWF and lead to cryptogenic, gastrointestinal, subcutaneous, and mucosal bleeding. Considering that VWF facilitates primary hemostasis and a local inflammatory response at high shear rates, its dysfunction may contribute to the development of coronary artery disease (CAD) and its complications. However, current diagnostic methods do not allow for an in-depth analysis of this contribution. The development of novel diagnostic techniques, primarily microfluidic, is underway. Such methods can provide physiologically relevant assessments of VWF function at high shear rates; however, they have not been introduced into clinical practice. The development and use of agents targeting VWF interaction with the vessel wall and/or platelets may be reasonable in prevention of CAD and its complications, given the prominent role of VWF in arterial thrombosis.
Collapse
Affiliation(s)
- Sergey Kozlov
- Department of Problems of Atherosclerosis, National Medical Research Centre of Cardiology Named After Academician E.I. Chazov of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Sergey Okhota
- Department of Problems of Atherosclerosis, National Medical Research Centre of Cardiology Named After Academician E.I. Chazov of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Yuliya Avtaeva
- Laboratory of Cell Hemostasis, National Medical Research Centre of Cardiology Named After Academician E.I. Chazov of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Ivan Melnikov
- Laboratory of Cell Hemostasis, National Medical Research Centre of Cardiology Named After Academician E.I. Chazov of the Ministry of Health of the Russian Federation, Moscow, Russia
- Laboratory of Gas Exchange, Biomechanics and Barophysiology, State Scientific Center of the Russian Federation—The Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Evgeny Matroze
- Laboratory of Cell Hemostasis, National Medical Research Centre of Cardiology Named After Academician E.I. Chazov of the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Innovative Pharmacy, Medical Devices and Biotechnology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Zufar Gabbasov
- Laboratory of Cell Hemostasis, National Medical Research Centre of Cardiology Named After Academician E.I. Chazov of the Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
177
|
Zhu B, Wang Y, Yuan J, Mu Y, Chen P, Srimoragot M, Li Y, Park CG, Reutrakul S. Associations between sleep variability and cardiometabolic health: A systematic review. Sleep Med Rev 2022; 66:101688. [PMID: 36081237 DOI: 10.1016/j.smrv.2022.101688] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/13/2022]
Abstract
This review explored the associations between sleep variability and cardiometabolic health. It was performed following PRISMA guidelines. We identified 63 studies. Forty-one studies examined the association between sleep variability and body composition, with 29 examined body mass index (BMI). Thirteen studies used social jet lag (SJL), n = 30,519, with nine reporting a null association. Eight studies used variability in sleep duration (n = 33,029), with five reporting a correlation with BMI. Fourteen studies (n = 133,403) focused on overweight/obesity; significant associations with sleep variability were found in 11 (n = 120,168). Sleep variability was associated with weight gain (seven studies; n = 79,522). Twenty-three studies examined glucose outcomes. The association with hemoglobin A1c (16 studies, n = 11,755) differed depending on populations, while associations with diabetes or glucose were mixed, and none were seen with insulin resistance (five studies; n = 6416). Sixteen studies examined cardiovascular-related outcomes, with inconsistent results. Overall significant associations were found in five studies focusing on metabolic syndrome (n = 7413). In summary, sleep variability was likely associated with obesity, weight gain, and metabolic syndrome. It might be associated with hemoglobin A1c in people with type 1 diabetes. The associations with other outcomes were mixed. This review highlighted the possible association between sleep variability and cardiometabolic health.
Collapse
Affiliation(s)
- Bingqian Zhu
- School of Nursing, Shanghai Jiao Tong University, Shanghai, China
| | - Yueying Wang
- School of Nursing, Shanghai Jiao Tong University, Shanghai, China
| | - Jinjin Yuan
- School of Nursing, Shanghai Jiao Tong University, Shanghai, China
| | - Yunping Mu
- School of Nursing, Shanghai Jiao Tong University, Shanghai, China
| | - Pei Chen
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Nursing, University of Illinois Chicago, Chicago, IL, USA
| | | | - Yan Li
- School of Nursing, Hong Kong Polytechnic University, Hong Kong, China
| | - Chang G Park
- College of Nursing, University of Illinois Chicago, Chicago, IL, USA
| | - Sirimon Reutrakul
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
178
|
Rosei CA, Gaggero A, Famà F, Malerba P, Chiarini G, Nardin M, Brami V, Rossini C, Coschignano MA, Porteri E, Salvetti M, Muiesan ML, Rizzoni D, De Ciuceis C. Skin capillary alterations in patients with acute SarsCoV2 infection. J Hypertens 2022; 40:2385-2393. [PMID: 35983856 PMCID: PMC9640263 DOI: 10.1097/hjh.0000000000003271] [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: 12/09/2021] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Acute SarsCov2 infection is associated with endothelial dysfunction and 'endothelitis', which might explain systemic microvascular impairment. The presence of endothelial damage may promote vasoconstriction with organ ischemia, inflammation, tissue oedema and a procoagulant state resulting in an increase in the incidence of cardiovascular and cerebrovascular events. Microvascular thrombosis has been demonstrated in postmortem autopsy of COVID-19 patients; however, few data are available about skin capillary alterations in these patients. MATERIALS AND METHODS We evaluated skin microvascular alteration in 22 patients admitted to our hospital with SarsCov2 infection. Capillary density was evaluated by capillaroscopy in the nailfold and the dorsum of the finger in the acute phase of the disease. Capillaroscopy was repeated after 3 months (recovery phase). In addition, blood chemistry parameters and inflammatory markers were obtained during acute infection and at the recovery after 3 months. RESULTS Patients with COVID-19 showed skin microvascular complications, such as thrombosis, microhaemorrhages and neoangiogenesis, which were not detected after 3 months from the discharge. A significant reduction of capillary density in the dorsum was observed after 3 months from the acute infection (97.2 ± 5.3 vs. 75.81 ± 3.9 n/mm 2P < 0.05). A significant inverse correlation between C-reactive protein and capillary density was observed in patients with acute SarsCov2 infection ( r = 0.44, P < 0.05). Conversely a direct correlation between capillary density during the acute phase and lymphocyte number was detected ( r = 0.49, P < 0.05). CONCLUSION This is the first in-vivo evidence of skin capillary thrombosis, microhaemorrhages and angiogenesis in patients with acute SarsCov2 infection, which disappeared after 3 months, supporting the presence of endothelial dysfunction and inflammation. Capillary alterations might reflect systemic vascular effects of viral infection.
Collapse
Affiliation(s)
- Claudia Agabiti Rosei
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | - Andrea Gaggero
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | - Francesca Famà
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | - Paolo Malerba
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | - Giulia Chiarini
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | - Matteo Nardin
- Third Division of Medicine, ASST Spedali Civili, Brescia
| | - Valeria Brami
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | - Claudia Rossini
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | | | - Enzo Porteri
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | - Massimo Salvetti
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | - Maria Lorenza Muiesan
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| | - Damiano Rizzoni
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
- Division of Medicine, ASST Spedali Civili di Brescia, Montichiari, Italy
| | - Carolina De Ciuceis
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia
| |
Collapse
|
179
|
Nayeem MA, Hanif A, Geldenhuys WJ, Agba S. Crosstalk between adenosine receptors and CYP450-derived oxylipins in the modulation of cardiovascular, including coronary reactive hyperemic response. Pharmacol Ther 2022; 240:108213. [PMID: 35597366 DOI: 10.1016/j.pharmthera.2022.108213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
Abstract
Adenosine is a ubiquitous endogenous nucleoside or autacoid that affects the cardiovascular system through the activation of four G-protein coupled receptors: adenosine A1 receptor (A1AR), adenosine A2A receptor (A2AAR), adenosine A2B receptor (A2BAR), and adenosine A3 receptor (A3AR). With the rapid generation of this nucleoside from cellular metabolism and the widespread distribution of its four G-protein coupled receptors in almost all organs and tissues of the body, this autacoid induces multiple physiological as well as pathological effects, not only regulating the cardiovascular system but also the central nervous system, peripheral vascular system, and immune system. Mounting evidence shows the role of CYP450-enzymes in cardiovascular physiology and pathology, and the genetic polymorphisms in CYP450s can increase susceptibility to cardiovascular diseases (CVDs). One of the most important physiological roles of CYP450-epoxygenases (CYP450-2C & CYP2J2) is the metabolism of arachidonic acid (AA) and linoleic acid (LA) into epoxyeicosatrienoic acids (EETs) and epoxyoctadecaenoic acid (EpOMEs) which generally involve in vasodilation. Like an increase in coronary reactive hyperemia (CRH), an increase in anti-inflammation, and cardioprotective effects. Moreover, the genetic polymorphisms in CYP450-epoxygenases will change the beneficial cardiovascular effects of metabolites or oxylipins into detrimental effects. The soluble epoxide hydrolase (sEH) is another crucial enzyme ubiquitously expressed in all living organisms and almost all organs and tissues. However, in contrast to CYP450-epoxygenases, sEH converts EETs into dihydroxyeicosatrienoic acid (DHETs), EpOMEs into dihydroxyoctadecaenoic acid (DiHOMEs), and others and reverses the beneficial effects of epoxy-fatty acids leading to vasoconstriction, reducing CRH, increase in pro-inflammation, increase in pro-thrombotic and become less cardioprotective. Therefore, polymorphisms in the sEH gene (Ephx2) cause the enzyme to become overactive, making it more vulnerable to CVDs, including hypertension. Besides the sEH, ω-hydroxylases (CYP450-4A11 & CYP450-4F2) derived metabolites from AA, ω terminal-hydroxyeicosatetraenoic acids (19-, 20-HETE), lipoxygenase-derived mid-chain hydroxyeicosatetraenoic acids (5-, 11-, 12-, 15-HETEs), and the cyclooxygenase-derived prostanoids (prostaglandins: PGD2, PGF2α; thromboxane: Txs, oxylipins) are involved in vasoconstriction, hypertension, reduction in CRH, pro-inflammation and cardiac toxicity. Interestingly, the interactions of adenosine receptors (A2AAR, A1AR) with CYP450-epoxygenases, ω-hydroxylases, sEH, and their derived metabolites or oxygenated polyunsaturated fatty acids (PUFAs or oxylipins) is shown in the regulation of the cardiovascular functions. In addition, much evidence demonstrates polymorphisms in CYP450-epoxygenases, ω-hydroxylases, and sEH genes (Ephx2) and adenosine receptor genes (ADORA1 & ADORA2) in the human population with the susceptibility to CVDs, including hypertension. CVDs are the number one cause of death globally, coronary artery disease (CAD) was the leading cause of death in the US in 2019, and hypertension is one of the most potent causes of CVDs. This review summarizes the articles related to the crosstalk between adenosine receptors and CYP450-derived oxylipins in vascular, including the CRH response in regular salt-diet fed and high salt-diet fed mice with the correlation of heart perfusate/plasma oxylipins. By using A2AAR-/-, A1AR-/-, eNOS-/-, sEH-/- or Ephx2-/-, vascular sEH-overexpressed (Tie2-sEH Tr), vascular CYP2J2-overexpressed (Tie2-CYP2J2 Tr), and wild-type (WT) mice. This review article also summarizes the role of pro-and anti-inflammatory oxylipins in cardiovascular function/dysfunction in mice and humans. Therefore, more studies are needed better to understand the crosstalk between the adenosine receptors and eicosanoids to develop diagnostic and therapeutic tools by using plasma oxylipins profiles in CVDs, including hypertensive cases in the future.
Collapse
Affiliation(s)
- Mohammed A Nayeem
- Faculties of the Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA.
| | - Ahmad Hanif
- Faculties of the Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Werner J Geldenhuys
- Faculties of the Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Stephanie Agba
- Graduate student, Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
180
|
Badour MI, Stone RM, Parikh KS, Lester NJ, Meloche OL, Wulterkens RN, Bain AR. Circulating Notch1 in response to altered vascular wall shear stress in adults. Exp Physiol 2022; 107:1426-1431. [PMID: 36116111 DOI: 10.1113/ep090749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS What is the central question of this study? Is the plasma concentration of Notch1 extracellular domain altered in response to decreased and increased vascular wall shear stress in the forearm in humans? What is the main finding and its importance? Notch1 extracellular domain is increased with acute increases in antegrade shear rate but does not change with 20 min of decreased shear rate caused by distal forearm occlusion. A novel and integral endothelial mechanosensor in humans that can help explain vascular endothelial adjustments in response to increases in antegrade shear stress was characterized. ABSTRACT Notch1 has been proposed as a novel endothelial mechanosensor that is central for signalling adjustments in response to changes in vascular wall shear stress. However, there remains no controlled in vivo study in humans. Accordingly, we sought to address the question of whether plasma concentrations of Notch1 extracellular domain (ECD) is altered in response to transient changes in vascular wall shear stress. In 10 young healthy adults (6M/4F), alterations in shear stress were induced by supra-systolic cuff inflation around the wrist. The opposite arm was treated as a time control with no wrist cuff inflation. Plasma was collected from an antecubital vein of both arms at baseline, 20 min of wrist cuff inflation (low shear), as well as 1-2 min (high shear) and 15 min following (recovery) wrist cuff release. The Notch1 ECD was quantified using a commercially available ELISA. Duplex ultrasound was used to confirm alterations in shear stress. In the experimental arm, concentrations of Notch1 ECD remained statistically similar to baseline at all time points except for immediately following cuff release where it was elevated by ∼50% (P = 0.033), coinciding with the condition of high antegrade shear rate. Concentrations of Notch1 ECD remained unchanged in the control arm through all time points. These data indicate that Notch1 is a viable biomarker for quantifying mechanotransduction in response to increased shear stress in humans, and it may underlie the vascular adaptations or mal-adaptations associated with conditions that impact antegrade shear.
Collapse
Affiliation(s)
- Matthew I Badour
- Faculty of Human Kinetics, University of Windsor, Ontario, Canada
| | - Rachel M Stone
- Faculty of Human Kinetics, University of Windsor, Ontario, Canada
| | | | | | - Olivia L Meloche
- Faculty of Human Kinetics, University of Windsor, Ontario, Canada
| | | | - Anthony R Bain
- Faculty of Human Kinetics, University of Windsor, Ontario, Canada
| |
Collapse
|
181
|
Consequences of COVID-19 on the cardiovascular and renal systems. Sleep Med 2022; 100:31-38. [PMID: 35994936 PMCID: PMC9345655 DOI: 10.1016/j.sleep.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 01/11/2023]
|
182
|
Bockus L, Kim F. Coronary endothelial dysfunction: from pathogenesis to clinical implications. Open Heart 2022; 9:e002200. [PMID: 36600608 PMCID: PMC9743399 DOI: 10.1136/openhrt-2022-002200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022] Open
Abstract
Endothelial dysfunction (ED) has a substantial role in the pathogenesis of atherosclerosis and other vascular diseases. Multiple risk factors, including smoking, hyperlipiadaemia and diabetes, can have associated ED, which is correlated with cardiac events. Measurement of coronary artery endothelial function requires the use of invasive techniques to assess both epicardial coronary artery and microvascular beds. Peripheral vascular techniques and endothelial biomarkers can be used to indirectly assess coronary ED. In this review of coronary artery ED, we discuss the current state of the field, the techniques used to measure ED and its clinical implications.
Collapse
Affiliation(s)
- Lee Bockus
- Deparment of Medicine, University of Washington, Seattle, Washington, USA
| | - Francis Kim
- Deparment of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
183
|
Van Nguyen D, Nguyen TLL, Jin Y, Kim L, Myung CS, Heo KS. 6′-Sialylactose abolished lipopolysaccharide-induced inflammation and hyper-permeability in endothelial cells. Arch Pharm Res 2022; 45:836-848. [DOI: 10.1007/s12272-022-01415-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022]
|
184
|
Circulating Serum Cystatin C as an Independent Risk Biomarker for Vascular Endothelial Dysfunction in Patients with COVID-19-Associated Multisystem Inflammatory Syndrome in Children (MIS-C): A Prospective Observational Study. Biomedicines 2022; 10:biomedicines10112956. [PMID: 36428524 PMCID: PMC9687890 DOI: 10.3390/biomedicines10112956] [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: 11/01/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Multisystem inflammatory syndrome in children (MIS-C) is a new clinical entity that has emerged in the context of the COVID-19 pandemic. Despite the less severe course of the disease, varying degrees of cardiovascular events may occur in MIS-C; however, data on vascular changes occurring in MIS-C are still lacking. Endothelial dysfunction (ED) is thought to be one of the key risk factors contributing to MIS-C. BACKGROUND We conducted a prospective observational study. We investigated possible manifestations of cardiac and endothelial involvement in MIS-C after the treatment of the acute stage and potential predictive biomarkers in patients with MIS-C. METHODS Twenty-seven consecutive pediatric subjects (≥9 years), at least three months post-treated MIS-C of varying severity, in a stable condition, and twenty-three age- and sex-matched healthy individuals (HI), were enrolled. A combined non-invasive diagnostic approach was used to assess endothelial function as well as markers of organ damage using cardiac examination and measurement of the reactive hyperemia index (RHI), by recording the post- to pre-occlusion pulsatile volume changes and biomarkers related to ED and cardiac disease. RESULTS MIS-C patients exhibited a significantly lower RHI (indicative of more severe ED) than those in HI (1.32 vs. 1.80; p = 0.001). The cutoff of RHI ≤ 1.4 was independently associated with a higher cardiovascular risk. Age and biomarkers significantly correlated with RHI, while serum cystatin C (Cys C) levels were independently associated with a diminished RHI, suggesting Cys C as a surrogate marker of ED in MIS-C. CONCLUSIONS Patients after MIS-C display evidence of ED, as shown by a diminished RHI and altered endothelial biomarkers. Cys C was identified as an independent indicator for the development of cardiovascular disease. The combination of these factors has the potential to better predict the cardiovascular consequences of MIS-C. Our study suggests that ED may be implicated in the pathophysiology of this disease.
Collapse
|
185
|
Is It All about Endothelial Dysfunction? Focusing on the Alteration in Endothelial Integrity as a Key Determinant of Different Pathological Mechanisms. Biomedicines 2022; 10:biomedicines10112757. [PMID: 36359277 PMCID: PMC9687329 DOI: 10.3390/biomedicines10112757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
|
186
|
Tain YL, Hsu CN. Cardiovascular Risks of Hypertension: Lessons from Children with Chronic Kidney Disease. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1650. [PMID: 36360378 PMCID: PMC9688449 DOI: 10.3390/children9111650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023]
Abstract
Hypertension is the most common complication of chronic kidney disease (CKD) in children, having a strong association with subsequential cardiovascular disease (CVD). In pediatric CKD, a considerable percentage of children with hypertension are undiagnosed or undertreated. Prior research has evaluated structural and functional markers of subclinical CVD and biomarkers in adults with CKD, while ideal biomarkers in pediatrics are still insufficiently studied. The ultimate goal of this review is to summarize what is currently known about state of hypertension, cardiovascular risk factors, and potential CVD markers/biomarkers in children with pre-dialysis CKD. We discuss omics-related biomarkers and the pathophysiologic processes of endothelial dysfunction, kidney injury, oxidative stress and inflammation that are classified by specific biomarkers. Moreover, we illustrate the existing challenges and highlight the paucity of pediatric CKD research to evaluate these CVD biomarkers for future clinical pediatric practice. Thus, achieving clinical utility of CVD biomarkers for use in pediatric CKD remains a significant challenge requiring additional efforts.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
187
|
Basilio E, Chen R, Fernandez AC, Padula AM, Robinson JF, Gaw SL. Wildfire Smoke Exposure during Pregnancy: A Review of Potential Mechanisms of Placental Toxicity, Impact on Obstetric Outcomes, and Strategies to Reduce Exposure. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13727. [PMID: 36360613 PMCID: PMC9657128 DOI: 10.3390/ijerph192113727] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/07/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023]
Abstract
Climate change is accelerating the intensity and frequency of wildfires globally. Understanding how wildfire smoke (WS) may lead to adverse pregnancy outcomes and alterations in placental function via biological mechanisms is critical to mitigate the harms of exposure. We aim to review the literature surrounding WS, placental biology, biological mechanisms underlying adverse pregnancy outcomes as well as interventions and strategies to avoid WS exposure in pregnancy. This review includes epidemiologic and experimental laboratory-based studies of WS, air pollution, particulate matter (PM), and other chemicals related to combustion in relation to obstetric outcomes and placental biology. We summarized the available clinical, animal, and placental studies with WS and other combustion products such as tobacco, diesel, and wood smoke. Additionally, we reviewed current recommendations for prevention of WS exposure. We found that there is limited data specific to WS; however, studies on air pollution and other combustion sources suggest a link to inflammation, oxidative stress, endocrine disruption, DNA damage, telomere shortening, epigenetic changes, as well as metabolic, vascular, and endothelial dysregulation in the maternal-fetal unit. These alterations in placental biology contribute to adverse obstetric outcomes that disproportionally affect the most vulnerable. Limiting time outdoors, wearing N95 respirator face masks and using high quality indoor air filters during wildfire events reduces exposure to related environmental exposures and may mitigate morbidities attributable to WS.
Collapse
Affiliation(s)
- Emilia Basilio
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Rebecca Chen
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | | | - Amy M. Padula
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Joshua F. Robinson
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
188
|
Choi KA, Kim JH, Ryu K, Kaushik N. Current Nanomedicine for Targeted Vascular Disease Treatment: Trends and Perspectives. Int J Mol Sci 2022; 23:12397. [PMID: 36293254 PMCID: PMC9604340 DOI: 10.3390/ijms232012397] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/04/2022] [Accepted: 10/14/2022] [Indexed: 12/19/2022] Open
Abstract
Nanotechnology has been developed to deliver cargos effectively to the vascular system. Nanomedicine is a novel and effective approach for targeted vascular disease treatment including atherosclerosis, coronary artery disease, strokes, peripheral arterial disease, and cancer. It has been well known for some time that vascular disease patients have a higher cancer risk than the general population. During atherogenesis, the endothelial cells are activated to increase the expression of adhesion molecules such as Intercellular Adhesion Molecule 1 (ICAM-1), Vascular cell adhesion protein 1 (VCAM-1), E-selectin, and P-selectin. This biological activation of endothelial cells gives a targetability clue for nanoparticle strategies. Nanoparticle formation has a passive targeting pathway due to the increased adhesion molecule expression on the cell surface as well as increased cell activation. In addition, the VCAM-1-targeting peptide has been widely used to target the inflamed endothelial cells. Biomimetic nanoparticles using platelet and leukocyte membrane fragment strategies have been promising techniques for targeted vascular disease treatment. Cyclodextrin, a natural oligosaccharide with a hydrophobic cavity, increase the solubility of cholesterol crystals at the atherosclerotic plaque site and has been used to deliver the hydrophobic drug statin as a therapeutic in a targeted manner. In summary, nanoparticles decorated with various targeting molecules will be an effective and promising strategy for targeted vascular disease treatment.
Collapse
Affiliation(s)
- Kyung-A Choi
- National Institute of Medical Welfare, Kangnam University, Yongin 16979, Korea
| | - June Hyun Kim
- Department of Biotechnology, The University of Suwon, Suwon 18323, Korea
| | - Kitae Ryu
- Department of Biotechnology, The University of Suwon, Suwon 18323, Korea
| | - Neha Kaushik
- Department of Biotechnology, The University of Suwon, Suwon 18323, Korea
| |
Collapse
|
189
|
Willems LH, Thijssen DHJ, Groh LA, Kooijman NI, Ten Cate H, Spronk HMH, Donders ART, van der Vijver-Coppen RJ, van Hoek F, Nagy M, Reijnen MMPJ, Warlé MC. Dual pathway inhibition as compared to acetylsalicylic acid monotherapy in relation to endothelial function in peripheral artery disease, a phase IV clinical trial. Front Cardiovasc Med 2022; 9:979819. [PMID: 36277757 PMCID: PMC9583941 DOI: 10.3389/fcvm.2022.979819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
Objective Dual pathway inhibition (DPI) by combining acetylsalicylic acid (ASA) with low-dose rivaroxaban has been shown to reduce cardiovascular events in patients with peripheral arterial disease (PAD) when compared to ASA monotherapy. A potential explanation is that inhibition of factor Xa improves endothelial function through crosstalk between coagulation and inflammatory pathways, subsequently attenuating the occurrence of cardiovascular events. We hypothesize that the addition of rivaroxaban to ASA in PAD patients leads to improved endothelial function. Design An investigator-initiated, multicentre trial investigating the effect of DPI on endothelial function. Methods Patients, diagnosed with PAD, were enrolled in two cohorts: cohort A (Rutherford I-III) and cohort B (Rutherford IV-VI). Participants received ASA monotherapy for a 4-weeks run-in period, followed by 12 weeks of DPI. Macro- and microvascular endothelial dysfunction were studied by measuring carotid artery reactivity upon sympathetic stimulus and by measuring plasma endothelin-1 concentrations, respectively. All measurements were performed during the use of ASA (baseline) and after 12 weeks of DPI. Results 159 PAD patients (111 cohort A, 48 cohort B) were enrolled. Twenty patients discontinued study drugs early. Carotid artery constriction upon sympathetic stimulation at baseline (ASA) and after 12 weeks of DPI was similar in the total group, 22.0 vs. 22.7% (p = 1.000), and in the subgroups (Cohort A 22.6 vs. 23.7%, p = 1.000; cohort B 20.5 vs. 20.5%, p = 1.000), respectively. The mean concentration of plasma endothelin-1 at baseline and after 12 weeks of DPI did not differ, 1.70 ± 0.5 vs. 1.66 ± 0.64 pmol/L (p = 0.440) in the total group, 1.69 ± 0.59 vs. 1.62 ± 0.55 pmol/L in cohort A (p = 0.202), and 1.73 ± 0.53 vs. 1.77 ± 0.82 pmol/L in cohort B (p = 0.682), respectively. Conclusion Macro- and microvascular endothelial dysfunction, as reflected by carotid artery reactivity and plasma endothelin-1 concentrations, are not influenced in PAD patients by addition of low-dose rivaroxaban to ASA monotherapy for 12 weeks. Trial registration https://clinicaltrials.gov/ct2/show/NCT04218656.
Collapse
Affiliation(s)
- Loes H. Willems
- Department of Surgery, Radboud University Medical Center, Nijmegen, Netherlands,*Correspondence: Loes H. Willems
| | - Dick H. J. Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands,Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Laszlo A. Groh
- Department of Surgery, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nina I. Kooijman
- Department of Surgery, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hugo Ten Cate
- Departments of Internal Medicine and Biochemistry, Maastricht University Medical Center (MUMC) and Cardiovascular Research Institute Maastricht (CARIM) School for Cardiovascular Diseases, Maastricht, Netherlands,Center for Thrombosis and Haemostasis, Gutenberg University Medical Center, Mainz, Germany
| | - Henri M. H. Spronk
- Departments of Internal Medicine and Biochemistry, Maastricht University Medical Center (MUMC) and Cardiovascular Research Institute Maastricht (CARIM) School for Cardiovascular Diseases, Maastricht, Netherlands
| | - A. Rogier T. Donders
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Frank van Hoek
- Department of Surgery, Radboud University Medical Center, Nijmegen, Netherlands
| | - Magdolna Nagy
- Departments of Internal Medicine and Biochemistry, Maastricht University Medical Center (MUMC) and Cardiovascular Research Institute Maastricht (CARIM) School for Cardiovascular Diseases, Maastricht, Netherlands
| | - Michel M. P. J. Reijnen
- Department of Surgery, Rijnstate Hospital, Arnhem, Netherlands,Multi-Modality Medical Imaging Group, Faculty of Science and Technology, University of Twente, Enschede, Netherlands
| | - Michiel C. Warlé
- Department of Surgery, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
190
|
Coscas R, Dubosq M, Charton J, El Batti S, Gaudric J, Koskas F, Chiche L, Couture T, Davaine JM, Castier Y, Cerceau P, EL Hajjam M, Samb P, Beauchet A, Grimaldi L, Javerliat I, Goeau-Brissonniere O, Alsac JM, Onorati I, Martinod E, Desgranges P, Touma J, Cochennec F, Pellenc Q, Julia P, Coggia M. Thrombosis of Medium-Sized and Large Arteries During Covid-19 Infection: Results of the COVIVASC Study. Ann Vasc Surg 2022; 86:35-42. [PMID: 35780947 PMCID: PMC9242891 DOI: 10.1016/j.avsg.2022.04.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/15/2022] [Accepted: 04/15/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND COVID-19 infection is associated not only with venous thromboses but also with arterial thromboses (COV-ATs) in relation with an endothelial dysfunction, a coagulopathy and rhythm disorders. The incidence, the topography, and the prognosis of COV-ATs remain poorly known. The objective of this study was to report the overall experience of the Greater Paris University Hospitals (Assistance Publique - Hopitaux de Paris, AP-HP) during the first pandemic wave of COVID-19 infection. METHODS After approval by the ethics committee, a study using the AP-HP clinical data warehouse was carried out between March and May 2020. Overall, 124,609 patients had a polymerase chain reaction for COVID-19 in our hospitals, of which 25,345 were positive. From 20,710 exploitable stays, patients tested positive for COVID who presented an episode of acute COV-AT (except coronary and intracranial arteries) were selected on the basis of the French medical classification for clinical procedures codes. The data are presented as absolute values with percentages and/or means with standard deviation. RESULTS Over the studied period, 60 patients (aged 71±14 years, 42 men) presented a COV-AT at the time of their hospitalization, an incidence of 0.2%. The arterial complication occurred 3±7 days after the COVID infection and was inaugural in 30% of the cases (n = 18). The sites of COV-AT were the lower extremities (n = 35%, 58%), the abdominal aorta (n = 10%, 17%), the thoracic aorta (n = 7%, 12%), the upper limbs (n = 7%, 12%), the cerebral arteries (n = 7%, 12%), the digestive arteries (n = 6%, 10%), the renal arteries (n = 2%, 3%), and the ophthalmic artery (n = 1%, 2%). Multiple COV-ATs were observed in 13 patients (22%). At the time of diagnosis, 20 (33%) patients were in intensive care, including six (10%) patients who were intubated. On computed tomography angiography, COVID lesions were classified as moderate and severe in 25 (42%) and 21 (35%) cases, respectively. Revascularization was attempted in 27 patients (45%), by open surgery in 16 cases, using endovascular techniques in 8 cases and with a hybrid approach in three cases. Six patients (22%) required reinterventions. The duration of hospitalization was 12±9 days. Early mortality (in-hospital or at 30 days) was 30% (n = 18). Nine (15%) patients presented severe nonlethal ischemic complications. CONCLUSIONS Arterial involvement is rare during COVID-19 infection. The aorta and the arteries of the limbs are the privileged sites. The morbi-mortality of these patients is high. Future studies will have to determine if the systematization of anticoagulation therapy decreases the incidence and the severity of the condition.
Collapse
Affiliation(s)
- Raphael Coscas
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France,Correspondence to: Raphaël Coscas, Department of Vascular Surgery, Ambroise Paré University Hospital, 9 avenue Charles de Gaulle, 92104 Boulogne Cedex, France
| | - Maxime Dubosq
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Johanna Charton
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Salma El Batti
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Julien Gaudric
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Fabien Koskas
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Laurent Chiche
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Thibault Couture
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Jean-Michel Davaine
- Department of Vascular and Endovascular Surgery–Tertiary Aortic Center, Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris and Sorbonne Médecine Universités, 75013 Paris, France
| | - Yves Castier
- Department of Vascular Surgery, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France and Paris-Diderot University, Paris, France
| | - Pierre Cerceau
- Department of Vascular Surgery, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France and Paris-Diderot University, Paris, France
| | - Mostafa EL Hajjam
- Department of Radiology, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France
| | - Patricia Samb
- Clinical Research Unit, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France,University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Alain Beauchet
- Clinical Research Unit, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France,University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Lamiae Grimaldi
- Clinical Research Unit, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France,University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Isabelle Javerliat
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Olivier Goeau-Brissonniere
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| | - Jean-Marc Alsac
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Ilaria Onorati
- Department of Thoracic and Vascular Surgery, Hôpitaux Universitaires Paris Seine-Saint-Denis, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, and Université Paris 13, Sorbonne Paris Cité, UFR Santé, Médecine et Biologie Humaine, Bobigny, France
| | - Emmanuel Martinod
- Department of Thoracic and Vascular Surgery, Hôpitaux Universitaires Paris Seine-Saint-Denis, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, and Université Paris 13, Sorbonne Paris Cité, UFR Santé, Médecine et Biologie Humaine, Bobigny, France
| | - Pascal Desgranges
- Department of Vascular Surgery, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France,Paris-Est Créteil Faculty of Medicine, Créteil, France
| | - Joseph Touma
- Department of Vascular Surgery, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France,Paris-Est Créteil Faculty of Medicine, Créteil, France
| | - Frédéric Cochennec
- Department of Vascular Surgery, Henri Mondor University Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France,Paris-Est Créteil Faculty of Medicine, Créteil, France
| | - Quentin Pellenc
- Department of Vascular Surgery, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France and Paris-Diderot University, Paris, France
| | - Pierre Julia
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Marc Coggia
- Department of Vascular Surgery, Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France,UMR 1018, Inserm-Paris11 - CESP, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Paul Brousse Hospital, 94807 Villejuif, France
| |
Collapse
|
191
|
Lakshmanan AP, Murugesan S, Al Khodor S, Terranegra A. The potential impact of a probiotic: Akkermansia muciniphila in the regulation of blood pressure—the current facts and evidence. Lab Invest 2022; 20:430. [PMID: 36153618 PMCID: PMC9509630 DOI: 10.1186/s12967-022-03631-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022]
Abstract
Akkermansia muciniphila (A. muciniphila) is present in the human gut microbiota from infancy and gradually increases in adulthood. The potential impact of the abundance of A. muciniphila has been studied in major cardiovascular diseases including elevated blood pressure or hypertension (HTN). HTN is a major factor in premature death worldwide, and approximately 1.28 billion adults aged 30–79 years have hypertension. A. muciniphila is being considered a next-generation probiotic and though numerous studies had highlighted the positive role of A. muciniphila in lowering/controlling the HTN, however, few studies had highlighted the negative impact of increased abundance of A. muciniphila in the management of HTN. Thus, in the review, we aimed to discuss the current facts, evidence, and controversy about the role of A. muciniphila in the pathophysiology of HTN and its potential effect on HTN management/regulation, which could be beneficial in identifying the drug target for the management of HTN.
Collapse
|
192
|
Kollár B, Blaho A, Valovičová K, Poddaný M, Valkovič P, Straka I, Turčáni P, Šiarnik P. Impairment of endothelial function in Parkinson's disease. BMC Res Notes 2022; 15:284. [PMID: 36064624 PMCID: PMC9446825 DOI: 10.1186/s13104-022-06176-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE There are conflicting data regarding the relationship between Parkinson's disease (PD) and the atherosclerotic process. This study aimed to compare endothelial function in patients with PD and matched controls. In PD subjects, we searched for factors contributing to endothelial dysfunction as well. Traditional vascular risk factors, PD characteristics, and PD medication were considered. RESULTS We prospectively enrolled 41 patients with PD and 41 controls matched for age, sex, body mass index, and vascular risk factors. Endothelial function (EF) was assessed using peripheral arterial tonometry (EndoPAT 2000 device) and expressed as reperfusion hyperemia index (RHI). Clinical characteristics including PD medication were recorded. RHI was non-significantly lower in the PD group than in controls (1.8 ± 0.5 vs. 1.9 ± 0.5, p = 0.478). In PD patients, in linear regression analysis, smoking (beta = -0.453, p = 0.008) and use of dopamine agonists (beta = -0.365, p = 0.030) were significant contributors in a model predicting RHI. Despite non-significant differences in endothelial dysfunction between PD patients and controls, our results suggest an association between smoking, dopamine agonists, and impaired EF in PD patients. The small sample size, as well as the absence of an extended search for traditional and non-traditional vascular risk factors, are the most important factors limiting the interpretation of the current results.
Collapse
Affiliation(s)
- Branislav Kollár
- 1st Department of Neurology, Faculty of Medicine, Comenius University, Mickiewiczova 13, 813 69, Bratislava, Slovakia
| | - Andrej Blaho
- AB Neuro, Jilemnickeho 547/10, 911 01, Trencin, Slovakia
| | - Katarína Valovičová
- 1st Department of Neurology, Faculty of Medicine, Comenius University, Mickiewiczova 13, 813 69, Bratislava, Slovakia
| | - Michal Poddaný
- Department of Neurology, General Hospital, Palucanska 25, 031 23, Liptovsky Mikulas, Slovakia
| | - Peter Valkovič
- 2nd Department of Neurology, Faculty of Medicine, Comenius University, Limbova 5, 83305, Bratislava, Slovakia.,Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Sienkiewiczova 1, 813 71, Bratislava, Slovakia
| | - Igor Straka
- 2nd Department of Neurology, Faculty of Medicine, Comenius University, Limbova 5, 83305, Bratislava, Slovakia
| | - Peter Turčáni
- 1st Department of Neurology, Faculty of Medicine, Comenius University, Mickiewiczova 13, 813 69, Bratislava, Slovakia
| | - Pavel Šiarnik
- 1st Department of Neurology, Faculty of Medicine, Comenius University, Mickiewiczova 13, 813 69, Bratislava, Slovakia.
| |
Collapse
|
193
|
Lim P, Bleich D. Revisiting cardiovascular risk reduction in type 2 diabetes and dyslipidemia. INTERNATIONAL JOURNAL OF CARDIOLOGY CARDIOVASCULAR RISK AND PREVENTION 2022; 14:200141. [PMID: 36060284 PMCID: PMC9434405 DOI: 10.1016/j.ijcrp.2022.200141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/10/2022] [Accepted: 06/16/2022] [Indexed: 11/25/2022]
|
194
|
Identification of Differentially Expressed Genes and Prediction of Expression Regulation Networks in Dysfunctional Endothelium. Genes (Basel) 2022; 13:genes13091563. [PMID: 36140731 PMCID: PMC9498925 DOI: 10.3390/genes13091563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/23/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
The detection of early coronary atherosclerosis (ECA) is still a challenge and the mechanism of endothelial dysfunction remains unclear. In the present study, we aimed to identify differentially expressed genes (DEGs) and the regulatory network of miRNAs as well as TFs in dysfunctional endothelium to elucidate the possible pathogenesis of ECA and find new potential markers. The GSE132651 data set of the GEO database was used for the bioinformatic analysis. Principal component analysis (PCA), the identification of DEGs, correlation analysis between significant DEGs, the prediction of regulatory networks of miRNA and transcription factors (TFs), the validation of the selected significant DEGs, and the receiver operating characteristic (ROC) curve analysis as well as area under the curve (AUC) values were performed. We identified ten genes with significantly upregulated signatures and thirteen genes with significantly downregulated signals. Following this, we found twenty-two miRNAs regulating two or more DEGs based on the miRNA–target gene regulatory network. TFs with targets ≥ 10 were E2F1, RBPJ, SSX3, MMS19, POU3F3, HOXB5, and KLF4. Finally, three significant DEGs (TOX, RasGRP3, TSPAN13) were selected to perform validation experiments. Our study identified TOX, RasGRP3, and TSPAN13 in dysfunctional endothelium and provided potential biomarkers as well as new insights into the possible molecular mechanisms of ECA.
Collapse
|
195
|
Luo JY, Cheng CK, He L, Pu Y, Zhang Y, Lin X, Xu A, Lau CW, Tian XY, Ma RCW, Jo H, Huang Y. Endothelial UCP2 Is a Mechanosensitive Suppressor of Atherosclerosis. Circ Res 2022; 131:424-441. [PMID: 35899624 PMCID: PMC9390236 DOI: 10.1161/circresaha.122.321187] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Inflamed endothelial cells (ECs) trigger atherogenesis, especially at arterial regions experiencing disturbed blood flow. UCP2 (Uncoupling protein 2), a key mitochondrial antioxidant protein, improves endothelium-dependent relaxation in obese mice. However, whether UCP2 can be regulated by shear flow is unknown, and the role of endothelial UCP2 in regulating inflammation and atherosclerosis remains unclear. This study aims to investigate the mechanoregulation of UCP2 expression in ECs and the effect of UCP2 on endothelial inflammation and atherogenesis. METHODS In vitro shear stress simulation system was used to investigate the regulation of UCP2 expression by shear flow. EC-specific Ucp2 knockout mice were used to investigate the role of UCP2 in flow-associated atherosclerosis. RESULTS Shear stress experiments showed that KLF2 (Krüppel-like factor 2) mediates fluid shear stress-dependent regulation of UCP2 expression in human aortic and human umbilical vein ECs. Unidirectional shear stress, statins, and resveratrol upregulate whereas oscillatory shear stress and proinflammatory stimuli inhibit UCP2 expression through altered KLF2 expression. KLF2 directly binds to UCP2 promoter to upregulate its transcription in human umbilical vein ECs. UCP2 knockdown induced expression of genes involved in proinflammatory and profibrotic signaling, resulting in a proatherogenic endothelial phenotype. EC-specific Ucp2 deletion promotes atherogenesis and collagen production. Additionally, we found endothelial Ucp2 deficiency aggravates whereas adeno-associated virus-mediated EC-Ucp2 overexpression inhibits carotid atherosclerotic plaque formation in disturbed flow-enhanced atherosclerosis mouse model. RNA-sequencing analysis revealed FoxO1 (forkhead box protein O1) as the major proinflammatory transcriptional regulator activated by UCP2 knockdown, and FoxO1 inhibition reduced vascular inflammation and disturbed flow-enhanced atherosclerosis. We showed further that UCP2 level is critical for phosphorylation of AMPK (AMP-activated protein kinase), which is required for UCP2-induced inhibition of FoxO1. CONCLUSIONS Altogether, our studies uncover that UCP2 is novel mechanosensitive gene under the control of fluid shear stress and KLF2 in ECs. UCP2 expression is critical for endothelial proinflammatory response and atherogenesis. Therapeutic strategies enhancing UCP2 level may have therapeutic potential against atherosclerosis.
Collapse
Affiliation(s)
- Jiang-Yun Luo
- Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China (J.-Y.L.)
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
| | - Chak Kwong Cheng
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| | - Lei He
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| | - Yujie Pu
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China (Y.Z.)
| | - Xiao Lin
- School of Life Sciences (X.L.), Chinese University of Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, China (A.X.)
| | - Chi Wai Lau
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
| | - Xiao Yu Tian
- Heart and Vascular Institute, Shenzhen Research Institute and School of Biomedical Sciences (J.-Y.L., C.K.C., L.H., Y.P., C.W.L., X.Y.T.), Chinese University of Hong Kong, China
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics (R.C.W.M.), Chinese University of Hong Kong, China
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta (H.J.)
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, China (C.K.C., L.H., Y.P., Y.H.)
| |
Collapse
|
196
|
Davis KM, Petersen KS, Bowen KJ, Jones PJH, Taylor CG, Zahradka P, Letourneau K, Perera D, Wilson A, Wagner PR, Kris-Etherton PM, West SG. Effects of Diets Enriched with Conventional or High-Oleic Canola Oils on Vascular Endothelial Function: A Sub-Study of the Canola Oil Multi-Centre Intervention Trial 2 (COMIT-2), a Randomized Crossover Controlled Feeding Study. Nutrients 2022; 14:nu14163404. [PMID: 36014910 PMCID: PMC9416081 DOI: 10.3390/nu14163404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Partial replacement of saturated fatty acids (SFA) with unsaturated fatty acids is recommended to reduce cardiovascular disease (CVD) risk. Monounsaturated fatty acids (MUFA), including oleic acid, are associated with lower CVD risk. Measurement of flow-mediated dilation of the brachial artery (FMD) is the gold standard for measuring endothelial function and predicts CVD risk. This study examined the effect of partially replacing SFA with MUFA from conventional canola oil and high-oleic acid canola oil on FMD. Participants (n = 31) with an elevated waist circumference plus ≥1 additional metabolic syndrome criterion completed FMD measures as part of the Canola Oil Multi-Centre Intervention Trial 2 (COMIT-2), a multi-center, double-blind, three-period crossover, controlled feeding randomized trial. Diet periods were 6 weeks, separated by ≥4-week washouts. Experimental diets were provided during all feeding periods. Diets only differed by the fatty acid profile of the oils: canola oil (CO; 17.5% energy from MUFA, 9.2% polyunsaturated fatty acids (PUFA), 6.6% SFA), high-oleic acid canola oil (HOCO; 19.1% MUFA, 7.0% PUFA, 6.4% SFA), and a control oil blend (CON; 11% MUFA, 10% PUFA, 12% SFA). Multilevel models were used to examine the effect of the diets on FMD. No significant between-diet differences were observed for average brachial artery diameter (CO: 6.70 ± 0.15 mm, HOCO: 6.57 ± 0.15 mm, CON: 6.73 ± 0.14 mm; p = 0.72), peak brachial artery diameter (CO: 7.11 ± 0.15 mm, HOCO: 7.02 ± 0.15 mm, CON: 6.41 ± 0.48 mm; p = 0.80), or FMD (CO: 6.32 ± 0.51%, HOCO: 6.96 ± 0.49%, CON: 6.41 ± 0.48%; p = 0.81). Partial replacement of SFA with MUFA from CO and HOCO had no effect on FMD in participants with or at risk of metabolic syndrome.
Collapse
Affiliation(s)
- Kristin M. Davis
- Department of Biobehavioral Health, Pennsylvania State University, State College, PA 16802, USA
| | - Kristina S. Petersen
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA 16802, USA
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Kate J. Bowen
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA 16802, USA
| | - Peter J. H. Jones
- Richardson Centre for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, MB R3T 6C5, Canada
| | - Carla G. Taylor
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
- The Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada
| | - Peter Zahradka
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
- The Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada
| | - Karen Letourneau
- The Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada
| | - Danielle Perera
- The Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada
| | - Angela Wilson
- The Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada
| | - Paul R. Wagner
- Department of Biobehavioral Health, Pennsylvania State University, State College, PA 16802, USA
| | - Penny M. Kris-Etherton
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA 16802, USA
- Correspondence:
| | - Sheila G. West
- Department of Biobehavioral Health, Pennsylvania State University, State College, PA 16802, USA
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA 16802, USA
| |
Collapse
|
197
|
Khanna NN, Maindarkar M, Puvvula A, Paul S, Bhagawati M, Ahluwalia P, Ruzsa Z, Sharma A, Munjral S, Kolluri R, Krishnan PR, Singh IM, Laird JR, Fatemi M, Alizad A, Dhanjil SK, Saba L, Balestrieri A, Faa G, Paraskevas KI, Misra DP, Agarwal V, Sharma A, Teji J, Al-Maini M, Nicolaides A, Rathore V, Naidu S, Liblik K, Johri AM, Turk M, Sobel DW, Pareek G, Miner M, Viskovic K, Tsoulfas G, Protogerou AD, Mavrogeni S, Kitas GD, Fouda MM, Kalra MK, Suri JS. Vascular Implications of COVID-19: Role of Radiological Imaging, Artificial Intelligence, and Tissue Characterization: A Special Report. J Cardiovasc Dev Dis 2022; 9:268. [PMID: 36005433 PMCID: PMC9409845 DOI: 10.3390/jcdd9080268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/30/2022] [Accepted: 08/09/2022] [Indexed: 12/15/2022] Open
Abstract
The SARS-CoV-2 virus has caused a pandemic, infecting nearly 80 million people worldwide, with mortality exceeding six million. The average survival span is just 14 days from the time the symptoms become aggressive. The present study delineates the deep-driven vascular damage in the pulmonary, renal, coronary, and carotid vessels due to SARS-CoV-2. This special report addresses an important gap in the literature in understanding (i) the pathophysiology of vascular damage and the role of medical imaging in the visualization of the damage caused by SARS-CoV-2, and (ii) further understanding the severity of COVID-19 using artificial intelligence (AI)-based tissue characterization (TC). PRISMA was used to select 296 studies for AI-based TC. Radiological imaging techniques such as magnetic resonance imaging (MRI), computed tomography (CT), and ultrasound were selected for imaging of the vasculature infected by COVID-19. Four kinds of hypotheses are presented for showing the vascular damage in radiological images due to COVID-19. Three kinds of AI models, namely, machine learning, deep learning, and transfer learning, are used for TC. Further, the study presents recommendations for improving AI-based architectures for vascular studies. We conclude that the process of vascular damage due to COVID-19 has similarities across vessel types, even though it results in multi-organ dysfunction. Although the mortality rate is ~2% of those infected, the long-term effect of COVID-19 needs monitoring to avoid deaths. AI seems to be penetrating the health care industry at warp speed, and we expect to see an emerging role in patient care, reduce the mortality and morbidity rate.
Collapse
Affiliation(s)
- Narendra N. Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi 110001, India
| | - Mahesh Maindarkar
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | - Anudeep Puvvula
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
- Annu’s Hospitals for Skin and Diabetes, Nellore 524101, India
| | - Sudip Paul
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | - Mrinalini Bhagawati
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India
| | - Puneet Ahluwalia
- Max Institute of Cancer Care, Max Super Specialty Hospital, New Delhi 110017, India
| | - Zoltan Ruzsa
- Invasive Cardiology Division, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Aditya Sharma
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22904, USA
| | - Smiksha Munjral
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| | - Raghu Kolluri
- Ohio Health Heart and Vascular, Columbus, OH 43214, USA
| | | | - Inder M. Singh
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| | - John R. Laird
- Heart and Vascular Institute, Adventist Health St. Helena, St Helena, CA 94574, USA
| | - Mostafa Fatemi
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Azra Alizad
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Surinder K. Dhanjil
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, 40138 Cagliari, Italy
| | - Antonella Balestrieri
- Cardiovascular Prevention and Research Unit, Department of Pathophysiology, National & Kapodistrian University of Athens, 15772 Athens, Greece
| | - Gavino Faa
- Department of Pathology, Azienda Ospedaliero Universitaria, 09124 Cagliari, Italy
| | | | - Durga Prasanna Misra
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Vikas Agarwal
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Aman Sharma
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Jagjit Teji
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Mustafa Al-Maini
- Allergy, Clinical Immunology and Rheumatology Institute, Toronto, ON L4Z 4C4, Canada
| | - Andrew Nicolaides
- Vascular Screening and Diagnostic Centre and University of Nicosia Medical School, 2408 Nicosia, Cyprus
| | - Vijay Rathore
- Nephrology Department, Kaiser Permanente, Sacramento, CA 95119, USA
| | - Subbaram Naidu
- Electrical Engineering Department, University of Minnesota, Duluth, MN 55812, USA
| | - Kiera Liblik
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Amer M. Johri
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Monika Turk
- The Hanse-Wissenschaftskolleg Institute for Advanced Study, 27753 Delmenhorst, Germany
| | - David W. Sobel
- Rheumatology Unit, National Kapodistrian University of Athens, 15772 Athens, Greece
| | - Gyan Pareek
- Minimally Invasive Urology Institute, Brown University, Providence, RI 02912, USA
| | - Martin Miner
- Men’s Health Centre, Miriam Hospital Providence, Providence, RI 02906, USA
| | - Klaudija Viskovic
- Department of Radiology and Ultrasound, University Hospital for Infectious Diseases, 10000 Zagreb, Croatia
| | - George Tsoulfas
- Department of Surgery, Aristoteleion University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Athanasios D. Protogerou
- Cardiovascular Prevention and Research Unit, Department of Pathophysiology, National & Kapodistrian University of Athens, 15772 Athens, Greece
| | - Sophie Mavrogeni
- Cardiology Clinic, Onassis Cardiac Surgery Centre, 17674 Athens, Greece
| | - George D. Kitas
- Academic Affairs, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, UK
- Arthritis Research UK Epidemiology Unit, Manchester University, Manchester M13 9PL, UK
| | - Mostafa M. Fouda
- Department of Electrical and Computer Engineering, Idaho State University, Pocatello, ID 83209, USA
| | - Manudeep K. Kalra
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jasjit S. Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| |
Collapse
|
198
|
da Silva RSN, da Silva DS, Waclawovsky G, Schaun MI. Effects of aerobic, resistance, and combined training on endothelial function and arterial stiffness in older adults: study protocol for a systematic review and meta-analysis. Syst Rev 2022; 11:171. [PMID: 35964075 PMCID: PMC9375352 DOI: 10.1186/s13643-022-02036-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Aging is an independent risk factor for cardiovascular events. It promotes vascular dysfunction which is associated with risk factors for cardiovascular diseases (CVDs). Exercise can modulate vascular function parameters, but little is known about the effects of different modalities of training (aerobic, resistance, and combined) on endothelial function and arterial stiffness in older adults. METHODS This systematic review study will include randomized controlled trials (RCTs) selected from the electronic databases MEDLINE (PubMed), Cochrane, LILACS, EMBASE, and Web of Science. We will follow the PRISMA guidelines and PICOS framework. Studies involving both male and female older adults (≥60 years old) with or without comorbidities undergoing aerobic, resistance, and/or combined training compared to a control group (no exercise) will be eligible. We will use the Cochrane Risk of Bias 2 (RoB 2) tool to evaluate the quality of individual studies and GRADE to assess the strength of evidence. Statistical analyses will be conducted with RStudio for Windows (v1.3.959) using R package meta. DISCUSSION A systematic review and meta-analysis involving data from studies of older adults would deepen our understanding of vascular adaptations to exercise training in this population. It could provide new insights into how health providers can improve patient management and prevention of cardiovascular events in older adults. SYSTEMATIC REVIEW REGISTRATION PROSPERO 42021275451.
Collapse
Affiliation(s)
- Raphael S N da Silva
- Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária de Cardiologia, Av. Princesa Isabel, 395 Santana, Porto Alegre, RS, 90620-001, Brazil
| | - Diego S da Silva
- Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária de Cardiologia, Av. Princesa Isabel, 395 Santana, Porto Alegre, RS, 90620-001, Brazil
| | - Gustavo Waclawovsky
- Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária de Cardiologia, Av. Princesa Isabel, 395 Santana, Porto Alegre, RS, 90620-001, Brazil
| | - Maximiliano I Schaun
- Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária de Cardiologia, Av. Princesa Isabel, 395 Santana, Porto Alegre, RS, 90620-001, Brazil.
| |
Collapse
|
199
|
Rehberger Likozar A, Šebeštjen M. Smoking and diabetes attenuate beneficial effects of PSCK9 inhibitors on arterial wall properties in patients with very high lipoprotein (a) levels. ATHEROSCLEROSIS PLUS 2022; 50:1-9. [PMID: 36643800 PMCID: PMC9833244 DOI: 10.1016/j.athplu.2022.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/08/2022] [Accepted: 07/15/2022] [Indexed: 01/18/2023]
Abstract
Background and aims Elevated lipoprotein (a) (Lp(a)) and low-density lipoprotein cholesterol levels (LDL-C) are significant residual risk factors for cardiovascular events. Treatment with protein convertase subtilisin kexin type 9 (PCSK9) inhibitors reduces the levels of both. Less is known about effects of PCSK9 inhibitors on functional and morphological properties of the arterial wall. The aim of the present study was to determine whether other factors besides decreased LDL-C and Lp(a) are associated with functional (flow-mediated dilation [FMD]) and morphological (carotid intima-media thickness [c-IMT], pulse-wave velocity [PWV]) changes of the arterial wall properties in patients with coronary artery disease (CAD) treated with alirocumab and evolocumab. Methods One hundred patients with CAD after myocardial infarction before 55 years and with high Lp(a) were randomised to lipid-lowering therapies without PCSK9 inhibitors (control; N = 31), or with alirocumab 150 mg SC (N = 35) or evolocumab 140 mg SC (N = 34), every 2 weeks. All patients underwent blood sampling for biochemical analyses and ultrasound measurements for FMD, c-IMT and PWV. Results There were no significant changes in FMD for the control (10.7% ± 6.6%-11.1% ± 4.4%, p = 0.716) and alirocumab (10.7% ± 5.9%-11.2% ± 5.3%, p = 0.547) groups, while evolocumab promoted significant increase (11.2% ± 6.8%-14.1% ± 6.6%, p < 0.0001). Only in non-smokers and non-diabetics significant improvements in FMD (p < 0.0001) after treatment with PCSK9 inhibitors were observed. Conclusion These data show that for patients with CAD and high Lp(a) levels, beneficial effects of PCSK9 inhibitors on the arterial wall properties can be attenuated by specific risk factors, such as smoking and diabetes.
Collapse
Affiliation(s)
| | - Miran Šebeštjen
- Department of Vascular Diseases, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia,Department of Cardiology, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia,University of Ljubljana, Faculty of Medicine, 1000, Ljubljana, Slovenia,Corresponding author. Department of Cardiology, University Medical Centre Ljubljana, Zaloška 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
200
|
Xue Y, Li X, Wang Z, Lv Q. Cilostazol regulates the expressions of endothelin‑1 and endothelial nitric oxide synthase via activation of the p38 MAPK signaling pathway in HUVECs. Biomed Rep 2022; 17:77. [DOI: 10.3892/br.2022.1560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/12/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Ying Xue
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xiaoye Li
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Zi Wang
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Qianzhou Lv
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|