1
|
Sahebkar A, Reiner Ž, Almahmeed W, Jamialahmadi T, Simental-Mendía LE. Effect of Statin Treatment on Lipoprotein-Associated Phospholipase A2 Mass and Activity: A Systematic Review and Meta-analysis of Randomized Placebo-Controlled Trials. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07634-5. [PMID: 39466484 DOI: 10.1007/s10557-024-07634-5] [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] [Accepted: 10/16/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE The goal of this meta-analysis was to establish whether statin treatment reduces Lp-PLA2 mass concentration and/or activity. METHODS PubMed, Scopus, Web of Science, ClinicalTrials.gov, and Google Scholar databases were searched using MESH terms and keywords. Randomized controlled trials (RCT) with either parallel or cross-over design examining the effect of statins on Lp-PLA2 mass and/or activity were included in meta-analysis. RESULTS Out of 256 articles, 10 RCT were selected for meta-analysis. Statin therapy significantly reduced both Lp-PLA2 mass (WMD -44.46 ng/mL; 95%CI -59.01, -29.90; p < 0.001; I2 = 93%) and activity (WMD -39.37 nmol/min/mL; 95%CI -69.99, -8.75; p = 0.01; I2 = 100%). The sensitivity analysis was robust for Lp-PLA2 mass and was also positive for two studies concerning Lp-PLA2 activity. CONCLUSION Statin therapy significantly reduced both Lp-PLA2 mass and activity.
Collapse
Affiliation(s)
- Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Center Zagreb, University of Zagreb, Kišpatićeva 12, Zagreb, Croatia
- Polish Mother's Memorial Hospital Research Institute, Lodz, 93-338, Poland
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Luis E Simental-Mendía
- Unidad de Investigación Biomédica, Delegación Durango, Instituto Mexicano del Seguro Social, Durango, Mexico.
| |
Collapse
|
2
|
Futatsugi A, Tozuka M, Horiuchi Y, Ohkawa R, Kosho T. High-density lipoprotein functionality in cholesterol efflux in early childhood is related to the content ratio of triglyceride to cholesterol. Sci Rep 2024; 14:23323. [PMID: 39375444 PMCID: PMC11458590 DOI: 10.1038/s41598-024-74699-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024] Open
Abstract
Cholesterol efflux capacity (CEC), commonly measured as a useful risk marker of atherosclerotic cardiovascular disease, depends on high-density lipoprotein (HDL) functionality and its concentration. We defined the relative HDL functionality in cholesterol efflux, not influenced by HDL concentration, as the ratio of measured CEC to standardized CEC (stCEC) based on HDL-cholesterol (HDL-C) of each individual using the curve regression equation obtained from the correlation. HDL-C, CEC, and CEC/stCEC levels in the < 28-day-old participants (neonates) were significantly low compared to those of the ≥ 28-day-old participants, indicating that the low CEC levels in the neonates depend on not only lower HDL-C but also lower HDL functionality. The low level of CEC/stCEC was remarkable in neonates born at < 34 weeks of gestation and did not improved to the reference level (1.000) until the infantile period. The relatively low or high CEC/stCEC ratios in neonates and infants were associated with lower or higher HDL-TG and HDL-TG/HDL-C ratio, respectively. However, no apparent effect of HDL-TG and HDL-TG/HDL-C ratio on CEC/stCEC was observed in the ≥ 1-year-old participants, indicating that HDL functionality in cholesterol efflux could be associated with the various HDL particles with various lipid compositions, but not just with HDL-TG and HDL-TG/HDL-C ratio.
Collapse
Affiliation(s)
- Akiko Futatsugi
- Department of Medical Genetics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Department of Clinical Laboratory, Nagano Children's Hospital, 3100 Toyoshina, Azumino, 399-8288, Japan
| | - Minoru Tozuka
- Department of Medical Genetics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
- Life Science Research Center, Nagano Children's Hospital, 3100, Toyoshina, Azumino, 399-8288, Japan.
| | - Yuna Horiuchi
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, 6-8-1 Hinode, Urayasu, 279-0013, Japan
| | - Ryunosuke Ohkawa
- Department of Clinical Bioanalysis and Molecular Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Tomoki Kosho
- Department of Medical Genetics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Center for Medical Genetics, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Research Center for Supports to Advanced Science, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- BioBank Shinshu, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| |
Collapse
|
3
|
Lian PA, Zhu WQ, Zhao WX, Huang PP, Ran JL, Tang YX, Huang XS, Li R. Lipoprotein(a) in atherosclerotic cardiovascular disease and proprotein convertase subtilisin/kexin-type 9 inhibitors. Clin Chim Acta 2024; 565:119982. [PMID: 39366516 DOI: 10.1016/j.cca.2024.119982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
High plasma lipoprotein(a) (Lp(a)) levels increase the cardiovascular risk in populations with atherosclerotic cardiovascular disease (ASCVD). Apolipoprotein (a) [apo(a)], a unique protein component of Lp(a), plays an important role in the pathogenesis of atherosclerosis. Statins, the primary medication in managing ASCVD, lower low-density lipoprotein cholesterol (LDL-C) but concurrently elevate plasma Lp(a) levels, contributing to an increased residual cardiovascular risk. In turn, proprotein convertase subtilisin/kexin-type 9 (PCSK9) inhibitors, a novel class of LDL-C lowering drugs, effectively reduce plasma Lp(a) levels, which is believed to decrease residual cardiovascular risk. However, the mechanism by which PCSK9 inhibitors reduce Lp(a) levels remains unknown. In addition, there are some clinical limitations of PCSK9 inhibitors. Here, we systematically review the past, present, and prospects of studies pertaining to Lp(a), PCSK9 inhibitors, and ASCVD.
Collapse
Affiliation(s)
- Ping-An Lian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen-Qiang Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei-Xin Zhao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Piao-Piao Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan-Li Ran
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-Xin Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xian-Sheng Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cardiovascular Medicine, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China
| | - Rong Li
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Stomatology, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China.
| |
Collapse
|
4
|
Jendrek ST, Schmelter F, Schinke S, Hackel A, Graßhoff H, Lamprecht P, Humrich JY, Sina C, Müller A, Günther U, Riemekasten G. Metabolomic signature identifies HDL and apolipoproteins as potential biomarker for systemic sclerosis with interstitial lung disease. Respir Med 2024; 234:107825. [PMID: 39357678 DOI: 10.1016/j.rmed.2024.107825] [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: 01/18/2024] [Revised: 06/18/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND High-density lipoproteins (HDL) affect endothelial functions such as the expression of endothelial cell adhesion molecules and exert anti-apoptotic/-thrombotic functionalities. Therefore, profound analysis of lipoproteins may unveil biomarkers for (micro-)vasculopathy in systemic sclerosis (SSc) and mortality determining disease manifestations like interstitial lung disease (SSc-ILD). Because nuclear magnetic resonance (NMR) spectroscopy provides a wide range of lipoprotein parameters beyond the capabilities of classical analyses it has been used herein to examine lipoprotein profiles in SSc. METHODS To detect the metabolic and lipidomic profile serum samples from clinically well-characterized SSc patients (n = 100) and age-and sex-matched healthy controls (n = 40) were analyzed by 1H NMR spectroscopy using Bruker's in-vitro diagnostic research (IVDr) protocol. Statistical analyses were performed to validate significant findings and to search for associations between lipoproteins and clinical phenotypes. RESULTS Patients with SSc-ILD and lung fibrosis displayed reduced HDL levels. Furthermore, a reduction in apolipoprotein A1 + A2 and its HDL fractions reflected a distinct lipoprotein profile for SSc-ILD patients. This association was independent of potential clinical confounders for dyslipidemia. Notably, in SSc-ILD HDL levels correlate with FVC (forced vital capacity), DLCO (diffusion capacity of the lungs for carbon monoxide), and the modified Rodnan-Skin-Score. CONCLUSION These results suggest HDL and its lipoproteins may be considered as potential new biomarkers for SSc-ILD. Immune-mediated HDL effects on the endothelium facilitate microvasculopathy - one of the pathophysiological hallmarks in SSc. Therefore, a closer prospective evaluation of the capability of HDL-determination and its lipoproteins regarding a more individualized evaluation of SSc-ILD is warranted.
Collapse
Affiliation(s)
- Sebastian T Jendrek
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany.
| | | | - Susanne Schinke
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Alexander Hackel
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Hanna Graßhoff
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Peter Lamprecht
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Jens Y Humrich
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Christian Sina
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany; Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Antje Müller
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Ulrich Günther
- Institute of Chemistry and Metabolomics, University of Lübeck, Lübeck, Germany
| | - Gabriela Riemekasten
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| |
Collapse
|
5
|
Yin Y, Liu J, Yu J, Dong D, Gao F, Yu L, Du X, Wu S. ASGR1 is a promising target for lipid reduction in pigs with PON2 as its inhibitor. iScience 2024; 27:110288. [PMID: 39055948 PMCID: PMC11269292 DOI: 10.1016/j.isci.2024.110288] [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: 03/01/2024] [Revised: 05/17/2024] [Accepted: 06/13/2024] [Indexed: 07/28/2024] Open
Abstract
Although the role of asialoglycoprotein receptor 1 (ASGR1) in lowering lipid levels is well established, recent studies indicate that ASGR1 inhibition can cause unexpected liver damage in pigs, raising a serious issue about whether ASGR1 can be a good target for treating ASCVD. Here, we utilized the CRISPR-Cas9 system to regenerate ASGR1-knockout pigs, who displayed decreased lipid profiles without observable liver damage. This was confirmed by the lower levels of serum ALT and AST, reduced expression of inflammation markers, and normal histological morphology. Also, we implemented immunoprecipitation combined with mass spectrometry (IP-MS) and discovered that paraoxonase-2 (PON2) can interact with and significantly degrade ASGR1 in a dose-dependent manner. This degradation reduced lipid levels in mice, accompanied by little inflammation. Our study highlights the effectiveness and safety of degrading ASGR1 to reduce lipid levels in pigs and provides a potential inhibitor of ASGR1.
Collapse
Affiliation(s)
- Yunjun Yin
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jun Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jia Yu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Dingcai Dong
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Fei Gao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
- Sanya Institute of China Agricultural University, Sanya 572024, China
| | - Libao Yu
- The Eighth Medical Center of PLA General Hospital, Beijing 100094, China
| | - Xuguang Du
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
- Sanya Institute of China Agricultural University, Sanya 572024, China
| | - Sen Wu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
- Sanya Institute of China Agricultural University, Sanya 572024, China
| |
Collapse
|
6
|
Zhang R, Hu K, Bai H, Liu H, Pu Y, Yang C, Liu Q, Fan P. Increased oxidative stress is associated with hyperandrogenemia in polycystic ovary syndrome evidenced by oxidized lipoproteins stimulating rat ovarian androgen synthesis in vitro. Endocrine 2024; 84:1238-1249. [PMID: 38374513 DOI: 10.1007/s12020-024-03726-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/02/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE To determine the relationship between serum total testosterone (TT) levels and oxidative stress indices in patients with polycystic ovary syndrome (PCOS), and to investigate the effect of oxidative stress on androgen synthesis and its mechanism in rat ovarian theca-interstitial (T-I) cells. METHODS Clinical, hormonal, metabolic, and oxidative stress parameters were analyzed in a cross-sectional case-control study including 626 patients with PCOS and 296 controls. The effects of oxidized low-density lipoprotein (ox-LDL) and oxidized high-density lipoprotein (ox-HDL) on cell proliferation, TT secretion, and expression of key enzymes involved in testosterone synthesis were evaluated in T-I cells. RESULTS Serum TT levels were elevated with an increase in ox-LDL levels, whereas glutathione concentrations were lower in the high-TT subgroup than in the low-TT subgroup. The average ovarian volume and ox-LDL and malondialdehyde levels were significant predictors of TT levels in the multivariate regression models. In a rat ovarian T-I cell model, lipoprotein and oxidized lipoprotein treatments stimulated proliferation and promoted testosterone secretion. The mRNA and protein levels of 17α-hydroxylase were significantly higher in oxidized lipoprotein-treated cells than those in lipoprotein-treated cells. The mRNA levels of cholesterol side chain cleavage enzyme and steroidogenic acute regulatory protein were also significantly higher in ox-HDL-treated cells than in HDL-treated cells. CONCLUSIONS Oxidative stress can promote androgen production by up-regulating the expression of testosterone synthesis-related enzymes in vitro and may be an essential factor in elevating serum TT levels in patients with PCOS.
Collapse
Affiliation(s)
- Renjiao Zhang
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaifeng Hu
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongwei Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yifu Pu
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyi Yang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingqing Liu
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Zhang L, Zhang T, Chen X, Wang F, Liu L, Yang Y, Zeng Y, Si Y, Yang N. Overexpression of CuZn superoxide dismutase improves high-density lipoprotein function in swine. Prostaglandins Other Lipid Mediat 2024; 172:106817. [PMID: 38331090 DOI: 10.1016/j.prostaglandins.2024.106817] [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: 10/12/2023] [Revised: 02/01/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
Cardiovascular disease (CVD) has been the leading cause of death worldwide. As a chronic inflammatory disease, atherosclerosis (AS) acts as the initiating factor for CVD and reactive oxygen species (ROS) play a vital role in its development. Superoxide dismutases (SOD) can alleviate the detrimental effects of ROS and serve as the first line of defense through detoxifying the products derived from oxidative stress in vivo. Considering the potential preventive effects of high-density lipoprotein (HDL) on AS and the close relationship between CuZn superoxide dismutase (CuZnSOD) and HDL, the present work investigated whether CuZnSOD overexpression in swine could improve the function of HDL. Seven CuZnSOD transgenic swine, constructed by sperm and magnetic nanoparticles, demonstrated overexpressed CuZnSOD in the liver (P < 0.01) but comparable level to control in plasma (P > 0.05). CuZnSOD overexpression significantly down-regulated the levels of triglyceride (TG), apolipoprotein A-I (apoA-I) (P < 0.05), and high-density lipoprotein cholesterol (HDL-C) (P < 0.01) in plasma. In the presence of CuZnSOD overexpression, HDL3 significantly inhibited levels of IL-6 and TNF-α induced by oxidized low-density lipoprotein (oxLDL) (P < 0.05), indicating enhanced anti-inflammatory activity of HDL. At the same time, HDL-mediated cholesterol efflux did not decrease (P > 0.05). CuZnSOD overexpression improves the anti-inflammatory function of HDL despite decreased levels of HDL-C. In Conclusion, CuZnSOD overexpression improves HDL function in swine.
Collapse
Affiliation(s)
- Lichun Zhang
- Medical Laboratory Animal Center, School of Life Science and Technology, Weifang Medical University, Weifang, Shandong, China
| | - Tianliang Zhang
- Experimental Center for Medical Research, Weifang Medical University, Weifang, Shandong, China
| | - Xiaofeng Chen
- Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Fengjiao Wang
- Medical Laboratory Animal Center, School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Li Liu
- Medical Laboratory Animal Center, School of Life Science and Technology, Weifang Medical University, Weifang, Shandong, China
| | - Yanmei Yang
- Medical Laboratory Animal Center, School of Life Science and Technology, Weifang Medical University, Weifang, Shandong, China
| | - Yongqing Zeng
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China.
| | - Yanhong Si
- Basic Medical College, Shandong First Medical University, Taian, Shandong, China.
| | - Nana Yang
- Medical Laboratory Animal Center, School of Life Science and Technology, Weifang Medical University, Weifang, Shandong, China; Weifang Key Laboratory of Animal Model Research on Cardiovascular and Cerebrovascular Diseases, Weifang, Shandong, China.
| |
Collapse
|
8
|
Madaudo C, Coppola G, Parlati ALM, Corrado E. Discovering Inflammation in Atherosclerosis: Insights from Pathogenic Pathways to Clinical Practice. Int J Mol Sci 2024; 25:6016. [PMID: 38892201 PMCID: PMC11173271 DOI: 10.3390/ijms25116016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
This comprehensive review explores the various scenarios of atherosclerosis, a systemic and chronic arterial disease that underlies most cardiovascular disorders. Starting from an overview of its insidious development, often asymptomatic until it reaches advanced stages, the review delves into the pathophysiological evolution of atherosclerotic lesions, highlighting the central role of inflammation. Insights into clinical manifestations, including heart attacks and strokes, highlight the disease's significant burden on global health. Emphasis is placed on carotid atherosclerosis, clarifying its epidemiology, clinical implications, and association with cognitive decline. Prevention strategies, lifestyle modifications, risk factor management, and nuanced antithrombotic treatment considerations are critical to managing cardiovascular complications, thus addressing a crucial aspect of cardiovascular health.
Collapse
Affiliation(s)
- Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| | - Giuseppe Coppola
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| | | | - Egle Corrado
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| |
Collapse
|
9
|
Mi Y, Zhu Q, Chen Y, Zheng X, Wan M, Li Y. Impact of Physical Activity Frequency, Duration, and Intensity on Senile Cataract Risk: A Mendelian Randomization Study. Transl Vis Sci Technol 2024; 13:26. [PMID: 38814746 PMCID: PMC11146084 DOI: 10.1167/tvst.13.5.26] [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: 12/05/2023] [Accepted: 04/29/2024] [Indexed: 06/01/2024] Open
Abstract
Purpose We aimed to determine the causal effects of physical activities with different frequencies, durations, and intensities on the risk of senile cataracts using Mendelian randomization (MR). Methods A bidirectional two-sample MR approach was used to determine the association between physical activity and senile cataract risk. Our primary analysis used the inverse variance weighted method, and secondary analyses included MR-Egger regression, MR-PRESSO, and Cochran's Q statistic to evaluate heterogeneity and pleiotropy. Causal estimates were presented as odds ratios (ORs) with 95% confidence intervals (95% CIs). Results Genetically predicted moderate physical activity ≥ 10 min/wk (OR = 0.765, 95% CI = 0.627-0.936, P = 8.73E-03), vigorous physical activity ≥ 10 min/wk (OR = 0.691, 95% CI = 0.521-0.917, P = 1.04E-02), moderate-to-vigorous physical activity levels (OR = 0.552, 95% CI = 0.369-0.823, P = 3.75E-03), and overall acceleration average (OR = 0.952, 95% CI = 0.926-0.978, P = 3.80E-04) were associated with a decreased risk of senile cataract while walking ≥ 10 min/wk (OR = 0.972, 95% CI = 0.741-1.275, P = 8.36E-01) had no significant correlation. The reverse MR analysis showed no reversal causality from senile cataract to physical activity except for walking ≥ 10 min/wk (OR = 0.951, 95% CI = 0.923-0.979, P = 7.30E-04). Conclusions Our findings suggest that moderate to vigorous physical activity with higher frequency and longer duration will causally reduce the risk of senile cataracts, and there is no reverse causal relationship. Translational Relevance These findings underscore the potential of incorporating physical activity into preventive health strategies for senile cataracts.
Collapse
Affiliation(s)
- Yuze Mi
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qinnan Zhu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuxiang Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xinni Zheng
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Minghui Wan
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yipao Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Lymperopoulos D, Dedemadi AG, Voulgari ML, Georgiou E, Dafnis I, Mountaki C, Panagopoulou EA, Karvelas M, Chiou A, Karathanos VT, Chroni A. Corinthian Currants Promote the Expression of Paraoxonase-1 and Enhance the Antioxidant Status in Serum and Brain of 5xFAD Mouse Model of Alzheimer's Disease. Biomolecules 2024; 14:426. [PMID: 38672443 PMCID: PMC11047902 DOI: 10.3390/biom14040426] [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: 03/07/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Paraoxonase-1 (PON1), a serum antioxidant enzyme, has been implicated in Alzheimer's disease (AD) pathogenesis that involves early oxidative damage. Corinthian currants and their components have been shown to display antioxidant and other neuroprotective effects in AD. We evaluated the effect of a Corinthian currant paste-supplemented diet (CurD), provided to 1-month-old 5xFAD mice for 1, 3, and 6 months, on PON1 activity and levels of oxidation markers in serum and the brain of mice as compared to a control diet (ConD) or glucose/fructose-matched diet (GFD). Administration of CurD for 1 month increased PON1 activity and decreased oxidized lipid levels in serum compared to ConD and GFD. Longer-term administration of CurD did not, however, affect serum PON1 activity and oxidized lipid levels. Furthermore, CurD administered for 1 and 3 months, but not for 6 months, increased PON1 activity and decreased free radical levels in the cortex of mice compared to ConD and GFD. To probe the mechanism for the increased PON1 activity in mice, we studied the effect of Corinthian currant polar phenolic extract on PON1 activity secreted by Huh-7 hepatocytes or HEK293 cells transfected with a PON1-expressing plasmid. Incubation of cells with the extract led to a dose-dependent increase of secreted PON1 activity, which was attributed to increased cellular PON1 expression. Collectively, our findings suggest that phenolics in Corinthian currants can increase the hepatic expression and activity of antioxidant enzyme PON1 and that a Corinthian currant-supplemented diet during the early stages of AD in mice reduces brain oxidative stress.
Collapse
Affiliation(s)
- Dimitris Lymperopoulos
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
- Department of Biology, National and Kapodistrian University of Athens, Zografou, 15784 Athens, Greece
| | - Anastasia-Georgia Dedemadi
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
- Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15784 Athens, Greece
| | - Maria-Lydia Voulgari
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
- Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15784 Athens, Greece
| | - Eirini Georgiou
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
- Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15784 Athens, Greece
| | - Ioannis Dafnis
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
| | - Christina Mountaki
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
| | - Eirini A. Panagopoulou
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Dietetics and Nutrition, Harokopio University, 17676 Kallithea, Greece (A.C.); (V.T.K.)
| | - Michalis Karvelas
- Research and Development Department, Agricultural Cooperatives’ Union of Aeghion, 25100 Aeghion, Greece;
| | - Antonia Chiou
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Dietetics and Nutrition, Harokopio University, 17676 Kallithea, Greece (A.C.); (V.T.K.)
| | - Vaios T. Karathanos
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Dietetics and Nutrition, Harokopio University, 17676 Kallithea, Greece (A.C.); (V.T.K.)
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
| |
Collapse
|
11
|
Fung KYY, Ho TWW, Xu Z, Neculai D, Beauchemin CAA, Lee WL, Fairn GD. Apolipoprotein A1 and high-density lipoprotein limit low-density lipoprotein transcytosis by binding SR-B1. J Lipid Res 2024; 65:100530. [PMID: 38479648 PMCID: PMC11004410 DOI: 10.1016/j.jlr.2024.100530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/09/2024] Open
Abstract
Atherosclerosis results from the deposition and oxidation of LDL and immune cell infiltration in the sub-arterial space leading to arterial occlusion. Studies have shown that transcytosis transports circulating LDL across endothelial cells lining blood vessels. LDL transcytosis is initiated by binding to either scavenger receptor B1 (SR-B1) or activin A receptor-like kinase 1 on the apical side of endothelial cells leading to its transit and release on the basolateral side. HDL is thought to partly protect individuals from atherosclerosis due to its ability to remove excess cholesterol and act as an antioxidant. Apolipoprotein A1 (APOA1), an HDL constituent, can bind to SR-B1, raising the possibility that APOA1/HDL can compete with LDL for SR-B1 binding, thereby limiting LDL deposition in the sub-arterial space. To examine this possibility, we used in vitro approaches to quantify the internalization and transcytosis of fluorescent LDL in coronary endothelial cells. Using microscale thermophoresis and affinity capture, we find that SR-B1 and APOA1 interact and that binding is enhanced when using the cardioprotective variant of APOA1 termed Milano (APOA1-Milano). In male mice, transiently increasing the levels of HDL reduced the acute deposition of fluorescently labeled LDL in the atheroprone inner curvature of the aorta. Reduced LDL deposition was also observed when increasing circulating wild-type APOA1 or the APOA1-Milano variant, with a more robust inhibition from the APOA1-Milano. The results suggest that HDL may limit SR-B1-mediated LDL transcytosis and deposition, adding to the mechanisms by which it can act as an atheroprotective particle.
Collapse
Affiliation(s)
- Karen Y Y Fung
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Tse Wing Winnie Ho
- Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Zizhen Xu
- Department of Cell Biology, and Department of Pathology Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dante Neculai
- Department of Cell Biology, and Department of Pathology Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Catherine A A Beauchemin
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Interdisciplinary Theoretical and Mathematical Sciences (iTHEMS) program, RIKEN, Wako, Saitama, Japan
| | - Warren L Lee
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
12
|
Zhou Y, Wu Q, Guo Y. Deciphering the emerging landscape of HOX genes in cardiovascular biology, atherosclerosis and beyond (Review). Int J Mol Med 2024; 53:17. [PMID: 38131178 PMCID: PMC10781420 DOI: 10.3892/ijmm.2023.5341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Atherosclerosis, a dominant driving force underlying multiple cardiovascular events, is an intertwined and chronic inflammatory disease characterized by lipid deposition in the arterial wall, which leads to diverse cardiovascular problems. Despite unprecedented advances in understanding the pathogenesis of atherosclerosis and the substantial decline in cardiovascular mortality, atherosclerotic cardiovascular disease remains a global public health issue. Understanding the molecular landscape of atherosclerosis is imperative in the field of molecular cardiology. Recently, compelling evidence has shown that an important family of homeobox (HOX) genes endows causality in orchestrating the interplay between various cardiovascular biological processes and atherosclerosis. Despite seemingly scratching the surface, such insight into the realization of biology promises to yield extraordinary breakthroughs in ameliorating atherosclerosis. Primarily recapitulated herein are the contributions of HOX in atherosclerosis, including diverse cardiovascular biology, knowledge gaps, remaining challenges and future directions. A snapshot of other cardiovascular biological processes was also provided, including cardiac/vascular development, cardiomyocyte pyroptosis/apoptosis, cardiac fibroblast proliferation and cardiac hypertrophy, which are responsible for cardiovascular disorders. Further in‑depth investigation of HOX promises to provide a potential yet challenging landscape, albeit largely undetermined to date, for partially pinpointing the molecular mechanisms of atherosclerosis. A plethora of new targeted therapies may ultimately emerge against atherosclerosis, which is rapidly underway. However, translational undertakings are crucially important but increasingly challenging and remain an ongoing and monumental conundrum in the field.
Collapse
Affiliation(s)
- Yu Zhou
- Medical College, Guizhou University, Guiyang, Guizhou 550025, P.R. China
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Qiang Wu
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Yingchu Guo
- Department of Clinical Laboratory, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
13
|
Xiao W, Liu S, Huang Z, Jin D, Yang Y, Li F, Duan J, Wang J, Li Y, Deng P, Wang W, Meng C, Wu J, Cai J, Li Y, Hong Y. Non-high-density lipoprotein cholesterol levels as a risk factor for short-term mortality in elderly Chinese: a large-scale, population-based cohort study. BMJ Open 2023; 13:e078216. [PMID: 38097241 PMCID: PMC10728999 DOI: 10.1136/bmjopen-2023-078216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
OBJECTIVES To explore the association between non-high-density lipoprotein (non-HDL) and mortality risk, both short-term and long-term, in Chinese people. DESIGN A prospective cohort study. SETTING The National Basic Public Health Service (BPHS) in China. PARTICIPANTS Including 621 164 elderly individuals around Hunan Province who underwent healthcare management receiving check-ups in China BPHS from 2010 to 2020. EXCLUSION CRITERIA (1) missing information on gender; (2) missing records of lipid screening; (3) missing information on key covariates; and (4) missing records of comorbidities (cardiovascular disease, hypertension, diabetes, cancer.) PRIMARY AND SECONDARY OUTCOME MEASURES: The study's primary endpoint was all-cause and cause-specific mortality, sourced from Hunan's CDC(Center for Disease Control and Prevention)-operated National Mortality Surveillance System, tracking participants until 24 February 2021. RESULTS 26 758 (4.3%) deaths were recorded, with a median follow-up of 0.83 years. Association between non-HDL and mortality was non-linear after multivariable adjustment, with the optimum concentration (OC) being 3.29 and 4.85 mmol/L. Compared with OC, the risk increased by 1.12-fold for non-HDL <3.29 mmol/L (HR: 1.12 (1.09 to 1.15)) and 1.08-fold for non-HDL ≥4.85 mmol/L (HR: 1.08 (1.02 to 1.13)) for all-cause mortality. Furthermore, there is also an increased risk of cardiovascular mortality (HR for non-HDL <3.29: 1.10 (1.06 to 1.32) and HR for non-HDL ≥4.85: 1.07 (1.01 to 1.14)). However, cancer mortality risk was significantly increased only for non-HDL <3.29 mmol/L (HR: 1.11 (1.04 to 1.18)). Non-optimum concentration of non-HDL had significant effects on both the long-term and the short-term risk of mortality, especially for risks of mortality for all-cause (log HR:0 .086 (0.038 to 0.134)), cardiovascular (log HR:0 .082 (0.021 to 0.144)), and cancer (log HR:0 .187 (0.058 to 0.315)) within 3 months. A two-sided value of p <0.05 was considered to be statistically significant. CONCLUSIONS Non-HDL was non-linearly associated with the risk of mortality, and non-optimal concentrations of non-HDL significantly increased short-term mortality in elderly Chinese, which needs more attention for cardiovascular disease prevention.
Collapse
Affiliation(s)
- Weiwei Xiao
- General Medicine Department, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Shiqi Liu
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Zheng Huang
- Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Donghui Jin
- Disease Control and Prevention of Hunan Province, Changsha, Hunan, China
| | - Yiping Yang
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Fei Li
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Jingwen Duan
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Jie Wang
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Yalan Li
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Peizhi Deng
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Wei Wang
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Changjiang Meng
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Jingjing Wu
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Jingjing Cai
- Central South University Third Xiangya Hospital, Changsha City, China
| | - Yan Li
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| | - Yuan Hong
- Chlinical Research Center, Central South University Third Xiangya Hospital, Changsha, Hunan, China
- Department of Cardiology, Central South University Third Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
14
|
Kumar M, Deshmukh P, Kumar M, Bhatt A, Sinha AH, Chawla P. Vitamin E Supplementation and Cardiovascular Health: A Comprehensive Review. Cureus 2023; 15:e48142. [PMID: 38046702 PMCID: PMC10692867 DOI: 10.7759/cureus.48142] [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: 08/21/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
This article conducts a thorough investigation into the potential role of vitamin E in preventing cardiovascular diseases (CVDs) in the context of shifting mortality patterns from infectious diseases to the continued prominence of CVDs in modern medicine. The primary focus is on vitamin E's antioxidant properties and its specific ability to counter lipid peroxidation, a pivotal process in the early stages of atherosclerosis, a precursor to CVDs. The research spans a wide range of methodologies, including in vitro, in vivo, clinical, and experimental studies, examining how vitamin E affects critical aspects of cardiovascular health, such as signaling pathways, gene expression, inflammation, and cholesterol metabolism. It also explores vitamin E's influence on complex processes like smooth muscle cell development, oxidative stress reduction, foam cell formation, and the stability of atherosclerotic plaques. In the context of clinical studies, the article presents findings that both support and yield inconclusive results regarding the impact of vitamin E supplementation on CVDs. It acknowledges the intricate interplay of factors such as patient selection, pathophysiological conditions, and genetic variations, all of which can significantly influence the efficacy of vitamin E. The article underscores the need for ongoing research, with a specific focus on understanding the regulatory metabolites of vitamin E and their roles in modulating cellular processes relevant to CVDs. It highlights the potential for innovative therapeutic approaches based on a deeper comprehension of vitamin E's multifaceted effects. However, it also candidly addresses the challenges of translating clinical trial findings into practical applications and emphasizes the importance of considering diverse variables to optimize therapeutic outcomes. In summary, this meticulously conducted study provides a comprehensive examination of vitamin E's potential as a preventive agent against CVDs, recognizing the complexity of the subject and the need for continued research to unlock its full potential in cardiovascular health.
Collapse
Affiliation(s)
- Mayank Kumar
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Prasad Deshmukh
- Otolaryngology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Mayank Kumar
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Asmi Bhatt
- Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Arya Harshyt Sinha
- Anatomy, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Parth Chawla
- Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
15
|
Rastgoo S, Fateh ST, Nikbaf-Shandiz M, Rasaei N, Aali Y, Zamani M, Shiraseb F, Asbaghi O. The effects of L-carnitine supplementation on inflammatory and anti-inflammatory markers in adults: a systematic review and dose-response meta-analysis. Inflammopharmacology 2023; 31:2173-2199. [PMID: 37656233 DOI: 10.1007/s10787-023-01323-9] [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: 10/25/2022] [Accepted: 07/31/2023] [Indexed: 09/02/2023]
Abstract
L-carnitine supplementation may be beneficial in improving inflammatory conditions and reducing the level of inflammatory cytokines. Therefore, according to the finding of randomized controlled trials (RCTs), the systematic review and meta-analysis aimed to investigate the effect of L-carnitine supplementation on inflammation in adults. To obtain acceptable articles up to October 2022, a thorough search was conducted in databases including PubMed, ISI Web of Science, the Cochrane Library, and Scopus. A random-effects model was used to estimate the weighted mean difference (WMD). We included the 48 RCTs (n = 3255) with 51 effect sizes in this study. L-carnitine supplementation had a significant effect on C-reactive protein (CRP) (p < 0.001), interleukin-6 (IL-6) (p = 0.001), tumor necrosis factor-α (TNF-α) (p = 0.002), malondialdehyde (MDA) (p = 0.001), total antioxidant capacity (TAC) (p = 0.029), alanine transaminase (ALT) (p < 0.001), and aspartate transaminase (AST) (p < 0.001) in intervention, compared to the placebo group. Subgroup analyses showed that L-carnitine supplementation had a lowering effect on CRP and TNF-α in trial duration ≥ 12 weeks in type 2 diabetes and BMI ≥ 25 kg/m2. L-carnitine supplementation reduced ALT levels in overweight and normal BMI subjects at any trial dose and trial duration ≥ 12 weeks and reduced AST levels in overweight subjects and trial dose ≥ 2 g/day. This meta-analysis revealed that L-carnitine supplementation effectively reduces the inflammatory state by increasing the level of TAC and decreasing the levels of CRP, IL-6, TNF-α and MDA in the serum.
Collapse
Affiliation(s)
- Samira Rastgoo
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Niloufar Rasaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Yasaman Aali
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Zamani
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Shiraseb
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Omid Asbaghi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Orellana-Urzúa S, Briones-Valdivieso C, Chichiarelli S, Saso L, Rodrigo R. Potential Role of Natural Antioxidants in Countering Reperfusion Injury in Acute Myocardial Infarction and Ischemic Stroke. Antioxidants (Basel) 2023; 12:1760. [PMID: 37760064 PMCID: PMC10525378 DOI: 10.3390/antiox12091760] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Stroke and acute myocardial infarction are leading causes of mortality worldwide. The latter accounts for approximately 9 million deaths annually. In turn, ischemic stroke is a significant contributor to adult physical disability globally. While reperfusion is crucial for tissue recovery, it can paradoxically exacerbate damage through oxidative stress (OS), inflammation, and cell death. Therefore, it is imperative to explore diverse approaches aimed at minimizing ischemia/reperfusion injury to enhance clinical outcomes. OS primarily arises from an excessive generation of reactive oxygen species (ROS) and/or decreased endogenous antioxidant potential. Natural antioxidant compounds can counteract the injury mechanisms linked to ROS. While promising preclinical results, based on monotherapies, account for protective effects against tissue injury by ROS, translating these models into human applications has yielded controversial evidence. However, since the wide spectrum of antioxidants having diverse chemical characteristics offers varied biological actions on cell signaling pathways, multitherapy has emerged as a valuable therapeutic resource. Moreover, the combination of antioxidants in multitherapy holds significant potential for synergistic effects. This study was designed with the aim of providing an updated overview of natural antioxidants suitable for preventing myocardial and cerebral ischemia/reperfusion injuries.
Collapse
Affiliation(s)
- Sofía Orellana-Urzúa
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile;
| | | | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy;
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380000, Chile;
| |
Collapse
|
17
|
Bai Z, Hu H, Hu F, Ji J, Ji Z. Bone marrow mesenchymal stem cellsderived exosomes stabilize atherosclerosis through inhibiting pyroptosis. BMC Cardiovasc Disord 2023; 23:441. [PMID: 37679676 PMCID: PMC10486039 DOI: 10.1186/s12872-023-03453-y] [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: 11/29/2022] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
OBJECTIVES This study aimed to determine the effects of bone marrow mesenchymal stem cells (BMSCs)-derived exosomes (BMSC-EXO) on atherosclerosis (AS), and its related underlying mechanisms. METHODS Exosomes were isolated from mouse BMSCs, and identified by transmission electron microscopy (TEM), Nanosight (NTA), and western blot. A mouse AS model was established, and exosomes were injected into the tail vein. Total cholesterol (TC) and triglycerides (TG) were detected using their corresponding assay kits. The contents of IL-1β and IL-18 in serum were detected by ELISA. The mRNA and protein expression levels of GSDMD, Caspase1, and NLRP3 were detected by qRT-PCR and Western blot. Finally, aortic tissues in the Model and BMSC-EXO groups were sent for sequencing. RESULTS TEM, NTA, and western blot indicated successful isolation of exosomes. Compared with the control group, the TC, TG contents, IL-1β and IL-18 concentrations of the mice in the Model group were significantly increased; nonetheless, were significantly lower after injected with BMSC-EXO than those in the Model group (p < 0.05). Compared with the control group, the expressions of NLRP3, caspase-1 and GSDMD were significantly up-regulated in the Model group (p < 0.05), while the expressions of NLRP3, caspase-1, and GSDMD were significantly down-regulated by BMSC-EXO. By sequencing, a total of 3852 DEGs were identified between the Model and BMSC-EXO group and were significantly enriched in various biological processes and pathways related to mitochondrial function, metabolism, inflammation, and immune response. CONCLUSION AS can induce pyroptosis, and BMSC-EXO can reduce inflammation and alleviate the progression of AS by inhibiting NLRP3/Caspase-1/GSDMD in the pyroptosis pathway.
Collapse
Affiliation(s)
- Zhibin Bai
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Medical School, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China
| | - Haolin Hu
- Department of General Surgery, Institute for Minimally Invasive Surgery, Medical School, ZhongDa Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China
| | - Fangfang Hu
- Department of General Surgery, Institute for Minimally Invasive Surgery, Medical School, ZhongDa Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China
| | - Jiajie Ji
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Medical School, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China
| | - Zhenling Ji
- Department of General Surgery, Institute for Minimally Invasive Surgery, Medical School, ZhongDa Hospital, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
18
|
Abstract
Atherosclerotic disease, including stroke and myocardial infarction, is the leading cause of morbidity and mortality worldwide. Atherosclerotic plaque formation occurs in the setting of excess oxidative and hemodynamic stress and is perpetuated by smoking, poor diet, dyslipidemia, hypertension, and diabetes. Plaque may rupture, resulting in acute thrombotic events. Smoking cessation, lifestyle modification, risk factor optimization, and antithrombotic therapies are the mainstays of atherosclerotic disease management and are the cornerstones to reduce morbidity and mortality in this high-risk patient population. Novel therapeutics are in development and will add to the growing armamentarium available to physicians who manage atherosclerotic disease.
Collapse
Affiliation(s)
- Amanda M Morrison
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA
| | - Alexander E Sullivan
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA
| | - Aaron W Aday
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 300, Nashville, TN 37203, USA.
| |
Collapse
|
19
|
Domingues N, Gaifem J, Matthiesen R, Saraiva DP, Bento L, Marques ARA, Soares MIL, Sampaio J, Klose C, Surma MA, Almeida MS, Rodrigues G, Gonçalves PA, Ferreira J, E Melo RG, Pedro LM, Simons K, Pinho E Melo TMVD, Cabral MG, Jacinto A, Silvestre R, Vaz W, Vieira OV. Cholesteryl hemiazelate identified in CVD patients causes in vitro and in vivo inflammation. J Lipid Res 2023; 64:100419. [PMID: 37482218 PMCID: PMC10450993 DOI: 10.1016/j.jlr.2023.100419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/25/2023] Open
Abstract
Oxidation of PUFAs in LDLs trapped in the arterial intima plays a critical role in atherosclerosis. Though there have been many studies on the atherogenicity of oxidized derivatives of PUFA-esters of cholesterol, the effects of cholesteryl hemiesters (ChEs), the oxidation end products of these esters, have not been studied. Through lipidomics analyses, we identified and quantified two ChE types in the plasma of CVD patients and identified four ChE types in human endarterectomy specimens. Cholesteryl hemiazelate (ChA), the ChE of azelaic acid (n-nonane-1,9-dioic acid), was the most prevalent ChE identified in both cases. Importantly, human monocytes, monocyte-derived macrophages, and neutrophils exhibit inflammatory features when exposed to subtoxic concentrations of ChA in vitro. ChA increases the secretion of proinflammatory cytokines such as interleukin-1β and interleukin-6 and modulates the surface-marker profile of monocytes and monocyte-derived macrophage. In vivo, when zebrafish larvae were fed with a ChA-enriched diet, they exhibited neutrophil and macrophage accumulation in the vasculature in a caspase 1- and cathepsin B-dependent manner. ChA also triggered lipid accumulation at the bifurcation sites of the vasculature of the zebrafish larvae and negatively impacted their life expectancy. We conclude that ChA behaves as an endogenous damage-associated molecular pattern with inflammatory and proatherogenic properties.
Collapse
Affiliation(s)
- Neuza Domingues
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Joana Gaifem
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Portugal and ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rune Matthiesen
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Diana P Saraiva
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Luís Bento
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - André R A Marques
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Maria I L Soares
- Department of Chemistry, Coimbra Chemistry Centre, Institute of Molecular Sciences, University of Coimbra, Coimbra, Portugal
| | | | | | | | - Manuel S Almeida
- Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Carnaxide, Portugal
| | - Gustavo Rodrigues
- Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Carnaxide, Portugal
| | | | - Jorge Ferreira
- Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Carnaxide, Portugal
| | - Ryan Gouveia E Melo
- Department of Vascular Surgery, Hospital de Santa Maria, Centro Hospitalar Universitario Lisboa Norte (CHULN), Lisboa, Portugal
| | - Luís Mendes Pedro
- Department of Vascular Surgery, Hospital de Santa Maria, Centro Hospitalar Universitario Lisboa Norte (CHULN), Lisboa, Portugal
| | | | - Teresa M V D Pinho E Melo
- Department of Chemistry, Coimbra Chemistry Centre, Institute of Molecular Sciences, University of Coimbra, Coimbra, Portugal
| | - M Guadalupe Cabral
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Antonio Jacinto
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Portugal and ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Winchil Vaz
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Otília V Vieira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, (NMS, FCM), Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
20
|
Oxidative stress, mitochondrial dysfunction, and respiratory chain enzyme defects in inflammatory myopathies. Autoimmun Rev 2023; 22:103308. [PMID: 36822387 DOI: 10.1016/j.autrev.2023.103308] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023]
Abstract
We investigated the relationship between oxidative stress and inflammatory myopathies. We searched in the current literature the role of mitochondria and respiratory chain defects as sources of oxidative stress and reactive oxygen species production that led to muscle weakness and fatigue. Different molecules and pathways contribute to redox milieu, reactive oxygen species generation, accumulation of misfolded and carbonylated proteins that lose their ability to fulfil cellular activities. Small peptides and physical techniques proved, in mice models, to reduce oxidative stress. We focused on inclusion body myositis, as a major expression of myopathy related to oxidative stress, where mitochondrial abnormalities are causative agents as well. We described the effect of physical exercise in inclusion body myositis that showed to increase strength and to reduce beta amyloid accumulation with subsequent improvement of the mitochondrial functions. We illustrated the influence of epigenetic control on the immune system by non-coding genetic material in the interaction between oxidative stress and inflammatory myopathies.
Collapse
|
21
|
Development and validation of novel automatable assay for cholesterol efflux capacity. Biosci Rep 2023; 43:232455. [PMID: 36645426 PMCID: PMC9905788 DOI: 10.1042/bsr20221519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/24/2022] [Accepted: 01/16/2023] [Indexed: 01/17/2023] Open
Abstract
During the past decade, evaluation of high-density lipoprotein (HDL) functionality has been well studied for predicting cardiovascular disease (CVD) risk. Cholesterol efflux capacity (CEC) is the strongest candidate as the biomarker out of various HDL antiatherosclerotic functions. However, CEC has not yet been introduced clinically because of several technical issues, including the use of radioactive materials and differentiated cells in the assay. Previously, our laboratory developed a radioisotope- and cell-free CEC assay called the immobilized liposome-bound gel beads (ILGs) method to replace the conventional method. However, the separation process of the supernatant was not suitable for installation in an automatic analyzer. The present study aims to develop a new method that is easier to operate. We assumed that the use of magnetic beads instead of gel beads would enable the skip of the centrifugal process. First, similar to the ILG method, porous magnetic beads were treated with liposomes containing fluorescently labeled cholesterol. Fluorescence was observed inside the magnetic beads, and almost the same amount of liposomes as in the ILG method was immobilized successfully. These immobilized liposome-bound magnetic beads (ILMs) were available for CEC assay when HDL and apolipoprotein B-100-depleted serum (BDS) were used as cholesterol acceptors. The ILM method showed sufficient basic performance and a good correlation with the ILG method. Furthermore, when the CEC of 15 serum samples from healthy subjects was measured, a good correlation between HDL-cholesterol level and the ILG method was confirmed. Thus, it was confirmed that the ILM method was successfully developed and could be automated.
Collapse
|
22
|
Attia AA, Sorour JM, Mohamed NA, Mansour TT, Al-Eisa RA, El-Shenawy NS. Biochemical, Histological, and Ultrastructural Studies of the Protective Role of Vitamin E on Cyclophosphamide-Induced Cardiotoxicity in Male Rats. Biomedicines 2023; 11:biomedicines11020390. [PMID: 36830928 PMCID: PMC9952974 DOI: 10.3390/biomedicines11020390] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Cyclophosphamide (CP) (Cytoxan or Endoxan) is an efficient anti-tumor agent, widely used for the treatment of various neoplastic diseases. The study aimed to investigate the protective role of vitamin E (vit E) in improving cardiotoxicity in rats induced by CP. MATERIALS AND METHODS Forty male Wistar rats were divided randomly into four experimental groups (each consisting of ten rats); the control group was treated with saline. The other three groups were treated with vit E, CP, and the combination of vit E and CP. Serum lipid profiles, enzyme cardiac biomarkers, and cardiac tissue antioxidants were evaluated, as well as histological and ultrastructure investigations. RESULTS CP-treated rats showed a significant increase in serum levels of cardiac markers (troponin, CK, LDH, AST, and ALT), lipid profiles, a reduction in the antioxidant enzyme activities (CAT, SOD, and GPx), and an elevation in the level of lipid peroxidation (LPO). The increase in the levels of troponin, LDH, AST, ALP, and triglycerides is a predominant indicator of cardiac damage due to the toxic effect of CP. The biochemical changes parallel cardiac injuries such as myocardial infarction, myocarditis, and heart failure. Vitamin E played a pivotal role, as it attenuated most of these changes because of its ability to scavenge free radicals and reduce LPO. In addition, vit E was found to improve the histopathological alterations caused by CP where no evidence of damage was observed in the cardiac architecture, and the cardiac fibers had regained their normal structure with minimal hemorrhage. CONCLUSIONS As a result of its antioxidant activity and its stabilizing impact on the cardiomyocyte membranes, vit E is recommended as a potential candidate in decreasing the damaging effects of CP.
Collapse
Affiliation(s)
- Azza A. Attia
- Zoology Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Jehan M. Sorour
- Zoology Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Neama A. Mohamed
- Zoology Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Tagreed T. Mansour
- Zoology Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Rasha A. Al-Eisa
- Biology Department, Main Campus, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Nahla S. El-Shenawy
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
- Correspondence:
| |
Collapse
|
23
|
HDL Functions-Current Status and Future Perspectives. Biomolecules 2023; 13:biom13010105. [PMID: 36671490 PMCID: PMC9855960 DOI: 10.3390/biom13010105] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in Western countries. A low HDL-C is associated with the development of CVD. However, recent epidemiology studies have shown U-shaped curves between HDL-C and CVD mortality, with paradoxically increased CVD mortality in patients with extremely high HDL-C levels. Furthermore, HDL-C raising therapy using nicotinic acids or CETP inhibitors mostly failed to reduce CVD events. Based on this background, HDL functions rather than HDL-C could be a novel biomarker; research on the clinical utility of HDL functionality is ongoing. In this review, we summarize the current status of HDL functions and their future perspectives from the findings of basic research and clinical trials.
Collapse
|
24
|
Finney AC, Das S, Kumar D, McKinney MP, Cai B, Yurdagul A, Rom O. The interplay between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease. Front Cardiovasc Med 2023; 10:1116861. [PMID: 37200978 PMCID: PMC10185914 DOI: 10.3389/fcvm.2023.1116861] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/23/2023] [Indexed: 05/20/2023] Open
Abstract
Therapeutic approaches that lower circulating low-density lipoprotein (LDL)-cholesterol significantly reduced the burden of cardiovascular disease over the last decades. However, the persistent rise in the obesity epidemic is beginning to reverse this decline. Alongside obesity, the incidence of nonalcoholic fatty liver disease (NAFLD) has substantially increased in the last three decades. Currently, approximately one third of world population is affected by NAFLD. Notably, the presence of NAFLD and particularly its more severe form, nonalcoholic steatohepatitis (NASH), serves as an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), thus, raising interest in the relationship between these two diseases. Importantly, ASCVD is the major cause of death in patients with NASH independent of traditional risk factors. Nevertheless, the pathophysiology linking NAFLD/NASH with ASCVD remains poorly understood. While dyslipidemia is a common risk factor underlying both diseases, therapies that lower circulating LDL-cholesterol are largely ineffective against NASH. While there are no approved pharmacological therapies for NASH, some of the most advanced drug candidates exacerbate atherogenic dyslipidemia, raising concerns regarding their adverse cardiovascular consequences. In this review, we address current gaps in our understanding of the mechanisms linking NAFLD/NASH and ASCVD, explore strategies to simultaneously model these diseases, evaluate emerging biomarkers that may be useful to diagnose the presence of both diseases, and discuss investigational approaches and ongoing clinical trials that potentially target both diseases.
Collapse
Affiliation(s)
- Alexandra C. Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Sandeep Das
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Dhananjay Kumar
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - M. Peyton McKinney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| |
Collapse
|
25
|
Slowey C, Nyhan D. The Vascular System: Anatomical, Physiological, Pathological, and Aging Considerations. Anesthesiol Clin 2022; 40:557-574. [PMID: 36328615 DOI: 10.1016/j.anclin.2022.08.004] [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: 06/16/2023]
Abstract
The vascular system is one of the earliest recognized anatomical systems. It is composed of 3 parts; arterial, capillary, and venous, each with their own unique anatomy and physiology. Blood flow through this system is compromised in aging, atherosclerosis and peripheral vascular disease, and the practicing anesthesiologist must understand both the physiology and pathophysiology of the vascular tree.
Collapse
Affiliation(s)
- Charlie Slowey
- Johns Hopkins Department of Anesthesiology and Critical Care Medicine, 600 North Wolf Street, Baltimore, MD 21287, USA.
| | - Daniel Nyhan
- Johns Hopkins Department of Anesthesiology and Critical Care Medicine, 600 North Wolf Street, Baltimore, MD 21287, USA
| |
Collapse
|
26
|
Li Z, Zhang R, Mu H, Zhang W, Zeng J, Li H, Wang S, Zhao X, Chen W, Dong J, Yang R. Oral Administration of Branched-Chain Amino Acids Attenuates Atherosclerosis by Inhibiting the Inflammatory Response and Regulating the Gut Microbiota in ApoE-Deficient Mice. Nutrients 2022; 14:5065. [PMID: 36501095 PMCID: PMC9739883 DOI: 10.3390/nu14235065] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that serves as a common pathogenic underpinning for various cardiovascular diseases. Although high circulating branched-chain amino acid (BCAA) levels may represent a risk factor for AS, it is unclear whether dietary BCAA supplementation causes elevated levels of circulating BCAAs and hence influences AS, and the related mechanisms are not well understood. Here, ApoE-deficient mice (ApoE-/-) were fed a diet supplemented with or without BCAAs to investigate the effects of BCAAs on AS and determine potential related mechanisms. In this study, compared with the high-fat diet (HFD), high-fat diet supplemented with BCAAs (HFB) reduced the atherosclerotic lesion area and caused a significant decrease in serum cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels. BCAA supplementation suppressed the systemic inflammatory response by reducing macrophage infiltration; lowering serum levels of inflammatory factors, including monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6); and suppressing inflammatory related signaling pathways. Furthermore, BCAA supplementation altered the gut bacterial beta diversity and composition, especially reducing harmful bacteria and increasing probiotic bacteria, along with increasing bile acid (BA) excretion. In addition, the levels of total BAs, primary BAs, 12α-hydroxylated bile acids (12α-OH BAs) and non-12α-hydroxylated bile acids (non-12α-OH BAs) in cecal and colonic contents were increased in the HFB group of mice compared with the HFD group. Overall, these data indicate that dietary BCAA supplementation can attenuate atherosclerosis induced by HFD in ApoE-/- mice through improved dyslipidemia and inflammation, mechanisms involving the intestinal microbiota, and promotion of BA excretion.
Collapse
Affiliation(s)
- Ziyun Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Ranran Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Hongna Mu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Wenduo Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jie Zeng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Hongxia Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Siming Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Xianghui Zhao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Wenxiang Chen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Jun Dong
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Ruiyue Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| |
Collapse
|
27
|
Pantazi D, Tellis C, Tselepis AD. Oxidized phospholipids and lipoprotein-associated phospholipase A 2 (Lp-PLA 2 ) in atherosclerotic cardiovascular disease: An update. Biofactors 2022; 48:1257-1270. [PMID: 36192834 DOI: 10.1002/biof.1890] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022]
Abstract
Inflammation and oxidative stress conditions lead to a variety of oxidative modifications of lipoprotein phospholipids implicated in the occurrence and development of atherosclerotic lesions. Lipoprotein-associated phospholipase A2 (Lp-PLA2 ) is established as an independent risk biomarker of atherosclerosis-related cardiovascular disease (ASCVD) and mediates vascular inflammation through the regulation of lipid metabolism in the blood and in atherosclerotic lesions. Lp-PLA2 is associated with low- and high-density lipoproteins and Lipoprotein (a) in human plasma and specifically hydrolyzes oxidized phospholipids involved in oxidative stress modification. Several oxidized phospholipids (OxPLs) subspecies can be detoxified through enzymatic degradation by Lp-PLA2 activation, forming lysophospholipids and oxidized non-esterified fatty acids (OxNEFAs). Lysophospholipids promote the expression of adhesion molecules, stimulate cytokines production (TNF-α, IL-6), and attract macrophages to the arterial intima. The present review article discusses new data on the functional roles of OxPLs and Lp-PLA2 associated with lipoproteins.
Collapse
Affiliation(s)
- Despoina Pantazi
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Constantinos Tellis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Alexandros D Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
28
|
Scisciola L, Cataldo V, Taktaz F, Fontanella RA, Pesapane A, Ghosh P, Franzese M, Puocci A, De Angelis A, Sportiello L, Marfella R, Barbieri M. Anti-inflammatory role of SGLT2 inhibitors as part of their anti-atherosclerotic activity: Data from basic science and clinical trials. Front Cardiovasc Med 2022; 9:1008922. [PMID: 36148061 PMCID: PMC9485634 DOI: 10.3389/fcvm.2022.1008922] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/16/2022] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is a progressive inflammatory disease leading to mortality and morbidity in the civilized world. Atherosclerosis manifests as an accumulation of plaques in the intimal layer of the arterial wall that, by its subsequent erosion or rupture, triggers cardiovascular diseases. Diabetes mellitus is a well-known risk factor for atherosclerosis. Indeed, Type 2 diabetes mellitus patients have an increased risk of atherosclerosis and its associated-cardiovascular complications than non-diabetic patients. Sodium-glucose co-transport 2 inhibitors (SGLT2i), a novel anti-diabetic drugs, have a surprising advantage in cardiovascular effects, such as reducing cardiovascular death in a patient with or without diabetes. Numerous studies have shown that atherosclerosis is due to a significant inflammatory burden and that SGLT2i may play a role in inflammation. In fact, several experiment results have demonstrated that SGLT2i, with suppression of inflammatory mechanism, slows the progression of atherosclerosis. Therefore, SGLT2i may have a double benefit in terms of glycemic control and control of the atherosclerotic process at a myocardial and vascular level. This review elaborates on the anti-inflammatory effects of sodium-glucose co-transporter 2 inhibitors on atherosclerosis.
Collapse
Affiliation(s)
- Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- *Correspondence: Lucia Scisciola
| | - Vittoria Cataldo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Fatemeh Taktaz
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Rosaria Anna Fontanella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Ada Pesapane
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Puja Ghosh
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Martina Franzese
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Armando Puocci
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli”, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Liberata Sportiello
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli”, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
29
|
Salekeen R, Haider AN, Akhter F, Billah MM, Islam ME, Didarul Islam KM. Lipid oxidation in pathophysiology of atherosclerosis: Current understanding and therapeutic strategies. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2022; 14:200143. [PMID: 36060286 PMCID: PMC9434419 DOI: 10.1016/j.ijcrp.2022.200143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/04/2022] [Accepted: 07/18/2022] [Indexed: 01/21/2023]
Abstract
A marked increase in the global prevalence of ischemic heart disease demands focused research for novel and more effective therapeutic strategies. At present, atherosclerotic cardiovascular disease (ACVD) is the leading cause of the global incidence of heart attacks and a major contributor to many peripheral cardiac diseases. Decades of research have unearthed the complex and multidimensional pathophysiology of ACVD encompassing oxidative stress, redox imbalance, lipid peroxidation, pro-inflammatory signaling, hyperglycemic stress and diabetes mellitus, chronic low-grade inflammation and aging, immune dysregulation, vascular dysfunction, loss of hemostasis, thrombosis, and fluid shear stress. However, the scientific basis of therapeutic interventions using conventional understandings of the disease mechanisms has been subject to renewed scrutiny with novel findings in recent years. This critical review attempts to revise the pathophysiological mechanisms of atherosclerosis using a recent body of literature, with a focus on lipid metabolism and associated cellular and biochemical processes. The comprehensive study encompasses different molecular perspectives in the development and progression of coronary atherosclerosis. The review also summarizes currently prescribed small molecule therapeutics in inflammation and ACVD, and overviews prospective management measures under development including peptides and microRNA therapeutics. The study provides updated insights into the current knowledge of coronary atherosclerosis, and highlights the need for effective prevention, management and development of novel intervention approaches to overcome this chronic epidemic.
Collapse
Affiliation(s)
- Rahagir Salekeen
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | - Abu Nasim Haider
- Biotechnology Program, Department of Mathematics and Natural Sciences, BRAC University, Dhaka, 1212, Bangladesh
| | - Fouzia Akhter
- Khulna Medical College Hospital, Khulna, 9000, Bangladesh
| | - Md Morsaline Billah
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | - Md Emdadul Islam
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | - Kazi Mohammed Didarul Islam
- Biotechnology and Genetic Engineering Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| |
Collapse
|
30
|
Mamillapalli R, Toffoloni N, Habata S, Qunhua H, Atwani R, Stachenfeld N, Taylor HS. Endometriosis promotes atherosclerosis in a murine model. Am J Obstet Gynecol 2022; 227:248.e1-248.e8. [PMID: 35351413 PMCID: PMC9308711 DOI: 10.1016/j.ajog.2022.03.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Epidemiologic studies have demonstrated an association between endometriosis and the subsequent development of cardiovascular disease. The direct effect of endometriosis on the progression of atherosclerotic, if any, has not been previously characterized. Endometriosis leads to systemic inflammation that could have consequences for cardiovascular health. Here, we reported the effects of endometriosis on the development of atherosclerosis in a murine model. OBJECTIVE This study aimed to determine the contribution of endometriosis in promoting cardiovascular disease in a murine model of endometriosis. STUDY DESIGN Endometriosis was induced in 18 apolipoprotein E-null mice, the standard murine model used to study atherosclerosis. Mice of the same strain were used as controls (n=18) and underwent sham surgery without inducing endometriosis. The formation of endometriotic lesions was confirmed after 25 weeks of induction. Atherosclerotic lesions were subjected to hematoxylin and eosin staining followed by measurement of the aortic root luminal area and wall thickness. The whole aorta was isolated, and Oil Red O staining was performed to quantify the lipid deposits or plaque formation; moreover, biochemical assays were carried out in serum to determine the levels of lipids and inflammatory-related cytokines. RESULTS Apolipoprotein E mice with endometriosis exhibited increased aortic atherosclerosis compared with controls as measured using Oil Red O staining (7.9% vs 3.1%, respectively; P=.0004). Mice with endometriosis showed a significant 50% decrease in the aortic luminal area compared with sham mice (0.85 mm2 vs 1.46 mm2; P=.03) and a significant increase in aortic root wall thickness (0.22 mm vs 0.15 mm; P=.04). There was no difference in the lipoprotein profile (P<.05) between mice with endometriosis and sham mice. The serum levels of inflammatory cytokines interleukin 1 alpha, interleukin 6, interferon gamma, and vascular endothelial growth factor were significantly (P<.05)increased in the endometriosis mice. CONCLUSION Our study used a murine model to determine the effect of endometriosis on atherosclerosis. Inflammation-related cytokines interleukin 1 alpha, interleukin 6, interferon gamma, and vascular endothelial growth factor (angiogenic factor) released by endometriotic lesions may contribute to the increased cardiovascular risks in women with endometriosis. To reduce the risk of cardiovascular disease, early identification and treatment of endometriosis are essential. Future treatments targeting inflammatory cytokines may help reduce the long-term risk of cardiovascular disease in women with endometriosis.
Collapse
Affiliation(s)
- Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT.
| | - Nikoletta Toffoloni
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Shutaro Habata
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Huang Qunhua
- Department of Surgery (Cardiac Surgery), Yale School of Medicine, New Haven, CT
| | - Rula Atwani
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Nina Stachenfeld
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT.
| |
Collapse
|
31
|
Dyslipidemia in Transplant Patients: Which Therapy? J Clin Med 2022; 11:jcm11144080. [PMID: 35887846 PMCID: PMC9318180 DOI: 10.3390/jcm11144080] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease is the most important cause of death worldwide in recent years; an increasing trend is also shown in organ transplant patients subjected to immunosuppressive therapies, in which cardiovascular diseases represent one of the most frequent causes of long-term mortality. This is also linked to immunosuppressant-induced dyslipidemia, which occurs in 27 to 71% of organ transplant recipients. The aim of this review is to clarify the pathophysiological mechanisms underlying dyslipidemia in patients treated with immunosuppressants to identify immunosuppressive therapies which do not cause dyslipidemia or therapeutic pathways effective in reducing hypercholesterolemia, hypertriglyceridemia, or both, without further adverse events.
Collapse
|
32
|
Koite NLN, Sanogo NI, Lépine O, Bard JM, Ouguerram K. Antioxidant Efficacy of a Spirulina Liquid Extract on Oxidative Stress Status and Metabolic Disturbances in Subjects with Metabolic Syndrome. Mar Drugs 2022; 20:md20070441. [PMID: 35877734 PMCID: PMC9318250 DOI: 10.3390/md20070441] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 01/30/2023] Open
Abstract
Lipid peroxidation is associated with the development of some pathologies, such as cardiovascular diseases. Reduction in oxidative stress by antioxidants, such as Arthrospira (formely Spirulina), helps improving this redox imbalance. The aim of the study was to evaluate the effect of the Arthrospira liquid extract “Spirulysat®” on oxidative markers—in particular, oxidized LDL (oxLDL)/total LDL cholesterol—and isoprostanes and to investigate its impact on lipid and glucose metabolism in the metabolic syndrome subject. A controlled, randomised, double-blind design was conducted in 40 subjects aged 18 to 65 years with metabolic syndrome after a daily intake of Spirulysat® or placebo for twelve weeks. Blood and urinary samples were collected at three visits (V1, V2, V3) in the two groups for parameters determination. Although the Spirulysat® group showed a decrease at all visits of the oxLDL/total cholesterol ratio, there was no significant difference compared to the placebo (p = 0.36). The urinary isoprostanes concentration in the Spirulysat® group was reduced (p = 0.014) at V3. Plasma triglycerides decreased at V3 (p = 0.003) and HDL-cholesterol increased (p = 0.031) at all visits with Spirulysat®. In conclusion, Spirulysat® did not change the oxidized LDL (oxLDL)/LDL ratio but decreased the urinary isoprostanes, plasma triglycerides and increased HDL cholesterol, suggesting a beneficial effect on metabolic syndrome.
Collapse
Affiliation(s)
- N’Deye Lallah Nina Koite
- Département de Recherche en Santé Publique, Faculté de Pharmacie, Université des Sciences, des Techniques et des Technologies, Bamako J287+PM5, Mali;
| | | | | | - Jean-Marie Bard
- Centre National de la Recherche Scientifique, ISOMer—UE 2160, IUML—Institut Universitaire Mer et Littoral, Nantes Université, 44035 Nantes, France;
- Institut de Cancérologie de l’Ouest, 44805 Saint-Herblain, France
- Centre de Recherche en Nutrition Humaine Ouest (CRNH-O), 44093 Nantes, France
| | - Khadija Ouguerram
- Centre de Recherche en Nutrition Humaine Ouest (CRNH-O), Unité Mixte de Recherche, Institut des Maladies de l’Appareil Digestif (IMAD), NRAE, Physiopathologie des Adaptations Nutritionnelles (PhAN), Nantes Université, 44093 Nantes, France
- Correspondence: ; Tel.: +33-240-084-485
| |
Collapse
|
33
|
Cuomo G, Cioffi G, Di Lorenzo A, Iannone FP, Cudemo G, Iannicelli AM, Pacileo M, D’Andrea A, Vigorito C, Iannuzzo G, Giallauria F. Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors Use for Atherogenic Dyslipidemia in Solid Organ Transplant Patients. J Clin Med 2022; 11:jcm11113247. [PMID: 35683632 PMCID: PMC9180971 DOI: 10.3390/jcm11113247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 01/27/2023] Open
Abstract
Dyslipidemia is a widespread risk factor in solid organ transplant patients, due to many reasons, such as the use of immunosuppressive drugs, with a consequent increase in cardiovascular diseases in this population. PCSK9 is an enzyme mainly known for its role in altering LDL levels, consequently increasing cardiovascular risk. Monoclonal antibody PCSK9 inhibitors demonstrated remarkable efficacy in the general population in reducing LDL cholesterol levels and preventing cardiovascular disease. In transplant patients, these drugs are still poorly used, despite having comparable efficacy to the general population and giving fewer drug interactions with immunosuppressants. Furthermore, there is enough evidence that PCSK9 also plays a role in other pathways, such as inflammation, which is particularly dangerous for graft survival. In this review, the current evidence on the function of PCSK9 and the use of its inhibitors will be discussed, particularly in transplant patients, in which they may provide additional benefits.
Collapse
Affiliation(s)
- Gianluigi Cuomo
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (G.C.); (G.C.); (A.D.L.); (G.C.); (A.M.I.); (C.V.)
| | - Giuseppe Cioffi
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (G.C.); (G.C.); (A.D.L.); (G.C.); (A.M.I.); (C.V.)
| | - Anna Di Lorenzo
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (G.C.); (G.C.); (A.D.L.); (G.C.); (A.M.I.); (C.V.)
| | - Francesca Paola Iannone
- Department of Clinical Medicine and Surgery, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (F.P.I.); (G.I.)
| | - Giuseppe Cudemo
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (G.C.); (G.C.); (A.D.L.); (G.C.); (A.M.I.); (C.V.)
| | - Anna Maria Iannicelli
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (G.C.); (G.C.); (A.D.L.); (G.C.); (A.M.I.); (C.V.)
| | - Mario Pacileo
- Unit of Cardiology and Intensive Care, Umberto I Hospital, 84014 Nocera Inferiore, Italy; (M.P.); (A.D.)
| | - Antonello D’Andrea
- Unit of Cardiology and Intensive Care, Umberto I Hospital, 84014 Nocera Inferiore, Italy; (M.P.); (A.D.)
| | - Carlo Vigorito
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (G.C.); (G.C.); (A.D.L.); (G.C.); (A.M.I.); (C.V.)
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (F.P.I.); (G.I.)
| | - Francesco Giallauria
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (G.C.); (G.C.); (A.D.L.); (G.C.); (A.M.I.); (C.V.)
- Correspondence:
| |
Collapse
|
34
|
Shishikura D, Octavia Y, Hayat U, Thondapu V, Barlis P. Atherogenesis and Inflammation. Interv Cardiol 2022. [DOI: 10.1002/9781119697367.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
35
|
Koschinsky ML, Boffa MB. Oxidized phospholipid modification of lipoprotein(a): Epidemiology, biochemistry and pathophysiology. Atherosclerosis 2022; 349:92-100. [DOI: 10.1016/j.atherosclerosis.2022.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/22/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023]
|
36
|
Li Y, Karim MR, Wang B, Peng J. Effects of Green Tea (-)-Epigallocatechin-3-Gallate (EGCG) on Cardiac Function - A Review of the Therapeutic Mechanism and Potentials. Mini Rev Med Chem 2022; 22:2371-2382. [PMID: 35345998 DOI: 10.2174/1389557522666220328161826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/10/2022] [Accepted: 02/09/2022] [Indexed: 11/22/2022]
Abstract
Heart disease, the leading cause of death globally, refers to various illnesses that affect heart structure and function. Specific abnormalities affecting cardiac muscle contractility and remodeling and common factors including oxidative stress, inflammation, and apoptosis underlie the pathogenesis of heart diseases. Epidemiology studies have associated green tea consumption with lower morbidity and mortality of cardiovascular diseases, including heart and blood vessel dysfunction. Among the various compounds found in green tea, catechins are believed to play a significant role in producing benefits to cardiovascular health. Comprehensive literature reviews have been published to summarize the tea catechins' antioxidative, anti-inflammatory, and anti-apoptosis effects in the context of various diseases, such as cardiovascular diseases, cancers, and metabolic diseases. However, recent studies on tea catechins, especially the most abundant (-)-Epigallocatechin-3-Gallate (EGCG), revealed their capabilities in regulating cardiac muscle contraction by directly altering myofilament Ca2+ sensitivity on force development and Ca2+ ion handling in cardiomyocytes under both physiological and pathological conditions. In vitro and in vivo data also demonstrated that green tea extract or EGCG protected or rescued cardiac function, independent of their well-known effects against oxidative stress and inflammation. This minireview will focus on the specific effects of tea catechins on heart muscle contractility at the molecular and cellular level, revisit their effects on oxidative stress and inflammation in a variety of heart diseases, and discuss EGCG's potential as one of the lead compounds for new drug discovery for heart diseases.
Collapse
Affiliation(s)
- Yuejin Li
- Department of Biology, Morgan State University, Baltimore
| | | | - Buheng Wang
- Department of Biology, Morgan State University, Baltimore
| | - Jiangnan Peng
- Department of Biology, Morgan State University, Baltimore
- Department of Chemistry, Morgan State University, Baltimore
| |
Collapse
|
37
|
Assessment of Ex Vivo Antioxidative Potential of Murine HDL in Atherosclerosis. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:283-292. [PMID: 35237971 DOI: 10.1007/978-1-0716-1924-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This chapter provides details on a simple and reproducible method used to determine the capacity of murine HDL to prevent the oxidation of LDL . The principle of the method is based on the rearrangement of double bonds of polyunsaturated fatty acids that occurs during the oxidation of human LDL , which generates a sigmoidal curve. The shape and length of the curve is modified in the presence of HDL , and such modifications are easily quantifiable by measuring the absorbance of conjugated dienes at 234 nm. The general technique described herein may be applied to evaluate the effect of HDL obtained from different experimental murine models of atherosclerosis.
Collapse
|
38
|
Akhmedov A, Crucet M, Simic B, Kraler S, Bonetti NR, Ospelt C, Distler O, Ciurea A, Liberale L, Jauhiainen M, Metso J, Miranda M, Cydecian R, Schwarz L, Fehr V, Zilinyi R, Amrollahi-Sharifabadi M, Ntari L, Karagianni N, Ruschitzka F, Laaksonen R, Vanhoutte PM, Kollias G, Camici GG, Lüscher TF. TNFα induces endothelial dysfunction in rheumatoid arthritis via LOX-1 and arginase 2: reversal by monoclonal TNFα antibodies. Cardiovasc Res 2022; 118:254-266. [PMID: 33483748 DOI: 10.1093/cvr/cvab005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/08/2021] [Indexed: 02/02/2023] Open
Abstract
AIMS Rheumatoid arthritis (RA) is a chronic inflammatory disease affecting joints and blood vessels. Despite low levels of low-density lipoprotein cholesterol (LDL-C), RA patients exhibit endothelial dysfunction and are at increased risk of death from cardiovascular complications, but the molecular mechanism of action is unknown. We aimed in the present study to identify the molecular mechanism of endothelial dysfunction in a mouse model of RA and in patients with RA. METHODS AND RESULTS Endothelium-dependent relaxations to acetylcholine were reduced in aortae of two tumour necrosis factor alpha (TNFα) transgenic mouse lines with either mild (Tg3647) or severe (Tg197) forms of RA in a time- and severity-dependent fashion as assessed by organ chamber myograph. In Tg197, TNFα plasma levels were associated with severe endothelial dysfunction. LOX-1 receptor was markedly up-regulated leading to increased vascular oxLDL uptake and NFκB-mediated enhanced Arg2 expression via direct binding to its promoter resulting in reduced NO bioavailability and vascular cGMP levels as shown by ELISA and chromatin immunoprecipitation. Anti-TNFα treatment with infliximab normalized endothelial function together with LOX-1 and Arg2 serum levels in mice. In RA patients, soluble LOX-1 serum levels were also markedly increased and closely related to serum levels of C-reactive protein. Similarly, ARG2 serum levels were increased. Similarly, anti-TNFα treatment restored LOX-1 and ARG2 serum levels in RA patients. CONCLUSIONS Increased TNFα levels not only contribute to RA, but also to endothelial dysfunction by increasing vascular oxLDL content and activation of the LOX-1/NFκB/Arg2 pathway leading to reduced NO bioavailability and decreased cGMP levels. Anti-TNFα treatment improved both articular symptoms and endothelial function by reducing LOX-1, vascular oxLDL, and Arg2 levels.
Collapse
MESH Headings
- Adult
- Animals
- Animals, Genetically Modified
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/immunology
- Aorta, Thoracic/physiopathology
- Arginase/genetics
- Arginase/metabolism
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/enzymology
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/physiopathology
- Case-Control Studies
- Disease Models, Animal
- Endothelial Cells/drug effects
- Endothelial Cells/enzymology
- Endothelial Cells/immunology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/physiopathology
- Female
- Humans
- Lipoproteins, LDL/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Middle Aged
- NF-kappa B/metabolism
- Scavenger Receptors, Class E/genetics
- Scavenger Receptors, Class E/metabolism
- Signal Transduction
- Tumor Necrosis Factor Inhibitors/therapeutic use
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Vasodilation/drug effects
- Mice
Collapse
Affiliation(s)
- Alexander Akhmedov
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
| | - Margot Crucet
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
| | - Branko Simic
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
| | - Simon Kraler
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
| | - Nicole R Bonetti
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
| | - Caroline Ospelt
- Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Adrian Ciurea
- Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
- Department of Internal Medicine and Medical Specialties, University of Genova, Genova, Italy
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Jari Metso
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Melroy Miranda
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
| | - Rose Cydecian
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
| | - Lena Schwarz
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
| | - Vera Fehr
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
| | - Rita Zilinyi
- Department of Pharmacology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | | | - Lydia Ntari
- Institute for Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - Niki Karagianni
- Institute for Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center, University Hospital, Zürich, Switzerland
| | - Reijo Laaksonen
- Zora Biosciences Oy, Espoo, Finland
- Finnish Cardiovascular Research Center, University of Tampere and Finnish Clinical Biobank Tampere, Tampere University Hospital, Tampere, Finland
| | - Paul M Vanhoutte
- Department of Pharmacology, Hong Kong University, Hong Kong, Peoples Republic of China
| | - George Kollias
- Institute for Immunology, Biomedical Sciences Research Center Alexander Fleming, Vari, Greece
| | - Giovanni G Camici
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, 8001 Zurich, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| |
Collapse
|
39
|
Feng J, Wang Y, Li W, Zhao Y, Liu Y, Yao X, Liu S, Yu P, Li R. High levels of oxidized fatty acids in HDL impair the antioxidant function of HDL in patients with diabetes. Front Endocrinol (Lausanne) 2022; 13:993193. [PMID: 36339401 PMCID: PMC9630736 DOI: 10.3389/fendo.2022.993193] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/05/2022] [Indexed: 11/22/2022] Open
Abstract
AIMS Previous studies demonstrate that the antioxidant functions of high-density lipoprotein (HDL) are impaired in diabetic patients. The composition of HDL plays an important role in maintaining the normal functionality of HDL. In this study, we compared the levels of oxidized fatty acids in HDL from diabetic subjects and non-diabetic healthy controls, aiming to investigate the role of oxidized fatty acids in the antioxidant property of HDL. METHODS HDL was isolated from healthy subjects (n=6) and patients with diabetes (n=6, hemoglobin A1c ≥ 9%, fasting glucose ≥ 7 mmol/L) using a dextran sulfate precipitation method. Cholesterol efflux capacity mediated by HDL was measured on THP-1 derived macrophages. The antioxidant capacity of HDL was evaluated with dichlorofluorescein-based cellular assay in human aortic endothelial cells. Oxidized fatty acids in HDL were determined by liquid chromatography-tandem mass spectrometry. The correlations between the levels of oxidized fatty acids in HDL and the endothelial oxidant index in cells treated with HDLs were analyzed through Pearson's correlation analyses, and the effects of oxidized fatty acids on the antioxidant function of HDL were verified in vitro. RESULTS The cholesterol efflux capacity of HDL and the circulating HDL-cholesterol were similar in diabetic patients and healthy controls, whereas the antioxidant capacity of HDL was significantly decreased in diabetic patients. There were higher levels of oxidized fatty acids in HDL isolated from diabetic patients, which were strongly positively correlated with the oxidant index of cells treated with HDLs. The addition of a mixture of oxidized fatty acids significantly disturbed the antioxidant activity of HDL from healthy controls, while the apolipoprotein A-I mimetic peptide D-4F could restore the antioxidant function of HDL from diabetic patients. CONCLUSION HDL from diabetic patients displayed substantially impaired antioxidant activity compared to HDL from healthy subjects, which is highly correlated with the increased oxidized fatty acids levels in HDL.
Collapse
Affiliation(s)
- Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen Guangdong, China
| | - Yunfeng Wang
- Department of Endocrinology, Shenzhen Sami Medical Center (The Fourth People’s Hospital of Shenzhen), Shenzhen Guangdong, China
| | - Weixi Li
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen Guangdong, China
| | - Yue Zhao
- Clinical Laboratory, Shenzhen Sami Medical Center (The Fourth People’s Hospital of Shenzhen), Shenzhen Guangdong, China
| | - Yi Liu
- Clinical Laboratory, Shenzhen Sami Medical Center (The Fourth People’s Hospital of Shenzhen), Shenzhen Guangdong, China
| | - Xingang Yao
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou Guangdong, China
| | - Shuwen Liu
- National Medical Products Administration Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou Guangdong, China
| | - Ping Yu
- Department of Endocrinology, Shenzhen Sami Medical Center (The Fourth People’s Hospital of Shenzhen), Shenzhen Guangdong, China
- *Correspondence: Ping Yu, ; Rongsong Li,
| | - Rongsong Li
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen Guangdong, China
- *Correspondence: Ping Yu, ; Rongsong Li,
| |
Collapse
|
40
|
Aluganti Narasimhulu C, Parthasarathy S. Preparation of LDL , Oxidation , Methods of Detection, and Applications in Atherosclerosis Research. Methods Mol Biol 2022; 2419:213-246. [PMID: 35237967 DOI: 10.1007/978-1-0716-1924-7_13] [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: 06/14/2023]
Abstract
The concept of lipid peroxidation has been known for a long time. It is now well established that LDL plays a major role in atherosclerosis. Oxidized low-density lipoprotein (Ox-LDL) has been studied for over 35 years. Numerous pro- and anti-atherogenic properties have been attributed to Ox-LDL. Component composition of Ox-LDL is complex due to the influence of various factors, including the source, method of preparation, storage and use. Hence, it is very difficult to clearly define and characterize Ox-LDL. It contains unoxidized and oxidized fatty acid derivatives both in the ester and free forms, their decomposition products, cholesterol and its oxidized products, proteins with oxidized amino acids and cross-links, polypeptides with varying extents of covalent modification with lipid oxidation products and many others. The measurement of lipid oxidation has been a great boon, not only to the understanding of the process but also in providing numerous serendipitous discoveries and methodologies. In this chapter, we outline the methodologies for the preparation and testing of various lipoproteins for oxidation studies.
Collapse
Affiliation(s)
| | - Sampath Parthasarathy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
41
|
Zhang W, Liu J, Hu D, Li L, Cui L, Xu J, Wang W, Deng F, Guo X. Joint effect of multiple air pollutants on lipid profiles in obese and normal-weight young adults: The key role of ozone. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 292:118247. [PMID: 34624398 DOI: 10.1016/j.envpol.2021.118247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 06/13/2023]
Abstract
Dyslipidemia may be a potential mechanism linking air pollution to adverse cardiovascular outcomes and this may differ among obese and normal-weight populations. However, the joint effect of multiple air pollutants on lipid profiles and the role of each pollutant are still unclear. This panel study aims to investigate and compare the overall associations of major air pollutants with lipid parameters in obese and normal-weight adults, and assess the relative importance of each pollutant for lipid parameters. Forty-four obese and 53 normal-weight young adults were recruited from December 2017 to June 2018 in Beijing, China. Their fasting blood was collected and serum lipid levels were measured in three visits. Six major air pollutants were included in this study, which were PM2.5, PM10, NO2, SO2, O3 and CO. Bayesian kernel machine regression (BKMR) was implemented to estimate the joint effect of the six air pollutants on various lipid parameters. We found that decreased high-density lipoprotein cholesterol (HDL-C) in the obese group and increased low-density lipoprotein cholesterol (LDL-C) and non-HDL-C in the normal-weight group were associated with the exposure to the mixture of six air pollutants above. Significant increases in total cholesterol (TC)/HDL-C and non-HDL-C/HDL-C were observed in both groups, and the effect was stronger in obese group. Of the six air pollutants above, O3 had the largest posterior inclusion probability in above lipid indices, ranging from 0.75 to 1.00. In the obese group, approximately linear exposure-response relationships were observed over the whole range of logarithmic O3-8 h max concentration, while in the normal-weight group, these relationships existed when the logarithmic concentration exceeded about 2.8. Therefore, lipid profiles of obese adults may be more sensitive to air pollution and this study highlights the importance of strengthening emissions control efforts for O3 in the future.
Collapse
Affiliation(s)
- Wenlou Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Junxiu Liu
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Dayu Hu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Luyi Li
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Junhui Xu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Wanzhou Wang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| | - Furong Deng
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China.
| | - Xinbiao Guo
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100191, China
| |
Collapse
|
42
|
Horiuchi Y, Lai SJ, Kameda T, Tozuka M, Ohkawa R. Novel cholesterol efflux assay using immobilized liposome-bound gel beads: Confirmation and improvement for application in clinical laboratory. Ann Clin Biochem 2021; 59:134-143. [PMID: 34719976 DOI: 10.1177/00045632211054406] [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] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Cholesterol efflux capacity (CEC), an atheroprotective function of high-density lipoprotein, is expected to be a potential biomarker for cardiovascular disease. However, CEC has not been widely introduced for application in clinical laboratories because of the complexity of the conventional CEC assay using cells and radioactive materials. Previously, we developed a novel CEC assay using immobilized liposome-bound gel beads (ILG), which solves these issues. We aimed to confirm the validation and further improve the ILG method for application in the clinical setting. METHODS Cholesterol efflux capacity values by the ILG method assayed for shorter incubation time (4 h) were compared to those assayed for 16 h (our previous ILG method). To investigate a reference material that can correct the variation between ILG manufacturing lots, bovine serum albumin, human gamma-globulins, and globulin complexes were evaluated. CEC values were also estimated in plasmas obtained with different anticoagulants, serum treated with freeze-thaw cycles, and serum mixed with several interference substances. RESULTS The CEC of 4- and 16-h incubation times were well correlated. Globulin complexes may be used as a reference material. Plasma can be used as the specimen. The serum and stored temperature of the specimen did not largely affect CEC. Hemoglobin and chyle did not have an effect on CEC, whereas high-bilirubin serum showed elevated CEC. The effect of bilirubin was nearly canceled by subtracting basal fluorescence intensity. CONCLUSIONS Present ILG method further fulfills some requirements for application in clinical laboratory. Using this reliable simple method, evaluation for clinical significance of CEC is expected.
Collapse
Affiliation(s)
- Yuna Horiuchi
- Analytical Laboratory Chemistry, 92190Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan.,Research Fellow of Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo, Japan
| | - Shao-Jui Lai
- Analytical Laboratory Chemistry, 92190Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Takahiro Kameda
- Analytical Laboratory Chemistry, 92190Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Minoru Tozuka
- Analytical Laboratory Chemistry, 92190Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan.,Life Science Research Center, Nagano Children's Hospital, Toyoshina, Azumino, Japan
| | - Ryunosuke Ohkawa
- Analytical Laboratory Chemistry, 92190Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
43
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
44
|
Zhang L, Xue S, Ren F, Huang S, Zhou R, Wang Y, Zhou C, Li Z. An atherosclerotic plaque-targeted single-chain antibody for MR/NIR-II imaging of atherosclerosis and anti-atherosclerosis therapy. J Nanobiotechnology 2021; 19:296. [PMID: 34583680 PMCID: PMC8479957 DOI: 10.1186/s12951-021-01047-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Background Oxidation-specific epitopes (OSEs) are rich in atherosclerotic plaques. Innate and adaptive immune responses to OSEs play an important role in atherosclerosis. The purpose of this study was to develop novel human single-chain variable fragment (scFv) antibody specific to OSEs to image and inhibit atherosclerosis. Results Here, we screened a novel scFv antibody, named as ASA6, from phage-displayed human scFv library. ASA6 can bind to oxidized LDL (Ox-LDL) and atherosclerotic plaques. Meanwhile, ASA6 can also inhibit the uptake of Ox-LDL into macrophage to reduce macrophage apoptosis. The atherosclerotic lesion area of ApoE−/− mice administrated with ASA6 antibody was significantly reduced. Transcriptome analysis reveals the anti-atherosclerosis effect of ASA6 is related to the regulation of fatty acid metabolism and inhibition of M1 macrophage polarization. Moreover, we conjugated ASA6 antibody to NaNdF4@NaGdF4 nanoparticles for noninvasive imaging of atherosclerotic plaques by magnetic resonance (MR) and near-infrared window II (NIR-II) imaging. Conclusions Together, these data demonstrate the potential of ASA6 antibody in targeted therapy and noninvasive imaging for atherosclerosis. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01047-4.
Collapse
Affiliation(s)
- Liwei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Sheng Xue
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Feng Ren
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Siyang Huang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Ruizhi Zhou
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Yu Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Changyong Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| |
Collapse
|
45
|
Intestinal and Hepatic Uptake of Dietary Peroxidized Lipids and Their Decomposition Products, and Their Subsequent Effects on Apolipoprotein A1 and Paraoxonase1. Antioxidants (Basel) 2021; 10:antiox10081258. [PMID: 34439506 PMCID: PMC8389297 DOI: 10.3390/antiox10081258] [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: 06/15/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 11/23/2022] Open
Abstract
Both pro- and antiatherosclerotic effects have been ascribed to dietary peroxidized lipids. Confusion on the role of peroxidized lipids in atherosclerotic cardiovascular disease is punctuated by a lack of understanding regarding the metabolic fate and potential physiological effects of dietary peroxidized lipids and their decomposition products. This study sought to determine the metabolic fate and physiological ramifications of 13-hydroperoxyoctadecadienoic acid (13-HPODE) and 13-HODE (13-hydroxyoctadecadienoic acid) supplementation in intestinal and hepatic cell lines, as well as any effects resulting from 13-HPODE or 13-HODE degradation products. In the presence of Caco-2 cells, 13-HPODE was rapidly reduced to 13-HODE. Upon entering the cell, 13-HODE appears to undergo decomposition, followed by esterification. Moreover, 13-HPODE undergoes autodecomposition to produce aldehydes such as 9-oxononanoic acid (9-ONA). Results indicate that 9-ONA was oxidized to azelaic acid (AzA) rapidly in cell culture media, but AzA was poorly absorbed by intestinal cells and remained detectable in cell culture media for up to 18 h. An increased apolipoprotein A1 (ApoA1) secretion was observed in Caco-2 cells in the presence of 13-HPODE, 9-ONA, and AzA, whereas such induction was not observed in HepG2 cells. However, 13-HPODE treatments suppressed paraoxonase 1 (PON1) activity, suggesting the induction of ApoA1 secretion by 13-HPODE may not represent functional high-density lipoprotein (HDL) capable of reducing oxidative stress. Alternatively, AzA induced both ApoA1 secretion and PON1 activity while suppressing ApoB secretion in differentiated Caco-2 cells but not in HepG2. These results suggest oxidation of 9-ONA to AzA might be an important phenomenon, resulting in the accumulation of potentially beneficial dietary peroxidized lipid-derived aldehydes.
Collapse
|
46
|
Xu S, Chaudhary O, Rodríguez-Morales P, Sun X, Chen D, Zappasodi R, Xu Z, Pinto AFM, Williams A, Schulze I, Farsakoglu Y, Varanasi SK, Low JS, Tang W, Wang H, McDonald B, Tripple V, Downes M, Evans RM, Abumrad NA, Merghoub T, Wolchok JD, Shokhirev MN, Ho PC, Witztum JL, Emu B, Cui G, Kaech SM. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8 + T cells in tumors. Immunity 2021; 54:1561-1577.e7. [PMID: 34102100 PMCID: PMC9273026 DOI: 10.1016/j.immuni.2021.05.003] [Citation(s) in RCA: 331] [Impact Index Per Article: 110.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/03/2021] [Accepted: 05/04/2021] [Indexed: 01/21/2023]
Abstract
A common metabolic alteration in the tumor microenvironment (TME) is lipid accumulation, a feature associated with immune dysfunction. Here, we examined how CD8+ tumor infiltrating lymphocytes (TILs) respond to lipids within the TME. We found elevated concentrations of several classes of lipids in the TME and accumulation of these in CD8+ TILs. Lipid accumulation was associated with increased expression of CD36, a scavenger receptor for oxidized lipids, on CD8+ TILs, which also correlated with progressive T cell dysfunction. Cd36-/- T cells retained effector functions in the TME, as compared to WT counterparts. Mechanistically, CD36 promoted uptake of oxidized low-density lipoproteins (OxLDL) into T cells, and this induced lipid peroxidation and downstream activation of p38 kinase. Inhibition of p38 restored effector T cell functions in vitro, and resolution of lipid peroxidation by overexpression of glutathione peroxidase 4 restored functionalities in CD8+ TILs in vivo. Thus, an oxidized lipid-CD36 axis promotes intratumoral CD8+ T cell dysfunction and serves as a therapeutic avenue for immunotherapies.
Collapse
Affiliation(s)
- Shihao Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Omkar Chaudhary
- Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06510, USA
| | - Patricia Rodríguez-Morales
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Xiaoli Sun
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dan Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ziyan Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Antonio F M Pinto
- Mass Spectrometry Core for Proteomics and Metabolomics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - April Williams
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Isabell Schulze
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yagmur Farsakoglu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Siva Karthik Varanasi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jun Siong Low
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA; Fondazione per l'istituto di ricerca in biomedicina, Bellinzona, Switzerland
| | - Wenxi Tang
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Haiping Wang
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Bryan McDonald
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Victoria Tripple
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nada A Abumrad
- Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maxim N Shokhirev
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ping-Chih Ho
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research at University of Lausanne, Lausanne, Switzerland
| | - Joseph L Witztum
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brinda Emu
- Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06510, USA
| | - Guoliang Cui
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
47
|
Tracz J, Luczak M. Applying Proteomics and Integrative "Omics" Strategies to Decipher the Chronic Kidney Disease-Related Atherosclerosis. Int J Mol Sci 2021; 22:7492. [PMID: 34299112 PMCID: PMC8305100 DOI: 10.3390/ijms22147492] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Patients with chronic kidney disease (CKD) are at increased risk of atherosclerosis and premature mortality, mainly due to cardiovascular events. However, well-known risk factors, which promote "classical" atherosclerosis are alone insufficient to explain the high prevalence of atherosclerosis-related to CKD (CKD-A). The complexity of the molecular mechanisms underlying the acceleration of CKD-A is still to be defied. To obtain a holistic picture of these changes, comprehensive proteomic approaches have been developed including global protein profiling followed by functional bioinformatics analyses of dysregulated pathways. Furthermore, proteomics surveys in combination with other "omics" techniques, i.e., transcriptomics and metabolomics as well as physiological assays provide a solid ground for interpretation of observed phenomena in the context of disease pathology. This review discusses the comprehensive application of various "omics" approaches, with emphasis on proteomics, to tackle the molecular mechanisms underlying CKD-A progression. We summarize here the recent findings derived from global proteomic approaches and underline the potential of utilizing integrative systems biology, to gain a deeper insight into the pathogenesis of CKD-A and other disorders.
Collapse
Affiliation(s)
| | - Magdalena Luczak
- European Centre for Bioinformatics and Genomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland;
| |
Collapse
|
48
|
Pluijmert NJ, de Jong RCM, de Vries MR, Pettersson K, Atsma DE, Jukema JW, Quax PHA. Phosphorylcholine antibodies restrict infarct size and left ventricular remodelling by attenuating the unreperfused post-ischaemic inflammatory response. J Cell Mol Med 2021; 25:7772-7782. [PMID: 34190404 PMCID: PMC8358891 DOI: 10.1111/jcmm.16662] [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: 01/29/2021] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 12/19/2022] Open
Abstract
Phosphorylcholine is a pro‐inflammatory epitope exposed on apoptotic cells, and phosphorylcholine monoclonal immunoglobulin (Ig)G antibodies (PC‐mAb) have anti‐inflammatory properties. In this study, we hypothesize that PC‐mAb treatment reduces adverse cardiac remodelling and infarct size (IS) following unreperfused transmural myocardial infarction (MI). Unreperfused MI was induced by permanent ligation of the left anterior descending (LAD) coronary artery in hypercholesterolaemic APOE*3‐Leiden mice. Three weeks following MI, cardiac magnetic resonance (CMR) imaging showed a reduced LV end‐diastolic volume (EDV) by 21% and IS by 31% upon PC‐mAb treatment as compared to the vehicle control group. In addition, the LV fibrous content was decreased by 27% and LV wall thickness was better preserved by 47% as determined by histological analysis. Two days following MI, CCL2 concentrations, assessed by use of ELISA, were decreased by 81% and circulating monocytes by 64% as assessed by use of FACS analysis. Additionally, local leucocyte infiltration determined by immunohistological analysis showed a 62% decrease after three weeks. In conclusion, the local and systemic inflammatory responses are limited by PC‐mAb treatment resulting in restricted adverse cardiac remodelling and IS following unreperfused MI. This indicates that PC‐mAb holds promise as a therapeutic agent following MI limiting adverse cardiac remodelling.
Collapse
Affiliation(s)
- Niek J Pluijmert
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob C M de Jong
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Margreet R de Vries
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
49
|
Shah S, Shiekh Y, Lawrence JA, Ezekwueme F, Alam M, Kunwar S, Gordon DK. A Systematic Review of Effects of Vitamin E on the Cardiovascular System. Cureus 2021; 13:e15616. [PMID: 34277234 PMCID: PMC8275884 DOI: 10.7759/cureus.15616] [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: 05/28/2021] [Accepted: 06/12/2021] [Indexed: 11/05/2022] Open
Abstract
Vitamin E is a fat-soluble vitamin and an antioxidant that prevents the peroxidation of lipid in vitro. The antioxidant role of vitamin E in preventing adverse cardiovascular outcomes is controversial as some studies support it, while others reject it. Therefore, this review aims to determine whether there is an association between vitamin E and cardiovascular diseases (CVDs). An electronic search was done to find out relevant articles. Papers were shortlisted after the initial title and abstract screen. A full-text study was done, and inclusion and exclusion criteria were applied before the quality assessment of each paper was done. Only high-quality papers were selected for analysis. Full-text articles of the last ten years were included, while non-English articles, gray literature, and animal studies were excluded. The majority of the papers, including 75% of the total population in this review, suggested no role of vitamin E in preventing CVD and CVD mortality. Some studies also suggested that a high level of vitamin E can be associated with adverse cardiovascular outcomes. Thus, one should be prudent about taking vitamin E supplementation for cardiovascular risk prevention.
Collapse
Affiliation(s)
- Sunil Shah
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Internal Medicine, Mulee Regional Hospital/Ministry of Health, Muli, MDV
| | - Yasir Shiekh
- Emergency Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Emergency Medicine, Hamad General Hospital, Doha, QAT
| | - Jannel A Lawrence
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Francis Ezekwueme
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mohammad Alam
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Saru Kunwar
- Pediatrics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Neonatal Intermediate Care Unit, Kanti Childrens Hospital, Kathmandu, NPL
| | - Domonick K Gordon
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Internal Medicine, Scarborough General Hospital, Scarborough, TTO
| |
Collapse
|
50
|
Singh H, Rai V, Agrawal DK. Discerning the promising binding sites of S100/calgranulins and their therapeutic potential in atherosclerosis. Expert Opin Ther Pat 2021; 31:1045-1057. [PMID: 34056993 DOI: 10.1080/13543776.2021.1937122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Atherosclerosis is a chronic inflammatory disease in which the members of S100 family proteins (calgranulins) bind with their receptors, particularly receptor for advanced glycation end products (RAGE) and toll-like receptor-4 (TLR-4) and play a key role in the pathogenesis and progression of disease. Thus, these proteins could be considered as potential biomarkers and therapeutic targets in the treatment of atherosclerotic inflammation. AREAS COVERED This review summarizes the pathology of S100A8, S100A9, and S100A12 in the development of atherosclerosis and reveals key structural features of these proteins which are potentially critical in their pathological effects. This article focuses on the translational significance of antagonizing these proteins by using small molecules in patent literature, clinical and preclinical studies and also discusses future approaches that could be employed to block these proteins in the treatment of atherosclerosis. EXPERT OPINION Based on the critical role of S100/calgranulins in the regulation of atherosclerosis, these proteins are potential targets to develop better therapeutic options in the treatment of inflammatory diseases. However, further research is still needed to clarify their exact molecular mechanism by analyzing their detailed structural features that can expedite future research to develop novel therapeutics against these proteins to treat atherosclerotic inflammation.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|