1
|
Shi S, Zheng Z, Chen W, Song Y, Dou K. Low density lipoprotein cholesterol but not statins is the direct cause of intracerebral hemorrhage. Eur J Pharmacol 2025; 998:177443. [PMID: 40023359 DOI: 10.1016/j.ejphar.2025.177443] [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: 11/12/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVE To investigate the direct and indirect relationships between statin use, low-density lipoprotein cholesterol (LDL-C) levels, and intracerebral hemorrhage (ICH), providing new insights into this complex scientific question. METHODS In this cohort study, UK Biobank data from 2006 to 2010 were used to construct Structural Equation Models of statin use, LDL-C, and ICH, including 414,253 participants with LDL-C data. Published Genome-Wide Association Studies data were used for drug-target Mendelian Randomization analysis. RESULTS The study included 414,253 participants, comprising 225,454 women (54.4%) with a mean age of 56.07 (8.11) years. During a median follow-up of 14.01 years, 2973 patients experienced ICH. Structural Equation Modelling showed the indirect effect (path a∗b) of statin on ICH was 0.003 (P < 0.001), the direct effect (path c') was -0.001 (P = 0.568), the total effect (path c) was 0.002 (P = 0.391), and the mediation proportion of LDL-C (a∗b/c) was 150.0%. Mendelian Randomization showed a negative association between LDL-C levels and ICH (β: -0.663, SE: 0.229, P = 0.004), with no causal relationship between statin use and ICH (β: -1.454, SE: 3.133, P = 0.643). Drug-targeted Mendelian Randomization revealed LDL-C levels, predicted by variants in or near HMGCR, PCSK9, CETP, ABCG8/5, and LAP, were negatively associated with ICH risk. CONCLUSIONS This study confirmed that statins increase the risk of ICH primarily through their LDL-C-lowering effects, rather than the direct effects of the statins themselves. LDL-C is negatively associated with ICH, an association not confined to the effects of the HMGCR loci. This advance provides evidence for the controversy between statin use, LDL-C levels, and ICH risk.
Collapse
Affiliation(s)
- Shanshan Shi
- Cardiometabolic Medicine Center, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Zhihao Zheng
- Cardiometabolic Medicine Center, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Weihua Chen
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China
| | - Yanjun Song
- Cardiometabolic Medicine Center, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Kefei Dou
- Cardiometabolic Medicine Center, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, 167, Beilishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
2
|
Srivastava SP, Kopasz-Gemmen O, Thurman A, Rajendran BK, Selvam MM, Kumar S, Srivastava R, Suresh MX, Kumari R, Goodwin JE, Inoki K. The molecular determinants regulating redox signaling in diabetic endothelial cells. Front Pharmacol 2025; 16:1563047. [PMID: 40290438 PMCID: PMC12023289 DOI: 10.3389/fphar.2025.1563047] [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: 01/18/2025] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
Oxidation and reduction are vital for keeping life through several prime mechanisms, including respiration, metabolism, and other energy supplies. Mitochondria are considered the cell's powerhouse and use nutrients to produce redox potential and generate ATP and H2O through the process of oxidative phosphorylation by operating electron transfer and proton pumping. Simultaneously, mitochondria also produce oxygen free radicals, called superoxide (O2 -), non-enzymatically, which interacts with other moieties and generate reactive oxygen species (ROS), such as hydrogen peroxide (H2O2), peroxynitrite (ONOO-), and hydroxyl radical (OH-). These reactive oxygen species modify nucleic acids, proteins, and carbohydrates and ultimately cause damage to organs. The nutrient-sensing kinases, such as AMPK and mTOR, function as a key regulator of cellular ROS levels, as loss of AMPK or aberrant activation of mTOR signaling causes ROS production and compromises the cell's oxidant status, resulting in various cellular injuries. The increased ROS not only directly damages DNA, proteins, and lipids but also alters cellular signaling pathways, such as the activation of MAPK or PI3K, the accumulation of HIF-1α in the nucleus, and NFkB-mediated transcription of pro-inflammatory cytokines. These factors cause mesenchymal activation in renal endothelial cells. Here, we discuss the biology of redox signaling that underlies the pathophysiology of diabetic renal endothelial cells.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, United States
| | | | - Aaron Thurman
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - Barani Kumar Rajendran
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - M. Masilamani Selvam
- Department of Pharmaceutical Technology, Paavai Engineering College, Namakkal, Tamil Nadu, India
| | - Sandeep Kumar
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Rohit Srivastava
- Laboratory of Medical Transcriptomics, Department of Endocrinology, Nephrology Services, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - M. Xavier Suresh
- School of Advanced Sciences and Languages, VIT Bhopal University, Sehore, Madhya Pradesh, India
| | - Reena Kumari
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, United States
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
3
|
Xu X, Hu B, Qu X. Effects of propolis intake on endurance exercise and molecular signaling related to inflammation and oxidative stress. Front Nutr 2025; 12:1539701. [PMID: 40078415 PMCID: PMC11896820 DOI: 10.3389/fnut.2025.1539701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Honey bees extract sticky material from the exudates of different plants which transform afterwards to propolis. Propolis from several global locations has been shown to contain a wide variety of polyphenolic chemicals. Recent studies have revealed that propolis possesses antioxidant, anti-inflammatory, and immunomodulatory abilities. In laboratory animal studies, it has been demonstrated that propolis can enhance the functioning of the antioxidant defense system and decrease the activity of nuclear factor-kappa B. As a result, they can effectively alleviate the damage caused by exercise. One of the main flavonoids found in propolis, quercetin, has been demonstrated to enhance muscle mitochondrial biogenesis and exercise capacity. Propolis may aid athletes in preventing oxidative and inflammatory damage to their muscles during exercise and enhance their athletic performance. The goal of the current review was to evaluate how propolis consumption affected the molecular signaling associated with antioxidant/oxidant state, pro/anti-inflammatory cytokines, and anaerobic/aerobic endurance.
Collapse
Affiliation(s)
- Xiaoying Xu
- Sports College, Yantai University, Yantai, Shandong, China
| | - Bing Hu
- Sports Industry Development Service Department, Yantai Sports Industry Development Service Center, Yantai, Shandong, China
| | - Xiaorong Qu
- Training Section, Yantai Shooting and Archery Sports Center, Yantai, Shandong, China
| |
Collapse
|
4
|
Shao H, Xu C, Zhang C, Li L, Wu P, Chen Z, Guan R. Genetic Insights Into Lipid Traits and Lipid-Modifying Drug Targets in Pregnancy Complications: A Two-Sample Mendelian Randomization Study. Int J Womens Health 2025; 17:221-234. [PMID: 39911358 PMCID: PMC11794394 DOI: 10.2147/ijwh.s496268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Background Dyslipidemia is linked to pregnancy complications, but its causal role remains uncertain. This two-sample Mendelian Randomization (MR) study investigated the causal relationship between lipid traits and pregnancy complications and evaluated the impact of lipid-modifying drug targets. Methods Genetic instruments for lipid traits and targets for lipid-modifying drugs were obtained from the Global Lipids Genetics Consortium. Three pregnancy complications' summary statistics came from the FinnGen R9 database. Significant drug targets underwent further analysis using Expression Quantitative Trait Loci data, and mediation analysis identified potential mediators. Results Increased high-density lipoprotein cholesterol (HDL-C) reduced the incidence of preeclampsia (OR: 0.755, 95% CI: 0.639-0.891, p=0.001, FDR=0.012) and gestational diabetes mellitus (GDM) (OR: 0.835, 95% CI: 0.741-0.942, p=0.003, FDR=0.018). Genetic proxies for cholesteryl ester transfer protein (CETP) inhibition correlated with a decreased risk of preeclampsia (OR: 0.863, 95% CI: 0.786-0.947, p=0.002, FDR=0.027), while genetic inhibition of HMG-CoA reductase (HMGCR) increased preeclampsia risk (OR: 1.700, 95% CI: 1.189-2.431, p=0.004, FDR=0.036). Genetically mimicking the enhancement of lipoprotein lipase (LPL) related to a reduced risk of GDM (OR: 0.681, 95% CI: 0.560-0.829, p=1.29×10-4, FDR=0.004). Higher LPL expression in subcutaneous adipose tissue also reduced GDM risk (OR: 0.642, 95% CI: 0.454-0.909, p=0.013). Waist circumference (4.2%) and waist-to-hip ratio adjusted by BMI (5.7%) partially mediated LPL's effect on GDM risk. Conclusion Elevated HDL-C levels help prevent preeclampsia and GDM. CETP and LPL could be therapeutic targets for preeclampsia and GDM, respectively. However, caution is advised with HMGCR-targeting drugs, as they may increase the preeclampsia risk.
Collapse
Affiliation(s)
- Huijing Shao
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, People’s Republic of China
| | - Chang Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Caihong Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Lirong Li
- Department of Traditional Chinese Gynecology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Pengfei Wu
- Department of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People’s Republic of China
| | - Zixi Chen
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People’s Republic of China
| | - Rui Guan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
5
|
Clim A, Maranduca MA, Filip N, Tănase DM, Floria M, Pinzariu AC, Popa IP, Nemteanu R, Cozma TC, Faur FI, Serban DN, Scripcariu DV, Serban IL. The Influence of Atorvastatin Treatment on Homocysteine Metabolism and Oxidative Stress in an Experimental Model of Diabetic Rats. Life (Basel) 2024; 14:1414. [PMID: 39598213 PMCID: PMC11595749 DOI: 10.3390/life14111414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVE In our experimental study, we evaluated the influence of treatment with atorvastatin on the antioxidant activity of intracellular and extracellular systems factors, homocysteine levels (Hcy), and lipid profiles in obese and diabetic rats. METHOD Twenty-one male Wistar rats, aged 6 months, 450-550 g, were allocated into three groups. From the beginning of the study, the first group (G-I, control) received only standard food, while the second and third groups (G II-obese, G III-diabetic) were administered a high-fat diet (HFD) with 2% cholesterol. After 2 weeks of accommodation, the specimens in G-III were injected intraperitoneal (i.p.) streptozotocin (35 mg of body weight, pH 4.5), intervention followed by the onset of type 2 diabetes mellitus. Following confirmation of diabetes onset, the specimens in G III were administered concomitantly with the HFD a daily gavage of atorvastatin 20 mg of body weight/day for 20 days. We measured, at the beginning and the end of the study, the Hcy levels, lipid profile, vitamin B12, B6, folic acid, and various parameters of oxidative stress (OS)-total antioxidant status (TAS), glutathione peroxidase (GPX) and superoxide dismutase (SOD). RESULTS After treatment with atorvastatin, the lipid profile in G III significantly improved compared to the other two groups, but enzymatic markers of oxidative stress did not closely parallel this trend. However, after the treatment of statin, we observed an important reduction in Hcy values. CONCLUSION Our results demonstrate that treatment with atorvastatin can be used not only for its lipid-lowering properties and antioxidant effects but also to reduce Hcy concentration in this experimental model of diabetic rats. Moreover, atorvastatin therapy improves lipid profiles, reduces inflammation, suppresses oxidation, and decreases Hcy levels, potentially preventing major adverse cardiovascular events.
Collapse
Affiliation(s)
- Andreea Clim
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (A.C.); (M.A.M.); (A.C.P.); (I.P.P.); (T.C.C.); (D.N.S.); (I.L.S.)
| | - Minela Aida Maranduca
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (A.C.); (M.A.M.); (A.C.P.); (I.P.P.); (T.C.C.); (D.N.S.); (I.L.S.)
- Internal Medicine Clinic, “St Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Nina Filip
- Department of Morpho-Functional Sciences II, Discipline of Biochemistry, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Daniela Maria Tănase
- Internal Medicine Clinic, “St Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Mariana Floria
- Internal Medicine Clinic, “St Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alin Constantin Pinzariu
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (A.C.); (M.A.M.); (A.C.P.); (I.P.P.); (T.C.C.); (D.N.S.); (I.L.S.)
| | - Irene Paula Popa
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (A.C.); (M.A.M.); (A.C.P.); (I.P.P.); (T.C.C.); (D.N.S.); (I.L.S.)
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Roxana Nemteanu
- Medical I Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania;
- Institute of Gastroenterology and Hepatology, “St. Spiridon” University Hospital, 700111 Iași, Romania
| | - Tudor Cristian Cozma
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (A.C.); (M.A.M.); (A.C.P.); (I.P.P.); (T.C.C.); (D.N.S.); (I.L.S.)
| | - Flaviu Ionut Faur
- 2nd Surgery Clinic, Timisoara Emergency County Hospital, 300723 Timisoara, Romania;
- X Department of General Surgery, “Victor Babes University of Medicine and Pharmacy”, 300041 Timisoara, Romania
- Multidisciplinary Doctoral School “Vasile Goldis”, Western University of Arad, 310025 Arad, Romania
| | - Dragomir Nicolae Serban
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (A.C.); (M.A.M.); (A.C.P.); (I.P.P.); (T.C.C.); (D.N.S.); (I.L.S.)
| | - Dragoș Viorel Scripcariu
- Surgery Department, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iași, Romania
- 1st Surgical Oncology Unit, Regional Institute of Oncology, 2–4 General Henri Mathias Berthelot Street, 700483 Iași, Romania
| | - Ionela Lacramioara Serban
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (A.C.); (M.A.M.); (A.C.P.); (I.P.P.); (T.C.C.); (D.N.S.); (I.L.S.)
| |
Collapse
|
6
|
Jiang R, Lou L, Shi W, Chen Y, Fu Z, Liu S, Sok T, Li Z, Zhang X, Yang J. Statins in Mitigating Anticancer Treatment-Related Cardiovascular Disease. Int J Mol Sci 2024; 25:10177. [PMID: 39337662 PMCID: PMC11432657 DOI: 10.3390/ijms251810177] [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: 08/20/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Certain anticancer therapies inevitably increase the risk of cardiovascular events, now the second leading cause of death among cancer patients. This underscores the critical need for developing effective drugs or regimens for cardiovascular protection. Statins possess properties such as antioxidative stress, anti-inflammatory effects, antifibrotic activity, endothelial protection, and immune modulation. These pathological processes are central to the cardiotoxicity associated with anticancer treatment. There is prospective clinical evidence confirming the protective role of statins in chemotherapy-induced cardiotoxicity. Numerous preclinical studies have demonstrated that statins can ameliorate heart and endothelial damage caused by radiotherapy, although clinical studies are scarce. In the animal models of trastuzumab-induced cardiomyopathy, statins provide protection through anti-inflammatory, antioxidant, and antifibrotic mechanisms. In animal and cell models, statins can mitigate inflammation, endothelial damage, and cardiac injury induced by immune checkpoint inhibitors. Chimeric antigen receptor (CAR)-T cell therapy-induced cardiotoxicity and immune effector cell-associated neurotoxicity syndrome are associated with uncontrolled inflammation and immune activation. Due to their anti-inflammatory and immunomodulatory effects, statins have been used to manage CAR-T cell therapy-induced immune effector cell-associated neurotoxicity syndrome in a clinical trial. However, direct evidence proving that statins can mitigate CAR-T cell therapy-induced cardiotoxicity is still lacking. This review summarizes the possible mechanisms of anticancer therapy-induced cardiotoxicity and the potential mechanisms by which statins may reduce related cardiac damage. We also discuss the current status of research on the protective effect of statins in anticancer treatment-related cardiovascular disease and provide directions for future research. Additionally, we propose further studies on using statins for the prevention of cardiovascular disease in anticancer treatment.
Collapse
Affiliation(s)
- Rong Jiang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lian Lou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Wen Shi
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuxiao Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhaoming Fu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shuo Liu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Thida Sok
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhihang Li
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jian Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
7
|
Li JN, Wang MY, Tan YR, Wang LL. Multidirectional Intervention of Chinese Herbal Medicine in the Prevention and Treatment of Atherosclerosis: From Endothelial Protection to Immunomodulation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:925-947. [PMID: 38798151 DOI: 10.1142/s0192415x24500381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Atherosclerosis is a significant risk factor for developing cardiovascular disease and a leading cause of death worldwide. The occurrence of atherosclerosis is closely related to factors such as endothelial injury, lipid deposition, immunity, and inflammation. Conventional statins, currently used in atherosclerosis treatment, have numerous adverse side effects that limit their clinical utility, prompting the urgent need to identify safer and more effective therapeutic alternatives. Growing evidence indicates the significant potential of Chinese herbs in atherosclerosis treatment. Herbal monomer components, such as natural flavonoid compounds extracted from herbs like Coptis chinensis and Panax notoginseng, have been utilized for their lipid-lowering and inflammation-inhibiting effects in atherosclerosis treatment. These herbs can be used as single components in treating diseases and with other Chinese medicines to form herbal combinations. This approach targets the disease mechanism in multiple ways, enhancing the therapeutic effects. Thus, this review examines the roles of Chinese herbal medicine monomers and Chinese herbal compounds in inhibiting atherosclerosis, including regulating lipids, improving endothelial function, reducing oxidative stress, regulating inflammation and the immune response, and apoptosis. By highlighting these roles, our study offers new perspectives on atherosclerosis treatment with Chinese herbs and is anticipated to contribute to advancements in related research fields.
Collapse
Affiliation(s)
- Jia-Ni Li
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| | - Meng-Yu Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| | - Yu-Rong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| | - Li-Li Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Xiangya Road 88, Changsha 410078, Hunan, P. R. China
| |
Collapse
|
8
|
Yamaji T, Yusoff FM, Kishimoto S, Kajikawa M, Yoshimura K, Nakano Y, Goto C, Harada T, Mizobuchi A, Tanigawa S, Maruhashi T, Higashi Y. Association of cumulative low-density lipoprotein cholesterol exposure with vascular function. J Clin Lipidol 2024; 18:e238-e250. [PMID: 38185588 DOI: 10.1016/j.jacl.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND The relationship between cumulative low-density lipoprotein cholesterol (LDL-C) exposure and progression of atherosclerosis remains uncertain. OBJECTIVE The aim of this study was to determine the relationship between cumulative LDL-C level and flow-mediated vasodilation (FMD), nitroglycerine-induced vasodilation (NID) and the presence of plaque in the common carotid artery (CCA). METHODS This was a cross-sectional study. We measured FMD in 8208 subjects, NID in 1822 subjects, and CCA plaque in 591 subjects who were not taking lipid-lowering drugs. The subjects were divided into four groups based on cumulative LDL-C exposure: <4000 mg·year/dL, 4000-4999 mg·year/dL, 5000-5999 mg·year/dL, and ≥6000 mg·year/dL. RESULTS The odds ratio of the lower quartile of FMD in the cholesterol-year-score <4000 mg·year/dL group was significantly higher than the odds ratios in the other groups. The odds ratio of the lower quartile of NID in the <4000 mg·year/dL group was significantly higher than the odds ratios in the 5000-5999 mg·year/dL and ≥6000 mg·year/dL groups. The odds ratio of the prevalence of CCA plaque in the <4000 mg·year/dL group was significantly higher than that in the ≥6000 mg·year/dL group. CONCLUSIONS Endothelial dysfunction occurred from cumulative LDL-C exposure of 4000 mg·year/dL, vascular smooth muscle dysfunction occurred from cumulative LDL-C exposure of 5000 mg·year/dL, and prevalence of CCA plaque occurred from cumulative LDL-C exposure of 6000 mg·year/dL. CLINICAL TRIAL REGISTRY INFORMATION: http://www.umin.ac.jp (UMIN000012950, UMIN000012951, and UMIN000012952, UMIN000003409).
Collapse
Affiliation(s)
- Takayuki Yamaji
- Center for Radiation Disaster Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji and Higashi); Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (Drs Kajikawa, Yoshimura, and Higashi)
| | - Kenichi Yoshimura
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (Drs Kajikawa, Yoshimura, and Higashi)
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan (Dr Nakano)
| | - Chikara Goto
- Hiroshima International University, Hiroshima, Japan (Dr Goto)
| | - Takahiro Harada
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Aya Mizobuchi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Shunsuke Tanigawa
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Tatsuya Maruhashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi)
| | - Yukihito Higashi
- Center for Radiation Disaster Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji and Higashi); Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan (Drs Yamaji, Yusoff, Kishimoto, Harada, Mizobuchi, Tanigawa, Maruhashi, and Higashi); Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan (Drs Kajikawa, Yoshimura, and Higashi).
| |
Collapse
|
9
|
Khatiwada N, Hong Z. Potential Benefits and Risks Associated with the Use of Statins. Pharmaceutics 2024; 16:214. [PMID: 38399268 PMCID: PMC10892755 DOI: 10.3390/pharmaceutics16020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
HMG-CoA reductase inhibitors, commonly known as statins, are the primary treatment choice for cardiovascular diseases, which stand as the leading global cause of mortality. Statins also offer various pleiotropic effects, including improved endothelial function, anti-inflammatory properties, reduced oxidative stress, anti-thrombotic effects, and the stabilization of atherosclerotic plaques. However, the usage of statins can be accompanied by a range of adverse effects, such as the development of type 2 diabetes mellitus, muscular symptoms, liver toxicity, kidney diseases, cataracts, hemorrhagic strokes, and psychiatric complications. These issues are referred to as statin-associated symptoms (SAS) and are relatively infrequent in clinical trials, making it challenging to attribute them to statin use definitively. Therefore, these symptoms can lead to significant problems, necessitating dose adjustments or discontinuation of statin therapy. This review aims to provide a comprehensive overview of the mechanism of action, potential advantages, and associated risks of statin utilization in clinical settings.
Collapse
Affiliation(s)
| | - Zhongkui Hong
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX 79409, USA;
| |
Collapse
|
10
|
Zhang J, Zhang S, Xu S, Zhu Z, Li J, Wang Z, Wada Y, Gatt A, Liu J. Oxidative Stress Induces E-Selectin Expression through Repression of Endothelial Transcription Factor ERG. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1835-1843. [PMID: 37930129 PMCID: PMC10694031 DOI: 10.4049/jimmunol.2300043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
Oxidative stress induces a prothrombotic state through enhancement of adhesion properties of the endothelium. E-selectin, an endothelial cell adhesion molecule, becomes a therapeutic target for venous thrombosis, whereas the regulatory mechanisms of its expression have not been fully understood. In the present study, we report that H2O2 treatment increases expression of E-selectin but decreases expression of the endothelial transcription factor ETS-related gene (ERG) in HUVECs in a dose- and time-dependent manner. In BALB/c mice treated with hypochlorous acid, E-selectin expression is increased and ERG expression is decreased in endothelial cells of the brain and lung. RNA interference of ERG upregulates E-selectin expression, whereas transfection of ERG-expressing plasmid downregulates E-selectin expression in HUVECs. Knockdown or overexpression of ERG comprises H2O2-induced E-selectin expression in HUVECs. Deletion of the Erg gene in mice results in embryonic lethality at embryonic days 10.5-12.5, and E-selectin expression is increased in the Erg-/- embryos. No chromatin loop was found on the E-selectin gene or its promoter region by capture high-throughput chromosome conformation capture. Chromatin immunoprecipitation and luciferase reporter assay determined that the -127 ERG binding motif mediates ERG-repressed E-selectin promoter activity. In addition, ERG decreases H2O2-induced monocyte adhesion. Together, ERG represses the E-selectin gene transcription and inhibits oxidative stress-induced endothelial cell adhesion.
Collapse
Affiliation(s)
- Jinjin Zhang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Shuo Zhang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Shanhu Xu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Zhiying Zhu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Jiang Li
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Zengjin Wang
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Youichiro Wada
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Alex Gatt
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Tal-Qroqq, Msida, Malta
- Hematology Laboratory, Department of Pathology, Mater Dei Hospital, Msida, Malta
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
11
|
Pogran E, Haller PM, Wegberger C, Tscharre M, Vujasin I, Kaufmann CC, Dick P, Jäger B, Wojta J, Huber K. The LIPL study: Postprandial lipid profile, inflammation, and platelet activity in patients with chronic coronary syndrome. ATHEROSCLEROSIS PLUS 2023; 54:14-21. [PMID: 37811126 PMCID: PMC10550804 DOI: 10.1016/j.athplu.2023.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/30/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023]
Abstract
Background and aims It is suggested that the changes in atherosclerosis happen mainly under the influence of non-fasting lipids. To date, the studies in the postprandial state were primarily performed on healthy subjects. This exploratory, cross-sectional study investigates the change in lipid profile, inflammation, and platelet activation in patients with different cardiovascular risk profiles in the postprandial state. Methods The studied population consists of 66 patients with different cardiovascular risks: patients with a history of the chronic coronary syndrome (CCS) and diabetes mellitus type 2 (DM2) (n = 20), CCS without DM2 (n = 25), and a healthy control group (n = 21). Lipid variables and markers of platelet function and inflammation were assessed during the fasting state and three and 5 h after a standardized fat meal using a standardized oral fat tolerance test (OFTT), a milkshake with 90 g of fat. Results Patients with CCS and DM2 were significantly older and had the highest BMI. All patients with CCS were on acetylsalicylic acid, and 95% of CCS patients were on high-dose statins. The absolute leukocyte and neutrophile count increased significantly in the control group during the OFTT in comparison to CCS subjects. There was a significant decrease of HDL and increase of triglycerides during the OFTT, however with no difference between groups. There was no difference in the change of platelet activity between all groups. Conclusion This study showed that OFTT leads to an increased postprandial inflammation response in healthy group compared to CCS ± DM2 while there was no change in lipid profile and platelet activity.
Collapse
Affiliation(s)
- Edita Pogran
- 3rd Medical Department for Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
- Sigmund Freud University, Medical Faculty, Vienna, Austria
| | - Paul M. Haller
- 3rd Medical Department for Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
- Ludwig Boltzmann Institute for Interventional Cardiology and Rhythmology, Austria
| | - Claudia Wegberger
- 3rd Medical Department for Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
| | - Maximilian Tscharre
- 3rd Medical Department for Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
| | - Irena Vujasin
- 3rd Medical Department for Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
| | - Christoph C. Kaufmann
- 3rd Medical Department for Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
- Sigmund Freud University, Medical Faculty, Vienna, Austria
| | - Petra Dick
- Klinik Ottakring, Department of Surgery with a Focus on Vascular Surgery- Vascular and Endovascular Surgery, Vienna, Austria
| | - Bernhard Jäger
- 3rd Medical Department for Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
- Ludwig Boltzmann Institute for Interventional Cardiology and Rhythmology, Austria
- Sigmund Freud University, Medical Faculty, Vienna, Austria
| | - Johann Wojta
- Ludwig Boltzmann Institute for Interventional Cardiology and Rhythmology, Austria
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Sigmund Freud University, Medical Faculty, Vienna, Austria
| | - Kurt Huber
- 3rd Medical Department for Cardiology and Intensive Care Medicine, Klinik Ottakring (Wilhelminenhospital), Vienna, Austria
- Ludwig Boltzmann Institute for Interventional Cardiology and Rhythmology, Austria
- Sigmund Freud University, Medical Faculty, Vienna, Austria
| |
Collapse
|
12
|
Yehia Abdelzaher W, A Abdel-Gaber S, Atef Fawzy M, Hamid Sayed Abo Bakr Ali A, Ezzat Attya M, Geddawy A. Atorvastatin protects against cyclophosphamide-induced thyroid injury in rats via modulation of JNK/ ERK/ p38 MAPK signaling pathway. Int Immunopharmacol 2023; 124:111061. [PMID: 37844467 DOI: 10.1016/j.intimp.2023.111061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Cancer chemotherapy is associated with various tissue toxicities that limit its use. Cyclophosphamide (CYC) is one of the most commonly used antineoplastic and immunosuppressive agent. Thyroid dysfunction is a critical side effect of anticancer drugs. Atorvastatin (ATV) is antihyperlipedemic drug with different tissue protective activities. The aim of this study was to determine the potential protective effect of ATV against CYC-induced thyroid injury in rats. METHODS ATV was administered in the presence and absence of CYC. Thirty-two adult Wistar rats were randomly divided into four groups: control group, ATV group (20 mg/kg/day, p.o. for 14 day), CYC group (200 mg/kg, i.p. on day 9) and ATV/CYC group. Triiodothyronine (T3), thyroxine (T4), reduced glutathione (GSH), malondialdehyde (MDA), total nitrite/nitrate (NOx), p38 mitogen-activated protein kinase (P38MAPK), extracellular signal-regulated kinase (ERK) and c-Jun N-terminal Kinase (JNK) were measured. In addition, thyroid histopathology and caspase 3 immunohistochemistry were performed. RESULTS CYC significantly increased thyroid MDA, NOx, P38MAPK, ERK and JNK with decrease in GSH, T3 and T4 levels. Histopathological features of thyroid lesions and increased caspase 3 immune expression were appeared. ATV significantly normalized distributed oxidative, inflammatory and apoptotic indicators, resulting in an improvement of histopathological features and reduction of caspase 3 immunoexpression. CONCLUSION These findings suggest that ATV protects against CYC-induced thyroid injury by regulating the JNK/ERK/p38-MAPK signaling pathway.
Collapse
Affiliation(s)
| | - Seham A Abdel-Gaber
- Department of Pharmacology, Faculty of Medicine Minia University, Minia 61111, Egypt.
| | - Michael Atef Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | | | - Mina Ezzat Attya
- Department of Pathology, Faculty of Medicine, Minia University, Minia 61111, Egypt.
| | - Ayman Geddawy
- Department of Pharmacology, Faculty of Medicine Minia University, Minia 61111, Egypt; Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia.
| |
Collapse
|
13
|
Kishi T. Clarification of hypertension mechanisms provided by the research of central circulatory regulation. Hypertens Res 2023; 46:1908-1916. [PMID: 37277436 DOI: 10.1038/s41440-023-01335-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023]
Abstract
Sympathoexcitation, under the regulatory control of the brain, plays a pivotal role in the etiology of hypertension. Within the brainstem, significant structures involved in the modulation of sympathetic nerve activity include the rostral ventrolateral medulla (RVLM), caudal ventrolateral medulla (CVLM), nucleus tractus solitarius (NTS), and paraventricular nucleus (paraventricular). The RVLM, in particular, is recognized as the vasomotor center. Over the past five decades, fundamental investigations on central circulatory regulation have underscored the involvement of nitric oxide (NO), oxidative stress, the renin-angiotensin system, and brain inflammation in regulating the sympathetic nervous system. Notably, numerous significant findings have come to light through chronic experiments conducted in conscious subjects employing radio-telemetry systems, gene transfer techniques, and knockout methodologies. Our research has centered on elucidating the role of NO and angiotensin II type 1 (AT1) receptor-induced oxidative stress within the RVLM and NTS in regulating the sympathetic nervous system. Additionally, we have observed that various orally administered AT1 receptor blockers effectively induce sympathoinhibition by reducing oxidative stress via blockade of the AT1 receptor in the RVLM of hypertensive rats. Recent advances have witnessed the development of several clinical interventions targeting brain mechanisms. Nonetheless, Future and further basic and clinical research are needed.
Collapse
Affiliation(s)
- Takuya Kishi
- Department of Graduate School of Medicine (Cardiology), International University of Health and Welfare, Okawa, Japan.
| |
Collapse
|
14
|
Zheng P, Wu H, Gu Y, Li L, Hu R, Ma W, Bian Z, Liu N, Yang D, Chen X. Atorvastatin ameliorates lipid overload-induced mitochondrial dysfunction and myocardial hypertrophy by decreasing fatty acid oxidation through inactivation of the p-STAT3/CPT1 pathway. Biomed Pharmacother 2023; 157:114024. [PMID: 36402030 DOI: 10.1016/j.biopha.2022.114024] [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: 09/01/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Although statins are shown to have cardiac pleiotropic effects independent of lowering cholesterol, the underlying mechanism remains unclear. Mitochondrial dysfunction induced by increased fatty acid oxidation (FAO) is the culprit in the development of cardiac hypertrophy and dysfunction. This study was to explore whether the cardiac pleiotropic effects of atorvastatin were associated with FAO regulation, with a specific focus on carnitine palmitoyltransferase 1 (CPT1). High-fat diet (HFD)-fed mice and palmitic acid (PA)-stimulated neonatal rat primary cardiomyocytes (NRCMs) were treated with atorvastatin, with or without FAO modulators, signal transducer and activator of transcription 3 (STAT3) agonist, and inhibitor. Atorvastatin (3 mg/kg) did not reduce serum cholesterol levels in HFD-fed mice but ameliorated mitochondrial dysfunction and cardiac hypertrophy. In vitro, atorvastatin and the FAO inhibitor alleviated PA-induced mitochondrial dysfunction and cardiomyocyte hypertrophy. However, the FAO enhancer eliminated atorvastatin's protective effects. Furthermore, atorvastatin decreased CPT1 and FAO levels and prevented STAT3 phosphorylation and nuclear translocation. STAT3 inhibitor had the same inhibitory effects as atorvastatin on CPT1, FAO levels, and cardiomyocyte hypertrophy, whereas STAT3 agonist disrupted these effects of atorvastatin. Our results demonstrate that atorvastatin decreases myocardial FAO by inactivating the p-STAT3/CPT1 signaling pathway, which improves lipid overload-induced mitochondrial dysfunction and cardiac hypertrophy in a cholesterol-independent manner. This is the first study to explore the cardiac pleiotropic effects of atorvastatin with respect to FAO. However, whether atorvastatin regulates FAO in the cardiac hypertrophy model induced by other variables has not been investigated in this work, and this is expected to be performed in the future.
Collapse
Affiliation(s)
- Peng Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Hengfang Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Yilu Gu
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Luo Li
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, 899 Pinghai Road, Suzhou 215123, Jiangsu, China; Suzhou Medical College, Soochow University, 899 Pinghai Road, Suzhou 215123, Jiangsu, China
| | - Ran Hu
- Core Facility of The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Wenjing Ma
- Core Facility of The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Zhiping Bian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Nannan Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Di Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China; Core Facility of The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China.
| | - Xiangjian Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
15
|
Facts and ideas on statins with respect to their lipophilicity: a focus on skeletal muscle cells and bone besides known cardioprotection. Mol Cell Biochem 2022:10.1007/s11010-022-04621-y. [PMID: 36471123 PMCID: PMC9734727 DOI: 10.1007/s11010-022-04621-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Statins are known to block cholesterol synthesis in the liver. They also exhibit non-lipid pleiotropic effects due to the inhibition of protein prenylation, thereby modulating various signaling pathways of cellular homeostasis and integrity. Both lipid control and pleiotropic action of statins are clinically used, mainly for treatment of hypercholesterolemia and primary and secondary prevention of cardiovascular diseases. Because the prescription of statins is increasing and statin therapy is often lifelong, in particular in patients with other risk factors, safety issues being associated with polymorbidity and polypragmasia as well as the persistence with and adherence to statins are specific points of attention of clinicians and clinical pharmacologists. Furthermore, because skeletal myocytes have a cholesterol inhibitory sensitivity greater than hepatocytes, a choice of an appropriate statin based on its lipophilicity and the associated likelihood of its side effects on skeletal muscle cells and bone is warranted in such polymorbid patients. These approaches can effectively modulate the risk: benefit ratio and highlight a need for personalized therapy as much as possible, thereby minimizing risk of discontinuation of therapy and poor compliance.
Collapse
|
16
|
Shaghaghi Z, Alvandi M, Farzipour S, Dehbanpour MR, Nosrati S. A review of effects of atorvastatin in cancer therapy. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:27. [PMID: 36459301 DOI: 10.1007/s12032-022-01892-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
Cancer is one of the most challenging diseases to manage. A sizeable number of researches are done each year to find better diagnostic and therapeutic strategies. At the present time, a package of chemotherapy, targeted therapy, radiotherapy, and immunotherapy is available to cope with cancer cells. Regarding chemo-radiation therapy, low effectiveness and normal tissue toxicity are like barriers against optimal response. To remedy the situation, some agents have been proposed as adjuvants to improve tumor responses. Statins, the known substances for reducing lipid, have shown a considerable capability for cancer treatment. Among them, atorvastatin as a reductase (HMG-CoA) inhibitor might affect proliferation, migration, and survival of cancer cells. Since finding an appropriate adjutant is of great importance, numerous studies have been conducted to precisely unveil antitumor effects of atorvastatin and its associated pathways. In this review, we aim to comprehensively review the most highlighted studies which focus on the use of atorvastatin in cancer therapy.
Collapse
Affiliation(s)
- Zahra Shaghaghi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Alvandi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran. .,Department of Nuclear Medicine and Molecular Imaging, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Soghra Farzipour
- Department of Cardiology, Cardiovascular Diseases Research Center, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Reza Dehbanpour
- Department of Radiology, School of Paramedicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sahar Nosrati
- Institute of Nuclear Chemistry and Technology, Dorodna 16 Str, 03-195, Warsaw, Poland
| |
Collapse
|
17
|
Leal DP, Gonçalinho GHF, Tavoni TM, Kuwabara KL, Paccanaro AP, Freitas FR, Strunz CMC, César LAM, Maranhão RC, Mansur ADP. The Interplay of Sirtuin-1, LDL-Cholesterol, and HDL Function: A Randomized Controlled Trial Comparing the Effects of Energy Restriction and Atorvastatin on Women with Premature Coronary Artery Disease. Antioxidants (Basel) 2022; 11:2363. [PMID: 36552571 PMCID: PMC9774144 DOI: 10.3390/antiox11122363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION HDL function has gained prominence in the literature as there is a greater predictive capacity for risk in early coronary artery disease when compared to the traditional parameters. However, it is unclear how dietary energy restriction and atorvastatin influence HDL function. METHODS A randomized controlled trial with 39 women with early CAD divided into three groups (n = 13): energy restriction (30% of VET), atorvastatin (80 mg), and control. Analyses of traditional biochemical markers (lipid and glucose profile), circulating Sirt-1, and HDL function (lipid composition, lipid transfer, and antioxidant capacity). RESULTS Participants' mean age was 50.5 ± 3.8 years. Energy restriction increased Sirt-1 by 63.6 pg/mL (95%CI: 1.5-125.7; p = 0.045) and reduced BMI by 0.8 kg/m2 (95%CI: -1.349--0.273; p = 0.004) in a manner independent of other cardiometabolic factors. Atorvastatin reduced LDL-c by 40.0 mg/dL (95%CI: -69.910--10.1; p = 0.010). Increased Sirt-1 and reduced BMI were independently associated with reduced phospholipid composition of HDL (respectively, β = -0.071; CI95%:-0.136--0.006; p = 0.033; β = 7.486; CI95%:0.350-14.622; p = 0.040). Reduction in BMI was associated with lower HDL-free cholesterol (β = 0.818; CI95%:0.044-1.593; p = 0.039). LDL-c reduction by statins was associated with reduced maximal lipid peroxide production rate of HDL (β = 0.002; CI95%:0.000-0.003; p = 0.022) and total conjugated diene generation (β = 0.001; CI95%:0.000-0.001; p = 0.029). CONCLUSION This study showed that energy restriction and atorvastatin administration were associated with changes in lipid profile, serum Sirt-1 concentrations, and HDL function.
Collapse
Affiliation(s)
- Dalila Pinheiro Leal
- Faculdade de Medicina da Universidade de São Paulo, University of São Paulo, Sao Paulo 05508-060, Brazil
- Servico de Prevencao, Cardiopatia da Mulher e Reabilitacao Cardiovascular, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
- Laboratorio de Metabolismo de Lipides, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
| | - Gustavo Henrique Ferreira Gonçalinho
- Faculdade de Medicina da Universidade de São Paulo, University of São Paulo, Sao Paulo 05508-060, Brazil
- Servico de Prevencao, Cardiopatia da Mulher e Reabilitacao Cardiovascular, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
| | - Thauany Martins Tavoni
- Laboratorio de Metabolismo de Lipides, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
| | - Karen Lika Kuwabara
- Faculdade de Medicina da Universidade de São Paulo, University of São Paulo, Sao Paulo 05508-060, Brazil
- Servico de Prevencao, Cardiopatia da Mulher e Reabilitacao Cardiovascular, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
| | - Ana Paula Paccanaro
- Laboratorio de Metabolismo de Lipides, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
| | - Fatima Rodrigues Freitas
- Laboratorio de Metabolismo de Lipides, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
| | - Célia Maria Cassaro Strunz
- Laboratorio de Analises Clinicas, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
| | - Luiz Antonio Machado César
- Faculdade de Medicina da Universidade de São Paulo, University of São Paulo, Sao Paulo 05508-060, Brazil
- Unidade Clinica de Coronariopatias Cronicas, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
| | - Raul Cavalcante Maranhão
- Laboratorio de Metabolismo de Lipides, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
- Faculdade de Ciencias Farmaceuticas da Universidade de Sao Paulo, University of São Paulo, Sao Paulo 05508-060, Brazil
| | - Antonio de Padua Mansur
- Faculdade de Medicina da Universidade de São Paulo, University of São Paulo, Sao Paulo 05508-060, Brazil
- Servico de Prevencao, Cardiopatia da Mulher e Reabilitacao Cardiovascular, Instituto do Coracao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-900, Brazil
| |
Collapse
|
18
|
Atorvastatin Inhibits Ferroptosis of H9C2 Cells by regulatingSMAD7/Hepcidin Expression to Improve Ischemia-Reperfusion Injury. Cardiol Res Pract 2022; 2022:3972829. [PMID: 36398315 PMCID: PMC9666047 DOI: 10.1155/2022/3972829] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
Background Ferroptosis plays a key role in cardiomyopathy. Atorvastatin (ATV) has a protective effect on ischemia-reperfusion (I/R) cardiomyopathy. The purpose of this study is to elucidate the mechanism of ATV in I/R injury. Methods H9C2 cells and cardiomyopathy rats were induced by hypoxia/reoxygenation (H/R) and I/R to construct in vitro and in vivo models. Cell viability was determined by CCK8. Cardiac histopathology was observed by HE staining. Transmission electron microscope (TEM) was used to observe the mitochondrial morphology. The reactive oxygen species (ROS) content in cells was analyzed by the biochemical method. ELISA was conducted to calculate the concentrations of total iron/Fe2+ and hepcidin. The expression of ferroptosis and SMAD pathway-related genes were detected by qPCR. Western blot was performed to detect the expression levels of ferroptosis and SMAD pathway-related proteins. Results In H9C2 cells, ATV reversed the decline in cell viability, mitochondrial shrinkage, and ROS elevation induced by erastin or H/R. The concentration of total iron and Fe2+ in H/R-induced H9C2 cells increased, and the protein expression of FPN1 decreased. After ATV treatment, the concentration of total iron and Fe2+ decreased, and the protein expression of FPN1 increased. The expression of the SMAD7 gene in H/R-induced H9C2 cells decreased, and the expression of the hepcidin gene increased, which were reversed by ATV. When SMAD7 was knocked down, ATV treatment failed to produce the above effect. ATV also improved ferroptosis in I/R rat myocardium through the SMAD7/hepcidin pathway. Conclusions ATV reversed the decline in H9C2 cell viability, mitochondrial shrinkage, and ROS elevation, and improved the myocardium ferroptosis through the SMAD7/hepcidin pathway in I/R rat.
Collapse
|
19
|
Statins Have an Anti-Inflammation in CKD Patients: A Meta-Analysis of Randomized Trials. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4842699. [PMID: 36317110 PMCID: PMC9617709 DOI: 10.1155/2022/4842699] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022]
Abstract
Background Persistent inflammation has been recognized as an important comorbid condition in patients with chronic kidney disease (CKD) and is associated with many complications, mortality, and progression of CKD. Previous studies have not drawn a clear conclusion about the anti-inflammatory effects of statins in CKD. This meta-analysis is aimed at assessing the anti-inflammatory effects of statins therapy in patients with CKD. Methods A comprehensive literature search was conducted in these databases (Medline, Embase, Cochrane library, and clinical trials) to identify the randomized controlled trials that assess the anti-inflammatory effects of statins. Subgroup, sensitivity, and trim-and-fill analysis were conducted to determine the robustness of pooled results of the primary outcome. Results 25 eligible studies with 7921 participants were included in this meta-analysis. The present study showed that statins therapy was associated with a decreased C-reactive protein (CRP) (-2.06 mg/L; 95% CI: -2.85 to -1.27, p < 0.01). Subgroup, sensitivity, and trim-and-fill analysis showed that the pooled results of CPR were stable. Conclusion This meta-analysis demonstrates that statins supplementation has anti-inflammatory effects in patients with CKD. Statins exert an anti-inflammatory effect that is clinically important in improving complications, reducing mortality, and slowing progression in CKD. We believe that the benefits of statins to CKD are partly due to their anti-inflammatory effects. However, stains usually are prescribed in the CKD patients with dyslipidemia, whether statins can reduce inflammation in CKD patients with normal serum lipid needed to explore in the future. Therefore, we suggest that randomized clinical trials need to assess the effect of statins in CKD patients with normal serum lipid. Whether statins can be prescribed for aiming to inhibit inflammation in CKD also needed further study. Trial Registration. The study protocol was registered in the International Prospective Register of Systematic Reviews (PROSPERO); registration number: CRD42022310334.
Collapse
|
20
|
Omolaoye TS, Halabi MO, Mubarak M, Cyril AC, Duvuru R, Radhakrishnan R, Du Plessis SS. Statins and Male Fertility: Is There a Cause for Concern? TOXICS 2022; 10:627. [PMID: 36287907 PMCID: PMC9607778 DOI: 10.3390/toxics10100627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 05/06/2023]
Abstract
The well-known 3-hydroxyl 3-methyl glutaryl-Coenzyme A reductase inhibitors, called statins, have been the main medication used in the treatment of hypercholesterolemia and some cases of cardiovascular diseases. The effectiveness of this drug in controlling cholesterol production is impeccable, however, patients often complain of a variety of side effects, such as myalgia, muscle atrophy, and in some cases, rhabdomyolysis. Not only has the use of statins caused the aforementioned side effects, but they are also shown to cause testicular discomfort, erectile dysfunction, altered semen parameters, and modified steroid hormone production. These reported adverse effects on male fertility are not generally agreed upon, as some have shown the use to be beneficial. Hence, this makes the aftermath effect of statin use on male fertility debatable and controversial. The negative effects have been associated with imbalanced or reduced steroid hormones, which are necessary for proper spermatogenesis and other sexual functions. Meanwhile, the beneficial effects are related to statin's anti-inflammatory and cardioprotective properties. These contradictory findings are in part due to the different age of users, concentrations of statins, the type and duration of treatment, and the underlying disease and/or comorbidities. Therefore, the current study aims to analyze the literature and gather evidence as to the effects of statin on male sexual health and reproductive parameters, and subsequently give recommendations for the direction of future studies.
Collapse
Affiliation(s)
- Temidayo S. Omolaoye
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Mouhammad Omar Halabi
- School of Medicine, Royal College of Surgeons Ireland-Bahrain, Busaiteen 15503, Bahrain
| | - Maitha Mubarak
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Asha Caroline Cyril
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Ruthwik Duvuru
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Rajan Radhakrishnan
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Stefan S. Du Plessis
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town P.O. Box 7505, South Africa
| |
Collapse
|
21
|
Tsai MH, Su FY, Chang HY, Su PC, Chiu LY, Nowicki M, Kao CC, Lin YC. The Effect of Statin on Anemia in Patients with Chronic Kidney Disease and End-Stage Kidney Disease: A Systematic Review and Meta-Analysis. J Pers Med 2022; 12:jpm12071175. [PMID: 35887674 PMCID: PMC9317421 DOI: 10.3390/jpm12071175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 11/20/2022] Open
Abstract
Although erythropoietin-stimulating agents are effective in treating anemia in patients with end-stage kidney disease (ESKD) undergoing hemodialysis, some ESKD patients, especially those with inflammation, continue to suffer from anemia. Statin, an inhibitor of hydroxymethylglutaryl-CoA (HMG-CoA) reductase with lipid-lowering effects, may have a pleiotropic effect in reducing inflammation, and thus increase hemoglobin (Hb) level. We searched the PubMed, Embase, and Cochrane databases for relevant studies. The population of interest comprised advanced chronic kidney disease (CKD) patients and ESKD patients receiving hemodialysis with statin treatment. The included study designs were randomized control trial/cohort study/pre-post observational study, and outcomes of interest were Hb, erythropoietin resistance index (ERI) and ferritin. PRISMA 2020 guidelines were followed, and risk of bias (RoB) was assessed using the RoB 2.0 tool in randomized controlled trials, and the Newcastle-Ottawa scale (NOS) in cohort studies. We eventually included ten studies (5258 participants), comprising three randomized controlled trials and seven cohort studies. Overall, Hb increased by 0.84 g/dL (95% confidence interval [CI]: −0.02 to 1.70) in all groups using statins, including single-arm cohorts, and by 0.72 g/dL (95% CI: −0.02 to 1.46) in studies with placebo control. Hb levels were higher in the study group than in the control group, with a mean difference of 0.18 g/dL (95% CI: 0.04–0.32) at baseline and 1.0 g/dL (95% CI: 0.13–1.87) at the endpoint. Ferritin increased by 9.97 ng/mL (95% CI: −5.36 to 25.29) in the study group and decreased by 34.01 ng/mL (95% CI: −148.16 to 80.14) in the control group; ferritin fluctuation was higher in the control group. In conclusion, statin may improve renal anemia in ESKD patients receiving hemodialysis and regular erythropoietin-stimulating agents. Future studies with more rigorous methodology and larger sample size study should be performed to confirm this beneficial effect.
Collapse
Affiliation(s)
- Meng-Hsu Tsai
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250, Wu-xing St., Taipei 110, Taiwan; (M.-H.T.); (F.-Y.S.); (H.-Y.C.); (P.-C.S.); (L.-Y.C.); (C.-C.K.)
- Department of Medical Education, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Fu-You Su
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250, Wu-xing St., Taipei 110, Taiwan; (M.-H.T.); (F.-Y.S.); (H.-Y.C.); (P.-C.S.); (L.-Y.C.); (C.-C.K.)
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, 252, Wu-xing St., Taipei 110, Taiwan
| | - Hao-Yun Chang
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250, Wu-xing St., Taipei 110, Taiwan; (M.-H.T.); (F.-Y.S.); (H.-Y.C.); (P.-C.S.); (L.-Y.C.); (C.-C.K.)
- Department of Medical Education, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Po-Cheng Su
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250, Wu-xing St., Taipei 110, Taiwan; (M.-H.T.); (F.-Y.S.); (H.-Y.C.); (P.-C.S.); (L.-Y.C.); (C.-C.K.)
- Department of Medical Education, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Li-Yun Chiu
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250, Wu-xing St., Taipei 110, Taiwan; (M.-H.T.); (F.-Y.S.); (H.-Y.C.); (P.-C.S.); (L.-Y.C.); (C.-C.K.)
- Department of Medical Education, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Michal Nowicki
- Department of Nephrology, Hypertension and Kidney Transplantation, Central University Hospital, Medical University of Lodz, 92-213 Lodz, Poland;
| | - Chih-Chin Kao
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250, Wu-xing St., Taipei 110, Taiwan; (M.-H.T.); (F.-Y.S.); (H.-Y.C.); (P.-C.S.); (L.-Y.C.); (C.-C.K.)
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, 252, Wu-xing St., Taipei 110, Taiwan
- TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei 110, Taiwan
| | - Yen-Chung Lin
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250, Wu-xing St., Taipei 110, Taiwan; (M.-H.T.); (F.-Y.S.); (H.-Y.C.); (P.-C.S.); (L.-Y.C.); (C.-C.K.)
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, 252, Wu-xing St., Taipei 110, Taiwan
- TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei 110, Taiwan
- Correspondence:
| |
Collapse
|
22
|
Kukula O, Günaydın C. Atorvastatin reduces alloxan-induced impairment of aversive stimulus memory in mice. ASIAN BIOMED 2022; 16:71-78. [PMID: 37551286 PMCID: PMC10321169 DOI: 10.2478/abm-2022-0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background An association between dysregulated glucose levels in patients with diabetes mellitus and detrimental effects on the central nervous system, particularly in Alzheimer disease, has been recognized. Atorvastatin treatment has improved memory and cognition in some patients with diabetes mellitus and Alzheimer disease. Objectives To determine possible neuroprotective effects of atorvastatin on memory and cognition by measuring changes in an adverse stimulus avoidance learning deficit induced by alloxan in a murine model of diabetes mellitus and impaired memory and cognition. Methods We administered 150 mg/kg and 100 mg/kg alloxan in saline (intraperitoneally, i.p.) at a 48 h interval to produce a model of diabetes mellitus in male BALB/c mice. An oral glucose tolerance test (OGTT) was used to assess blood glucose regulation. After demonstrating hyperglycemia in mice (n = 7 per group) we administered vehicle (saline, i.p.), atorvastatin (10 mg/kg, i.p.), or liraglutide (200 μg/kg, i.p.) for 28 d except for those in a negative control group, which were given saline instead of alloxan, and a group administered atorvastatin alone, which were given saline instead of alloxan followed by atorvastatin (10 mg/kg, i.p.) for 28 d. Locomotor activity was measured 24 h after the final drug treatments, and subsequently their learned behavioral response to an adverse electrical stimulus to their plantar paw surface in a dark compartment was measured using a passive avoidance apparatus (Ugo Basile) in a model of impaired memory and cognition associated with Alzheimer disease. To determine any deficit in their learned avoidance of the adverse stimulus, we measured the initial latency or time mice spent in an illuminated white compartment before entering the dark compartment in the learning trial, and on the day after learning to avoid the adverse stimulus, the retention period latency in the light compartment and time spent in the dark compartment. Results Atorvastatin alone produced no significant change in blood glucose levels (F4,10 = 0.80, P = 0.55) within 2 h. Liraglutide decreased blood glucose levels after 0.5 h (F4,10 = 11.7, P < 0.001). We found no significant change in locomotor activity in any group. In mice with alloxan-induced diabetes, atorvastatin significantly attenuated the decreased avoidance associated with the diabetes (F4,30 = 38.0, P = 0.02) and liraglutide also significantly attenuated the decreased avoidance (F4,30 = 38.0, P < 0.001). Atorvastatin alone had no significant effect on the adversive learned response compared with vehicle treatment (F4,30 = 38.0, P > 0.05). Atorvastatin significantly decreased the time mice with alloxan-induced diabetes spent in the dark compartment compared with mice in the diabetes group without atorvastatin treatment (F4,30 = 53.9, P = 0.046). Liraglutide also significantly reduced the time mice with alloxan-induced diabetes spent in the dark compartment compared with vehicle-treated mice with alloxan-induced diabetes (F4,30 = 53.9, P < 0.001). Atorvastatin treatment alone had no significant effect on the time mice spent in dark compartment compared with the control group (F4,30 = 53.9, P > 0.05). Conclusion Atorvastatin significantly attenuated the adverse stimulus avoidance learning deficit in the alloxan-induced murine model of diabetes suggesting decreased impairment of memory and cognition.
Collapse
Affiliation(s)
- Osman Kukula
- Department of Pharmacology, Ondokuz Mayıs University, Faculty of Medicine, Atakum, Samsun55139, Turkey
| | - Caner Günaydın
- Department of Pharmacology, Ondokuz Mayıs University, Faculty of Medicine, Atakum, Samsun55139, Turkey
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Chronic inflammation has been recognized as one of the most important pathophysiological mechanisms' initiation and progression of atherosclerosis. Statins belong to most successful therapeutic agents in the prevention and treatment of atherothrombotic vascular disease. Their non-lipid related effects including suppression of inflammation have been repeatedly proven in both experimental and clinical settings. RECENT FINDINGS Recently, the importance of inflammation in the process of atherosclerosis has been confirmed by interventions targeting inflammation selectively. Clinical trial with selective inhibitor of a principal inflammatory mediator interleukin 1-beta - canakinumab - confirmed the notion of direct vasculoprotective effects of primarily targeting inflammation. This has increased interest in the non-lipid, pleiotropic and, particularly, anti-inflammatory effects of statins. Anti-inflammatory effects of statins have been proven both experimentally and in clinical settings beyond any doubt. They comprise a direct positive effect on not only many cell types and pathways that are lipid independent but, also, some that are mediated by lipid modification. Undoubtedly, suppression of inflammatory response by statins contributes to their generally positive action in atherosclerosis and represents an important part of the vasculo- and atheroprotective effect of this drug class.
Collapse
Affiliation(s)
- Martin Satny
- First Faculty of Medicine, Charles Univesity, U Nemocnice 1, 128 08, Prague, Czech Republic
| | - Jaroslav A Hubacek
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Michal Vrablik
- First Faculty of Medicine, Charles Univesity, U Nemocnice 1, 128 08, Prague, Czech Republic. .,3rd Department of Internal Medicine, General University Hospital, U Nemocnice 1, 128 08, Prague 2, Czech Republic.
| |
Collapse
|
24
|
Association between Statin Use and Sensorineural Hearing Loss in Type 2 Diabetic Patients: A Hospital-Based Study. Pharmaceuticals (Basel) 2021; 14:ph14111076. [PMID: 34832858 PMCID: PMC8625623 DOI: 10.3390/ph14111076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 12/30/2022] Open
Abstract
Statins have emerged as protective agents against sensorineural hearing loss (SNHL) associated with dyslipidemia, but the effects of statins on SNHL are not consistent. The purpose of this study was to investigate the association between statin use and the risk of SNHL using a hospital cohort. This nested case-control study included type 2 diabetic patients over the age of 18 years without a history of hearing loss. Of these, 1379 patients newly diagnosed with SNHL or tinnitus were classified as cases, and 5512 patients matched to the cases based on age, sex, and index year were classified as controls. Chi-squared tests were used to compare categorical variables between the two groups. Odds ratios (ORs) and adjusted odds ratios (AOR) were calculated from univariate and multivariable unconditional logistic regression analyses, respectively. There was a significant difference in the prevalence of statin use between the cases and controls (53.7% vs. 61.2%, respectively; p < 0.001). The use of statins in type 2 diabetic patients significantly reduced the risk of SNHL or tinnitus by 24.8% (95% CI 14.2–34.1%, p < 0.001) after controlling for confounders. Similar results were found for the association between statin use and SNHL (AOR = 0.706; 95% CI 0.616–0.811, p < 0.001). The protective effects of statins against SNHL were consistent regardless of age and sex. The use of statins for type 2 diabetic patients was significantly associated with a reduced risk of SNHL, regardless of age and sex. Further studies are needed, especially large cohort studies, to evaluate the long-term protective effects of statins.
Collapse
|
25
|
Zheng P, Ding Y, Lu F, Liu N, Wu H, Bian Z, Chen X, Yang D. Atorvastatin reverses high cholesterol-induced cardiac remodelling and regulates mitochondrial quality-control in a cholesterol-independent manner: An experimental study. Clin Exp Pharmacol Physiol 2021; 48:1150-1161. [PMID: 33891707 DOI: 10.1111/1440-1681.13507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/09/2021] [Accepted: 04/17/2021] [Indexed: 01/03/2023]
Abstract
Mitochondria are key regulators of cell fate, maintaining self-stability by a fine-tuned quality-control network including mitophagy, biogenesis, fission and fusion processes. Myocardial mitochondria can be impaired by hypercholesterolemia. Statins, such as atorvastatin, are considered the cornerstone in the management of hypercholesterolaemia primarily due to their marked cholesterol-lowering ability. The direct effect of atorvastatin on myocardial mitochondria remains unclear. We aimed to explore whether atorvastatin could attenuate myocardial mitochondrial defects induced by high cholesterol, and whether cycloastragenol, a potent telomerase activator, could be used as a potential complementary bioactive compound for obesity and hypercholesterolaemia treatment. We found that atorvastatin at a low dose (3 mg/kg) did not reduce elevated serum cholesterol, but reversed cardiac remodelling and dysfunction in C57BL/6J mice fed with high-fat diet (HFD). Atorvastatin reversed the upregulated mitophagy, mitochondrial fission and fusion, accompanied by mitochondrial biogenesis activation in HFD-fed mice hearts. Mitochondrial structural impairments were attenuated by atorvastatin in HFD-fed mice and oxidized low-density lipoprotein (ox-LDL) exposed HL-1 cardiomyocytes. The depolarized mitochondrial membrane potential and increased mitochondrial oxygen consumption rates in ox-LDL exposed HL-1 cells were recovered by atorvastatin. Furthermore, atorvastatin co-treated with cycloastragenol had better effects on reducing body weight, improving cardiac remodelling and dysfunction, and protecting mitochondria in high cholesterol. Conclusively, low-dose atorvastatin exhibited a cholesterol-independent cardioprotective effect through improving the mitochondrial quality-control network and repairing mitochondrial ultrastructure in high cholesterol. Atorvastatin plus cycloastragenol supplement therapy has a better effect on treating obesity and hypercholesterolaemia.
Collapse
Affiliation(s)
- Peng Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanzi Ding
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Feiyan Lu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Nannan Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hengfang Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiping Bian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiangjian Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Di Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Science and Technology Office, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Molecules and Mechanisms to Overcome Oxidative Stress Inducing Cardiovascular Disease in Cancer Patients. Life (Basel) 2021; 11:life11020105. [PMID: 33573162 PMCID: PMC7911715 DOI: 10.3390/life11020105] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) are molecules involved in signal transduction pathways with both beneficial and detrimental effects on human cells. ROS are generated by many cellular processes including mitochondrial respiration, metabolism and enzymatic activities. In physiological conditions, ROS levels are well-balanced by antioxidative detoxification systems. In contrast, in pathological conditions such as cardiovascular, neurological and cancer diseases, ROS production exceeds the antioxidative detoxification capacity of cells, leading to cellular damages and death. In this review, we will first describe the biology and mechanisms of ROS mediated oxidative stress in cardiovascular disease. Second, we will review the role of oxidative stress mediated by oncological treatments in inducing cardiovascular disease. Lastly, we will discuss the strategies that potentially counteract the oxidative stress in order to fight the onset and progression of cardiovascular disease, including that induced by oncological treatments.
Collapse
|
27
|
McCarty MF, Assanga SI, Lujan LL. Age-adjusted mortality from pancreatic cancer increased NINE-FOLD in japan from 1950 to 1995 - Was a low-protein quasi-vegan diet a key factor in their former low risk? Med Hypotheses 2021; 149:110518. [PMID: 33582316 DOI: 10.1016/j.mehy.2021.110518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/23/2021] [Indexed: 12/18/2022]
Abstract
During the last half of the twentieth century, age-adjusted mortality from pancreatic cancer in Japan rose about nine-fold in both sexes. Well-characterized risk factors such as smoking, obesity/metabolic syndrome, and heavy alcohol use appear to explain only a modest part of this rise. It is proposed that a diet relatively low in protein, and particularly low in animal protein, was a key determinant of the low risk for pancreatic cancer in mid-century Japan. It is further proposed that pancreatic acinar cells, owing to their extraordinarily high rate of protein synthesis, are at high risk for ER stress; that such stress plays a fundamental role in the induction of most pancreatic cancers; and that low-protein diets help to offset such stress by modulating activities of the kinases GCN2 and mTORC1 while increasing autocrine and systemic production of fibroblast growth factor 21. This model appears to clarify the role of various risk factors and protective factors in pancreatic cancer induction. A vegan or quasi-vegan low-protein diet may have broader potential for decreasing risk for a range of common "Western" cancers.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity Foundation, San Diego, CA, United States.
| | | | | |
Collapse
|
28
|
Tursunova NV, Klinnikova MG, Babenko OA, Lushnikova EL. [Molecular mechanisms of the cardiotoxic action of anthracycline antibiotics and statin-induced cytoprotective reactions of cardiomyocytes]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 66:357-371. [PMID: 33140729 DOI: 10.18097/pbmc20206605357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The manifestation of the side cardiotoxic effect of anthracycline antibiotics limits their use in the treatment of malignant processes in some patients. The review analyzes the main causes of the susceptibility of cardiomyocytes to the damaging effect of anthracyclines, primarily associated with an increase in the processes of free radical oxidation. Currently, research is widely carried out to find ways to reduce anthracycline cardiotoxicity, in particular, the use of cardioprotective agents in the complex treatment of tumors. Hydroxymethylglutaryl coenzyme A reductase inhibitors (statins) have been shown to improve the function and metabolism of the cardiovascular system under various pathological impacts, therefore, it is proposed to use them to reduce cardiotoxic complications of chemotherapy. Statins exhibit direct (hypolipidemic) and pleiotropic effects due to the blockade of mevalonic acid synthesis and downward biochemical cascades that determine their cardioprotective properties. The main point of intersection of the pharmacological activity of anthracyclines and statins is the ability of both to regulate the functioning of small GTPase from the Rho family, and their effect in this regard is the opposite. The influence of statins on the modification and membrane dislocation of Rho proteins mediates the indirect antioxidant, anti-inflammatory, endothelioprotective, antiapoptotic effect. The mechanism of statin inhibition of doxorubicin blockade of the DNA-topoisomerase complex, which may be important in preventing cardiotoxic damage during chemotherapy, is discussed. At the same time, it should be noted that the use of statins can be accompanied by adverse side effects: a provocation of increased insulin resistance and glucose tolerance, which often causes them to be canceled in patients with impaired carbohydrate metabolism, so further studies are needed here. The review also analyzes data on the antitumor effect of statins, their ability to sensitize the tumor to treatment with cytostatic drug. It has been shown that the relationship between anthracycline antibiotics and statins is characterized not only by antagonism, but also in some cases by synergism. Despite some adverse effects, statins are one of the most promising cardio- and vasoprotectors for use in anthracycline cardiomyopathy.
Collapse
Affiliation(s)
- N V Tursunova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - M G Klinnikova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - O A Babenko
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - E L Lushnikova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| |
Collapse
|
29
|
Bonacina F, Da Dalt L, Catapano AL, Norata GD. Metabolic adaptations of cells at the vascular-immune interface during atherosclerosis. Mol Aspects Med 2020; 77:100918. [PMID: 33032828 PMCID: PMC7534736 DOI: 10.1016/j.mam.2020.100918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022]
Abstract
Metabolic reprogramming is a physiological cellular adaptation to intracellular and extracellular stimuli that couples to cell polarization and function in multiple cellular subsets. Pathological conditions associated to nutrients overload, such as dyslipidaemia, may disturb cellular metabolic homeostasis and, in turn, affect cellular response and activation, thus contributing to disease progression. At the vascular/immune interface, the site of atherosclerotic plaque development, many of these changes occur. Here, an intimate interaction between endothelial cells (ECs), vascular smooth muscle cells (VSMCs) and immune cells, mainly monocytes/macrophages and lymphocytes, dictates physiological versus pathological response. Furthermore, atherogenic stimuli trigger metabolic adaptations both at systemic and cellular level that affect the EC layer barrier integrity, VSMC proliferation and migration, monocyte infiltration, macrophage polarization, lymphocyte T and B activation. Rewiring cellular metabolism by repurposing “metabolic drugs” might represent a pharmacological approach to modulate cell activation at the vascular immune interface thus contributing to control the immunometabolic response in the context of cardiovascular diseases.
Collapse
Affiliation(s)
- F Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - L Da Dalt
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - A L Catapano
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCSS Multimedica, Milan, Italy.
| | - G D Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCCS, Ospedale Bassini, Cinisello Balsamo, Italy.
| |
Collapse
|
30
|
Lee YY, Choo OS, Kim YJ, Gil ES, Jang JH, Kang Y, Choung YH. Atorvastatin prevents hearing impairment in the presence of hyperlipidemia. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118850. [PMID: 32918982 DOI: 10.1016/j.bbamcr.2020.118850] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023]
Abstract
It is known that hyperlipidemia is a risk factor for sensorineural hearing loss. However, the biological mechanisms underlying hyperlipidemia and hearing impairment have not been completely elucidated in the cochlea. Based on our previous study of human subjects, elderly people taking drugs for hyperlipidemia showed better hearing than those not taking any medications. We hypothesized that drugs for hyperlipidemia, such as statins, may have the potential to prevent hearing impairment. The aim of this study was to investigate the correlation between hyperlipidemia and hearing impairment and the hearing preservation effect of atorvastatin using a hyperlipidemic mouse model with diet-induced obesity (DIO). Here, we demonstrate that DIO mice had a significant hearing impairment as well as increased levels of reactive oxygen species (ROS) and hair cell death due to reduced levels of pAKT and superoxide dismutase 2 (SOD2). However, these changes were significantly prevented by atorvastatin. Oxidative stress-induced intrinsic apoptosis was decreased by the high expression of Nrf2 and antioxidant genes, which improved mitochondrial function and ROS via activation of the PI3K-pAKT pathway by atorvastatin. Therefore, atorvastatin has the potential to prevent hearing impairment via redox balance in the presence of hyperlipidemia.
Collapse
Affiliation(s)
- Yun Yeong Lee
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea
| | - Oak-Sung Choo
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea
| | - Yeon Ju Kim
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea
| | - Eun Sol Gil
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea; Department of Biomedical Science, Ajou University Graduate School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea
| | - Jeong Hun Jang
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea
| | - Yup Kang
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea; Department of Physiology, Ajou University School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea.
| | - Yun-Hoon Choung
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea; Department of Biomedical Science, Ajou University Graduate School of Medicine, Suwon, Gyunggi-do 443-749, Republic of Korea.
| |
Collapse
|
31
|
Abstract
BACKGROUND Paraquat poisoning is one of leading intoxication worldwide without an effective antidote and treatment protocol. Among the other organs, cardiotoxicity of paraquat has been frequently reported. AIM The protective effects of atorvastatin (STN) on paraquat-induced cardiotoxicity and the role of peroxisome proliferator-activated receptors γ in the mediation of STN effects were investigated. METHODS Forty-two male Wistar rats were aliquoted into control or test groups. The animals in test groups in addition of paraquat received saline normal (PQ), pioglitazone (PGT), atorvastatin (STN), PGT + STN, PGT + GW9662, and/or STN + GW9662 for 14 days. RESULTS PGT and STN lowered lipid peroxidation rate, nitric oxide concentration, and activity of myeloperoxidase and CK/MB in the heart. PGT and STN protected from thiol molecules reduction and PQ-induced histopathological injuries. STN regulated the PQ-induced upregulation of COX-II expression in the heart. All STN-related protective effects were reversed by GW9662 as PPARγ antagonist. CONCLUSIONS These data suggest a cardioprotective effect for STN against the PQ-induced inflammation and oxidative stress. The pharmacologic approach of these findings indicates that STN through PPARγ pathway lowered the PQ-induced cardiotoxicity.
Collapse
|
32
|
Yorulmaz H, Ozkok E, Demir G, Ertugrul Yalcin I, Ates G, Olgac V, Tamer S. Pretreatment of simvastatin on liver trace element levels during endotoxemia. Arch Physiol Biochem 2020; 126:196-200. [PMID: 30450988 DOI: 10.1080/13813455.2018.1508234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
There are a number of studies investigating anti-inflammatory effects of simvastatin in patients with sepsis and animal models. There are a few studies which investigated effect of simvastatin on elements in sepsis. In the present study, the impact of pretreatment with simvastatin on element levels was evaluated in liver during endotoxemia. Rats were divided into control, LPS, simvastatin, and simvastatin + LPS. The histopathologic examination of the liver was performed using hematoxylin and eosin. Selenium, zinc, iron, manganese, magnesium, and copper were analyzed using inductively coupled plasma - optical emission spectroscopy. In the LPS, the hepatocyte cell structure was damaged. In the simvastatin + LPS, hepatocyte, and sinusoidal cord damage were partially smaller than LPS. Levels of selenium, and copper significantly decreased in both of LPS and simvastatin + LPS. In the LPS group, iron was found to increase. In the simvastatin + LPS, zinc was increased. Simvastatin partially smaller liver damage by increasing zinc levels during endotoxemia.
Collapse
Affiliation(s)
| | - Elif Ozkok
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Goksel Demir
- Department of Urban, and Regional Planning, Faculty of Architecture, Kirklareli University, Kirklareli, Turkey
| | - Ibrahim Ertugrul Yalcin
- Department of Molecular Biology, and Genetics, Faculty of Engineering and Natural Sciences, Bahcesehir University, Istanbul, Turkey
| | - Gulten Ates
- Department of Physiology, Faculty of Medicine, Istanbul Yeni Yuzyil University, Istanbul, Turkey
- Department of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Vakur Olgac
- Department of Pathology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Sule Tamer
- Department of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
33
|
Williams EA, Russo V, Ceraso S, Gupta D, Barrett-Jolley R. Anti-arrhythmic properties of non-antiarrhythmic medications. Pharmacol Res 2020; 156:104762. [PMID: 32217149 PMCID: PMC7248574 DOI: 10.1016/j.phrs.2020.104762] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/10/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023]
Abstract
Traditional anti-arrhythmic drugs are classified by the Vaughan-Williams classification scheme based on their mechanisms of action, which includes effects on receptors and/or ion channels. Some known anti-arrhythmic drugs do not perfectly fit into this classification scheme. Other medications/molecules with established non-anti-arrhythmic indications have shown anti-arrhythmic properties worth exploring. In this narrative review, we discuss the molecular mechanisms and evidence base for the anti-arrhythmic properties of traditional non-antiarrhythmic drugs such as inhibitors of the renin angiotensin system (RAS), statins and polyunsaturated fatty acids (PUFAs). In summary, RAS antagonists, statins and PUFAs are 'upstream target modulators' that appear to have anti-arrhythmic roles. RAS blockers prevent the downstream arrhythmogenic effects of angiotensin II - the main effector peptide of RAS - and the angiotensin type 1 receptor. Statins have pleiotropic effects including anti-inflammatory, immunomodulatory, modulation of autonomic nervous system, anti-proliferative and anti-oxidant actions which appear to underlie their anti-arrhythmic properties. PUFAs have the ability to alter ion channel function and prevent excessive accumulation of calcium ions in cardiac myocytes, which might explain their benefits in certain arrhythmic conditions. Clearly, whilst a number of anti-arrhythmic drugs exist, there is still a need for randomised trials to establish whether additional agents, including those already in clinical use, have significant anti-arrhythmic effects.
Collapse
Affiliation(s)
- Emmanuel Ato Williams
- Department of Cardiology, Liverpool Heart and Chest Hospital, Thomas Drive, Liverpool, L14 3PE, United Kingdom; Institute of Aging and Chronic Disease, University of Liverpool, United Kingdom
| | - Vincenzo Russo
- Chair of Cardiology, Department of Medical Translational Sciences, University of Campania "Luigi Vanvitelli", Monaldi Hospital, Naples, Italy
| | - Sergio Ceraso
- Specialization Fellow in Cardiology, Department of Medical Translational Sciences, University of Campania "Luigi Vanvitelli" - Monaldi Hospital, Naples, Italy
| | - Dhiraj Gupta
- Department of Cardiology, Liverpool Heart and Chest Hospital, Thomas Drive, Liverpool, L14 3PE, United Kingdom
| | - Richard Barrett-Jolley
- Chair Neuropharmacology, Institute of Aging and Chronic Disease, University of Liverpool, United Kingdom.
| |
Collapse
|
34
|
Manfrini O, Amaduzzi P, Bergami M, Cenko E. Effects of Statin Treatment on Patients with Angina and Normal or Nearly Normal Angiograms. Eur Cardiol 2020; 15:e15. [PMID: 32373188 PMCID: PMC7199123 DOI: 10.15420/ecr.2019.15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/08/2020] [Indexed: 11/25/2022] Open
Abstract
This article offers an updated and comprehensive overview of major findings on the effects of statin treatment in patients with chronic angina but without any epicardial coronary artery with obstructive lesion.
Collapse
Affiliation(s)
- Olivia Manfrini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna Bologna, Italy
| | - Peter Amaduzzi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna Bologna, Italy
| | - Maria Bergami
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna Bologna, Italy
| | - Edina Cenko
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna Bologna, Italy
| |
Collapse
|
35
|
Liberale L, Carbone F, Montecucco F, Sahebkar A. Statins reduce vascular inflammation in atherogenesis: A review of underlying molecular mechanisms. Int J Biochem Cell Biol 2020; 122:105735. [PMID: 32126319 DOI: 10.1016/j.biocel.2020.105735] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 01/23/2020] [Accepted: 02/28/2020] [Indexed: 01/09/2023]
Abstract
Chronic inflammation enhances the detrimental role of dyslipidaemia during atherogenesis. Statins are among the most effective anti-atherosclerotic medications, being able to impact on both cardiovascular morbidity and mortality. Although these molecules have been first described as lipid-lowering medications, several lines of evidence suggest additional benefits through their "pleiotropic" anti-atherosclerotic activities. Specifically, statins can modulate vascular atherosclerotic inflammation by directly improving functions of endothelial cells, vascular smooth muscle cells, platelets, and immune cells. Here, we discuss basic and clinical evidence to provide an update on the molecular mechanisms underlying the protective anti-inflammatory role of statins in atherogenesis.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland.
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
36
|
Yan L, Jiaqiong L, Yue G, Xiaoyong L, Xuexian T, Ming L, Yinglan L, Xinxue L, Zena H. Atorvastatin protects against contrast-induced acute kidney injury via upregulation of endogenous hydrogen sulfide. Ren Fail 2020; 42:270-281. [PMID: 33685337 PMCID: PMC7144258 DOI: 10.1080/0886022x.2020.1740098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background Contrast-induced acute kidney injury (CIAKI) is the third leading cause of acute renal failure in hospitalized patients. This study was aimed to investigate whether atorvastatin could upregulate the expression of hydrogen sulfide (H2S) and hence protect against CIAKI. Methods We treated male rats and NRK-52E cells by iopromide to establish in vivo and in vitro models of CIAKI. Pretreatment with atorvastatin was given in CIAKI rats to investigate its effect on CIAKI. We collected serum and urine samples to detect renal function. We obtained kidney tissue for histological analysis and detection of protein concentration. We tested the serum concentration of H2S and renal expression of two H2S synthetases [cystathionine γ-lyase (CSE) and cystathionine-β synthase (CBS)]. NaHS was pretreated in NRK-52E cells to testify its underlying effect on contrast-induced injury. Results Atorvastatin significantly ameliorated renal dysfunction and morphological changes in CIAKI rats, as well as inflammation, apoptosis, and excessive oxidative stress. Atorvastatin also markedly increased the serum concentration of H2S and renal expression of CSE and CBS. Moreover, pretreatment with NaHS in NRK-52E cells considerably attenuated contrast-induced cell death and inflammation. Conclusion Atorvastatin protects against CIAKI via upregulation of endogenous hydrogen sulfide.
Collapse
Affiliation(s)
- Lin Yan
- Department of Nephrology, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lin Jiaqiong
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guo Yue
- Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Xiaoyong
- Department of General Surgery, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tan Xuexian
- Department of Pathology, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Long Ming
- Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Yinglan
- Department of Endocrine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Liao Xinxue
- Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huang Zena
- Department of General Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
37
|
Tran V, De Silva TM, Sobey CG, Lim K, Drummond GR, Vinh A, Jelinic M. The Vascular Consequences of Metabolic Syndrome: Rodent Models, Endothelial Dysfunction, and Current Therapies. Front Pharmacol 2020; 11:148. [PMID: 32194403 PMCID: PMC7064630 DOI: 10.3389/fphar.2020.00148] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/04/2020] [Indexed: 12/30/2022] Open
Abstract
Metabolic syndrome is characterized by visceral obesity, dyslipidemia, hyperglycemia and hypertension, and affects over one billion people. Independently, the components of metabolic syndrome each have the potential to affect the endothelium to cause vascular dysfunction and disrupt vascular homeostasis. Rodent models of metabolic syndrome have significantly advanced our understanding of this multifactorial condition. In this mini-review we compare the currently available rodent models of metabolic syndrome and consider their limitations. We also discuss the numerous mechanisms by which metabolic abnormalities cause endothelial dysfunction and highlight some common pathophysiologies including reduced nitric oxide production, increased reactive oxygen species and increased production of vasoconstrictors. Additionally, we explore some of the current therapeutics for the comorbidities of metabolic syndrome and consider how these benefit the vasculature.
Collapse
Affiliation(s)
- Vivian Tran
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Kyungjoon Lim
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Antony Vinh
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Maria Jelinic
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
38
|
Yu B, Yu M, Zhang H, Xie D, Nie W, Shi K. Suppression of miR-143-3p contributes to the anti-fibrosis effect of atorvastatin on myocardial tissues via the modulation of Smad2 activity. Exp Mol Pathol 2020; 112:104346. [PMID: 31758917 DOI: 10.1016/j.yexmp.2019.104346] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 08/20/2019] [Accepted: 11/19/2019] [Indexed: 01/17/2023]
Abstract
Atorvastatin is a commonly prescribed statin drug for the control of lipid synthesis and recent studies have shown the cardiac protection potential of atorvastatin. Cardiac fibrosis is a critical process that impairs heart function. In the current study, the anti-fibrosis potential of atorvastatin was assessed and the mechanism associated with the treatment was explored. Fibrotic symptoms were induced using transverse aortic constriction (TAC) method in vivo and using TGF-β1 in vitro. The effect of atorvastatin on the development of cardiac fibrosis was firstly measured. Moreover, the influence of miR-143-3p induction on the anti-fibrosis function of atorvastatin was determined. TAC administration induced cardiac fibrosis and heart weight increase, which was associated with the induced expressions of TGF-β1, miR-143-3p, p-Smad2, and collagens. Atorvastatin restored the levels of TGF-β1, miR-143-3p, p-Smad2, and collagens. The administration of TGF-β1 induced the expressions of miR-143-3p, p-Smad2, and collagens in cardiac fibroblasts (CFs) and the effect was inhibited by atorvastatin. However, the function of atorvastatin was blocked by miR-143-3p mimics. The current study demonstrated that the suppression of miR-143-3p contributed to the anti-fibrosis effect of atorvastatin on myocardial tissues, which subsequently inhibited Smad2-mediated production of collagens.
Collapse
Affiliation(s)
- Bo Yu
- Department of Cardiology, Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Ming Yu
- Department of Cardiology, Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Hongli Zhang
- Department of Cardiology, Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Di Xie
- Department of Cardiology, Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Wei Nie
- Department of Cardiology, Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Kaiyao Shi
- Department of Cardiology, Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| |
Collapse
|
39
|
Ruszkowski P, Masajtis-Zagajewska A, Nowicki M. Effects of combined statin and ACE inhibitor therapy on endothelial function and blood pressure in essential hypertension - a randomised double-blind, placebo controlled crossover study. J Renin Angiotensin Aldosterone Syst 2020; 20:1470320319868890. [PMID: 31486700 PMCID: PMC6728690 DOI: 10.1177/1470320319868890] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background: The aim of this study was to compare the influence of
3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors on endothelial
function and blood pressure in patients with essential hypertension on
long-term angiotensin-converting enzyme inhibitor therapy. Method: The study was designed as a prospective, double-blind, randomised, placebo
controlled, crossover clinical trial. Twenty patients with essential
hypertension were treated with an angiotensin-converting enzyme inhibitor;
the control group included 10 healthy subjects. Hypertensive patients
received in random order 80 mg of fluvastatin daily or placebo for 6 weeks.
The following parameters were assessed at baseline and after each treatment
period: serum lipids, flow-mediated vasodilation, activity of von Willebrand
factor, concentration of vascular endothelial growth factor, C-reactive
protein and 24-hour blood pressure profile. Results: Hypertensive patients did not differ from healthy subjects with respect to
age, body mass and biochemical parameters, with the exception of C-reactive
protein, which was higher in hypertensive patients
(P=0.02). After statin therapy, low-density lipoprotein
cholesterol (P<0.0001), C-reactive protein
(P=0.03), von Willebrand factor
(P=0.03) and vascular endothelial growth factor
(P<0.01) decreased and flow-mediated vasodilation
improved (P<0.001). Statins had no significant effect on
blood pressure. Conclusions: Statins added to angiotensin-converting enzyme inhibitors may improve
endothelial function and ameliorate inflammation independently of blood
pressure.
Collapse
Affiliation(s)
- Piotr Ruszkowski
- Department of Nephrology, Hypertension and Kidney Transplantation, Medical University of Lodz, Poland
| | - Anna Masajtis-Zagajewska
- Department of Nephrology, Hypertension and Kidney Transplantation, Medical University of Lodz, Poland
| | - Michał Nowicki
- Department of Nephrology, Hypertension and Kidney Transplantation, Medical University of Lodz, Poland
| |
Collapse
|
40
|
Healy A, Berus JM, Christensen JL, Lee C, Mantsounga C, Dong W, Watts JP, Assali M, Ceneri N, Nilson R, Neverson J, Wu WC, Choudhary G, Morrison AR. Statins Disrupt Macrophage Rac1 Regulation Leading to Increased Atherosclerotic Plaque Calcification. Arterioscler Thromb Vasc Biol 2020; 40:714-732. [PMID: 31996022 DOI: 10.1161/atvbaha.119.313832] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Calcification of atherosclerotic plaque is traditionally associated with increased cardiovascular event risk; however, recent studies have found increased calcium density to be associated with more stable disease. 3-hydroxy-3-methylglutaryl coenzymeA reductase inhibitors or statins reduce cardiovascular events. Invasive clinical studies have found that statins alter both the lipid and calcium composition of plaque but the molecular mechanisms of statin-mediated effects on plaque calcium composition remain unclear. We recently defined a macrophage Rac (Ras-related C3 botulinum toxin substrate)-IL-1β (interleukin-1 beta) signaling axis to be a key mechanism in promoting atherosclerotic calcification and sought to define the impact of statin therapy on this pathway. Approach and Results: Here, we demonstrate that statin therapy is independently associated with elevated coronary calcification in a high-risk patient population and that statins disrupt the complex between Rac1 and its inhibitor RhoGDI (Rho GDP-dissociation inhibitor), leading to increased active (GTP bound) Rac1 in primary monocytes/macrophages. Rac1 activation is prevented by rescue with the isoprenyl precursor geranylgeranyl diphosphate. Statin-treated macrophages exhibit increased activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), increased IL-1β mRNA, and increased Rac1-dependent IL-1β protein secretion in response to inflammasome stimulation. Using an animal model of calcific atherosclerosis, inclusion of statin in the atherogenic diet led to a myeloid Rac1-dependent increase in atherosclerotic calcification, which was associated with increased serum IL-1β expression, increased plaque Rac1 activation, and increased plaque expression of the osteogenic markers, alkaline phosphatase and RUNX2 (Runt-related transcription factor 2). CONCLUSIONS Statins are capable of increasing atherosclerotic calcification through disinhibition of a macrophage Rac1-IL-1β signaling axis.
Collapse
Affiliation(s)
- Abigail Healy
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Joshua M Berus
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Jared L Christensen
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Cadence Lee
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Chris Mantsounga
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Willie Dong
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Jerome P Watts
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Maen Assali
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Nicolle Ceneri
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Rachael Nilson
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Jade Neverson
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Wen-Chih Wu
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Gaurav Choudhary
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| | - Alan R Morrison
- From the Department of Medicine (Section of Cardiovascular Medicine) and Research Services, Providence VA Medical Center, RI; and Department of Internal Medicine (Section of Cardiovascular Medicine), Alpert Medical School at Brown University, Providence, RI
| |
Collapse
|
41
|
Tentolouris A, Eleftheriadou I, Tzeravini E, Tsilingiris D, Paschou SA, Siasos G, Tentolouris N. Endothelium as a Therapeutic Target in Diabetes Mellitus: From Basic Mechanisms to Clinical Practice. Curr Med Chem 2020; 27:1089-1131. [PMID: 30663560 DOI: 10.2174/0929867326666190119154152] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/28/2018] [Accepted: 01/09/2019] [Indexed: 12/12/2022]
Abstract
Endothelium plays an essential role in human homeostasis by regulating arterial blood pressure, distributing nutrients and hormones as well as providing a smooth surface that modulates coagulation, fibrinolysis and inflammation. Endothelial dysfunction is present in Diabetes Mellitus (DM) and contributes to the development and progression of macrovascular disease, while it is also associated with most of the microvascular complications such as diabetic retinopathy, nephropathy and neuropathy. Hyperglycemia, insulin resistance, hyperinsulinemia and dyslipidemia are the main factors involved in the pathogenesis of endothelial dysfunction. Regarding antidiabetic medication, metformin, gliclazide, pioglitazone, exenatide and dapagliflozin exert a beneficial effect on Endothelial Function (EF); glimepiride and glibenclamide, dipeptidyl peptidase-4 inhibitors and liraglutide have a neutral effect, while studies examining the effect of insulin analogues, empagliflozin and canagliflozin on EF are limited. In terms of lipid-lowering medication, statins improve EF in subjects with DM, while data from short-term trials suggest that fenofibrate improves EF; ezetimibe also improves EF but further studies are required in people with DM. The effect of acetylsalicylic acid on EF is dose-dependent and lower doses improve EF while higher ones do not. Clopidogrel improves EF, but more studies in subjects with DM are required. Furthermore, angiotensin- converting-enzyme inhibitors /angiotensin II receptor blockers improve EF. Phosphodiesterase type 5 inhibitors improve EF locally in the corpus cavernosum. Finally, cilostazol exerts favorable effect on EF, nevertheless, more data in people with DM are required.
Collapse
Affiliation(s)
- Anastasios Tentolouris
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Ioanna Eleftheriadou
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Evangelia Tzeravini
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Dimitrios Tsilingiris
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Stavroula A Paschou
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Gerasimos Siasos
- First Department of Cardiology, Hippokration Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Nikolaos Tentolouris
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
42
|
Verma MK, Jaiswal A, Sharma P, Kumar P, Singh AN. Oxidative stress and biomarker of TNF-α, MDA and FRAP in hypertension. J Med Life 2019; 12:253-259. [PMID: 31666827 PMCID: PMC6814874 DOI: 10.25122/jml-2019-0031] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Concurrent with the misbalance of oxidizing agents and antioxidants, high blood pressure is a major physical burden condition in the current scenario. Tumor necrosis factor-α (TNF-α) plays a vital role in the pathogenesis of hypertension. Tumor necrosis factor-α, inhibitor improves clinical symptoms however their outcome on high blood pressure has not been investigated. We investigated the inflammatory marker TNF-α, malondialdehyde (MDA) and ferric reducing antioxidant power (FRAP) in hypertensive patients. We measured randomly blood pressure using an ambulatory observe in hypertensive patients, measured systolic BP X 140 mmHg and/or diastolic BP X 90 mmHg were considered hypertensive. Total 60 cases were considered in the study that involves 30 hypertensive patients and 30 normal control. Measurements of serum concentrations of TNF-α, MDA, FRAP in hypertension patients was done in both the groups. Serum TNF-α was found to be remarkably increased in study subjects as compared to normal group (r=0.32, p<0.0001*). Serum MDA was also raised in hypertensive as compared to control (r=0.99**, p<0.0001*). While Serum FRAP was found to be decreased in hypertensive group in comparison to healthy control (r=0.23, p<0.0001*). It is concluded that high blood pressure leads to generation of oxidative stress with remarkable elevation of TNF-α and malondialdehyde levels. While reduced FRAP indicates its probable role in lipid peroxidation and in the pathogenesis of hypertension.
Collapse
Affiliation(s)
| | - Anoop Jaiswal
- Department of Biochemistry, Moti Lal Nehru Medical College, Allahabad, India
| | - Preeti Sharma
- Department of Biochemistry, Santosh Medical College and Hospital (Santosh University), Ghaziabad, India
| | - Pradeep Kumar
- Department of Biochemistry, Santosh Medical College and Hospital (Santosh University), Ghaziabad, India
| | | |
Collapse
|
43
|
Guo DW, Wang CY, Shih HC. N-acetylcysteine and atorvastatin alleviates lung injury due to ischemia-reperfusion injury in rats. J Chin Med Assoc 2019; 82:909-914. [PMID: 31567653 DOI: 10.1097/jcma.0000000000000193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Acute lung injury is a major cause of death following severe injury and ischemia-reperfusion (IR). We investigated the protective effect of pretreatment with N-acetylcysteine (NAC) and atorvastatin (ATOR) in a mesenteric IR rat model. METHODS Male rats were randomly divided into five experimental groups: sham; mesenteric IR; and ATOR, NAC, ATOR + NAC (A + N) pretreatment followed by IR. Blood gas and cytokine levels, biochemistry, and cell count were analyzed. Lung injury was evaluated through histopathology and by using the wet-to-dry lung weight (W/D) ratio. RESULTS Following IR, significant changes were noted in biochemistry, cytokine, and lung injury. Compared with those in the IR group, neutrophil-to-lymphocyte ratio, lactate and alanine aminotransferase (ALT) levels were lower in all pretreatment groups, and creatinine and alkaline phosphatase (ALKP) levels were lower only in the A + N group. Blood pH and base excess (BE) were higher, and partial pressure of carbon dioxide in venous blood (PvCO2) lowered significantly in the ATOR and A + N groups than those in the IR group, and bicarbonate (HCO3-) levels increased only in the A + N group. Lung injury scores and W/D indicated significant attenuation in the A + N group. Compared with those in the IR group, tissue tumor necrosis factor-α levels were significantly lower in all the pretreatment groups and interleukin-1β levels were lower in the A + N group. CONCLUSION NAC and ATOR decreased inflammation and lung injury following mesenteric IR in rats. NAC and ATOR may alleviate lung injury more efficiently in combination than individually.
Collapse
Affiliation(s)
- Da-Wei Guo
- Institute of Emergency and Critical Care Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Emergency, Tri-Service General Hospital, Taipei, Taiwan, ROC
| | - Chien-Ying Wang
- Division of Trauma, Department of Emergency, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hsin-Chin Shih
- Institute of Emergency and Critical Care Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Trauma, Department of Emergency, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
44
|
Piccirillo F, Carpenito M, Verolino G, Chello C, Nusca A, Lusini M, Spadaccio C, Nappi F, Di Sciascio G, Nenna A. Changes of the coronary arteries and cardiac microvasculature with aging: Implications for translational research and clinical practice. Mech Ageing Dev 2019; 184:111161. [PMID: 31647940 DOI: 10.1016/j.mad.2019.111161] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 12/28/2022]
Abstract
Aging results in functional and structural changes in the cardiovascular system, translating into a progressive increase of mechanical vessel stiffness, due to a combination of changes in micro-RNA expression patterns, autophagy, arterial calcification, smooth muscle cell migration and proliferation. The two pivotal mechanisms of aging-related endothelial dysfunction are oxidative stress and inflammation, even in the absence of clinical disease. A comprehensive understanding of the aging process is emerging as a primary concern in literature, as vascular aging has recently become a target for prevention and treatment of cardiovascular disease. Change of life-style, diet, antioxidant regimens, anti-inflammatory treatments, senolytic drugs counteract the pro-aging pathways or target senescent cells modulating their detrimental effects. Such therapies aim to reduce the ineluctable burden of age and contrast aging-associated cardiovascular dysfunction. This narrative review intends to summarize the macrovascular and microvascular changes related with aging, as a better understanding of the pathways leading to arterial aging may contribute to design new mechanism-based therapeutic approaches to attenuate the features of vascular senescence and its clinical impact on the cardiovascular system.
Collapse
Affiliation(s)
| | | | | | - Camilla Chello
- Dermatology, Università "La Sapienza" di Roma, Rome, Italy
| | | | - Mario Lusini
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | | | - Francesco Nappi
- Cardiac surgery, Centre Cardiologique du Nord de Saint Denis, Paris, France
| | | | - Antonio Nenna
- Cardiovascular surgery, Università Campus Bio-Medico di Roma, Rome, Italy.
| |
Collapse
|
45
|
NADPH oxidases and oxidase crosstalk in cardiovascular diseases: novel therapeutic targets. Nat Rev Cardiol 2019; 17:170-194. [PMID: 31591535 DOI: 10.1038/s41569-019-0260-8] [Citation(s) in RCA: 368] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS)-dependent production of ROS underlies sustained oxidative stress, which has been implicated in the pathogenesis of cardiovascular diseases such as hypertension, aortic aneurysm, hypercholesterolaemia, atherosclerosis, diabetic vascular complications, cardiac ischaemia-reperfusion injury, myocardial infarction, heart failure and cardiac arrhythmias. Interactions between different oxidases or oxidase systems have been intensively investigated for their roles in inducing sustained oxidative stress. In this Review, we discuss the latest data on the pathobiology of each oxidase component, the complex crosstalk between different oxidase components and the consequences of this crosstalk in mediating cardiovascular disease processes, focusing on the central role of particular NADPH oxidase (NOX) isoforms that are activated in specific cardiovascular diseases. An improved understanding of these mechanisms might facilitate the development of novel therapeutic agents targeting these oxidase systems and their interactions, which could be effective in the prevention and treatment of cardiovascular disorders.
Collapse
|
46
|
Antioxidant properties of probucol released from mesoporous silica. Eur J Pharm Sci 2019; 138:105038. [PMID: 31398394 DOI: 10.1016/j.ejps.2019.105038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/15/2019] [Accepted: 08/05/2019] [Indexed: 11/21/2022]
Abstract
Antioxidants play a vital role in scavenging reactive oxygen species (ROS) produced by the reduction of molecular oxygen from various cellular mechanisms. Under oxidative stress, an increase in the levels of ROS overwhelms the antioxidant response, causing oxidative damage to biological molecules, and leading to the development of various diseases. Drug compounds with potent antioxidant properties are typically poorly water soluble and highly hydrophobic. An extreme case is Probucol (PB), a potent antioxidant with reported water solubility of 5 ng/ml, and oral bioavailiability of <10%. In this study, PB was loaded in mesoporous silica at various drug loadings to understand the changes to the physical properties of the loaded drug, and it's in vitro drug release. Further in vitro studies were conducted in endothelial and microglia cell models to compare the free radical scavening efficiency of ascorbic acid, PB, and PB release from mesoporous silica particles. Out of the three different mesostructured particles studied, the maximum loading of PB was achieved for large pore mesoporous particles (SBA-15) at 50 wt% drug loading, before complete pore filling was observed. For all materials, loadings above complete pore filling resulted in the recrystallization of PB on the external surface. In vitro drug release measurements showed a rapid dissolution rate at low drug loadings compared to a bimodal release profile of amorphous and crystalline drug at higher drug loadings. PB loaded in mesoporous particle was shown to enhance the antioxidant response to extracellular ROS in the endothelial cell line model, and to intracellular ROS in the microglia cell model. Our results indicate that the antioxidant properties of PB can be significantly improved by using mesoporous silica as a delivery vehicle.
Collapse
|
47
|
Zinellu A, Paliogiannis P, Usai MF, Carru C, Mangoni AA. Effect of statin treatment on circulating malondialdehyde concentrations: a systematic review and meta-analysis. Ther Adv Chronic Dis 2019; 10:2040622319862714. [PMID: 31367297 PMCID: PMC6643183 DOI: 10.1177/2040622319862714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/17/2019] [Indexed: 12/30/2022] Open
Abstract
Background The effect of statins on oxidative stress markers, such as malondialdehyde (MDA), is still a matter of debate. We sought to address this issue by conducting a systematic review and meta-analysis of published data on the effect of statin treatment on systemic MDA concentrations. Methods A literature search was conducted on MEDLINE/PubMed, ISI Web of Sciences and Scopus. Data were pooled using a random-effects model. Results A total of 35 studies assessing MDA concentrations before and after statin treatment in 1512 participants (mean age 53.6 years, 48.7% males) were identified. Extreme between-study heterogeneity was observed (I2 = 96.0%, p < 0.001). Pooled standardized mean difference (SMD) showed a significant reduction in plasma MDA concentrations after treatment (SMD = -1.47 µmol/l, 95% confidence interval = -1.89 to -1.05 μmol/l; p < 0.001). Similarly, a subgroup analysis of 10 studies that also included a placebo group showed a significant reduction in plasma MDA concentrations with statins (-1.03 μmol/l, 95% confidence interval = -1.52 to -0.29 μmol/l; p = 0.036). Conclusions This systematic review and meta-analysis showed that statin treatment significantly reduces systemic MDA concentrations. However, the results should be interpreted with caution because of extreme between-study heterogeneity, which warrants further intervention studies.
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Maria Franca Usai
- Department of Chemistry and Pharmacy, University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, SA 5042, Australia
| |
Collapse
|
48
|
Hirunpattarasilp C, Attwell D, Freitas F. The role of pericytes in brain disorders: from the periphery to the brain. J Neurochem 2019; 150:648-665. [PMID: 31106417 DOI: 10.1111/jnc.14725] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/15/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022]
Abstract
It is becoming increasingly apparent that disorders of the brain microvasculature contribute to many neurological disorders. In recent years it has become clear that a major player in these events is the capillary pericyte which, in the brain, is now known to control the blood-brain barrier, regulate blood flow, influence immune cell entry and be crucial for angiogenesis. In this review we consider the under-explored possibility that peripheral diseases which affect the microvasculature, such as hypertension, kidney disease and diabetes, produce central nervous system (CNS) dysfunction by mechanisms affecting capillary pericytes within the CNS. We highlight how cellular messengers produced peripherally can act via signalling pathways within CNS pericytes to reshape blood vessels, restrict blood flow or compromise blood-brain barrier function, thus causing neuronal dysfunction. Increased understanding of how renin-angiotensin, Rho-kinase and PDGFRβ signalling affect CNS pericytes may suggest novel therapeutic approaches to reducing the CNS effects of peripheral disorders.
Collapse
Affiliation(s)
- Chanawee Hirunpattarasilp
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - David Attwell
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - Felipe Freitas
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| |
Collapse
|
49
|
Gu X, Xie S, Hong D, Ding Y. An in vitro model of foam cell formation induced by a stretchable microfluidic device. Sci Rep 2019; 9:7461. [PMID: 31097769 PMCID: PMC6522483 DOI: 10.1038/s41598-019-43902-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/13/2019] [Indexed: 11/17/2022] Open
Abstract
Although a variety of animal models of atherosclerosis have been developed, these models are time-consuming and costly. Here, we describe an in vitro model to induce foam cell formation in the early stage of atherosclerosis. This model is based on a three-dimension co-culture system in a stretchable microfluidic device. An elastic membrane embedded in the microfluidic device is capable of delivering nonuniform strain to vascular smooth muscle cells, endothelial cells and monocytes adhering thereto, which are intended to mimic the biological environment of blood vessels. Under low-density lipoprotein and stretch treatment, foam cell formation was successfully induced in co-culture with changes in mRNA and protein expression of some related key factors. Subsequently, the model was used to assess the inhibitory effect of atorvastatin on foam cell formation. The results obtained indicate that atorvastatin has a significantly dose-dependent inhibition of foam cell formation, which can be explained by the changes in mRNA and protein expression of the related factors. In principle, the model can be used to study the role of different types of cells in the formation of foam cells, as well as the evaluation of anti-atherosclerotic drugs.
Collapse
Affiliation(s)
- Xiaoyang Gu
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China
| | - Shijie Xie
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China
| | - Dandan Hong
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China
| | - Yongsheng Ding
- College of Life Sciences, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
50
|
Zhang QS, Deater M, Phan N, Marcogliese A, Major A, Guinan EC, Grompe M. Combination therapy with atorvastatin and celecoxib delays tumor formation in a Fanconi anemia mouse model. Pediatr Blood Cancer 2019; 66:e27460. [PMID: 30255556 PMCID: PMC6249055 DOI: 10.1002/pbc.27460] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/17/2018] [Accepted: 08/14/2018] [Indexed: 11/05/2022]
Abstract
BACKGROUND Fanconi anemia is an inherited bone marrow failure disorder associated with a high incidence of leukemia and solid tumors. Currently, no interventions to prevent or delay the formation of solid tumors are available. PROCEDURE Two of the most important hallmarks of Fanconi anemia are inflammation and oxidative stress. In this study, we administrated the antioxidant atorvastatin and the anti-inflammatory drug celecoxib to cohorts of Fancd2-/- /Trp53+/- mice, a model of Fanconi anemia. Treatment started at weaning and continued until the mice developed a palpable mass or suffered from >20% weight loss. Tumor samples and selected tissues were subjected to histopathological examination. χ2 test was performed to analyze tumor incidence, and Kaplan-Meier survival curves were evaluated with log-rank test. In addition, a small cohort of mice was monitored for the safety of the drugs. RESULTS The combined oral administration of both drugs significantly delayed tumor onset in Fancd2-/- /Trp53+/- mice. Specifically, the treatment delayed the onset of ovarian tumors in Fancd2-/- /Trp53+/- mice and increased the mean ovarian tumor-free survival time by 17%, whereas this combinatorial drug regimen did not have a significant effect on other tumor types. In addition, no detrimental effects on hematopoiesis from the drug treatment were observed during a 12-month safety monitoring. CONCLUSIONS The data presented here suggest that a combination of atorvastatin and celecoxib may be a good candidate for chemoprevention in Fanconi anemia.
Collapse
Affiliation(s)
- Qing-shuo Zhang
- Oregon Stem Cell Center, Department of Pediatrics, Oregon Health and Science University, Portland, USA
| | - Matthew Deater
- Oregon Stem Cell Center, Department of Pediatrics, Oregon Health and Science University, Portland, USA
| | - Ngoc Phan
- Oregon Stem Cell Center, Department of Pediatrics, Oregon Health and Science University, Portland, USA
| | | | - Angela Major
- Department of Pathology, Baylor College of Medicine, Houston, USA
| | - Eva C. Guinan
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA,Department of Radiation Oncology, Harvard Medical School, Boston, USA
| | - Markus Grompe
- Oregon Stem Cell Center, Department of Pediatrics, Oregon Health and Science University, Portland, USA
| |
Collapse
|