1
|
Zhang Y, Chai S, Dai H, Chen X, Meng Z, Ying X. Vascular endothelial function and its response to moderate-intensity aerobic exercise in trained and untrained healthy young men. Sci Rep 2024; 14:20450. [PMID: 39242762 PMCID: PMC11379850 DOI: 10.1038/s41598-024-71471-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024] Open
Abstract
30 min of moderate-intensity aerobic exercise per day is recommended, but the response and adaptation of endothelial function (EF) to this exercise remains controversial. The purpose of this study was to determine the changes in EF in endurance trained and untrained individuals before and after this exercise and to compare the differences between trained and untrained individuals. Twelve endurance-trained male college athletes (trained group) and 12 untrained male college students (untrained group) performed a 30-min run at an intensity of 60% VO2max. Brachial artery flow-mediated dilation (FMD) was measured before exercise, 30 min and 60 min after exercise, and the following morning. Resting diameter and maximum diameter showed large time effects (p < 0.001, η2 = 0.533; p < 0.001, η2 = 0.502). Resting diameters at 30 and 60 min after exercise were higher than before exercise in both the untrained and trained groups (p < 0.05), and maximum diameters at 30 min after exercise were higher than before exercise in both the untrained and trained groups (p < 0.01). Resting diameter and maximum diameter also exhibited some group effects (p = 0.055, η2 = 0.157; p = 0.041, η2 = 0.176). Resting diameters and maximum diameters were higher in the trained group than in the untrained group before exercise (p < 0.05). FMD (%) showed no time, group, or time-group interaction effects. 30 min of moderate-intensity aerobic exercise can increase resting and maximal arterial diameters in both trained and untrained young men, but has no effect on FMD. Long-term endurance training has the potential to increase resting and maximal arterial diameters in young men, but not necessarily FMD.
Collapse
Affiliation(s)
- Yong Zhang
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Shiyi Chai
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Hailun Dai
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Xiaofei Chen
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Zhaofeng Meng
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China.
| | - Xiaofang Ying
- School of Business, Shaoxing University, Shaoxing, China.
| |
Collapse
|
2
|
Kishimoto S, Hashimoto Y, Maruhashi T, Kajikawa M, Mizobuchi A, Harada T, Yamaji T, Nakano Y, Goto C, Yusoff FM, Iwanaga Y, Kanaoka K, Yada T, Itarashiki T, Higashi Y. New device for assessment of endothelial function: plethysmographic flow-mediated vasodilation (pFMD). Hypertens Res 2024; 47:2471-2477. [PMID: 38951680 PMCID: PMC11374665 DOI: 10.1038/s41440-024-01770-z] [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: 02/28/2024] [Revised: 05/30/2024] [Accepted: 06/06/2024] [Indexed: 07/03/2024]
Abstract
Measurement of flow-mediated vasodilation (FMD) in the brachial artery by using ultrasound is a well-established technique for evaluating endothelial function. To make the measurement quicker and simpler than the measurements of conventional ultrasound FMD (uFMD), we have developed a new noninvasive method, plethysmographic FMD (pFMD), to assess vascular response to reactive hyperemia in the brachial artery. The aim of this study was to determine the accuracy of measurement of pFMD in comparison to that of measurement of conventional uFMD. This study was a multi-center, cross-sectional study. We compared pFMD by a new device using cuff pressure and volume with conventional uFMD using ultrasound in 50 men (mean age, 41 ± 9 years). pFMD significantly correlated with conventional uFMD (β = 0.59, P < 0.001). In Bland-Altman plot analysis of pFMD and conventional uFMD, the mean difference of pFMD and conventional uFMD was 0.78%, and limits of agreement (mean difference ±2 standard deviations of the difference) ranged from -4.53% to 6.11%. We demonstrated validity of the new method for measurement of pFMD, which can automate the evaluation of endothelial function in a short time. Measurement of pFMD is simpler than measurement of conventional uFMD and may have reduced artificial bias compared to that of conventional uFMD measurement (URL for Clinical Trial: https://ethics.hiroshima-u.ac.jp/site/wp-content/uploads/2022/12/eki_giji20221213.pdf . Registration Number for Clinical Trial: E2022-0131).
Collapse
Affiliation(s)
- Shinji Kishimoto
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yu Hashimoto
- Department of Cardiovascular Medicine, Medical Corporation JR Hiroshima Hospital, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Aya Mizobuchi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Takahiro Harada
- Center for Cause of Death Investigation Research, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takayuki Yamaji
- Center for Radiation Disaster Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chikara Goto
- Dpartment of Rehabilitation, Faculty of General Rehabilitation, Hiroshima International University, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yoshitaka Iwanaga
- Department of Medical and Health Information Management, National Cerebral and Cardiovascular Center, Osaka, Japan
- Department of Cardiology, Sakurabashi Watanabe Hospital, Osaka, Japan
| | - Koshiro Kanaoka
- Department of Medical and Health Information Management, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | | | | - Yukihito Higashi
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan.
| |
Collapse
|
3
|
Sinha Roy S, Sondararajan R, Sharma A, Singhal M, Sharma S, Malhotra P, Singh C, Jain A, Khadwal A, Prakash G. Non-invasive Estimation of Microvasculopathy & Endothelial Dysfunction in Stem Cell Transplant Recipients and its Relationship with GVHD. BLOOD CELL THERAPY 2024; 7:79-86. [PMID: 39263621 PMCID: PMC11384125 DOI: 10.31547/bct-2023-041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/11/2024] [Indexed: 09/13/2024]
Abstract
Introduction Microvasculopathy and endothelial dysfunction play important roles in the development of post-transplant complications, including graft-versus-host disease (GVHD). We assessed structural microvasculopathy by employing nailfold video capillaroscopy (NFVC) and endothelial dysfunction via flow-mediated dilatation (FMD) of the brachial artery in recipients of hematopoietic stem cell transplantation. Patients and methods Recipients of stem cell transplantation were included in this study post day+100 and divided into two cohorts. The first cohort consisted of 35 recipients of allogeneic hematopoietic stem cell transplantation (HCT) and the second cohort was comprised of 31 recipients of autologous HCT. A third cohort included 35 healthy individuals. NFVC was conducted on the second to fifth fingers of both hands using an Optilia video capillaroscope at 200× magnification, and the images were analyzed according to the European Alliance of Associations for Rheumatology (EULAR) criteria. The following parameters were used to measure vasculopathy: (a) median capillary density, derived from the capillary density of eight fingers, (b) median capillary diameter, derived from maximum capillary apical diameters of eight fingers, and (c) significant neoangiogenesis (neoangiogenesis present in ≥2 fingers). FMD of the right brachial artery was observed by high-resolution ultrasonography using the principle of post-occlusive reactive hyperemia, and video images were analyzed using edge-detecting software. Results The median capillary diameter was significantly higher in the allo-HCT cohort (20.56±5.17 micrometer) compared to the auto-HCT cohort (16.19±3.31 micrometer; p<0.001) and healthy controls (14.66±2.61 micrometer; p<0.001). The median capillary density was significantly lower in the allo-HCT cohort (median: 6 capillaries/mm, range: 5-9 capillaries/mm) compared to the auto-HCT cohort (median: 8.5 capillaries, range: 5-12 capillaries/mm; p<0.001) and healthy controls (median: 8 capillaries/mm, range: 7-10.5 capillaries/mm; p<0.001). The allo-HCT cohort had a higher proportion of patients with significant neoangiogenesis (86%) than the auto-HCT cohort (10%) and healthy controls (9%). The presence of significant neoangiogenesis was more frequent in the subgroup of patients with a history of GVHD (93%) compared to the subgroup of patients without any history of GVHD (57%; p=0.044). No significant differences in NFVC parameters or FMD were observed between recipients of myeloablative and reduced-intensity conditioning regimens. There was no significant difference in NFVC parameters between the auto-HCT cohort and healthy controls. There was no significant difference in FMD among the three cohorts; however, a higher proportion of patients in the allo-HCT cohort (28%) had lower FMD than those in the auto-HCT cohort (3%) and healthy controls (6%), suggesting endothelial dysfunction. Conclusions Our findings demonstrate the presence of structural microvasculopathy in allo-HCT recipients and suggest a possible role of alloreactivity in the pathogenesis of post-HCT microvasculopathy.
Collapse
Affiliation(s)
- Sayan Sinha Roy
- Department of Clinical Hematology and Medical Oncology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Raghuraman Sondararajan
- Department of Radiodiagnosis and Imaging, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Arun Sharma
- Department of Radiodiagnosis and Imaging, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Manphool Singhal
- Department of Radiodiagnosis and Imaging, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shefali Sharma
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Malhotra
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Charanpreet Singh
- Department of Clinical Hematology and Medical Oncology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Arihant Jain
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Khadwal
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Gaurav Prakash
- Department of Clinical Hematology and Medical Oncology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
4
|
Zhang H, Sun J, Zhang Y, Xiao K, Wang Y, Si J, Li Y, Sun L, Zhao T, Yi M, Chu X, Li J. Association between exposure to air pollution and arterial stiffness in participants with and without atherosclerotic cardiovascular disease. Clin Res Cardiol 2024:10.1007/s00392-024-02506-2. [PMID: 39105787 DOI: 10.1007/s00392-024-02506-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
AIMS To assess the association of air pollution exposure at different time scales with arterial stiffness in participants with and without atherosclerotic cardiovascular disease (ASCVD). METHODS We measured participants' arterial stiffness with brachial-ankle pulse wave velocity (baPWV) from October 2016 to January 2020. Concentrations of air pollutants including fine particles < 2.5 μm aerodynamic diameter (PM2.5), inhalable particles < 10 μm aerodynamic diameter (PM10), sulfur dioxide (SO2), nitrogen dioxide (NO2), carbon monoxide (CO), and ozone (O3) measured by fixed ambient air monitoring stations were collected for short- (7-day) and long-term (365-day) exposure assessment. We used generalized estimating equations (GEEs) to analyze and further explored the modification effects between ASCVD and air pollutants. RESULTS Seven hundred sixty-five participants were finally included and four hunderd sixty (60.1%) participants had a history of ASCVD. Based on the partial regression coefficients (β) and 95% confidence intervals (95% CI) calculated from GEEs using linear regression, each 10 μg/m3 increase in long-term exposure to PM2.5 and PM10 was associated with 31.85 cm/s (95% CI, 17.97 to 45.73) and 35.93 cm/s (95% CI, 21.01 to 50.84) increase in baPWV. There was no association between short-term exposure to air pollution and arterial stiffness. Although no significant interaction effect was observed between air pollution and ASCVD, baPWV showed a greater increment in the subgroup without ASCVD. CONCLUSION Long-term exposure to air pollution is closely associated with higher arterial stiffness in participants with and without ASCVD. Reducing air pollution exposure is essential in the primary and secondary prevention of ASCVD.
Collapse
Affiliation(s)
- Haoyu Zhang
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Jinghao Sun
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yinghua Zhang
- Department of Cardiology, Chuiyangliu Hospital Affiliated to Tsinghua University, Beijing, 100021, China
| | - Keling Xiao
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yang Wang
- Medical Research and Biometrics Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jin Si
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yan Li
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Lijie Sun
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Ting Zhao
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Ming Yi
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Xi Chu
- Health Management Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Jing Li
- Department of Geriatrics, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
| |
Collapse
|
5
|
Huang X, Huang X, Pan M, Lin J, Xie L. Effect of early endothelial function improvement on subclinical target organ damage in hypertensives. Sci Rep 2024; 14:16078. [PMID: 38992162 PMCID: PMC11239846 DOI: 10.1038/s41598-024-67143-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
Endothelial dysfunction is acknowledged as a marker for subclinical target organ damage (STOD) in hypertension, though its therapeutic potential has not yet been clarified. This study assessed whether early endothelial function improvement (EEFI) reduced STOD in patients with essential hypertension (EH). We conducted a retrospective cohort analysis of 456 EH patients initially free from STOD. Endothelial function was assessed using brachial artery flow-mediated dilation (FMD), with values ≤ 7.1% indicating dysfunction. Patients were initially categorized by endothelial status (dysfunction: n = 180, normal: n = 276), and further divided into improved or unimproved groups based on changes within three months post-enrollment. During a median follow-up of 25 months, 177 patients developed STOD. The incidence of STOD was significantly higher in patients with initial dysfunction compared to those with normal function. Kaplan-Meier analysis indicated that the improved group had a lower cumulative incidence of STOD compared to the unimproved group (p < 0.05). Multivariable Cox regression confirmed EEFI as an independent protective factor against STOD in EH patients (p < 0.05), regardless of their baseline endothelial status, especially in those under 65 years old, non-smokers, and with low-density lipoprotein cholesterol levels ≤ 3.4 mmol/L. In conclusion, EEFI significantly reduces STOD incidence in EH patients, particularly in specific subgroups, emphasizing the need for early intervention in endothelial function to prevent STOD.
Collapse
Affiliation(s)
- Xiaodong Huang
- Department of Emergency, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Xiamen Key Laboratory for Clinical Efficacy and Evidence-Based Research of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Xianwei Huang
- Department of Emergency, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Xiamen Key Laboratory for Clinical Efficacy and Evidence-Based Research of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Mandong Pan
- Department of Emergency, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Xiamen Key Laboratory for Clinical Efficacy and Evidence-Based Research of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Jiyan Lin
- Department of Emergency, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
- Xiamen Key Laboratory for Clinical Efficacy and Evidence-Based Research of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Liangdi Xie
- Department of Geriatrics, Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China.
| |
Collapse
|
6
|
Angjelova A, Jovanova E, Polizzi A, Laganà L, Santonocito S, Ragusa R, Isola G. Impact of Periodontitis on Endothelial Risk Dysfunction and Oxidative Stress Improvement in Patients with Cardiovascular Disease. J Clin Med 2024; 13:3781. [PMID: 38999345 PMCID: PMC11242897 DOI: 10.3390/jcm13133781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Periodontitis is a multifactorial chronic inflammatory disease that affects the periodontium and overall oral health and is primarily caused by a dysbiotic gingival biofilm, which includes, among others, Gram-negative bacteria such as Porphyromonas gingivalis, Actinobacillus actinomycetemcomitans, and Tannerella forsythensis that colonize gingival tissues and that can lead, if not properly treated, to periodontal tissue destruction and tooth loss. In the last few decades, several large-scale epidemiological studies have evidenced that mild and severe forms of periodontitis are strictly bilaterally associated with several cardiovascular diseases (CVDs), stroke, and endothelial dysfunction. Specifically, it is hypothesized that patients with severe periodontitis would have compromised endothelial function, a crucial step in the pathophysiology of atherosclerosis and several CVD forms. In this regard, it was postulated that periodontal treatment would ameliorate endothelial dysfunction, hence bolstering the notion that therapeutic approaches targeted at diminishing cardiovascular risk factors and different forms of periodontal treatment could improve several CVD biomarker outcomes in the short- and long-term in CVD patients. The aim of this review is to update and analyze the link between periodontitis and CVD, focusing on the inflammatory nature of periodontitis and its correlation with CVD, the effects of periodontal therapy on endothelial dysfunction and oxidative stress, and the impact of such therapy on CVD biomarkers and outcomes. The article also discusses future research directions in this field.
Collapse
Affiliation(s)
- Angela Angjelova
- University Dental Clinical Center St. Pantelejmon, Skopje, Faculty of Dentistry, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia
| | - Elena Jovanova
- University Dental Clinical Center St. Pantelejmon, Skopje, Faculty of Dentistry, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia
| | - Alessandro Polizzi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Ludovica Laganà
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Rosalia Ragusa
- Health Direction of Policlinic Hospital, 95100 Catania, Italy
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| |
Collapse
|
7
|
Yamaji T, Harada T, Hashimoto Y, Nakano Y, Kajikawa M, Yoshimura K, Goto C, Han Y, Mizobuchi A, Yusoff FM, Kishimoto S, Maruhashi T, Nakashima A, Higashi Y. Effects of BNT162b2 mRNA Covid-19 vaccine on vascular function. PLoS One 2024; 19:e0302512. [PMID: 38687730 PMCID: PMC11060586 DOI: 10.1371/journal.pone.0302512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
The effects of Covid-19 vaccines on vascular function are still controversial. We evaluated the effects of BNT162b2 vaccine (BioNTech and Pfizer) on endothelial function assessed by flow-mediated vasodilation (FMD) and vascular smooth muscle function assessed by nitroglycerine-induced vasodilation (NID). This study was a prospective observational study. A total of 23 medical staff at Hiroshima University Hospital were enrolled in this study. FMD and NID were measured before vaccination and two weeks and six months after the 2nd dose of vaccination. FMD was significantly smaller two weeks after the 2nd dose of vaccination than before vaccination (6.5±2.4% and 8.2±2.6%, p = 0.03). FMD was significantly larger at six months than at two weeks after the 2nd dose of vaccination (8.2±3.0% and 6.5±2.4%, p = 0.03). There was no significant difference between FMD before vaccination and that at six months after the 2nd dose of vaccination (8.2±2.6% to 8.2±3.0%, p = 0.96). NID values were similar before vaccination and at two weeks, and six months after vaccination (p = 0.89). The BNT162b2 Covid-19 vaccine temporally impaired endothelial function but not vascular smooth muscle function, and the impaired endothelial function returned to the baseline level within six months after vaccination.
Collapse
Affiliation(s)
- Takayuki Yamaji
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yu Hashimoto
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Kenichi Yoshimura
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
- Department of Biostatistics, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Chikara Goto
- Department of Rehabilitation, Faculty of General Rehabilitation, Hiroshima International University, Hiroshima, Japan
| | - Yiming Han
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Aya Mizobuchi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
8
|
Mohammadi N, Farrell M, O'Sullivan L, Langan A, Franchin M, Azevedo L, Granato D. Effectiveness of anthocyanin-containing foods and nutraceuticals in mitigating oxidative stress, inflammation, and cardiovascular health-related biomarkers: a systematic review of animal and human interventions. Food Funct 2024; 15:3274-3299. [PMID: 38482946 DOI: 10.1039/d3fo04579j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cardiovascular diseases (CVDs) are a group of chronic health disorders prevalent worldwide that claim millions of lives yearly. Inflammation and oxidative stress are intricately associated with myocardial tissue damage, endothelial dysfunction, and increased odds of heart failure. Thus, dietary strategies aimed at decreasing the odds of CVDs are paramount. In this regard, the consumption of anthocyanins, natural pigments found in edible flowers, fruits, and vegetables, has attracted attention due to their potential to promote cardiovascular health. The main mechanisms of action linked with their protective effects on antioxidant and anti-inflammatory activities, serum lipid profile modulation, and other cardiovascular health parameters are explained and exemplified. However, little is known about the dose-dependency nature of the effects, which anthocyanin has better efficiency, and whether anthocyanin-containing foods display better in vivo efficacy than nutraceuticals (i.e., concentrated extracts containing higher levels of anthocyanins than foods). Thus, this systematic review focused on determining the effects of anthocyanin-containing foods and nutraceuticals on biomarkers associated with CVDs using animal studies and human interventions supported by in vitro mechanistic insights. Overall, the results showed that the regular consumption of anthocyanin-containing foods and nutraceuticals improved vascular function, lipid profile, and antioxidant and anti-inflammatory effects. The daily dosage, the participants' health status, and the duration of the intervention also significantly influenced the results.
Collapse
Affiliation(s)
- Nima Mohammadi
- University of Limerick, School of Natural Sciences, Faculty of Science and Engineering, Department of Biological Sciences, Bioactivity and Applications Lab, V94 T9PX Limerick, Ireland.
| | - Michelle Farrell
- University of Limerick, School of Natural Sciences, Faculty of Science and Engineering, Department of Biological Sciences, Bioactivity and Applications Lab, V94 T9PX Limerick, Ireland.
| | - Laura O'Sullivan
- University of Limerick, School of Natural Sciences, Faculty of Science and Engineering, Department of Biological Sciences, Bioactivity and Applications Lab, V94 T9PX Limerick, Ireland.
| | - Andrea Langan
- University of Limerick, School of Natural Sciences, Faculty of Science and Engineering, Department of Biological Sciences, Bioactivity and Applications Lab, V94 T9PX Limerick, Ireland.
| | - Marcelo Franchin
- University of Limerick, School of Natural Sciences, Faculty of Science and Engineering, Department of Biological Sciences, Bioactivity and Applications Lab, V94 T9PX Limerick, Ireland.
| | - Luciana Azevedo
- Federal University of Alfenas, In Vitro and In Vivo Nutritional and Toxicological Analysis Laboratory, Av. Jovino Fernandes Sales, 2600, Bairro Santa Clara - CEP 37133-840, Alfenas, Minas Gerais, Brazil
| | - Daniel Granato
- University of Limerick, School of Natural Sciences, Faculty of Science and Engineering, Department of Biological Sciences, Bioactivity and Applications Lab, V94 T9PX Limerick, Ireland.
- Health Research Institute, University of Limerick, V94 T9PX Limerick, Ireland
| |
Collapse
|
9
|
Yamaji T, Harada T, Kajikawa M, Maruhashi T, Kishimoto S, Yusoff FM, Chayama K, Goto C, Nakashima A, Tomiyama H, Takase B, Kohro T, Suzuki T, Ishizu T, Ueda S, Yamazaki T, Furumoto T, Kario K, Inoue T, Watanabe K, Takemoto Y, Hano T, Sata M, Ishibashi Y, Node K, Maemura K, Ohya Y, Furukawa T, Ito H, Yamashina A, Koba S, Higashi Y. Role of Small Dense Low-density Lipoprotein Cholesterol in Cardiovascular Events in Patients with Coronary Artery Disease and Type 2 Diabetes Mellitus Receiving Statin Treatment. J Atheroscler Thromb 2024; 31:478-500. [PMID: 37926523 PMCID: PMC10999715 DOI: 10.5551/jat.64416] [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: 06/04/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
AIM There is little information on the relationships of serum small dense low-density lipoprotein cholesterol (sdLDL-C) levels and serum triglyceride (TG) levels with cardiovascular events in patients with coronary artery disease (CAD) and type 2 diabetes mellitus (DM) who are receiving statins. The aim of this study was to evaluate the relationships of serum TG levels and sdLDL-C levels as residual risks for cardiovascular events in patients with CAD and type 2 DM who were being treated with statins. METHODS The subjects were divided into four groups based on TG levels and sdLDL-C levels: sdLDL-C of <40.0 mg/dL and TG of <150 mg/dL, sdLDL-C of ≥ 40.0 mg/dL and TG of <150 mg/dL, sdLDL-C of <40.0 mg/dL and TG of ≥ 150 mg/dL, and sdLDL-C of ≥ 40.0 mg/dL and TG of ≥ 150 mg/dL. During a median follow-up period of 1419 days, cardiovascular events occurred in 34 patients. RESULTS The incidences of cardiovascular events were significantly higher in patients with sdLDL-C of ≥ 40.0 mg/dL and TG of <150 mg/dL and in patients with sdLDL-C of ≥ 40.0 mg/dL and TG of ≥ 150 mg/dL, but not in patients with sdLDL-C of <40.0 mg/dL and TG of ≥ 150 mg/dL, than in patients with sdLDL-C of <40.0 mg/dL and TG of <150 mg/dL. CONCLUSIONS Under the condition of treatment with statins, patients with CAD and type 2 DM who had sdLDL-C levels of ≥ 40.0 mg/dL had a high risk for cardiovascular events even though serum TG levels were controlled at <150 mg/dL.
Collapse
Affiliation(s)
- Takayuki Yamaji
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chikara Goto
- Department of Physical Therapy, Hiroshima International University, Hiroshima, Japan
| | - Ayumu Nakashima
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | | | - Bonpei Takase
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Tokorozawa, Japan
| | - Takahide Kohro
- Department of Clinical Informatics, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Toru Suzuki
- Cardiovascular Medicine, University of Leicester, Leicester, UK
| | - Tomoko Ishizu
- Cardiovascular Division, Institute of Clinical Medicine, University of Tsukuba, Ibaraki, Japan
| | - Shinichiro Ueda
- Department of Clinical Pharmacology and Therapeutics, University of the Ryukyu School of Medicine, Okinawa, Japan
| | - Tsutomu Yamazaki
- Department of Clinical Epidemiology and Systems, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoo Furumoto
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Teruo Inoue
- Department of Cardiovascular Medicine, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Kentaro Watanabe
- Department of Neurology, Hematology, Metabolism, Endocrinology and Diabetelogy (DNHMED), Yamagata University School of Medicine, Yamagata, Japan
| | - Yasuhiko Takemoto
- Department of Internal Medicine and Cardiology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Takuzo Hano
- Department of Medical Education and Population-based Medicine, Postgraduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yutaka Ishibashi
- Department of General Medicine, Shimane University Faculty of Medicine, Izumo, Japan
| | - Koichi Node
- Department of Cardiovascular and Renal Medicine, Saga University, Saga, Japan
| | - Koji Maemura
- Department of Cardiovascular Medicine, Course of Medical and Dental Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yusuke Ohya
- The Third Department of Internal Medicine, University of the Ryukyus, Okinawa, Japan
| | - Taiji Furukawa
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan
| | - Akira Yamashina
- Department of Cardiology, Tokyo Medical University, Tokyo, Japan
| | - Shinji Koba
- Department of Medicine, Division of Cardiology, Showa University School of Medicine, Tokyo, Japan
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
10
|
Kim H, Choi CU, Rhew K, Park J, Lim Y, Kim MG, Kim K. Comparative effects of glucose-lowering agents on endothelial function and arterial stiffness in patients with type 2 diabetes: A network meta-analysis. Atherosclerosis 2024; 391:117490. [PMID: 38452432 DOI: 10.1016/j.atherosclerosis.2024.117490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND AND AIMS Despite accumulating evidence on the potential of glucose-lowering agents (GLAs) to prevent cardiovascular events, the comparative effects of GLAs on vascular function remain unclear. This study utilized validated indicators such as flow-mediated dilation (FMD; positive value favors) and pulse wave velocity (PWV; negative value favors) to uncover the comparative effects of GLAs on vascular function. METHODS Randomized controlled trials (RCTs) comparing the effects of GLAs on FMD or PWV with placebo or other GLAs in patients with type 2 diabetes (T2DM) were searched through PubMed and Embase. The frequentist method of network meta-analysis (NMA) was conducted using a random effects model, and standardized mean differences (SMDs) with 95% confidence intervals (CIs) were calculated. RESULTS The NMA included 38 RCTs with 2,065 patients. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) and sodium glucose cotransporter-2 inhibitors (SGLT-2Is) had significantly more positive effects on FMD improvement and PWV reduction than placebo. Thiazolidinedione (TZD) treatment resulted in significantly improved FMD compared to other GLAs as well as placebo (SMD: 1.14; 95% CI: 0.84 to 1.43). Both pioglitazone and rosiglitazone were discovered to have considerably more favorable effects on improving FMD and reducing PWV compared to placebo and other GLAs, as a result of the analysis incorporating each drug in the TZD class. The sensitivity analysis results corroborated the main findings. CONCLUSIONS This NMA showed more favorable effects of GLP-1RAs and SGLT-2Is than placebo in improving both arterial stiffness and endothelial function in patients with T2DM. In addition, TZDs showed superior effects in improving endothelial function as compared with the other GLAs and placebo.
Collapse
Affiliation(s)
- Hayeon Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Cheol Ung Choi
- Cardiovascular Center, Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Kiyon Rhew
- College of Pharmacy, Dongduk Women's University, Seoul, 02748, Republic of Korea
| | - Jiwon Park
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Yejee Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, 13620, Republic of Korea
| | - Myeong Gyu Kim
- College of Pharmacy, Ewha Womans University, Seoul, 03760, Republic of Korea; Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
| | - Kyungim Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea; Institute of Pharmaceutical Science, Korea University, Sejong, 30019, Republic of Korea.
| |
Collapse
|
11
|
Yamaji T, Yusoff FM, Kishimoto S, Kajikawa M, Yoshimura K, Nakano Y, Goto C, Harada T, Mizobuchi A, Tanigawa S, Maruhashi T, Higashi Y. Association of cumulative low-density lipoprotein cholesterol exposure with vascular function. J Clin Lipidol 2024; 18:e238-e250. [PMID: 38185588 DOI: 10.1016/j.jacl.2023.12.006] [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: 10/04/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND The relationship between cumulative low-density lipoprotein cholesterol (LDL-C) exposure and progression of atherosclerosis remains uncertain. OBJECTIVE The aim of this study was to determine the relationship between cumulative LDL-C level and flow-mediated vasodilation (FMD), nitroglycerine-induced vasodilation (NID) and the presence of plaque in the common carotid artery (CCA). METHODS This was a cross-sectional study. We measured FMD in 8208 subjects, NID in 1822 subjects, and CCA plaque in 591 subjects who were not taking lipid-lowering drugs. The subjects were divided into four groups based on cumulative LDL-C exposure: <4000 mg·year/dL, 4000-4999 mg·year/dL, 5000-5999 mg·year/dL, and ≥6000 mg·year/dL. RESULTS The odds ratio of the lower quartile of FMD in the cholesterol-year-score <4000 mg·year/dL group was significantly higher than the odds ratios in the other groups. The odds ratio of the lower quartile of NID in the <4000 mg·year/dL group was significantly higher than the odds ratios in the 5000-5999 mg·year/dL and ≥6000 mg·year/dL groups. The odds ratio of the prevalence of CCA plaque in the <4000 mg·year/dL group was significantly higher than that in the ≥6000 mg·year/dL group. CONCLUSIONS Endothelial dysfunction occurred from cumulative LDL-C exposure of 4000 mg·year/dL, vascular smooth muscle dysfunction occurred from cumulative LDL-C exposure of 5000 mg·year/dL, and prevalence of CCA plaque occurred from cumulative LDL-C exposure of 6000 mg·year/dL. CLINICAL TRIAL REGISTRY INFORMATION: http://www.umin.ac.jp (UMIN000012950, UMIN000012951, and UMIN000012952, UMIN000003409).
Collapse
Affiliation(s)
- Takayuki Yamaji
- Center for Radiation Disaster Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji and Higashi); Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (Drs Kajikawa, Yoshimura, and Higashi)
| | - Kenichi Yoshimura
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (Drs Kajikawa, Yoshimura, and Higashi)
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan (Dr Nakano)
| | - Chikara Goto
- Hiroshima International University, Hiroshima, Japan (Dr Goto)
| | - Takahiro Harada
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Aya Mizobuchi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Shunsuke Tanigawa
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Yukihito Higashi
- Center for Radiation Disaster Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji and Higashi); Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi); Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (Drs Kajikawa, Yoshimura, and Higashi).
| |
Collapse
|
12
|
Toprak K, Balaban İ, Pala S. The effect of successful lower extremity revascularization on aortic stiffness in patients with peripheral arterial disease. Vascular 2023; 31:1253-1261. [PMID: 36796873 DOI: 10.1177/17085381231153223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
OBJECTIVE Peripheral arterial disease (PAD) is a global health problem and associated with poor outcomes. It causes increased arterial stiffness. The association of PAD with aortic arterial stiffness was investigated in previous studies. However, there is limited data regarding the effect of peripheral revascularization on arterial stiffness. The aim of our study is to investigate the effect of peripheral revascularization on aortic stiffness parameters in patients with symptomatic PAD. METHODS A total of 48 patients with PAD who underwent peripheral revascularization were included in the study. Echocardiography was performed before and after the procedure, and aortic stiffness parameters were obtained by using aortic diameters and arterial blood pressure measurements. RESULTS Post-procedural aortic strain (5.1 [1.3-14] vs. 6.3 [2.8-6.3], p = 0.009) and aortic distensibility (0.2 [0.0-0.9] vs. 0.3 [0.1-1.1], p = 0.001) measurements were significantly increased compared to pre-procedural values. Patients were also compared according to the lesion laterality, site and treatment methods. It was found that the change in aortic strain (p = 0.031) and distensibility (p = 0.043) were significantly higher in unilateral lesion compared to bilateral lesion. Also, the change in aortic strain (p = 0.042) and distensibility (p = 0.033) were significantly higher in iliac site lesion compared to superficial femoral artery (SFA) site lesion. Moreover, the change in aortic strain was significantly higher (p = 0.013) in patients treated with stent compared to only balloon angioplasty. CONCLUSION Our study showed that successful percutaneous revascularization significantly reduced aortic stiffness in PAD. The change in aortic stiffness was significantly higher in unilateral lesions, iliac site lesions and stent-treated lesions.
Collapse
Affiliation(s)
- Kenan Toprak
- Department of Cardiology, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - İsmail Balaban
- Clinic of Cardiology, Kartal Koşuyolu High Specialization Training and Research Hospital, İstanbul, Turkey
| | - Selçuk Pala
- Clinic of Cardiology, Kartal Koşuyolu High Specialization Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
13
|
Numazaki H, Nasu T, Satoh M, Kotozaki Y, Tanno K, Asahi K, Ohmomo H, Shimizu A, Omama S, Morino Y, Sobue K, Sasaki M. Association between vascular endothelial dysfunction and stroke incidence in the general Japanese population: Results from the tohoku medical megabank community-based cohort study. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2023; 19:200216. [PMID: 37780457 PMCID: PMC10539892 DOI: 10.1016/j.ijcrp.2023.200216] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/12/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
Background Flow-mediated dilation (FMD) measures vascular endothelial function by evaluating the vasodilatory response of blood vessels to increased blood flow. Nevertheless, the association between FMD and stroke incidence in a general population remains unclear. This study investigated the association between vascular endothelial function and stroke incidence in the general Japanese population. Methods Based on cohort data from the Tohoku Medical Megabank Community-based Cohort Study, participants aged ≥18 years were recruited from Iwate Prefecture, with the final sample comprising 2952 subjects. Results The FMD level was 0.5%-27.1%, with a median of 5.0% (interquartile, 4.2%-11.3%). The mean follow-up period was 5.5 ± 1.8 years (range, 0.6-6.9 years). After dividing the participants into two subgroups according to the median FMD value, a multivariate Cox regression analysis adjusting for gender, age, smoking, alcohol consumption, systolic blood pressure, low-density lipoprotein cholesterol, estimated glomerular filtration rate, N-terminal pro-brain natriuretic peptide, high-sensitivity cardiac troponin T and hemoglobin A1c revealed that a lower FMD value was strongly associated with incidences of total stroke (hazard ratio[HR] = 2.13, 95% confidence interval[CI] = 1.48-3.07, p < 0.001), ischemic stroke (HR = 3.33, 95%CI = 2.00-5.52, p < 0.001), nonlacunar stroke (HR = 2.77, 95%CI = 1.49-5.16, p = 0.001), and lacunar stroke (HR = 5.12, 95%CI = 1.74-16.05, p = 0.003). Conclusions This study showed that a low FMD value might reflect vascular endothelial dysfunction and then was associated with ischemic stroke incidence in the general Japanese population, suggesting that FMD can be used as a tool to identify future stroke risk.
Collapse
Affiliation(s)
- Harutomo Numazaki
- Division of Cardiology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Takahito Nasu
- Division of Cardiology, Department of Internal Medicine, Iwate Medical University, Japan
- Department of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Japan
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Japan
| | - Mamoru Satoh
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Japan
| | - Yuka Kotozaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Japan
- Department of Hygiene and Preventive Medicine, Iwate Medical University, Japan
| | - Kozo Tanno
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Japan
- Department of Hygiene and Preventive Medicine, Iwate Medical University, Japan
| | - Koichi Asahi
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Japan
- Division of Nephrology and Hypertension, Department of Internal Medicine, Iwate Medical University, Japan
| | - Hideki Ohmomo
- Department of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Japan
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Japan
| | - Atsushi Shimizu
- Department of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Japan
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Japan
| | - Shinichi Omama
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Japan
- Division of General Medicine, Department of Critical Care, Disaster, And General Medicine, Iwate Medical University, Japan
| | - Yoshihiro Morino
- Department of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Japan
| | - Kenji Sobue
- Department of Neuroscience, Institute for Biomedical Sciences, Iwate Medical University, Japan
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Japan
- Division of Ultrahigh Field MRI, Institute for Biomedical Sciences, Iwate Medical University, Japan
| |
Collapse
|
14
|
Bosman M, Krüger DN, Favere K, De Meyer GRY, Franssen C, Van Craenenbroeck EM, Guns PJ. Dexrazoxane does not mitigate early vascular toxicity induced by doxorubicin in mice. PLoS One 2023; 18:e0294848. [PMID: 38015959 PMCID: PMC10684076 DOI: 10.1371/journal.pone.0294848] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/08/2023] [Indexed: 11/30/2023] Open
Abstract
Apart from cardiotoxicity, the chemotherapeutic agent doxorubicin (DOX) provokes acute and long-term vascular toxicity. Dexrazoxane (DEXRA) is an effective drug for treatment of DOX-induced cardiotoxicity, yet it remains currently unknown whether DEXRA prevents vascular toxicity associated with DOX. Accordingly, the present study aimed to evaluate the protective potential of DEXRA against DOX-related vascular toxicity in a previously-established in vivo and ex vivo model of vascular dysfunction induced by 16 hour (h) DOX exposure. Vascular function was evaluated in the thoracic aorta in organ baths, 16h after administration of DOX (4 mg/kg) or DOX with DEXRA (40 mg/kg) to male C57BL6/J mice. In parallel, vascular reactivity was evaluated after ex vivo incubation (16h) of murine aortic segments with DOX (1 μM) or DOX with DEXRA (10 μM). In both in vivo and ex vivo experiments, DOX impaired acetylcholine-stimulated endothelium-dependent vasodilation. In the ex vivo setting, DOX additionally attenuated phenylephrine-elicited vascular smooth muscle cell (VSMC) contraction. Importantly, DEXRA failed to prevent DOX-induced endothelial dysfunction and hypocontraction. Furthermore, RT-qPCR and Western blotting showed that DOX decreased the protein levels of topoisomerase-IIβ (TOP-IIβ), a key target of DEXRA, in the heart, but not in the aorta. Additionally, the effect of N-acetylcysteine (NAC, 10 μM), a reactive oxygen species (ROS) scavenger, was evaluated ex vivo. NAC did not prevent DOX-induced impairment of acetylcholine-stimulated vasodilation. In conclusion, our results show that DEXRA fails to prevent vascular toxicity resulting from 16h DOX treatment. This may relate to DOX provoking vascular toxicity in a ROS- and TOP-IIβ-independent way, at least in the evaluated acute setting. However, it is important to mention that these findings only apply to the acute (16h) treatment period, and further research is warranted to delineate the therapeutic potential of DEXRA against vascular toxicity associated with longer-term repetitive DOX dosing.
Collapse
Affiliation(s)
- Matthias Bosman
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
| | - Dustin N. Krüger
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
| | - Kasper Favere
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Guido R. Y. De Meyer
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Emeline M. Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
15
|
Bosman M, Krüger D, Van Assche C, Boen H, Neutel C, Favere K, Franssen C, Martinet W, Roth L, De Meyer GRY, Cillero-Pastor B, Delrue L, Heggermont W, Van Craenenbroeck EM, Guns PJ. Doxorubicin-induced cardiovascular toxicity: a longitudinal evaluation of functional and molecular markers. Cardiovasc Res 2023; 119:2579-2590. [PMID: 37625456 PMCID: PMC10676457 DOI: 10.1093/cvr/cvad136] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 06/19/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
AIMS Apart from cardiotoxicity, the chemotherapeutic doxorubicin (DOX) induces vascular toxicity, represented by arterial stiffness and endothelial dysfunction. Both parameters are of interest for cardiovascular risk stratification as they are independent predictors of future cardiovascular events in the general population. However, the time course of DOX-induced cardiovascular toxicity remains unclear. Moreover, current biomarkers for cardiovascular toxicity prove insufficient. Here, we longitudinally evaluated functional and molecular markers of DOX-induced cardiovascular toxicity in a murine model. Molecular markers were further validated in patient plasma. METHODS AND RESULTS DOX (4 mg/kg) or saline (vehicle) was administered intra-peritoneally to young, male mice weekly for 6 weeks. In vivo cardiovascular function and ex vivo arterial stiffness and vascular reactivity were evaluated at baseline, during DOX therapy (Weeks 2 and 4) and after therapy cessation (Weeks 6, 9, and 15). Left ventricular ejection fraction (LVEF) declined from Week 4 in the DOX group. DOX increased arterial stiffness in vivo and ex vivo at Week 2, which reverted thereafter. Importantly, DOX-induced arterial stiffness preceded reduced LVEF. Further, DOX impaired endothelium-dependent vasodilation at Weeks 2 and 6, which recovered at Weeks 9 and 15. Conversely, contraction with phenylephrine was consistently higher in the DOX-treated group. Furthermore, proteomic analysis on aortic tissue identified increased thrombospondin-1 (THBS1) and alpha-1-antichymotrypsin (SERPINA3) at Weeks 2 and 6. Up-regulated THBS1 and SERPINA3 persisted during follow-up. Finally, THBS1 and SERPINA3 were quantified in plasma of patients. Cancer survivors with anthracycline-induced cardiotoxicity (AICT; LVEF < 50%) showed elevated THBS1 and SERPINA3 levels compared with age-matched control patients (LVEF ≥ 60%). CONCLUSIONS DOX increased arterial stiffness and impaired endothelial function, which both preceded reduced LVEF. Vascular dysfunction restored after DOX therapy cessation, whereas cardiac dysfunction persisted. Further, we identified SERPINA3 and THBS1 as promising biomarkers of DOX-induced cardiovascular toxicity, which were confirmed in AICT patients.
Collapse
Affiliation(s)
- Matthias Bosman
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Dustin Krüger
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Charles Van Assche
- Research Group M4I—Imaging Mass Spectrometry (IMS); Faculty of Health, Medicine and Life Sciences, Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Hanne Boen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Cédric Neutel
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Kasper Favere
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Lynn Roth
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Berta Cillero-Pastor
- Research Group M4I—Imaging Mass Spectrometry (IMS); Faculty of Health, Medicine and Life Sciences, Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- Department of Cell Biology-Inspired Tissue Engineering, Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER Maastricht/Room C3.577, PO Box 616, Maastricht 6200 MD, The Netherlands
| | - Leen Delrue
- Department of Cardiology, Cardiovascular Center OLV Hospital Aalst, Moorselbaan 164, Aalst B-9300, Belgium
| | - Ward Heggermont
- Department of Cardiology, Cardiovascular Center OLV Hospital Aalst, Moorselbaan 164, Aalst B-9300, Belgium
- Department of Cardiology, Center for Molecular and Vascular Biology, KU Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| |
Collapse
|
16
|
Masuda S, Hara T, Yamagami H, Mitsui Y, Kurahashi K, Yoshida S, Harada T, Otoda T, Yuasa T, Nakamura S, Kuroda A, Endo I, Matsumoto T, Matsuhisa M, Abe M, Aihara KI. Vascular Endothelial Function Is Associated with eGFR Slope in Female and Non-Smoking Male Individuals with Cardiovascular Risk Factors: A Pilot Study on the Predictive Value of FMD for Renal Prognosis. J Atheroscler Thromb 2023; 30:1727-1741. [PMID: 37081616 PMCID: PMC10627745 DOI: 10.5551/jat.63987] [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: 10/19/2022] [Accepted: 03/16/2023] [Indexed: 04/22/2023] Open
Abstract
AIMS It is known that there are sex differences in vascular endothelial function and the development of chronic kidney diseases; however, it remains unclear whether sex differences influence the association between vascular endothelial function and renal prognosis. METHODS To clarify the relationship between vascular endothelial function and longitudinal eGFR changes in male and female patients with cardiovascular risk factors, we retrospectively evaluated 341 patients (176 males and 165 females) with cardiovascular risk factors in whom vascular function was assessed by flow-mediated dilation (FMD) and brachial-ankle pulse wave velocity (baPWV) and in whom 24-month longitudinal eGFR values were recorded after the vascular function examinations. Associations of values of FMD and baPWV with values of eGFR slope were statistically analyzed. RESULTS Simple regression analysis showed that the value of FMD was positively associated with eGFR slope in females (p=0.001) and non-smoking males (p=0.033) but not in smoking males. Multiple regression analysis showed that the value of FMD remains a positive contributor for eGFR slope in females (p=0.001) and non-smoking males (p=0.045) but not in smoking males. In contrast, values of baPWV had no significant association with eGFR slope regardless of sex and cigarette smoking. CONCLUSIONS In individuals with cardiovascular risk factors, evaluation of vascular endothelial function enables prediction of renal prognosis in females and non-smoking males.
Collapse
Affiliation(s)
- Shiho Masuda
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tomoyo Hara
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hiroki Yamagami
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yukari Mitsui
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kiyoe Kurahashi
- Department of Community Medicine for Respirology, Hematology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Sumiko Yoshida
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takeshi Harada
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Toshiki Otoda
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tomoyuki Yuasa
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Shingen Nakamura
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Akio Kuroda
- Diabetes Therapeutics and Research Center, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Itsuro Endo
- Department of Bioregulatory Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Munehide Matsuhisa
- Diabetes Therapeutics and Research Center, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Masahiro Abe
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ken-ichi Aihara
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Department of Internal Medicine, Anan Medical Center, Tokushima, Japan
| |
Collapse
|
17
|
Stone K, Veerasingam D, Meyer ML, Heffernan KS, Higgins S, Maria Bruno R, Bueno CA, Döerr M, Schmidt-Trucksäss A, Terentes-Printzios D, Voicehovska J, Climie RE, Park C, Pucci G, Bahls M, Stoner L. Reimagining the Value of Brachial-Ankle Pulse Wave Velocity as a Biomarker of Cardiovascular Disease Risk-A Call to Action on Behalf of VascAgeNet. Hypertension 2023; 80:1980-1992. [PMID: 37470189 PMCID: PMC10510846 DOI: 10.1161/hypertensionaha.123.21314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
This review critiques the literature supporting clinical assessment and management of cardiovascular disease and cardiovascular disease risk stratification with brachial-ankle pulse wave velocity (baPWV). First, we outline what baPWV actually measures-arterial stiffness of both large central elastic arteries and medium-sized muscular peripheral arteries of the lower limb. Second, we argue that baPWV is not a surrogate for carotid-femoral pulse wave velocity. While both measures are dependent on the properties of the aorta, baPWV is also strongly dependent on the muscular arteries of the lower extremities. Increased lower-extremity arterial stiffness amplifies and hastens wave reflections at the level of the aorta, widens pulse pressure, increases afterload, and reduces coronary perfusion. Third, we used an established evaluation framework to identify the value of baPWV as an independent vascular biomarker. There is sufficient evidence to support (1) proof of concept; (2) prospective validation; (3) incremental value; and (4) clinical utility. However, there is limited or no evidence to support (5) clinical outcomes; (6) cost-effectiveness; (8) methodological consensus; or (9) reference values. Fourth, we address future research requirements. The majority of the evaluation criteria, (1) proof of concept, (2) prospective validation, (3) incremental value, (4) clinical utility and (9) reference values, can be supported using existing cohort datasets, whereas the (5) clinical outcomes and (6) cost-effectiveness criteria require prospective investigation. The (8) methodological consensus criteria will require an expert consensus statement. Finally, we finish this review by providing an example of a future clinical practice model.
Collapse
Affiliation(s)
- Keeron Stone
- Centre for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, Wales, United Kingdom (K.S.)
- National Cardiovascular Research Network, Wales (K.S.)
| | - Dave Veerasingam
- Cardiothoracic Surgery, Galway University Hospital, Ireland (D.V.)
| | - Michelle L Meyer
- Department of Emergency Medicine, University of North Carolina at Chapel Hill (M.L.M.)
| | | | - Simon Higgins
- Department of Exercise and Sport Science, University of North Carolina, Chapel Hill (S.H., L.S.)
| | - Rosa Maria Bruno
- Université Paris Cité, Inserm, PARCC, France (R.M.B.)
- Clinical Pharmacology Unit, AP-HP, Hôpital européen Georges Pompidou, Paris, France (R.M.B.)
| | - Celia Alvarez Bueno
- Health and Social Research Center, Universidad de Castilla La Mancha, Cuenca, Spain (C.A.B.)
- Universidad Politécnica y Artística del Paraguay, Asunción, Paraguay (C.A.B.)
| | - Marcus Döerr
- Department of Internal Medicine B, University Medicine Greifswald, Germany (M.D., M.B.)
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany (M.D., M.B.)
| | - Arno Schmidt-Trucksäss
- Department of Sport, Exercise, and Health (A.S.-T.), University of Basel, Switzerland
- Department of Clinical Research, University Hospital Basel (A.S.-T.), University of Basel, Switzerland
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Athens Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Greece (D.T.-P.)
| | - Jūlija Voicehovska
- Internal Diseases Department, Riga Stradins University, Latvia (J.V.)
- Nephrology and Renal Replacement Clinics, Riga East University Hospital, Latvia (J.V.)
| | - Rachel E Climie
- Menzies Institute for Medical Research, University of Tasmania (R.E.C.)
| | - Chloe Park
- MRC Unit for Lifelong Health and Ageing at UCL, Institute of Cardiovascular Science, London, United Kingdom (C.P.)
| | - Giacomo Pucci
- Department of Medicine, University of Perugia, Unit of Internal Medicine, "Santa Maria" Terni Hospital, Italy (G.P.)
| | - Martin Bahls
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Germany (M.D., M.B.)
| | - Lee Stoner
- Department of Exercise and Sport Science, University of North Carolina, Chapel Hill (S.H., L.S.)
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill (L.S.)
- Center for Health Promotion and Disease Prevention, University of North Carolina at Chapel Hill (L.S.)
| |
Collapse
|
18
|
Tahsin CT, Michopoulos V, Powers A, Park J, Ahmed Z, Cullen K, Jenkins NDM, Keller-Ross M, Fonkoue IT. Sleep efficiency and PTSD symptom severity predict microvascular endothelial function and arterial stiffness in young, trauma-exposed women. Am J Physiol Heart Circ Physiol 2023; 325:H739-H750. [PMID: 37505472 PMCID: PMC10642999 DOI: 10.1152/ajpheart.00169.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/13/2023] [Accepted: 07/28/2023] [Indexed: 07/29/2023]
Abstract
Posttraumatic stress disorder (PTSD) is linked to sleep disturbances and significantly higher risk of developing cardiovascular disease (CVD). Furthermore, vascular dysfunction and sleep are independently associated with CVD. Uncovering the link between PTSD symptom severity, sleep disturbances, and vascular function could shine a light on mechanisms of CVD risk in trauma-exposed young women. The purpose of the present study was to investigate the individual and combined effects of sleep efficiency and PTSD symptom severity on vascular function. We recruited 60 otherwise healthy women [age, 26 ± 7 yr and body mass index (BMI), 27.7 ± 6.5 kg/m2] who had been exposed to trauma. We objectively quantified sleep efficiency (SE) using actigraphy, microvascular endothelial function via Framingham reactive hyperemia index (fRHI), and arterial stiffness via pulse-wave velocity (PWV). PTSD symptom severity was assessed using the PTSD checklist for fifth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5) (PCL5). PWV was correlated with age (r = 0.490, P < 0.001) and BMI (r = 0.484, P < 0.001). In addition, fRHI was positively correlated with SE (r = 0.409, P = 0.001) and negatively correlated with PTSD symptoms (r = -0.382, P = 0.002). To explore the predictive value of SE and PTSD symptoms on PWV and fRHI, we conducted two multivariate linear regression models. The model predicting PWV was significant (R2 = 0.584, P < 0.001) with age, BMI, blood pressure, and SE emerging as predictors. Likewise, the model predicting fRHI was significant (R2 = 0.360, P < 0.001) with both PTSD symptoms and SE as significant predictors. Our results suggest that although PTSD symptoms mainly impact microvascular endothelial function, sleep efficiency is additionally associated with arterial stiffness in young trauma-exposed women, after controlling for age and BMI.NEW & NOTEWORTHY This is the first study to investigate the individual and combined impacts of objective sleep and PTSD symptoms severity on arterial stiffness and microvascular endothelial function in young premenopausal women. We report that in young trauma-exposed women, although low sleep efficiency is associated with overall vascular function (i.e., microvascular endothelial function and arterial stiffness), the severity of PTSD symptoms is specifically associated with microvascular endothelial function, after accounting for age and body mass index.
Collapse
Affiliation(s)
- Chowdhury Tasnova Tahsin
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Vasiliki Michopoulos
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Abigail Powers
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Jeanie Park
- Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Veterans Affairs, Research Service Line, Atlanta Veterans Affairs Healthcare Systems, Decatur, Georgia, United States
| | - Zynab Ahmed
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Kathryn Cullen
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Nathaniel D M Jenkins
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa, United States
- Abboud Cardiovascular Research Center, University of Iowa, Iowa City, Iowa, United States
| | - Manda Keller-Ross
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Ida T Fonkoue
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| |
Collapse
|
19
|
Tomiyama H. Vascular function: a key player in hypertension. Hypertens Res 2023; 46:2145-2158. [PMID: 37369849 DOI: 10.1038/s41440-023-01354-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/06/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023]
Abstract
The major functions of the arterial system are to "efficiently deliver blood to the peripheral organs and maintain vascular homeostasis". Both the endothelial and medial layer contribute to the three major functions, namely, conversion of pulsatile to steady blood flow, appropriate distribution of blood flow to the target organs, and vascular protection and homeostasis. Vascular dysfunction contributes to the development of cardiovascular diseases through a combination of several mechanisms, including impaired coronary perfusion, cardiac systolic/diastolic dysfunction, microvascular damage, and abnormal hemodynamics in the arterial tree. The representative marker of endothelial function is flow-mediated vasodilatation and that of the medial layer function is pulse wave velocity, and that of the blood supply function of the arterial tree is the ankle-brachial pressure index. In hypertension, vascular dysfunction could also lead to the development of isolated systolic hypertension, isolated diastolic hypertension, and systolic/diastolic hypertension. Vascular dysfunction is involved in a vicious cycle with abnormal blood pressure variability. Furthermore, a vicious cycle may also exist between vascular dysfunction and hypertension. While the significances of vascular function tests to predict future cardiovascular events has been established in cases of hypertension, their usefulness in assessing the effectiveness of management of the vascular functions in hypertension on the cardiovascular outcomes has not yet been fully clarified. Thus, vascular dysfunction plays crucial roles in the pathophysiology of hypertension, and further research is warranted to establish strategies to improve vascular dysfunction in cases of hypertension. Vascular functions in the pathophysiology of hypertension. Vascular dysfunction and elevation of blood pressure are components of a vicious cycle even from their early stages, which including abnormal blood pressure variabilities. This vicious cycle is associated with hypertensive organ damage and also adverse cardiovascular outcomes. Strategies to break this vicious cycle have not yet been fully established.
Collapse
Affiliation(s)
- Hirofumi Tomiyama
- Department of Cardiology, Tokyo Medical University, Tokyo, Japan.
- Division of Preemptive Medicine for Vascular Damage, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
20
|
Climie RE, Alastruey J, Mayer CC, Schwarz A, Laucyte-Cibulskiene A, Voicehovska J, Bianchini E, Bruno RM, Charlton PH, Grillo A, Guala A, Hallab M, Hametner B, Jankowski P, Königstein K, Lebedeva A, Mozos I, Pucci G, Puzantian H, Terentes-Printzios D, Yetik-Anacak G, Park C, Nilsson PM, Weber T. Vascular ageing: moving from bench towards bedside. Eur J Prev Cardiol 2023; 30:1101-1117. [PMID: 36738307 PMCID: PMC7614971 DOI: 10.1093/eurjpc/zwad028] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023]
Abstract
Prevention of cardiovascular disease (CVD) remains one of the largest public health challenges of our time. Identifying individuals at increased cardiovascular risk at an asymptomatic, sub-clinical stage is of paramount importance for minimizing disease progression as well as the substantial health and economic burden associated with overt CVD. Vascular ageing (VA) involves the deterioration in vascular structure and function over time and ultimately leads to damage in the heart, brain, kidney, and other organs. Vascular ageing encompasses the cumulative effect of all cardiovascular risk factors on the arterial wall over the life course and thus may help identify those at elevated cardiovascular risk, early in disease development. Although the concept of VA is gaining interest clinically, it is seldom measured in routine clinical practice due to lack of consensus on how to characterize VA as physiological vs. pathological and various practical issues. In this state-of-the-art review and as a network of scientists, clinicians, engineers, and industry partners with expertise in VA, we address six questions related to VA in an attempt to increase knowledge among the broader medical community and move the routine measurement of VA a little closer from bench towards bedside.
Collapse
Affiliation(s)
- Rachel E. Climie
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool St, 7000 Hobart, Australia
- Sports Cardiology, Baker Heart and Diabetes Institute, 99 Commercial Rd, Melbourne 3000, Australia
- Integrative Epidemiology of Cardiovascular Disease, Université de Paris, INSERM, U970, Paris Cardiovascular Research Center (PARCC), 56 rue Leblanc, 75015 Paris, France
| | - Jordi Alastruey
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King’s College London, 249 Westminster Bridge Rd, London SE1 7EH, UK
| | - Christopher C. Mayer
- Medical Signal Analysis, Center for Health & Bioresources, AIT Austrian Institute of Technology, Giefinggasse 4, 1210 Vienna, Austria
| | - Achim Schwarz
- ALF Distribution GmbH, Stephanstrasse 19, 52064 Aachen, Germany
| | - Agne Laucyte-Cibulskiene
- Department of Clinical Sciences, Lund University, Skane University Hospital, Sölvegatan 19 - BMC F12, 221 84 Lund, Malmö, Sweden
- Faculty of Medicine, Vilnius University, M. K. C iurlionio g. 21, 03101 Vilnius, Lithuania
| | - Julija Voicehovska
- Department of Internal Diseases, Riga Stradins University, Dzirciema str. 16, Riga, L-1007, Latvia
- Nephrology and Renal Replacement Therapy Clinics, Riga East University Hospital, Hipokrata str. 2, Riga, LV-1079, Latvia
| | - Elisabetta Bianchini
- Institute of Clinical Physiology, Italian National Research Council (CNR), Via Moruzzi, 1, 56124 Pisa (PI), Italy
| | - Rosa-Maria Bruno
- Integrative Epidemiology of Cardiovascular Disease, Université de Paris, INSERM, U970, Paris Cardiovascular Research Center (PARCC), 56 rue Leblanc, 75015 Paris, France
| | - Peter H. Charlton
- Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, 2 Worts Causeway, Cambridge CB1 8RN, UK
| | - Andrea Grillo
- Medicina Clinica, Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Andrea Guala
- Vall d’Hebron Institut de Recerca (VHIR), Paseo de la Vall d’Hebron, 129, 08035 Barcelona, Spain
| | - Magid Hallab
- Clinique Bizet, 23 Georges Bizet, 75116 Paris, France
| | - Bernhard Hametner
- Medical Signal Analysis, Center for Health & Bioresources, AIT Austrian Institute of Technology, Giefinggasse 4, 1210 Vienna, Austria
| | - Piotr Jankowski
- Department of Internal Medicine and Geriatric Cardiology, Centre of Postgraduate Medical Education, 231 Czerniakowska St., 00-416 Warsaw, Poland
| | - Karsten Königstein
- Department of Sport, Exercise and Health (DSBG) University of Basel, Grosse Allee 6, 4052 Basel, Switzerland
| | - Anna Lebedeva
- Department of Internal Medicine and Cardiology, Dresden Heart Centre, Dresden University of Technology, Fetscher str. 76, 01307 Dresden, Germany
| | - Ioana Mozos
- Department of Functional Sciences-Pathophysiology, Center for Translational Research and Systems Medicine, ‘Victor Babes’ University of Medicine and Pharmacy, T. Vladimirescu Street 14, 300173 Timisoara, Romania
| | - Giacomo Pucci
- Unit of Internal Medicine, Terni University Hospital - Department of Medicine and Surgery, University of Perugia, Terni, Italy
| | - Houry Puzantian
- Hariri School of Nursing, American University of Beirut, P.O. Box 11-0236, Riad El Solh 1107 2020, Beirut, Lebanon
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, 114 Vasilissis Sofias Avenue, 11527 Athens, Greece
| | - Gunay Yetik-Anacak
- Department of Pharmacology, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, Kayisdagi Cad. No:32 Atasehir, 34752 Istanbul, Turkey
| | - Chloe Park
- MRC Unit for Lifelong Health and Ageing at UCL, 1-19 Torrington Place, London WC1E 7HB, UK; and
| | - Peter M. Nilsson
- Department of Clinical Sciences, Lund University, Skane University Hospital, Sölvegatan 19 - BMC F12, 221 84 Lund, Malmö, Sweden
| | - Thomas Weber
- Cardiology Department, Klinikum Wels-Grieskirchen, Grieskirchnerstrasse 42, 4600 Wels, Austria
| |
Collapse
|
21
|
Yanai H, Adachi H, Hakoshima M, Katsuyama H. Significance of Endothelial Dysfunction Amelioration for Sodium-Glucose Cotransporter 2 Inhibitor-Induced Improvements in Heart Failure and Chronic Kidney Disease in Diabetic Patients. Metabolites 2023; 13:736. [PMID: 37367894 DOI: 10.3390/metabo13060736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Beyond lowering plasma glucose levels, sodium-glucose cotransporter 2 inhibitors (SGLT2is) significantly reduce hospitalization for heart failure (HF) and retard the progression of chronic kidney disease (CKD) in patients with type 2 diabetes. Endothelial dysfunction is not only involved in the development and progression of cardiovascular disease (CVD), but is also associated with the progression of CKD. In patients with type 2 diabetes, hyperglycemia, insulin resistance, hyperinsulinemia and dyslipidemia induce the development of endothelial dysfunction. SGLT2is have been shown to improve endothelial dysfunction, as assessed by flow-mediated vasodilation, in individuals at high risk of CVD. Along with an improvement in endothelial dysfunction, SGLT2is have been shown to improve oxidative stress, inflammation, mitochondrial dysfunction, glucotoxicity, such as the advanced signaling of glycation end products, and nitric oxide bioavailability. The improvements in endothelial dysfunction and such endothelium-derived factors may play an important role in preventing the development of coronary artery disease, coronary microvascular dysfunction and diabetic cardiomyopathy, which cause HF, and play a role in retarding CKD. The suppression of the development of HF and the progression of CKD achieved by SGLT2is might have been largely induced by their capacity to improve vascular endothelial function.
Collapse
Affiliation(s)
- Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| | - Hiroki Adachi
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| | - Mariko Hakoshima
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| | - Hisayuki Katsuyama
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| |
Collapse
|
22
|
da Silva DVT, Baião DDS, Almeida CC, Paschoalin VMF. A Critical Review on Vasoactive Nutrients for the Management of Endothelial Dysfunction and Arterial Stiffness in Individuals under Cardiovascular Risk. Nutrients 2023; 15:nu15112618. [PMID: 37299579 DOI: 10.3390/nu15112618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Pathophysiological conditions such as endothelial dysfunction and arterial stiffness, characterized by low nitric oxide bioavailability, deficient endothelium-dependent vasodilation and heart effort, predispose individuals to atherosclerotic lesions and cardiac events. Nitrate (NO3-), L-arginine, L-citrulline and potassium (K+) can mitigate arterial dysfunction and stiffness by intensifying NO bioavailability. Dietary compounds such as L-arginine, L-citrulline, NO3- and K+ exert vasoactive effects as demonstrated in clinical interventions by noninvasive flow-mediated vasodilation (FMD) and pulse-wave velocity (PWV) prognostic techniques. Daily L-arginine intakes ranging from 4.5 to 21 g lead to increased FMD and reduced PWV responses. Isolated L-citrulline intake of at least 5.6 g has a better effect compared to watermelon extract, which is only effective on endothelial function when supplemented for longer than 6 weeks and contains at least 6 g of L-citrulline. NO3- supplementation employing beetroot at doses greater than 370 mg promotes hemodynamic effects through the NO3--NO2-/NO pathway, a well-documented effect. A potassium intake of 1.5 g/day can restore endothelial function and arterial mobility, where decreased vascular tone takes place via ATPase pump/hyperpolarization and natriuresis, leading to muscle relaxation and NO release. These dietary interventions, alone or synergically, can ameliorate endothelial dysfunction and should be considered as adjuvant therapies in cardiovascular diseases.
Collapse
Affiliation(s)
- Davi Vieira Teixeira da Silva
- Instituto de Química, Programa de Pós-Graduação em Ciência de Alimentos e Programa de Pós-Graduação em Química, Universidade Federal do Rio de Janeiro, Av. Athos da Silveira Ramos 149, sala 545, Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
| | - Diego Dos Santos Baião
- Instituto de Química, Programa de Pós-Graduação em Ciência de Alimentos e Programa de Pós-Graduação em Química, Universidade Federal do Rio de Janeiro, Av. Athos da Silveira Ramos 149, sala 545, Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
| | - Cristine Couto Almeida
- Instituto de Química, Programa de Pós-Graduação em Ciência de Alimentos e Programa de Pós-Graduação em Química, Universidade Federal do Rio de Janeiro, Av. Athos da Silveira Ramos 149, sala 545, Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
| | - Vania Margaret Flosi Paschoalin
- Instituto de Química, Programa de Pós-Graduação em Ciência de Alimentos e Programa de Pós-Graduação em Química, Universidade Federal do Rio de Janeiro, Av. Athos da Silveira Ramos 149, sala 545, Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
| |
Collapse
|
23
|
Huang XD, Lin JY, Huang XW, Zhou TT, Xie LD. A nomogram based on endothelial function and conventional risk factors predicts coronary artery disease in hypertensives. BMC Cardiovasc Disord 2023; 23:217. [PMID: 37118701 PMCID: PMC10148409 DOI: 10.1186/s12872-023-03235-6] [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: 12/15/2022] [Accepted: 04/11/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND There is currently a lack of a precise, concise, and practical clinical prediction model for predicting coronary artery disease (CAD) in patients with essential hypertension (EH). This study aimed to construct a nomogram to predict CAD in patients with EH based on flow-mediated dilation (FMD) of brachial artery and traditional risk factors. METHODS Clinical data of 1752 patients with EH were retrospectively collected. High-resolution vascular ultrasound was used to detect FMD in all patients at the Fujian Hypertension Research Institute, China. Patients were divided into two groups, i.e. training group (n = 1204, from August 2000 to December 2013) and validation group (n = 548, from January 2014 to May 2016) according to the time of enrollment. Independent predictors of CAD were analyzed by multivariable logistic regression in the training group, and a nomogram was constructed accordingly. Finally, we evaluated the discrimination, calibration, and clinical applicability of the model using the area under curve (AUC) of receiver operating characteristic analysis, calibration curve combined with Hosmer-Lemeshow test, and decision curve, respectively. RESULTS There were 263 (21.8%) cases of EH combined with CAD in the training group. Multivariate logistic regression showed that FMD, age, duration of EH, waist circumference, and diabetes mellitus were independent influencing factors for CAD in EH patients. Smoking which was close to statistical significance (P = 0.062) was also included in the regression model to increase the accuracy. Ultimately, the nomogram for predicting CAD in EH patients was constructed according to above predictors after proper transformation. The AUC values of the training group and the validation group were 0.799 (95%CI 0.770-0.829) and 0.836 (95%CI 0.787-0.886), respectively. Calibration curve and Hosmer-Lemeshow test showed that the model had good calibration (training group: χ2 = 0.55, P = 0.759; validation group: χ2 = 1.62, P = 0.446). The decision curve also verified the clinical applicability of the nomogram. CONCLUSION The nomogram based on FMD and traditional risk factors (age, duration of EH disease, smoking, waist circumference and diabetes mellitus) can predict CAD high-risk group among patients with EH.
Collapse
Affiliation(s)
- Xiao-Dong Huang
- Department of Emergency, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Xiamen Key Laboratory for Clinical Efficacy and Evidence-Based Research of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Ji-Yan Lin
- Department of Emergency, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
- Xiamen Key Laboratory for Clinical Efficacy and Evidence-Based Research of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Xian-Wei Huang
- Department of Emergency, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
- Xiamen Key Laboratory for Clinical Efficacy and Evidence-Based Research of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Ting-Ting Zhou
- Department of Cardiovascular Medicine, Xiamen Haicang Hospital, Xiamen, 361026, China
| | - Liang-Di Xie
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
24
|
Xiao K, Liu M, Sun X, Zhang Y, Si J, Shi N, Sun L, Miao Z, Zhang H, Zhao T, Liu Z, Fan Z, Gao J, Li J. Association of vascular endothelial function and quality of life in patients with ischemia and non-obstructive coronary artery disease. Heart Vessels 2023; 38:617-625. [PMID: 36598570 DOI: 10.1007/s00380-022-02213-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 12/01/2022] [Indexed: 01/05/2023]
Abstract
Improvements are required in the quality of life (QoL) of patients with ischemia and non-obstructive coronary artery disease (INOCA). Several patients with INOCA experience vascular endothelial dysfunction. However, the relationship between endothelial function and QoL remains unelucidated. This study aimed to initially investigate the relationship between endothelial function and QoL in patients with INOCA. This prospective observational study included 121 patients with INOCA (aged 31-85 years). Vascular endothelial function was assessed by flow-mediated dilatation (FMD) of the peripheral brachial artery. QoL was evaluated using the 36-Item Short-Form Health Survey (SF-36). Patients with INOCA were divided into two groups according to the median FMD change during the 1-year follow-up (group A, ≥ median FMD change cut-off; group B, < median FMD change cut-off). The median change in FMD was 0.92%. The mean baseline SF-36 scores were comparable between the two groups (53.95 ± 6.46 vs. 53.92 ± 4.29, p = 0.98). The QoL at follow-up was better in group A than in group B (56.61 ± 5.50 vs. 53.32 ± 5.58, p = 0.002). The change in FMD (r = 0.34, p < 0.01), rather than FMD at baseline (r = - 0.01, p = 0.89) or follow-up (r = 0.13, p = 0.15), was related to the follow-up SF-36 scores. FMD improvement was an independent predictor of increased QoL (odds ratio, 3.90; 95% confidence interval: 1.59-9.53, p = 0.003). Endothelial function change is associated with QoL, and patients with improved endothelial function have a better QoL than those without.
Collapse
Affiliation(s)
- Keling Xiao
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Machao Liu
- Division of Cardiology, Xuanwu Hospital Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Xipeng Sun
- Division of Cardiology, Xuanwu Hospital Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Yinghua Zhang
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Jin Si
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Ning Shi
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Lijie Sun
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Zupei Miao
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Haoyu Zhang
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Ting Zhao
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Zhi Liu
- Division of Cardiology, Xuanwu Hospital Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Zhenxing Fan
- Division of Cardiology, Xuanwu Hospital Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Jing Gao
- Division of Cardiology, Xuanwu Hospital Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
| | - Jing Li
- Department of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
25
|
Kishimoto S, Oda N, Maruhashi T, Tanigawa S, Mizobuchi A, Yusoff FM, Fujita A, Uchiki T, Kajikawa M, Yoshimura K, Yamaji T, Harada T, Hashimoto Y, Nakano Y, Hirota S, Yoshinaga S, Goto C, Nakashima A, Higashi Y. Effect of exposure to radiation caused by an atomic bomb on endothelial function in atomic bomb survivors. Front Cardiovasc Med 2023; 10:1122794. [PMID: 36873409 PMCID: PMC9981625 DOI: 10.3389/fcvm.2023.1122794] [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: 12/13/2022] [Accepted: 02/03/2023] [Indexed: 02/19/2023] Open
Abstract
Background The purpose of this study was to evaluate the effects of exposure to radiation caused by an atomic bomb in atomic bomb survivors on vascular function and vascular structure and to evaluate the relationships of radiation dose from the atomic bomb with vascular function and vascular structure in atomic bomb survivors. Methods Flow-mediated vasodilation (FMD) and nitroglycerine-induced vasodilation (NID) as indices of vascular function, brachial-ankle pulse wave velocity (baPWV) as an index of vascular function and vascular structure, and brachial artery intima-media thickness (IMT) as an index of vascular structure were measured in 131 atomic bomb survivors and 1,153 control subjects who were not exposed to the atomic bomb. Ten of the 131 atomic bomb survivors with estimated radiation dose in a cohort study of Atomic Bomb Survivors in Hiroshima were enrolled in the study to evaluate the relationships of radiation dose from the atomic bomb with vascular function and vascular structure. Results There was no significant difference in FMD, NID, baPWV, or brachial artery IMT between control subjects and atomic bomb survivors. After adjustment of confounding factors, there was still no significant difference in FMD, NID, baPWV, or brachial artery IMT between control subjects and atomic bomb survivors. Radiation dose from the atomic bomb was negatively correlated with FMD (ρ = -0.73, P = 0.02), whereas radiation dose was not correlated with NID, baPWV or brachial artery IMT. Conclusion There were no significant differences in vascular function and vascular structure between control subjects and atomic bomb survivors. Radiation dose from the atomic bomb might be negatively correlated with endothelial function.
Collapse
Affiliation(s)
- Shinji Kishimoto
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Nozomu Oda
- Department of Cardiovascular Medicine, Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shunsuke Tanigawa
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Aya Mizobuchi
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Asuka Fujita
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.,Plastic and Reconstructive Surgery, Hiroshima University Hospital, Hiroshima, Japan
| | - Toshio Uchiki
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.,Plastic and Reconstructive Surgery, Hiroshima University Hospital, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Kenichi Yoshimura
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Takayuki Yamaji
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yu Hashimoto
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Seiko Hirota
- Department of Environmetrics and Biometrics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shinji Yoshinaga
- Department of Environmetrics and Biometrics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Chikara Goto
- Department of Rehabilitation, Faculty of General Rehabilitation, Hiroshima International University, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukihito Higashi
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.,Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
26
|
Santos V, Massuça LM, Angarten V, Melo X, Pinto R, Fernhall B, Santa-Clara H. Arterial Stiffness Response to Acute Combined Training with Different Volumes in Coronary Artery Disease and Heart Failure Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14994. [PMID: 36429714 PMCID: PMC9690817 DOI: 10.3390/ijerph192214994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
Resistance training has been shown to acutely increase arterial stiffness (AS), while endurance training appears to decrease AS. However, the findings are from studies in apparently healthy subjects and have limited applicability to patients at low and high cardiovascular risk, for whom combined exercise is recommended. We compared the time course of changes in local and regional indices of AS in response to high-volume combined endurance training (CET) and high-volume combined resistance training (CRT) in patients with coronary artery disease (CAD) and heart failure (HF). We studied 20 men with CAD and HF (10 each) aged 68.3 ± 9.6 years. AS was measured by pulse wave velocity (PWV), and brachial and central blood pressure (BP) were determined after 15 min of rest and 5 and 15 min after the exercise session. All patients completed two sessions on nonconsecutive days. A protocol by time interaction effect was observed for carotid (η2 = 0.21, p = 0.02), aortic (η2 = 0.60, p < 0.001), and femoral (η2 = 0.46, p = 0.01) PWV after CET and CRT, suggesting that PWV decreased after CET and increased after CRT. Decreases in the brachial and central variables of BP across time points were observed in both protocols. CET decreased whereas CRT increased carotid, aortic, and femoral PWV at 15 min after exercise in patients with CAD and HF.
Collapse
Affiliation(s)
- Vanessa Santos
- Exercise and Health Laboratory, CIPER, Faculdade de Motricidade Humana, Universidade de Lisboa, Cruz Quebrada, 1649-004 Lisboa, Portugal
- KinesioLab, Research Unit in Human Movement Analysis, Instituto Piaget, 2805-059 Almada, Portugal
| | - Luís Miguel Massuça
- ICPOL Research Center, Higher Institute of Police Sciences and Internal Security, 1300-352 Lisbon, Portugal
- CIDEFES—Research Center in Sport, Physical Education, Exercise and Health, Lusófona University, 1749-024 Lisbon, Portugal
| | - Vitor Angarten
- Exercise and Health Laboratory, CIPER, Faculdade de Motricidade Humana, Universidade de Lisboa, Cruz Quebrada, 1649-004 Lisboa, Portugal
| | - Xavier Melo
- Egas Moniz Interdisciplinary Research Center (CiiEM), Egas Moniz School of Health, 2829-511 Almada, Portugal
| | - Rita Pinto
- Structural and Coronary Heart Disease Unit, Centro Cardiovascular da Universidade de Lisboa (CCUL@RISE), Faculty of Medicine, University of Lisbon, 1649-004 Lisboa, Portugal
| | - Bo Fernhall
- College of Nursing and Health Sciences, University of Massachusetts, 100 Morrissey Boulevard, Boston, MA 02125, USA
| | - Helena Santa-Clara
- Exercise and Health Laboratory, CIPER, Faculdade de Motricidade Humana, Universidade de Lisboa, Cruz Quebrada, 1649-004 Lisboa, Portugal
| |
Collapse
|
27
|
Ziganshina MM, Ziganshin AR, Khalturina EO, Baranov II. Arterial hypertension as a consequence of endothelial glycocalyx dysfunction: a modern view of the problem of cardiovascular diseases. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2022. [DOI: 10.15829/1728-8800-2022-3316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Arterial hypertension (AH) is a leading risk factor for the development of cardiovascular, cerebrovascular, and renal diseases, which are among the top 10 most common causes of death in the world. The etiology of hypertension has not been fully elucidated, but it has been established that endothelial dysfunction is the most significant pathogenetic link in the formation and progression of the disease. The data obtained in the last 10-15 years on endothelial glycocalyx (eGC) studies indicate that endothelial dysfunction is preceded by destabilization and shedding of eGC with the appearance of its soluble components in the blood, which is equivalent to a process that can be designated as eGC dysfunction. Signs of eGC dysfunction are expressed in the development of hypertension, diseases of the cardiovascular system, and their complications. The purpose of this review is to analyze and substantiate the pathophysiological role of eGC dysfunction in hypertension and cardiovascular diseases and to describe approaches for its assessment and pharmacological correction. Abstracts and full-size articles of 425 publications in Pubmed/MEDLINE databases over 20 years were studied. The review discusses the role of eGC in the regulation of vascular tone, endothelial barrier function, and anti-adhesive properties of eGC. Modifications of eGC under the influence of pro-inflammatory stimuli, changes in eGC with age, and with increased salt load are considered. The aspect associated with eGC dysfunction in atherosclerosis, hyperglycemia and hypertension is covered. Assessment of eGC dysfunction is difficult but can be performed by indirect methods, in particular by detecting eGC components in blood. A brief description of the main approaches to pharmacoprevention and pharmacocorrection of hypertension is given from the position of exposure effects on eGC, which currently has more a fundamental than practical orientation. This opens up great opportunities for clinical studies of eGC dysfunction for the prevention and treatment of hypertension and justifies a new direction in the clinical pharmacology of antihypertensive drugs.
Collapse
Affiliation(s)
- M. M. Ziganshina
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology
| | - A. R. Ziganshin
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology
| | - E. O. Khalturina
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology;
I. M. Sechenov First Moscow State Medical University (Sechenov University)
| | - I. I. Baranov
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology
| |
Collapse
|
28
|
Higashi Y. Beneficial Effects of Supplementation on Endothelial Function: What Mechanisms? J Atheroscler Thromb 2022; 29:1268-1270. [PMID: 34955494 PMCID: PMC9444803 DOI: 10.5551/jat.ed189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Affiliation(s)
- Yukihito Higashi
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital
| |
Collapse
|
29
|
Abe S, Haruyama Y, Kobashi G, Toyoda S, Inoue T, Tomiyama H, Ishizu T, Kohro T, Higashi Y, Takase B, Suzuki T, Ueda S, Yamazaki T, Furumoto T, Kario K, Koba S, Takemoto Y, Hano T, Sata M, Ishibashi Y, Node K, Maemura K, Ohya Y, Furukawa T, Ito H, Yamashina A. Effect of Novel Stratified Lipid Risk by "LDL-Window" and Flow-Mediated Dilation on the Prognosis of Coronary Artery Disease Using the FMD-J Study A Data. Circ J 2022; 86:1444-1454. [PMID: 35871575 DOI: 10.1253/circj.cj-21-1068] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
BACKGROUND Elevated levels of triglyceride (TG) and non-high-density lipoprotein cholesterol (non-HDL-C) are regarded as a residual lipid risk in low-density lipoprotein cholesterol (LDL-C)-lowering therapy. This study investigated the association between lipid risk stratified by TG and non-HDL-C and the prognosis of patients with coronary artery disease (CAD), and the association between stratified lipid risk and flow-mediated dilatation (FMD) index. METHODS AND RESULTS The 624 CAD patients enrolled in flow-mediated dilation (FMD)-J study A were divided into 4 groups: low-risk group (n=413) with TG <150 mg/dL and non-HDL-C <170 mg/dL; hyper-TG group (n=180) with TG ≥150 mg/dL and non-HDL-C <170 mg/dL; hyper-non-HDL group (n=12) with TG <150 mg/dL and non-HDL-C ≥170 mg/dL; and high-risk group (n=19) with TG ≥150 mg/dL and non-HDL-C ≥170 mg/dL. Comparison of the groups showed the cumulative incidence of a 3-point major adverse cardiovascular event (MACE) was different and highest in the high-risk group in all the patients (P=0.009), and in patients with a FMD index ≥7.0% (P=0.021), but not in those with a FMD index <7.0%. Multivariable regression analysis showed that high lipid risk (P=0.019) and FMD <7.0% (P=0.040) were independently correlated with the incidence of a 3-point MACE. CONCLUSIONS Novel stratification of lipid risk, simply using TG and non-HDL-C levels, combined with FMD measurement, is useful for predicting cardiovascular outcomes in patients with CAD.
Collapse
Affiliation(s)
- Shichiro Abe
- Department of Cardiovascular Medicine, Dokkyo Medical University School of Medicine
| | - Yasuo Haruyama
- Center for Research Collaboration and Support, Dokkyo Medical University School of Medicine
- Department of Public Health, Dokkyo Medical University School of Medicine
| | - Gen Kobashi
- Center for Research Collaboration and Support, Dokkyo Medical University School of Medicine
- Department of Public Health, Dokkyo Medical University School of Medicine
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University School of Medicine
| | - Teruo Inoue
- Department of Cardiovascular Medicine, Dokkyo Medical University School of Medicine
- Nasu Red Cross Hospital
| | | | - Tomoko Ishizu
- Cardiovascular Division, Institute of Clinical Medicine, University of Tsukuba
| | - Takahide Kohro
- Department of Hospital Planning and Management, Medical Informatics, Jichi Medical University School of Medicine
| | - Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine
| | - Bonpei Takase
- Division of Biomedical Engineering, National Defense Medical College Research Institute
| | - Toru Suzuki
- Cardiovascular Medicine, University of Leicester
| | - Shinichiro Ueda
- Department of Clinical Pharmacology and Therapeutics, University of the Ryukyus School of Medicine
| | - Tsutomu Yamazaki
- Department of Clinical Epidemiology and Systems, Faculty of Medicine, The University of Tokyo
| | | | - Kazuomi Kario
- Division of Cardiovascular Medicine, Jichi Medical University School of Medicine
| | - Shinji Koba
- Department of Medicine, Division of Cardiology, Showa University School of Medicine
| | - Yasuhiko Takemoto
- Department of Internal Medicine and Cardiology, Osaka City University Graduate School of Medicine
| | - Takuzo Hano
- Department of Medical Education and Population-based Medicine, Postgraduate School of Medicine, Wakayama Medical University
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School
| | - Yutaka Ishibashi
- Department of General Medicine, Shimane University Faculty of Medicine
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University
| | - Koji Maemura
- Department of Cardiovascular Medicine, Course of Medical and Dental Sciences, Graduate School of Biomedical Sciences, Nagasaki University
| | - Yusuke Ohya
- The Third Department of Internal Medicine, University of the Ryukyus
| | - Taiji Furukawa
- Department of Internal Medicine, Teikyo University School of Medicine
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | | |
Collapse
|
30
|
Long-Term Prognosis after Coronary Artery Bypass Grafting: The Impact of Arterial Stiffness and Multifocal Atherosclerosis. J Clin Med 2022; 11:jcm11154585. [PMID: 35956199 PMCID: PMC9369624 DOI: 10.3390/jcm11154585] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/24/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
The aim of the study was to study the effect of arterial stiffness and multifocal atherosclerosis on the 10-year prognosis of patients after coronary artery bypass grafting. Methods. Patients with coronary artery disease (n = 274) who underwent coronary artery bypass grafting (CABG), in whom cardio-ankle vascular index (CAVI) was assessed using the VaSera VS-1000 device and the presence of peripheral atherosclerosis in Doppler ultrasound. Groups were distinguished with normal CAVI (<9.0, n = 163) and pathological CAVI (≥9.0, n = 111). To assess the prognosis, coronary and non-coronary death, myocardial infarction, acute cerebrovascular accident/transient ischemic attack, repeated CABG, percutaneous coronary intervention, carotid endarterectomy, peripheral arterial surgery, pacemaker implantation were analyzed. Results. During the observation period, mortality was 27.7%. A fatal outcome from all causes was in 37 (22.7%) patients in the group with normal CAVI and in 39 (35.14%) in the group with pathological CAVI (p = 0.023). Death from cardiac causes was more common in the group with CAVI ≥ 9.0—in 25 cases (22.52%) than in the group with CAVI < 9.0—in 19 (11.6%, p = 0.016). The combined endpoint in patients with pathological CAVI was detected in 66 (59.46%) cases, with normal CAVI values—in 76 (46.63%) cases (p = 0.03). The presence of diabetes mellitus, multifocal atherosclerosis (p = 0.004), pathological CAVI (p = 0.063), and male gender were independent predictors of death at 10-year follow-up after CABG. The presence of multifocal atherosclerosis and pathological CAVI during the preoperative examination of patients were independent predictors of the combined endpoint development. Findings. Patients with coronary artery disease with pathological CAVI before CABG were more likely to experience adverse events and death in the long-term follow-up than patients with normal CAVI. Further studies are needed to investigate the possibility of correcting pathological CAVI after CABG after secondary prevention and the possible impact of this correction on prognosis.
Collapse
|
31
|
Zhang J, Tan Q, Lina W, Zhaoqian Z. Endothelial dysfunction predicted cardiovascular events in patients with paroxysmal atrial fibrillation. Saudi Med J 2022; 43:708-714. [PMID: 35830994 PMCID: PMC9749696 DOI: 10.15537/smj.2022.43.7.20220214] [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: 03/27/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES To examine the relationship between endothelial dysfunction and adverse cardiovascular events in patients with paroxysmal atrial fibrillation (AF). METHODS In this prospective observational study, flow-mediated dilation (FMD) was measured by ultrasound in 291 patients with paroxysmal AF. Then, the patients were divided into low FMD group (n=97, FMD of <5.9%) or high FMD group (n=194, FMD of ≥5.9%). Patients were followed up for at least 30 months. Primary end point was cardiovascular events (stroke, heart failure hospitalization, cardiovascular death, and non-fatal myocardial infarction) and second endpoint was heart failure hospitalization, or stroke. RESULTS Rate of cardiovascular events was higher in low FMD group compared with high FMD group (37.1% versus 18%, p<0.001). This higher risk of cardiovascular events in patients with low FMD was primarily due to a higher risk of heart failure hospitalization compared with patients with high FMD (19.6% versus 10.8%, p<0.05). There was no significant difference of stroke between both groups. Cox proportional hazards ratio (HR) analysis showed that low FMD (HR: 3.036, 95% CI: [1.546-5.963], CHA2DS2-VASc scores (HR: 1.383, 95% CI: [1.035-1.847]), and left atrial diameter (HR: 1.304, 95% CI: [1.001-1.069]) were independent predictors for composite cardiovascular events. CONCLUSION In patients with paroxysmal AF, endothelial dysfunction was associated with composite cardiovascular events. Flow-mediated dilation was a significant predictor of cardiovascular events in patients with paroxysmal AF.
Collapse
Affiliation(s)
- Jing Zhang
- From the Department of Cardiology, Qinhuangdao First Hospital, Hebei Medical University, Qinhuangdao, China
| | - Qiang Tan
- From the Department of Cardiology, Qinhuangdao First Hospital, Hebei Medical University, Qinhuangdao, China
- Address correspondence and reprint request to: Dr. Qiang Tan, Department of Cardiology, Qinhuangdao First Hospital, Hebei Medical University, Qinhuangdao, China. E-mail: ORCID ID: https://orcid.org/0000-0003-1621-4428
| | - Wang Lina
- From the Department of Cardiology, Qinhuangdao First Hospital, Hebei Medical University, Qinhuangdao, China
| | - Zhang Zhaoqian
- From the Department of Cardiology, Qinhuangdao First Hospital, Hebei Medical University, Qinhuangdao, China
| |
Collapse
|
32
|
Cilostazol improves endothelial function in patients with ischemic stroke: A randomized controlled single-center study with flow-mediated dilation technique. J Neurol Sci 2022; 439:120318. [PMID: 35749837 DOI: 10.1016/j.jns.2022.120318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/10/2022] [Accepted: 06/05/2022] [Indexed: 11/22/2022]
|
33
|
Radial Peripapillary Capillary Plexus Perfusion and Endothelial Dysfunction in Early Post-SARS-CoV-2 Infection. Vision (Basel) 2022; 6:vision6020026. [PMID: 35645380 PMCID: PMC9149887 DOI: 10.3390/vision6020026] [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: 03/28/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Endothelial cells damage and thromboinflammation are considered key elements in the generation of organ impairment in patients with COVID-19 disease. The endothelial function is evaluated by measuring flow-mediated dilation (FMD). We aimed to analyze the association between FMD impairment and retinal vascular parameters in early post-COVID-19 patients. 00118-00199Tomography (OCT), OCT Angiography (OCTA) and slit lamp examination were performed. FMD ≤ 7% was considered as pathological. Our primary outcome was to assess potential differences in the radial peripapillary capillary plexus flow index (RPCP-FI) and RPCP density (RPCP-D) values between post-COVID-19 patients with and without FMD impairment. The associations of other retinal vascular parameters with FMD impairment were assessed as secondary endpoints. Results: FMD impairment was detected in 31 patients (37.8%). RPCP-FI (p = 0.047), age (p = 0.048) and prevalence of diabetes (p = 0.046) significantly differed in patients with FMD ≤ 7% in regression analysis. RPCP-FI was linearly correlated with FMD values (R = 0.244, p =0.027). SCT was found to be lower in patients with impaired FMD (p = 0.004), although this difference was only a trend in binary logistic regression output (p = 0.07). Conclusions: Early post-COVID-19 patients showed a higher prevalence of FMD impairment compared to the general population. Age, diabetes and RPCP-FI were independently correlated with the presence of endothelial impairment in the early post-infective period.
Collapse
|
34
|
Self-reported total sitting time on a non-working day is associated with blunted flow-mediated vasodilation and blunted nitroglycerine-induced vasodilation. Sci Rep 2022; 12:6366. [PMID: 35430619 PMCID: PMC9012897 DOI: 10.1038/s41598-022-10242-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 04/05/2022] [Indexed: 12/26/2022] Open
Abstract
We divided the 466 subjects into two groups based on information on sitting time on a non-working day and evaluated flow-mediated vasodilation (FMD) and nitroglycerine-induced vasodilation (NID). FMD was smaller in subjects with sitting time on a non-working day of ≥6 h/day than in subjects with sitting time on a non-working day of <6 h/day (2.5 ± 2.6% vs. 3.7 ± 2.9%; p < 0.001). NID was smaller in subjects with sitting time at non-working day of ≥ 8 h/day than in subjects with sitting time on a non-working day of < 8 h/day (10.1 ± 5.6% vs. 11.5 ± 5.0%; p = 0.01). After adjustment for confounding factors for vascular function, the odds of having the lowest tertile of FMD was significantly higher in subjects with sitting time on a non-working day of ≥6 h/day than in subjects with sitting time on a non-working day of <6 h/day. The odds of having the lowest tertile of NID was significant higher in subjects with sitting time on a non-working day of ≥ 8 h/day than in subjects with sitting time on a non-working day of < 8 h/day. These findings suggest that prolonged sitting time on a non-working day is associated with blunted FMD and blunted NID.
Collapse
|
35
|
Knowles KA, Stewart KJ, Tejan J, Ouyang P, Ratchford EV, Sullam L, Magliato K, Whitt MD, Silber HA. A novel operator-independent noninvasive device for assessing arterial reactivity. IJC HEART & VASCULATURE 2022; 39:100960. [PMID: 35402694 PMCID: PMC8984635 DOI: 10.1016/j.ijcha.2022.100960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/21/2021] [Accepted: 01/18/2022] [Indexed: 11/23/2022]
Abstract
Background Endothelial dysfunction is associated with increased risk of cardiovascular disease (CVD). Currently available noninvasive methods of measuring endothelial function have limitations. We tested a novel device that provides an automated measurement of the difference between baseline and post-ischemic, hyperemia-induced, brachial arterial compliance, a phenomenon known to be endothelium-dependent. The association between the calculated index, Flow-mediated Compliance Response (FCR), and established CVD risk indices was determined. Methods Adults with CVD risk factors or known coronary artery disease (CAD) were enrolled. Framingham Risk Score (FRS) was calculated and presence of metabolic syndrome (MetSyn) was assessed. Carotid artery plaques were identified by ultrasound. Cardiorespiratory fitness was assessed by 6-minute walk test (6MWT). FCR was measured using the device. Results Among 135 participants, mean age 49.3 +/- 17.9 years, characteristics included: 48% female, 7% smokers, 7% CAD, 10% type 2 diabetes, 34% MetSyn, and 38% with carotid plaque. Those with MetSyn had 24% lower FCR than those without (p < 0.001). Lower FCR was associated with higher FRS percentile (r = -0.29, p < 0.001), more MetSyn factors (r = -0.30, p < 0.001), more carotid plaques (r = -0.22, p = 0.01), and lower 6MWT (r = 0.34, p < 0.0001). Conclusion FCR, an index of arterial reactivity obtained automatically using a novel, operator-independent device, was inversely associated with established CVD risk indices, increased number of carotid plaques, and lower cardiorespiratory fitness. Whether measuring FCR could play a role in screening for CVD risk and assessing whether endothelial function changes in response to treatments aimed at CVD risk reduction, warrants further study.
Collapse
Affiliation(s)
- Kellen A. Knowles
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kerry J. Stewart
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Joseph Tejan
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Pamela Ouyang
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elizabeth V. Ratchford
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laura Sullam
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kathy Magliato
- Division of Cardiothoracic Surgery, Providence St. John’s Health Center, Santa Monica, CA, United States
| | - Michael D. Whitt
- Department of Biomedical Engineering, California Polytechnic State University, San Luis Obispo, CA, United States
| | - Harry A. Silber
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Corresponding author at: Cardiology, Suite 2400, 301 Building, Johns Hopkins Bayview Medical Center, 4940 Eastern Avenue, Baltimore, MD 21224, United States.
| |
Collapse
|
36
|
Li X, Liu H, Zhang Y, Gu Y, Sun L, Yu H, Bai W. A Prediction Equation to Estimate Vascular Endothelial Function in Different Body Mass Index Populations. Front Cardiovasc Med 2022; 9:766565. [PMID: 35360015 PMCID: PMC8960173 DOI: 10.3389/fcvm.2022.766565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Vascular endothelial dysfunction is considered an early predictor of endothelial injury and the initiating factor of atherosclerosis (AS). Brachial artery flow-mediated dilation (FMD) can detect endothelial injury early and provide important prognostic information beyond traditional cardiovascular (CV) risk factors. This study aimed to find the influencing factors of FMD and develop a simple prediction model in populations with different body mass indices (BMIs). Methods In total, 420 volunteers with different BMIs were recruited in our study. Subjects were randomly assigned to the derivation and validation cohorts (the ratio of the two was 1:2) with simple random sampling. The former was used for influencing factors searching and model construction of FMD and the latter was used for verification and performance evaluation. Results The population was divided into two groups, i.e., 140 people in the derivation group and 280 people in the verification group. Analyzing in the training data, we found that females had higher FMD than males (p < 0.05), and FMD decreased with age (p < 0.05). In people with diabetes, hypertension or obesity, FMD was lower than that in normal individuals (p < 0.05). Through correlation analysis and linear regression, we found the main influencing factors of FMD: BMI, age, waist-to-hip radio (WHR), aspartate aminotransferase (AST) and low-density lipoprotein (LDL). And we developed a simple FMD prediction model: FMD = −0.096BMI−0.069age−4.551WHR−0.015AST−0.242LDL+17.938, where R2 = 0.599, and adjusted R2 = 0.583. There was no statistically significant difference between the actual FMD and the predicted FMD in the verification group (p > 0.05). The intra-class correlation coefficient (ICC) was 0.77. In a Bland-Altman plot, the actual FMD and the predicted FMD also showed good agreement. This prediction model had good hints in CV risk stratification (area under curve [AUC]: 0.780, 95 % confidence intervals [95% CI]: 0.708–0.852, p < 0.001), with a sensitivity and specificity of 73.8 and 72.1%, respectively. Conclusions Males, older, obesity, hypertension, diabetes, smoking, etc. were risk factors for FMD, which was closely related to CV disease (CVD). We developed a simple equation to predict FMD, which showed good agreement between the training and validation groups. And it would greatly simplify clinical work and may help physicians follow up the condition and monitor therapeutic effect. But further validation and modification bears great significance.
Collapse
Affiliation(s)
- Xiao Li
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated 6th People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, China
| | - Hanying Liu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Institute of Shanghai Diabetes, Shanghai, China
| | - Yan Zhang
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated 6th People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, China
| | - Yanting Gu
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated 6th People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, China
| | - Lianjie Sun
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haoyong Yu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Institute of Shanghai Diabetes, Shanghai, China
- *Correspondence: Haoyong Yu
| | - Wenkun Bai
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated 6th People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai, China
- Department of Ultrasound in Medicine, South Hospital of Shanghai Jiao Tong University Affiliated 6th People's Hospital, Shanghai, China
- Wenkun Bai
| |
Collapse
|
37
|
Isolated diastolic hypertension is not associated with endothelial dysfunction. Hypertens Res 2022; 45:698-707. [PMID: 35115698 DOI: 10.1038/s41440-022-00854-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/10/2021] [Accepted: 12/20/2021] [Indexed: 11/08/2022]
Abstract
Hypertension is associated with an impairment of endothelial function. However, it is unclear whether isolated diastolic hypertension is associated with endothelial dysfunction. The purpose of this study was to investigate the association of endothelial function with isolated diastolic hypertension diagnosed by using two major hypertension guidelines: systolic blood pressure (BP) < 130 mmHg and diastolic BP ≥ 80 mmHg according to the 2017 American College of Cardiology/American Heart Association (ACC/AHA) hypertension guideline and systolic BP < 140 mmHg and diastolic BP ≥ 90 mmHg according to the 2018 European Society of Cardiology/European Society of Hypertension (ESC/ESH) hypertension guideline. We measured the flow-mediated vasodilation (FMD) of the brachial artery in subjects without systolic hypertension who were not treated with antihypertensive drugs. Of 3727 subjects (2813 men; mean age: 41.3 ± 10.9 years), 749 (20.1%) had isolated diastolic hypertension according to the 2017 ACC/AHA definition. Multiple logistic regression analysis revealed that isolated diastolic hypertension was not associated with endothelial dysfunction, defined as FMD < 7.0% (OR, 1.15; 95% CI, 0.98-1.35; P = 0.09). Of 4747 subjects (3727 men; mean age: 45.1 ± 10.8 years), 314 subjects (6.6%) had isolated diastolic hypertension according to the ESC/ESH definition. Multiple logistic regression analysis revealed that isolated diastolic hypertension was not associated with endothelial dysfunction after adjusting for age and sex (OR, 1.04; 95% CI, 0.82-1.32; P = 0.76). Isolated diastolic hypertension was not associated with endothelial dysfunction in individuals who were not treated with antihypertensive drugs regardless of the major hypertension guideline definition used.
Collapse
|
38
|
Zhaojun LMDP, Cuiqin SMS, Qingqing CMB, Lei SMB, Xianghong LMDP, Lianfang DMDP. Shear Wave Dispersion Imaging for Measuring Carotid Elasticity and Viscosity. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2022. [DOI: 10.37015/audt.2021.200071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
39
|
Wang Y, Yao M, Wang J, Liu H, Zhang X, Zhao L, Hu X, Guan H, Lyu Z. Effects of Antidiabetic Drugs on Endothelial Function in Patients With Type 2 Diabetes Mellitus: A Bayesian Network Meta-Analysis. Front Endocrinol (Lausanne) 2022; 13:818537. [PMID: 35370959 PMCID: PMC8969579 DOI: 10.3389/fendo.2022.818537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/14/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The changes of endothelial function in type 2 diabetes mellitus (T2DM) patients are closely associated with the development of cardiovascular disease (CVD). However, it is still unclear whether commonly used antidiabetic drugs can improve endothelial function. Flow-mediated dilation (FMD) is a noninvasive tool for evaluating endothelial function, which typically examines changes in the brachial artery diameter in response to ischemia using ultrasound. We performed a network meta-analysis (NMA) to explore the associations between changes in endothelial function and antidiabetic drugs by evaluating FMD in T2DM patients. METHODS We systematically searched several electronic databases for randomized controlled trials (RCTs) published from inception until January 25, 2022 with no language restriction. The primary outcome was FMD change in all studies, and we performed subgroup analysis in T2DM patients without CVD. NMA was performed to calculate the mean differences (MDs) with 95% confidence intervals (CIs). RESULTS From the 1,987 candidate articles identified in the initial search, 30 RCTs were eventually included in the analysis. In all studies, glucagon-like peptide-1 receptor (GLP-1R) agonists [MD = 3.70 (1.39-5.97)], TZD [MD = 1.96 (0.006-3.89)] produced improvement of FMD change compared to lifestyle intervention. GLP-1R agonists [MD = 3.33 (1.36-5.34) and MD = 3.30 (1.21-5.43)] showed significantly greater improvements in FMD change in pairwise comparisons with sulfonylureas and placebo. SGLT-2i also showed efficacy compared to sulfonylureas (MD = 1.89, 95% CI, 0.10, 3.75). In studies of T2DM patients without CVD, GLP-1R agonists [MD = 3.53 (1.24-5.76)], and TZD [MD = 2.30 (0.27-3.24)] produced improvements in FMD change compared to lifestyle treatment. GLP-1R agonists [MD = 3.25 (1.13-5.40), and MD = 3.85 (1.68-6.13)] showed significantly greater improvements in pairwise comparisons with sulfonylureas, and placebo. CONCLUSION In T2DM patients, both GLP-1R agonists, SGLT-2i and TZD have favorable effects to improve endothelial function in T2DM patients. In T2DM patients without CVD, GLP-1R agonists had a greater effect to improve endothelial function than sulfonylureas. These suggested that GLP-1R agonists are associated with significantly improved endothelial function in T2DM patients.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mingyan Yao
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jincheng Wang
- Department of Radiology, Peking University Cancer Hospital, Beijing, China
| | - Hongzhou Liu
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xuelian Zhang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ling Zhao
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaodong Hu
- Department of Endocrinology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Haixia Guan
- Department of Endocrinology Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Haixia Guan, ; Zhaohui Lyu,
| | - Zhaohui Lyu
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Haixia Guan, ; Zhaohui Lyu,
| |
Collapse
|
40
|
Maruhashi T, Kajikawa M, Kishimoto S, Takaeko Y, Yamaji T, Harada T, Hashimoto Y, Han Y, Aibara Y, Yusoff FM, Nakano Y, Chayama K, Nakashima A, Goto C, Yoshimura K, Higashi Y. Serum Potassium Levels of 4.5 to Less Than 5.0 mmol/L Are Associated with Better Vascular Function. J Atheroscler Thromb 2021; 29:1588-1602. [PMID: 34937833 PMCID: PMC9623076 DOI: 10.5551/jat.63285] [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] [Indexed: 11/23/2022] Open
Abstract
Aim: An experimental study showed that potassium inhibits the formation of reactive oxygen species by vascular cells. The purpose of this study was to investigate the association between serum potassium levels and vascular function.
Methods: We measured flow-mediated vasodilation (FMD) as an index of endothelial function in 3045 subjects (1964 men; mean age, 62.3±13.8 years) and nitroglycerine-induced vasodilation (NID) as an index of vascular smooth muscle function in 1578 subjects (1001 men; mean age, 61.8±16.3 years).
Results: In the 3045 subjects, there was a significant positive correlation between FMD and serum potassium levels (r=0.09,P<0.001). Multivariate analysis revealed that serum potassium levels were significantly associated with FMD (β=0.109,P<0.001). When subjects were divided into two groups based on treatment with drugs that could alter serum potassium levels, the association between serum potassium levels and FMD was significant both in subjects with (β=0.096,P<0.001) and subjects without (β=0.123,P<0.001) treatment with drugs that could alter serum potassium levels. In the 1578 subjects, there was a significant positive correlation between NID and serum potassium levels (r=0.11,P<0.001). Multivariate analysis revealed that serum potassium levels were significantly associated with NID (β=0.098,P<0.001). The association between serum potassium levels and NID was significant both in subjects with (β=0.121,P=0.001) and subjects without (β=0.083,P=0.03) treatment with drugs that could alter serum potassium levels.
Conclusions: Serum potassium levels of 4.5-<5.0 mmol/L are associated with better vascular function regardless of the presence or absence of treatment with drugs that could alter serum potassium levels.
Collapse
Affiliation(s)
- Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| | - Yuji Takaeko
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Takayuki Yamaji
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Yu Hashimoto
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Yiming Han
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| | - Yoshiki Aibara
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Biomedical Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical and Sciences, Hiroshima University
| | - Chikara Goto
- Department of Rehabilitation, Faculty of general Rehabilitation, Hirohsima International University
| | - Kenichi Yoshimura
- Department of Biostatistics, Medical Center for Translational and Clinical Research, Hiroshima University Hospital
| | - Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University.,Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital
| |
Collapse
|
41
|
Zhang J, Liang J, Zhang X, Su C, He J, Qiu Y, Zhou Z, Wang Z, Dong B, Tu Q, Xu S, Xia W, Tao J. Non-invasive Systemic Hemodynamic Index in Vascular Risk Stratification Tailored for Hypertensives. Front Cardiovasc Med 2021; 8:744349. [PMID: 34881303 PMCID: PMC8645861 DOI: 10.3389/fcvm.2021.744349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/18/2021] [Indexed: 11/26/2022] Open
Abstract
Vascular dysfunction is a key hallmark of hypertension and related cardiovascular outcomes. As a well-known hemodynamic disease, hypertension is characterized by abnormal ventricular-vascular interactions. Complementing non-invasive systemic hemodynamics in hypertensive vascular risk assessment is of promising significance. We aimed to investigate the effects of abnormal hemodynamic states other than elevated blood pressure on vascular damage and establish a united index of systemic hemodynamics for generalized vascular risk evaluation. Non-invasive systemic hemodynamics, assessed by impedance cardiography, was compared among blood pressure stages. Vascular function was evaluated by flow-mediated dilation (FMD) and brachial-ankle pulse wave velocity (baPWV). Systemic hemodynamics was obtained from a total of 88 enrollees with a mean (±SD) systolic blood pressure 140 (±17) mm Hg, and aged 17 to 91 years. Both stroke systemic vascular resistance index and left stroke work index exhibited a significant alteration among blood pressure stages (p < 0.001; p = 0.01, respectively), whereas heterogeneous hemodynamic and vascular function subsets existed within similar blood pressure. In addition, blood pressure categories failed to recognize between-group differences in endothelial dysfunction (p = 0.88) and arterial stiffness (p = 0.26). An increase in myocardial contractility and a parallel decrease in afterload was associated with the decline of vascular dysfunction. Systemic Hemodynamic Index (SHI), as a surrogate marker, demonstrated a significantly negative correlation with vascular damage index (VDI, r = −0.49, p < 0.001). These findings illustrate that systemic hemodynamics underlying hypertensives provides more vascular information. The SHI/VDI score may be a feasible tool for cardiovascular function assessment.
Collapse
Affiliation(s)
- Jianning Zhang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiawen Liang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyu Zhang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chen Su
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiang He
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yumin Qiu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhe Zhou
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhichao Wang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bing Dong
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiang Tu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shiyue Xu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenhao Xia
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Tao
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Key Laboratory on Assisted Circulation of Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
42
|
Kim HL, Weber T. Pulsatile Hemodynamics and Coronary Artery Disease. Korean Circ J 2021; 51:881-898. [PMID: 34595882 PMCID: PMC8558570 DOI: 10.4070/kcj.2021.0227] [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: 06/28/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Coronary artery disease (CAD) is the leading cause of human death and has a high prevalence throughout the world. Therefore, it is important to detect CAD early and to apply individualized therapy according to the patients' risk. There is an increasing interest in pulsatile arterial hemodynamics in the cardiovascular area. Widely used measurements of arterial pulsatile hemodynamics include pulse pressure, pulse wave velocity and augmentation index. Here, we will review underlying pathophysiology linking the association of arterial pulsatile hemodynamics with CAD, and the usefulness of the measurements of pulsatile hemodynamics in the prediction of future cardiovascular events of CAD patients. Clinical and therapeutic implications will be also addressed.
Collapse
Affiliation(s)
- Hack-Lyoung Kim
- Division of Cardiology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Thomas Weber
- Department of Cardiology, Klinikum Wels-Grieskirchen GmbH, Wels, Austria.
| |
Collapse
|
43
|
Kajikawa M, Higashi Y. Promising Assessment of Vascular Function for Future Cardiovascular Events. J Atheroscler Thromb 2021; 28:1123-1125. [PMID: 33361649 PMCID: PMC8592704 DOI: 10.5551/jat.ed154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital.,Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| |
Collapse
|
44
|
Mahmoud M, Cancel L, Tarbell JM. Matrix Stiffness Affects Glycocalyx Expression in Cultured Endothelial Cells. Front Cell Dev Biol 2021; 9:731666. [PMID: 34692689 PMCID: PMC8530223 DOI: 10.3389/fcell.2021.731666] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 09/15/2021] [Indexed: 11/13/2022] Open
Abstract
Rationale: The endothelial cell glycocalyx (GCX) is a mechanosensor that plays a key role in protecting against vascular diseases. We have previously shown that age/disease mediated matrix stiffness inhibits the glycocalyx glycosaminoglycan heparan sulfate and its core protein Glypican 1 in human umbilical vein endothelial cells, rat fat pad endothelial cells and in a mouse model of age-mediated stiffness. Glypican 1 inhibition resulted in enhanced endothelial cell dysfunction. Endothelial cell culture typically occurs on stiff matrices such as plastic or glass. For the study of the endothelial GCX specifically it is important to culture cells on soft matrices to preserve GCX expression. To test the generality of this statement, we hypothesized that stiff matrices inhibit GCX expression and consequently endothelial cell function in additional cell types: bovine aortic endothelial cells, mouse aortic endothelial cell and mouse brain endothelial cells. Methods and Results: All cell types cultured on glass showed reduced GCX heparan sulfate expression compared to cells cultured on either soft polyacrylamide (PA) gels of a substrate stiffness of 2.5 kPa (mimicking the stiffness of young, healthy arteries) or on either stiff gels 10 kPa (mimicking the stiffness of old, diseased arteries). Specific cell types showed reduced expression of GCX protein Glypican 1 (4 of 5 cell types) and hyaluronic acid (2 of 5 cell types) on glass vs soft gels. Conclusion: Matrix stiffness affects GCX expression in endothelial cells. Therefore, the study of the endothelial glycocalyx on stiff matrices (glass/plastic) is not recommended for specific cell types.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Tarbell Lab, Department of Biomedical Engineering, The City University of New York, New York, NY, United States
| | | | - John M. Tarbell
- Tarbell Lab, Department of Biomedical Engineering, The City University of New York, New York, NY, United States
| |
Collapse
|
45
|
Yamaji T, Harada T, Hashimoto Y, Nakano Y, Kajikawa M, Yoshimura K, Chayama K, Goto C, Han Y, Mizobuchi A, Yusoff FM, Kishimoto S, Maruhashi T, Nakashima A, Higashi Y. Stair climbing activity and vascular function in patients with hypertension. Hypertens Res 2021; 44:1274-1282. [PMID: 34272476 DOI: 10.1038/s41440-021-00697-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023]
Abstract
We evaluated the relationship between daily stair climbing activity and vascular function as assessed by flow-mediated vasodilation (FMD) and nitroglycerine-induced vasodilation (NID). This study was a cross-sectional study. A total of 374 patients with hypertension were enrolled. The subjects were divided into three groups based on their daily stair climbing habit: no stairs group, climbing stairs to the 2nd-floor group, and climbing stairs to the ≥3rd-floor group. There was a significant difference in FMD between the ≥3rd-floor group and the other two groups (3.3 ± 2.5% vs. 2.3 ± 2.7% and 2.4 ± 2.7%, p = 0.02, respectively). FMD values were similar in the no stairs group and the 2nd-floor group (p = 0.96). There was a significant difference in NID between the no stairs group and the other two groups (7.4 ± 4.2% vs. 10.9 ± 5.3% and 11.3 ± 5.1%, p < 0.001, respectively). NID values were similar in the second-floor group and the ≥3rd-floor group (p = 0.86). These findings suggest that both endothelial function and vascular smooth muscle function are impaired in individuals who do not climb stairs and that endothelial function but not vascular smooth muscle function is impaired in individuals who climb stairs to the second floor compared with individuals who climb stairs to the ≥3rd floor. Stair climbing activity, a simple method for assessing daily physical activity, may reflect vascular function in patients with hypertension.
Collapse
Affiliation(s)
- Takayuki Yamaji
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yu Hashimoto
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Kenichi Yoshimura
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan.,Department of Biostatistics, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chikara Goto
- Department of Rehabilitation, Faculty of Rehabilitation, Hiroshima International University, Hiroshima, Japan
| | - Yiming Han
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Aya Mizobuchi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan. .,Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
46
|
The impact of antimalarial agents on traditional and non-traditional subclinical atherosclerosis biomarkers in systemic lupus erythematosus: A systematic review and meta-analysis. Autoimmun Rev 2021; 20:102887. [PMID: 34237422 DOI: 10.1016/j.autrev.2021.102887] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Cardiovascular (CV) morbidity is a well-established problem in systemic lupus erythematosus (SLE). Antimalarial (AM) therapy has been seen as a potential atheroprotective agent. The aim was to assess the impact of AM therapy on traditional and novel atherosclerosis (AT) biomarkers in patients with SLE. METHODS A search of MEDLINE, EMbase, and Cochrane library for studies evaluating the impact of AM on AT biomarkers in SLE was conducted. Data extraction included serum, functional and structural traditional and novel biomarkers. A narrative synthesis of the findings and a meta-analysis with random effects was conducted estimating mean differences (MD), OR, HR and 95% CIs. RESULTS The search strategy produced 148 articles, of which 64 were extracted for analysis. The MD in VLDL-cholesterol (-10.29, 95% CI -15.35, 5.24), triglycerides (-15.68, 95% CI -27.51, -3.86), and diastolic BP (-3.42, 95% CI -5.62, -1.23) differed significantly in patients on AM therapy compared with those without AM therapy. Patients on AM had a lower prevalence and incidence of diabetes mellitus than patients not on AM (HR: 0.39, 95% CI 0.17, 0.88). HCQ use was associated with lower blood pressure (BP) variability. Structural markers like carotid intima-media thickness (IMT), carotid plaque (CP) and coronary artery calcification (CAC) were not influenced by AM. For functional markers like endothelial and arterial stiffness the benefit was unclear. The GRADE approach showed a very low-to-low quality of evidence (QoE) per outcome. CONCLUSIONS There is some evidence on the associations between AM therapy and some AT markers. However, the data on which this conclusion was based was of low to very low evidence.
Collapse
|
47
|
Zhang Y, Zhang YJ, Zhang HW, Ye WB, Korivi M. Low-to-Moderate-Intensity Resistance Exercise Is More Effective than High-Intensity at Improving Endothelial Function in Adults: A Systematic Review and Meta-Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18136723. [PMID: 34206463 PMCID: PMC8297299 DOI: 10.3390/ijerph18136723] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/29/2022]
Abstract
Aerobic exercise has been confirmed to improve endothelial function (EF). However, the effect of resistance exercise (RE) on EF remains controversial. We conducted this systematic review and meta-analysis on randomized controlled trials (RCTs) to determine the effect of RE and its intensities on EF. We searched Web of Science, PubMed/MEDLINE, Scopus, and Wiley Online Library, and included 15 articles (17 trials) for the synthesis. Overall, RE intervention significantly improved flow-mediated dilatation (FMD) in brachial artery (SMD = 0.76; 95% CI: 0.47, 1.05; p < 0.00001), which represents improved EF. Meta-regression showed that the RE intensity was correlated with changes in FMD (Coef. = −0.274, T = −2.18, p = 0.045). We found both intensities of RE improved FMD, but the effect size for the low- to moderate-intensity (30–70%1RM) was bigger (SMD = 1.02; 95% CI: 0.60, 1.43; p < 0.0001) than for the high-intensity (≥70%1RM; SMD = 0.48; 95% CI: 0.21, 0.74; p = 0.005). We further noticed that RE had a beneficial effect (SMD = 0.61; 95% CI: 0.13, 1.09; p = 0.01) on the brachial artery baseline diameter at rest (BADrest), and the age variable was correlated with the changes in BADrest after RE (Coef. = −0.032, T = −2.33, p = 0.038). Young individuals (<40 years) presented with a bigger effect size for BADrest (SMD = 1.23; 95% CI: 0.30, 2.15; p = 0.009), while middle-aged to elderly (≥40 years) were not responsive to RE (SMD = 0.07; 95% CI: −0.28, 0.42; p = 0.70). Based on our findings, we conclude that RE intervention can improve the EF, and low- to moderate-intensity is more effective than high-intensity.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Rehabilitation Medicine, Shaoxing University, Shaoxing 312000, China;
| | - Ya-Jun Zhang
- Department of Rehabilitation Medicine, Shaoxing University, Shaoxing 312000, China;
- Correspondence: (Y.-J.Z.); (H.-W.Z.)
| | - Hong-Wei Zhang
- Department of Rehabilitation Medicine, Shaoxing University, Shaoxing 312000, China;
- Correspondence: (Y.-J.Z.); (H.-W.Z.)
| | - Wei-Bing Ye
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua 321004, China; (W.-B.Y.); (M.K.)
| | - Mallikarjuna Korivi
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua 321004, China; (W.-B.Y.); (M.K.)
| |
Collapse
|
48
|
Li X, Gao M, Zhu S, Yin L, Zhang B, Qi Y, Zhao Y, Yu Y, Xu L. Hengshun Aromatic Vinegar Ameliorates Vascular Endothelial Injury via Regulating PKCζ-Mediated Oxidative Stress and Apoptosis. Front Nutr 2021; 8:635232. [PMID: 34124116 PMCID: PMC8193096 DOI: 10.3389/fnut.2021.635232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/23/2021] [Indexed: 01/14/2023] Open
Abstract
Vascular endothelial injury (VEI) is an early event of atherosclerosis, and reversing endothelial dysfunction has become a new trend in the prevention and treatment of cardiovascular diseases. Hengshun aromatic vinegar (HSAV), a traditional vinegar, has been reported to have many pharmacological activities, but its effect against VEI and the molecular mechanism are still unknown. In this study, effects of HSAV on VEI were evaluated in H2O2-induced human umbilical vein endothelial cells (HUVECs) and methionine-induced VEI in rats. Results showed that HSAV significantly increased cell viability, inhibited apoptosis, and reduced the generation of reactive oxygen species (ROS) in H2O2-induced HUVECs. Meanwhile, HSAV decreased serum homocysteine (Hcy), endothelin 1 (ET-1), and oxidized low-density lipoprotein (ox-LDL) levels, increased nitric oxide (NO) and endothelin nitric oxide synthase (eNOS) levels, ameliorated pathological changes in rats with VEI induced by methionine. In parallel, HSAV relieved oxidative stress by decreasing malondialdehyde (MDA) level and increasing superoxide dismutase (SOD), glutathione (GSH), and glutathione peroxidase (GSH-Px) levels in rats with VEI. Mechanism studies indicated that HSAV markedly downregulated the expression of protein kinase C zeta (PKCζ), and consequently regulated sirtuin 1 (Sirt1)-mediated oxidative stress signal pathway, and protein inhibitor of activated STATy (PIASy)-mediated apoptosis pathway, leading to the alleviation of oxidative stress and inhibition of apoptosis. These regulative effects of HSAV were further validated by knockdown and overexpression of PKCζ in vitro. In conclusion, HSAV showed protective effect against VEI by inhibiting PKCζ and, thereby, ameliorating oxidative stress and inhibiting apoptosis. This study not only provides guidance for the consumption of vinegar in daily life but also promotes the development of diet supplement for disease prevention.
Collapse
Affiliation(s)
- Xin Li
- Jiangsu Hengshun Vinegar Industry Co., Ltd., Zhenjiang, China
| | - Meng Gao
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Shenghu Zhu
- Jiangsu Hengshun Vinegar Industry Co., Ltd., Zhenjiang, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Bao Zhang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yanyan Zhao
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yongjian Yu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Dalian, China
| |
Collapse
|
49
|
Mahmoud M, Mayer M, Cancel LM, Bartosch AM, Mathews R, Tarbell JM. The glycocalyx core protein Glypican 1 protects vessel wall endothelial cells from stiffness-mediated dysfunction and disease. Cardiovasc Res 2021; 117:1592-1605. [PMID: 32647868 PMCID: PMC8152694 DOI: 10.1093/cvr/cvaa201] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/22/2020] [Accepted: 07/02/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Arterial stiffness is an underlying risk factor and a hallmark of cardiovascular diseases. The endothelial cell (EC) glycocalyx is a glycan rich surface layer that plays a key role in protecting against EC dysfunction and vascular disease. However, the mechanisms by which arterial stiffness promotes EC dysfunction and vascular disease are not fully understood, and whether the mechanism involves the protective endothelial glycocalyx is yet to be determined. We hypothesized that endothelial glycocalyx protects the endothelial cells lining the vascular wall from dysfunction and disease in response to arterial stiffness. METHODS AND RESULTS Cells cultured on polyacrylamide (PA) gels of substrate stiffness 10 kPa (mimicking the subendothelial stiffness of aged, unhealthy arteries) showed a significant inhibition of glycocalyx expression compared to cells cultured on softer PA gels (2.5 kPa, mimicking the subendothelial stiffness of young, healthy arteries). Specifically, gene and protein analyses revealed that a glycocalyx core protein Glypican 1 was inhibited in cells cultured on stiff PA gels. These cells had enhanced endothelial cell dysfunction as determined by enhanced cell inflammation (enhanced inflammatory gene expression, monocyte adhesion, and inhibited nitric oxide expression), proliferation, and EndMT. Removal of Glypican 1 using gene-specific silencing with siRNA or gene overexpression using a plasmid revealed that Glypican 1 is required to protect against stiffness-mediated endothelial cell dysfunction. Consistent with this, using a model of age-mediated stiffness, older mice exhibited a reduced expression of Glypican 1 and enhanced endothelial cell dysfunction compared to young mice. Glypican 1 gene deletion in knockout mice (GPC1-/-) exacerbated endothelial dysfunction in young mice, which normally had high endothelial expression, but not in old mice that normally expressed low levels. Endothelial cell dysfunction was exacerbated in young, but not aged, Glypican 1 knockout mice (GPC1-/-). CONCLUSION Arterial stiffness promotes EC dysfunction and vascular disease at least partly through the suppression of the glycocalyx protein Glypican 1. Glypican 1 contributes to the protection against endothelial cell dysfunction and vascular disease in endothelial cells.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Mariya Mayer
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Limary M Cancel
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Anne Marie Bartosch
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Rick Mathews
- Oregon Health & Science University, School of Medicine, Portland, OR, USA
| | - John M Tarbell
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| |
Collapse
|
50
|
Takaeko Y, Maruhashi T, Kajikawa M, Kishimoto S, Yamaji T, Harada T, Hashimoto Y, Han Y, Kihara Y, Chayama K, Goto C, Yusoff FM, Yoshimura K, Nakashima A, Higashi Y. Lower triglyceride levels are associated with better endothelial function. J Clin Lipidol 2021; 15:500-511. [PMID: 34006457 DOI: 10.1016/j.jacl.2021.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 03/12/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Increased serum triglyceride levels are independently associated with endothelial dysfunction. However, there is little evidence to define normal levels of triglycerides and there is little information on endothelial function in subjects with extremely low levels of triglycerides. OBJECTIVE The purpose of this study was to determine the relationship between triglycerides, especially low levels of triglycerides, and vascular function. METHODS We measured flow-mediated vasodilation (FMD) in 7047 subjects and nitroglycerine-induced vasodilation (NID) in 1017 subjects. We divided the subjects into eight groups by triglyceride levels: <50 mg/dL, 50-69 mg/dL, 70-89 mg/dL, 90-109 mg/dL, 110-129 mg/dL, 130-149 mg/dL, 150-199 mg/dL, and ≥200 mg/dL. RESULTS FMD was significantly higher in subjects with triglyceride levels of <50 mg/dL than in subjects with triglyceride levels of 50-69 mg/dL, 70-89 mg/dL, 90-109 mg/dL, 110-129 mg/dL, 130-149 mg/dL, 150-199 mg/dL, and ≥200 mg/dL (p=0.002, p<0.001, p<0.001, p<0.001, p<0.001, p<0.001, and p<0.001, respectively). Using triglyceride levels of >200 mg/dL as a reference, the odds ratios for a lower quartile of FMD were significantly lower in the <50 mg/dL group, 50-69 mg/dL group, 70-89 mg/dL group, and 90-109 mg/dL group after adjustment for age, gender and other cardiovascular risk factors. There was a slight negative correlation between NID and triglycerides (r=-0.074; p=0.019). However, there was no significant differences in NID among the eight groups. CONCLUSIONS FMD values were highest in subjects with extremely low levels of triglycerides (<50 mg/dL). Lower triglyceride levels were associated with better endothelial function. CLINICAL TRIAL REGISTRATION INFORMATION http://www.umin.ac.jp (University Hospital Medical Information Network Clinical Trials Registry) (UMIN000012950).
Collapse
Affiliation(s)
- Yuji Takaeko
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan (Dr. Takaeko, Yamaji, Harada, Hashimoto and Kihara)
| | - Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Dr Maruhashi, Kishimoto, Han, Yusoff and Higashi)
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (Dr Kajikawa and Higashi)
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Dr Maruhashi, Kishimoto, Han, Yusoff and Higashi)
| | - Takayuki Yamaji
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan (Dr. Takaeko, Yamaji, Harada, Hashimoto and Kihara)
| | - Takahiro Harada
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan (Dr. Takaeko, Yamaji, Harada, Hashimoto and Kihara)
| | - Yu Hashimoto
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan (Dr. Takaeko, Yamaji, Harada, Hashimoto and Kihara)
| | - Yiming Han
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Dr Maruhashi, Kishimoto, Han, Yusoff and Higashi)
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan (Dr. Takaeko, Yamaji, Harada, Hashimoto and Kihara)
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan (Dr Chayama)
| | - Chikara Goto
- Department of Physical Therapy, Hiroshima International University, Hiroshima, Japan (Dr Goto)
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Dr Maruhashi, Kishimoto, Han, Yusoff and Higashi)
| | - Kenichi Yoshimura
- Department of Biostatistics, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (Dr Yoshimura)
| | - Ayumu Nakashima
- Department of Stem Cell Biology and Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan (Dr Nakashima)
| | - Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Dr Maruhashi, Kishimoto, Han, Yusoff and Higashi); Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (Dr Kajikawa and Higashi).
| |
Collapse
|