1
|
Parker LE, Papanicolaou KN, Zalesak-Kravec S, Weinberger EM, Kane MA, Foster DB. Retinoic acid signaling and metabolism in heart failure. Am J Physiol Heart Circ Physiol 2025; 328:H792-H813. [PMID: 39933792 DOI: 10.1152/ajpheart.00871.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 12/24/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Nearly 70 years after studies first showed that the offspring of vitamin A (retinol, ROL)-deficient rats exhibit structural cardiac defects and over 20 years since the role of vitamin A's potent bioactive metabolite hormone, all-trans retinoic acid (ATRA), was elucidated in embryonic cardiac development, the role of the vitamin A metabolites, or retinoids, in adult heart physiology and heart and vascular disease, remains poorly understood. Studies have shown that low serum levels of retinoic acid correlate with higher all-cause and cardiovascular mortality, though the relationship between circulating retinol and ATRA levels, cardiac tissue ATRA levels, and intracellular cardiac ATRA signaling in the context of heart and vascular disease has only begun to be addressed. We have recently shown that patients with idiopathic dilated cardiomyopathy show a nearly 40% decline of in situ cardiac ATRA levels, despite adequate local stores of retinol. Moreover, we and others have shown that the administration of ATRA forestalls the development of heart failure (HF) in rodent models. In this review, we summarize key facets of retinoid metabolism and signaling and discuss mechanisms by which impaired ATRA signaling contributes to several HF hallmarks including hypertrophy, contractile dysfunction, poor calcium handling, redox imbalance, and fibrosis. We highlight unresolved issues in cardiac ATRA metabolism whose pursuit will help refine therapeutic strategies aimed at restoring ATRA homeostasis.
Collapse
Affiliation(s)
- Lauren E Parker
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Kyriakos N Papanicolaou
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | | | - Eva M Weinberger
- School of Medicine, Imperial College London, London, United Kingdom
| | - Maureen A Kane
- School of Pharmacy, University of Maryland, Baltimore, Maryland, United States
| | - D Brian Foster
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
2
|
Mawson AR. Understanding health disparities affecting people of West Central African descent in the United States: An evolutionary perspective. Evol Appl 2023; 16:963-978. [PMID: 37216026 PMCID: PMC10197229 DOI: 10.1111/eva.13549] [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: 01/28/2022] [Revised: 03/19/2023] [Accepted: 04/02/2023] [Indexed: 05/24/2023] Open
Abstract
Human populations adapting to diverse aspects of their environment such as climate and pathogens leave signatures of genetic variation. This principle may apply to people of West Central African descent in the United States, who are at increased risk of certain chronic conditions and diseases compared to their European counterparts. Less well known is that they are also at reduced risk of other diseases. While discriminatory practices in the United States continue to affect access to and the quality of healthcare, the health disparities affecting African Americans may also be due in part to evolutionary adaptations to the original environment of sub-Saharan Africa, which involved continuous exposure to the vectors of potentially lethal endemic tropical diseases. Evidence is presented that these organisms selectively absorb vitamin A from the host, and its use in parasite reproduction contributes to the signs and symptoms of the respective diseases. These evolutionary adaptations included (1) sequestering vitamin A away from the liver to other organs, to reduce accessibility to the invaders; and (2) reducing the metabolism and catabolism of vitamin A (vA), causing it to accumulate to subtoxic concentrations and weaken the organisms, thereby reducing the risk of severe disease. However, in the environment of North America, lacking vA-absorbing parasites and with a mainly dairy-based diet that is high in vA, this combination of factors is hypothesized to lead to the accumulation of vA and to increased sensitivity to vA as a toxin, which contribute to the health disparities affecting African Americans. vA toxicity is linked to numerous acute and chronic conditions via mitochondrial dysfunction and apoptosis. Subject to testing, the hypothesis suggests that the adoption of traditional or modified West Central African-style diets that are low in vA and high in vA-absorbing fiber hold promise for disease prevention and treatment, and as a population-based strategy for health maintenance and longevity.
Collapse
Affiliation(s)
- Anthony R. Mawson
- Department of Epidemiology and Biostatistics, School of Public Health, College of Health SciencesJackson State UniversityJacksonMississippiUSA
| |
Collapse
|
3
|
Xu M, Shen YM, Han XY, Liu C, Jiang Q, Cao X, Yan B. "One stone and two birds" strategy to treat neovascular age-related macular degeneration by a novel retinoid drug, EYE-101. Exp Eye Res 2023; 227:109385. [PMID: 36638858 DOI: 10.1016/j.exer.2023.109385] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/20/2022] [Accepted: 01/07/2023] [Indexed: 01/11/2023]
Abstract
Choroidal neovascularization (CNV) is a typical pathological feature of neovascular age-related macular degeneration and has become a major cause of vision loss in the elderly. Current therapies require repeated intraocular injections of anti-VEGF drugs by inhibiting endothelial angiogenic effects, which is painful and may cause adverse effects on normal vascular and neuronal functions. Herein, we designed a novel retinoid drug, EYE-101, determined its therapeutic effects on CNV, and clarified the anti-angiogenic mechanism. The results show that administration of EYE-101 did not cause obvious cytotoxicity and ocular tissue toxicity at the concentrations less than 5 μM. Topical administration of EYE-101 could reduce choroidal sprouting, suppress laser-induced CNV formation, and decrease pericyte coverages on ocular vessels. Administration of EYE-101 also suppressed endothelial cell proliferation, migration, and tube formation and reduced pericyte proliferation, migration, recruitment towards endothelial cells. EYE-101 exerted its anti-angiogenic effects by targeting endothelial cells and pericytes via antagonizing Wnt/β-catenin signaling and PDGF signaling. Thus, EYE-101 administration may offer an"one stone and two birds" strategy for the prevention and treatment of ocular neovascular disorders.
Collapse
Affiliation(s)
- Miao Xu
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China; The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Ya-Ming Shen
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China; The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Xiao-Yan Han
- Eye Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Chang Liu
- Eye Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China; The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China.
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Biao Yan
- Eye Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Chang SF, Tsai HE, Kuo JT, Ruan YR, Chen CY, Wang SY, Liu PY, Lee DY. Blood Reflux-Induced Epigenetic Factors HDACs and DNMTs Are Associated with the Development of Human Chronic Venous Disease. Int J Mol Sci 2022; 23:12536. [PMID: 36293392 PMCID: PMC9603923 DOI: 10.3390/ijms232012536] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/04/2023] Open
Abstract
Blood reflux and metabolic regulation play important roles in chronic venous disease (CVD) development. Histone deacetylases (HDACs) and DNA methyltransferases (DNMTs) serve as repressors that inhibit metabolic signaling, which is induced by proatherogenic flow to promote aortic endothelial cell (EC) dysfunction and atherosclerosis. The aim of this study was to elucidate the relationship between blood reflux and epigenetic factors HDACs and DNMTs in CVD. Human varicose veins with different levels of blood reflux versus normal veins with normal venous flow were examined. The results show that HDAC-1, -2, -3, -5, and -7 are overexpressed in the endothelium of varicose veins with blood reflux. Blood reflux-induced HDACs are enhanced in the varicose veins with a longer duration time of blood reflux. In contrast, these HDACs are rarely expressed in the endothelium of the normal vein with normal venous flow. Similar results are obtained for DNMT1 and DNMT3a. Our findings suggest that the epigenetic factors, HDACs and DNMTs, are induced in venous ECs in response to blood reflux but are inhibited in response to normal venous flow. Blood reflux-induced HDACs and DNMTs could inhibit metabolic regulation and promote venous EC dysfunction, which is highly correlated with CVD pathogenesis.
Collapse
Affiliation(s)
- Shun-Fu Chang
- Department of Medical Research and Development, Chiayi Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- Center for General Education, Chiayi Chang Gung University of Science and Technology, Chiayi 613, Taiwan
| | - Hsiao-En Tsai
- Division of Cardiovascular Surgery, Department of Surgery, National Taiwan University Hsin-Chu Hospital, Hsinchu 300, Taiwan
| | - Jong-Tar Kuo
- Department of Biological Science and Technology, China University of Science and Technology, Taipei 115, Taiwan
| | - Yu-Rong Ruan
- Department of Biological Science and Technology, China University of Science and Technology, Taipei 115, Taiwan
| | - Chiu-Yen Chen
- Department of Biological Science and Technology, China University of Science and Technology, Taipei 115, Taiwan
| | - Shin-Yi Wang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 202, Taiwan
| | - Po-Yu Liu
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Ding-Yu Lee
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 202, Taiwan
| |
Collapse
|
5
|
Histopathological evaluation of a retinoic acid eluting stent in a rabbit iliac artery model. Sci Rep 2022; 12:13305. [PMID: 35922518 PMCID: PMC9349184 DOI: 10.1038/s41598-022-16025-5] [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: 03/16/2022] [Accepted: 07/04/2022] [Indexed: 11/08/2022] Open
Abstract
This study aimed to evaluate the safety and efficacy of innovative retinoic acid (RA) eluting stents with bioabsorbable polymer. Sixty stents divided in ten groups were implanted in the iliac arteries of 30 rabbits. Two polymers ("A", poly (lactic-co-glycolic acid) and "B", polylactic acid), and three doses ("Low", "Medium" and "High") of RA (groups: AL, AM, AH, BL, BM, BH) were used on cobalt chromium stents (Rontis Corporation), one group of bare stent (C), one group (D) of Everolimus eluting stent (Xience-Pro, Abbot Vascular), and two groups of Rontis Everolimus eluting stents coated with polymer A (EA) and B (EB). Treated arteries were explanted after 4 weeks, processed by methyl methacrylate resin and evaluated by histopathology. None of the implanted stents was related with thrombus formation or extensive inflammation. Image analysis showed limited differences between groups regarding area stenosis (BH, D and EB groups had the lower values). Group BH had lower intimal mean thickness than AH (105.1 vs 75.3 μm, p = 0.024). Stents eluting RA, a non-cytotoxic drug, were not related with thrombus formation and had an acceptable degree of stenosis 4 weeks post implantation. RA dose and type of polymer may play role in the biocompatibility of the stents.
Collapse
|
6
|
Deng Q, Chen J. Potential Therapeutic Effect of All-Trans Retinoic Acid on Atherosclerosis. Biomolecules 2022; 12:869. [PMID: 35883425 PMCID: PMC9312697 DOI: 10.3390/biom12070869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/08/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is a major risk factor for myocardial infarction and ischemic stroke, which are the leading cause of death worldwide. All-trans retinoic acid (ATRA) is a natural derivative of essential vitamin A. Numerous studies have shown that ATRA plays an important role in cell proliferation, cell apoptosis, cell differentiation, and embryonic development. All-trans retinoic acid (ATRA) is a ligand of retinoic acid receptors that regulates various biological processes by activating retinoic acid signals. In this paper, the metabolic processes of ATRA were reviewed, with emphasis on the effects of ATRA on inflammatory cells involved in the process of atherosclerosis.
Collapse
Affiliation(s)
| | - Jixiang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| |
Collapse
|
7
|
Hadi Y, Or T, Moady G, Atar S. Psoriasis and coronary heart disease-not as severe as predicted. QJM 2022; 115:388-392. [PMID: 34165570 DOI: 10.1093/qjmed/hcab173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/12/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Psoriasis is a systemic disorder involved in several disease processes, including cancer, metabolic syndrome and cardiovascular disease (CVD). Previous studies showed that psoriasis is most likely an independent risk factor for CVD, yet the extent of its impact on CVD and the extent of coronary artery disease (CAD) remains unclear. We investigated the correlation of psoriasis to the severity of CAD in age and gender-matched patients with CAD with and without psoriasis. METHODS This is a retrospective, case-control study of 59 patients with psoriasis who underwent coronary angiography were matched using a computer software to 59 patients without psoriasis according to age, gender, smoking status, hyperlipidemia, hypertension and diabetes. CAD severity was defined according to number of affected vessels (single vs. multiple) and location of lesions (proximal vs. distal). RESULTS CAD severity was significantly higher in the control group compared to the psoriasis group (P = 0.038). Among patients with psoriasis, 20.3% were disease free or with low severity (42.4%), while only 37.3% had severe CAD. Among patients without psoriasis, the majority had severe CAD (57.6%), followed by low severity (30.5%) or disease free (11.9%). We did not find an association of prior treatment with anti-inflammatory medications and the severity of CAD. CONCLUSIONS Our results show that although psoriasis may be a risk factor for CAD, psoriatic patients have a less severe CAD compared to the general population. The use of anti-inflammatory medications does not explain this finding.
Collapse
Affiliation(s)
- Y Hadi
- Department of Cardiology, Galilee Medical Center, 1 Ben Tzvi Blvd., Nahariya 2210001, Israel
| | - T Or
- From the Azrieli Faculty of Medicine, 8 Szold St., Safed 1311502, Israel
- Department of Cardiology, Galilee Medical Center, 1 Ben Tzvi Blvd., Nahariya 2210001, Israel
| | - G Moady
- From the Azrieli Faculty of Medicine, 8 Szold St., Safed 1311502, Israel
- Department of Cardiology, Galilee Medical Center , 1 Ben Tzvi Blvd., Nahariya 2210001, Israel
| | - S Atar
- From the Azrieli Faculty of Medicine , 8 Szold St., Safed 1311502, Israel
- Department of Cardiology, Galilee Medical Center, 1 Ben Tzvi Blvd., Nahariya 2210001, Israel
| |
Collapse
|
8
|
Yang Z, Yu M, Li X, Tu Y, Wang C, Lei W, Song M, Wang Y, Huang Y, Ding F, Hao K, Han X, Ni X, Qu L, Shen Z, Hu S. Retinoic acid inhibits the angiogenesis of human embryonic stem cell-derived endothelial cells by activating FBP1-mediated gluconeogenesis. Stem Cell Res Ther 2022; 13:239. [PMID: 35672803 PMCID: PMC9171939 DOI: 10.1186/s13287-022-02908-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Endothelial cells are located in the inner lumen of blood and lymphatic vessels and exhibit the capacity to form new vessel branches from existing vessels through a process called angiogenesis. This process is energy intensive and tightly regulated. Glycolysis is the main energy source for angiogenesis. Retinoic acid (RA) is an active metabolite of vitamin A and exerts biological effects through its receptor retinoic acid receptor (RAR). In the clinic, RA is used to treat acne vulgaris and acute promyelocytic leukemia. Emerging evidence suggests that RA is involved in the formation of the vasculature; however, its effect on endothelial cell angiogenesis and metabolism is unclear. METHODS Our study was designed to clarify the abovementioned effect with human embryonic stem cell-derived endothelial cells (hESC-ECs) employed as a cell model. RESULTS We found that RA inhibits angiogenesis, as manifested by decreased proliferation, migration and sprouting activity. RNA sequencing revealed general suppression of glycometabolism in hESC-ECs in response to RA, consistent with the decreased glycolytic activity and glucose uptake. After screening glycometabolism-related genes, we found that fructose-1,6-bisphosphatase 1 (FBP1), a key rate-limiting enzyme in gluconeogenesis, was significantly upregulated after RA treatment. After silencing or pharmacological inhibition of FBP1 in hESC-ECs, the capacity for angiogenesis was enhanced, and the inhibitory effect of RA was reversed. ChIP-PCR demonstrated that FBP1 is a target gene of RAR. When hESC-ECs were treated with the RAR inhibitor BMS493, FBP1 expression was decreased and the effect of RA on angiogenesis was partially blocked. CONCLUSIONS The inhibitory role of RA in glycometabolism and angiogenesis is RAR/FBP1 dependent, and FBP1 may be a novel therapeutic target for pathological angiogenesis.
Collapse
Affiliation(s)
- Zhuangzhuang Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Miao Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Xuechun Li
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Yuanyuan Tu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Chunyan Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Min Song
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Yong Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Ying Huang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Fengyue Ding
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Kaili Hao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Xinglong Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Xuan Ni
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Lina Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
9
|
Pouso MR, Cairrao E. Effect of retinoic acid on the neurovascular unit: A review. Brain Res Bull 2022; 184:34-45. [DOI: 10.1016/j.brainresbull.2022.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/08/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
|
10
|
Chen C, Ke L, Chan H, Chu C, Lee A, Lin K, Lee M, Hsiao P, Chen C, Shin S. Electronegative low-density lipoprotein of patients with metabolic syndrome induces pathogenesis of aorta through disruption of the stimulated by retinoic acid 6 cascade. J Diabetes Investig 2020; 11:535-544. [PMID: 31597015 PMCID: PMC7232312 DOI: 10.1111/jdi.13158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 10/01/2019] [Accepted: 10/06/2019] [Indexed: 12/26/2022] Open
Abstract
AIMS/INTRODUCTION Electronegative low-density lipoprotein (L5) is the most atherogenic fraction of low-density lipoprotein and is elevated in people with metabolic syndrome (MetS), whereas the retinol-binding protein 4 receptor (stimulated by retinoic acid 6 [STRA6]) cascade is disrupted in various organs of patients with obesity-related diseases. Our objective was to investigate whether L5 from MetS patients capably induces pathogenesis of aorta through disrupting the STRA6 cascade. MATERIAL AND METHODS We examined the in vivo and in vitro effects of L5 on the STRA6 cascade and aortic atherogenic markers. To investigate the role of this cascade on atherosclerotic formation, crbp1 transfection was carried out in vitro. RESULTS This study shows that L5 activates atherogenic markers (p38 mitogen-activated protein kinases, pSmad2 and matrix metallopeptidase 9) and simultaneously suppresses STRA6 signals (STRA6, cellular retinol-binding protein 1, lecithin-retinol acyltransferase, retinoic acid receptor-α and retinoid X receptor-α) in aortas of L5-injected mice and L5-treated human aortic endothelial cell lines and human aortic smooth muscle cell lines. These L5-induced changes of the STRA6 cascade and atherogenic markers were reversed in aortas of LOX1-/- mice and in LOX1 ribonucleic acid-silenced human aortic endothelial cell lines and human aortic smooth muscle cell lines. Furthermore, crbp1 gene transfection reversed the disruption of the STRA6 cascade, the phosphorylation of p38 mitogen-activated protein kinases and Smad2, and the elevation of matrix metallopeptidase 9 in L5-treated human aortic endothelial cell lines. CONCLUSIONS This study shows that L5 from MetS patients induces atherogenic markers by disrupting STRA6 signaling. Suppression of STRA6 might be one novel pathogenesis of aorta in patients with MetS.
Collapse
Affiliation(s)
- Chao‐Hung Chen
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Liang‐Yin Ke
- Lipid Science and Aging Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Medical Laboratory Science and BiotechnologyCollege of Health SciencesKaohsiung Medical UniversityKaohsiungTaiwan
| | - Hua‐Chen Chan
- Department of Medical Laboratory Science and BiotechnologyCollege of Health SciencesKaohsiung Medical UniversityKaohsiungTaiwan
| | - Chih‐Sheng Chu
- Division of CardiologyDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - An‐Sheng Lee
- Department of MedicineMackay Medical CollegeNew TaipeiTaiwan
| | - Kun‐Der Lin
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
- Vascular and Medical ResearchTexas Heart InstituteHoustonTexasUSA
| | - Mei‐Yueh Lee
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Pi‐Jung Hsiao
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Chu‐Huang Chen
- Lipid Science and Aging Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
- Vascular and Medical ResearchTexas Heart InstituteHoustonTexasUSA
- Department of Internal MedicineKaohsiung Ta‐Tung Municipal HospitalKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Shyi‐Jang Shin
- School of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Divison of Endocrinology and MetabolismDepartment of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
- Department of Medical Laboratory Science and BiotechnologyCollege of Health SciencesKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
11
|
Olsen T, Blomhoff R. Retinol, Retinoic Acid, and Retinol-Binding Protein 4 are Differentially Associated with Cardiovascular Disease, Type 2 Diabetes, and Obesity: An Overview of Human Studies. Adv Nutr 2020; 11:644-666. [PMID: 31868199 PMCID: PMC7231588 DOI: 10.1093/advances/nmz131] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/12/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
Vitamin A is a fat-soluble essential nutrient obtained from plant- and animal-based sources that has roles in growth, vision, and metabolism. Vitamin A circulates mainly as retinol bound to retinol-binding protein 4 (RBP4), and is delivered to tissues and converted to retinoic acid, which is a ligand for several nuclear receptors. In recent years, aspects of vitamin A metabolism have been under scrutiny with regards to the development of metabolic and lifestyle diseases including cardiovascular disease (CVD), type 2 diabetes mellitus (T2DM), and overweight and obesity in humans. Studies have mainly focused on RBP4 in this context, whereas the major circulating form, retinol, and the major bioactive form, retinoic acid, have been overlooked in this regard until recently. As one of the main roles of RBP4 is to deliver retinol to tissues for biological action, the associations of retinol and retinoic acid with these diseases must also be considered. In this review, we summarize and discuss recent and available evidence from human studies with focus on retinol, retinoic acid, and RBP4 and provide an overview of these crucial components of vitamin A metabolism in CVD, T2DM, and obesity. In summary, retinol was found to be both inversely and positively associated with CVD whereas the associations with T2DM and obesity were less clear. Although only a few studies have been published on retinoic acid, it was inversely associated with CVD. In contrast, serum RBP4 was mostly found to be positively associated with CVD, T2DM, and obesity. At present, it is difficult to ascertain why the reported associations differ depending on the compound under study, but there is a clear imbalance in the literature in disfavor of retinol and retinoic acid, which needs to be considered in future human studies.
Collapse
Affiliation(s)
- Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway,Address correspondence to TO (e-mail: )
| | - Rune Blomhoff
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway,Department of Clinical Service, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
Rogers MA, Chen J, Nallamshetty S, Pham T, Goto S, Muehlschlegel JD, Libby P, Aikawa M, Aikawa E, Plutzky J. Retinoids Repress Human Cardiovascular Cell Calcification With Evidence for Distinct Selective Retinoid Modulator Effects. Arterioscler Thromb Vasc Biol 2020; 40:656-669. [PMID: 31852220 PMCID: PMC7047603 DOI: 10.1161/atvbaha.119.313366] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Retinoic acid (RA) is a ligand for nuclear receptors that modulate gene transcription and cell differentiation. Whether RA controls ectopic calcification in humans is unknown. We tested the hypothesis that RA regulates osteogenic differentiation of human arterial smooth muscle cells and aortic valvular interstitial cells that participate in atherosclerosis and heart valve disease, respectively. Approach and Results: Human cardiovascular tissue contains immunoreactive RAR (RA receptor)-a retinoid-activated nuclear receptor directing multiple transcriptional programs. RA stimulation suppressed primary human cardiovascular cell calcification while treatment with the RAR inhibitor AGN 193109 or RARα siRNA increased calcification. RA attenuated calcification in a coordinated manner, increasing levels of the calcification inhibitor MGP (matrix Gla protein) while decreasing calcification-promoting TNAP (tissue nonspecific alkaline phosphatase) activity. Given that nuclear receptor action varies as a function of distinct ligand structures, we compared calcification responses to cyclic retinoids and the acyclic retinoid peretinoin. Peretinoin suppressed human cardiovascular cell calcification without inducing either secretion of APOC3 (apolipoprotein-CIII), which promotes atherogenesis, or reducing CYP7A1 (cytochrome P450 family 7 subfamily A member 1) expression, which occurred with cyclic retinoids all-trans RA, 9-cis RA, and 13-cis RA. Additionally, peretinoin did not suppress human femur osteoblast mineralization, whereas all-trans RA inhibited osteoblast mineralization. CONCLUSIONS These results establish retinoid regulation of human cardiovascular calcification, provide new insight into mechanisms involved in these responses, and suggest selective retinoid modulators, like acyclic retinoids may allow for treating cardiovascular calcification without the adverse effects associated with cyclic retinoids.
Collapse
MESH Headings
- Alkaline Phosphatase
- Aortic Valve/drug effects
- Aortic Valve/metabolism
- Aortic Valve/pathology
- Apolipoprotein C-III/genetics
- Apolipoprotein C-III/metabolism
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Carotid Arteries/drug effects
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cells, Cultured
- Cholesterol 7-alpha-Hydroxylase/genetics
- Cholesterol 7-alpha-Hydroxylase/metabolism
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Heart Valve Diseases/genetics
- Heart Valve Diseases/metabolism
- Heart Valve Diseases/pathology
- Heart Valve Diseases/prevention & control
- Humans
- Isotretinoin/pharmacology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Osteogenesis/drug effects
- Receptors, Retinoic Acid/agonists
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoids/pharmacology
- Retinoids/toxicity
- Signal Transduction
- Tretinoin/pharmacology
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/prevention & control
- Matrix Gla Protein
Collapse
Affiliation(s)
- Maximillian A. Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Jiaohua Chen
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Shriram Nallamshetty
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Tan Pham
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Shinji Goto
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Jochen D. Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Peter Libby
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| | - Jorge Plutzky
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, U.S.A
| |
Collapse
|
13
|
Nagpal I, Wei LN. All- trans Retinoic Acid as a Versatile Cytosolic Signal Modulator Mediated by CRABP1. Int J Mol Sci 2019; 20:ijms20153610. [PMID: 31344789 PMCID: PMC6696438 DOI: 10.3390/ijms20153610] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 12/12/2022] Open
Abstract
All-trans retinoic acid (AtRA), an active metabolite of vitamin A, is recognized for its classical action as an endocrine hormone that triggers genomic effects mediated through nuclear receptors RA receptors (RARs). New evidence shows that atRA-mediated cellular responses are biphasic with rapid and delayed responses. Most of these rapid atRA responses are the outcome of its binding to cellular retinoic acid binding protein 1 (CRABP1) that is predominantly localized in cytoplasm and binds to atRA with a high affinity. This review summarizes the most recent studies of such non-genomic outcomes of atRA and the role of CRABP1 in mediating such rapid effects in different cell types. In embryonic stem cells (ESCs), atRA-CRABP1 dampens growth factor sensitivity and stemness. In a hippocampal neural stem cell (NSC) population, atRA-CRABP1 negatively modulates NSC proliferation and affects learning and memory. In cardiomyocytes, atRA-CRABP1 prevents over-activation of calcium-calmodulin-dependent protein kinase II (CaMKII), protecting heart function. These are supported by the fact that CRABP1 gene knockout (KO) mice exhibit multiple phenotypes including hippocampal NSC expansion and spontaneous cardiac hypertrophy. This indicates that more potential processes/signaling pathways involving atRA-CRABP1 may exist, which remain to be identified.
Collapse
Affiliation(s)
- Isha Nagpal
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
14
|
Interscapular and Perivascular Brown Adipose Tissue Respond Differently to a Short-Term High-Fat Diet. Nutrients 2019; 11:nu11051065. [PMID: 31086124 PMCID: PMC6566556 DOI: 10.3390/nu11051065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
Brown adipose tissue (BAT) function may depend on its anatomical location and developmental origin. Interscapular BAT (iBAT) regulates acute macronutrient metabolism, whilst perivascular BAT (PVAT) regulates vascular function. Although phenotypically similar, whether these depots respond differently to acute nutrient excess is unclear. Given their distinct anatomical locations and developmental origins and we hypothesised that iBAT and PVAT would respond differently to brief period of nutrient excess. Sprague-Dawley rats aged 12 weeks (n=12) were fed either a standard (10% fat, n=6) or high fat diet (HFD: 45% fat, n=6) for 72h and housed at thermoneutrality. Following an assessment of whole body physiology, fat was collected from both depots for analysis of gene expression and the proteome. HFD consumption for 72h induced rapid weight gain (c. 2.6%) and reduced serum non-esterified fatty acids (NEFA) with no change in either total adipose or depot mass. In iBAT, an upregulation of genes involved in insulin signalling and lipid metabolism was accompanied by enrichment of lipid-related processes and functions, plus glucagon and peroxisome proliferator-activated receptor (PPAR) signalling pathways. In PVAT, HFD induced a pronounced down-regulation of multiple metabolic pathways which was accompanied with increased abundance of proteins involved in apoptosis (e.g., Hdgf and Ywaq) and toll-like receptor signalling (Ube2n). There was also an enrichment of DNA-related processes and functions (e.g., nucleosome assembly and histone exchange) and RNA degradation and cell adhesion pathways. In conclusion, we show that iBAT and PVAT elicit divergent responses to short-term nutrient excess highlighting early adaptations in these depots before changes in fat mass.
Collapse
|
15
|
Abstract
Much evidence has accumulated in the literature over the last fifteen years that indicates vitamin A has a role in metabolic disease prevention and causation. This literature proposes that vitamin A can affect obesity development and the development of obesity-related diseases including insulin resistance, type 2 diabetes, hepatic steatosis and steatohepatitis, and cardiovascular disease. Retinoic acid, the transcriptionally active form of vitamin A, accounts for many of the reported associations. However, a number of proteins involved in vitamin A metabolism, including retinol-binding protein 4 (RBP4) and aldehyde dehydrogenase 1A1 (ALDH1A1, alternatively known as retinaldehyde dehydrogenase 1 or RALDH1), have also been identified as being associated with metabolic disease. Some of the reported effects of these vitamin A-related proteins are proposed to be independent of their roles in assuring normal retinoic acid homeostasis. This review will consider both human observational data as well as published data from molecular studies undertaken in rodent models and in cells in culture. The primary focus of the review will be on the effects that vitamin A per se and proteins involved in vitamin A metabolism have on adipocytes, adipose tissue biology, and adipose-related disease, as well as on early stage liver disease, including non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH).
Collapse
Affiliation(s)
- William S Blaner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032.
| |
Collapse
|
16
|
Lee DY, Chiu JJ. Hemodynamics-Based Strategy of Using Retinoic Acid Receptor and Retinoid X Receptor Agonists to Induce MicroRNA-10a and Inhibit Atherosclerotic Lesion. Methods Mol Biol 2019; 2019:143-169. [PMID: 31359395 DOI: 10.1007/978-1-4939-9585-1_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The protocols in this chapter describe methods for identifying the functional roles of retinoic acid receptor (RAR) and retinoid X receptor (RXR) signaling in atherosclerosis and developing RARα/RXRα-specific agonists as hemodynamics-based therapeutic components for atherosclerosis treatment. In vitro cell culture flow system is used to elucidate the effects of different flow patterns and shear stresses, i.e., atherogenic oscillatory shear stress (OS) vs. atheroprotective pulsatile shear stress (PS), on RAR/RXR signaling and inflammatory responses in vascular endothelial cells (ECs). Western blotting, nuclear and cytoplasmic protein extraction, immunoprecipitation, and in situ proximity ligation assay are used to examine the expression, location, and association of RARs (i.e., RARα, RARβ, and RARγ) and RXRs (i.e., RXRα, RXRβ, and RXRγ) in ECs in response to OS vs. PS. Chromatin immunoprecipitation is used to examine the binding activity of RARα/RA-responsive elements (RARE). RT-microRNA (miR) quantitative real-time PCR and RT-PCR are used to detect the expressions of miR-10a and pro-inflammatory molecules, respectively. Specific siRNAs of RARα and RXRα, precursor miR-10a (PreR-10a), and antagomiR-10a (AMR-10a) are used to elucidate the regulatory roles of RARα, RXRα, and miR-10a in pro-inflammatory signaling in ECs. RARα/RXRα-specific agonists are used to induce miR-10a expression and inhibit OS-induced pro-inflammatory signaling in ECs in vitro. Apolipoprotein E-deficient (ApoE-/-) mice are used as an atherosclerotic animal model. Administration of ApoE-/- mice with RARα/RXRα-specific agonists results in inhibitions in atherosclerotic lesion formation. Co-administration of ApoE-/- mice with RARα/RXRα agonists and AMR-10a is performed to identify the role of miR-10a in RARα/RXRα agonists-mediated inhibition in atherosclerotic lesions. Oil Red O staining and H&E staining are used to examine the levels of atherosclerotic lesions in the vessel wall. In situ miR hybridization and immunohistochemical staining are used to detect the expression of miR-10a and pro-inflammatory molecules and the infiltration of inflammatory cells in the vessel wall. RARα/RXRα-specific agonists are used to mimic the atheroprotective effects of PS to induce endothelial miR-10a and hence repress OS-induced pro-inflammatory signaling and atherosclerotic lesion formation in vivo. The results indicate that RAR/RXR-specific agonists have great potential to be developed as hemodynamics-based therapeutic components for atherosclerosis treatment.
Collapse
Affiliation(s)
- Ding-Yu Lee
- Departments of Food Science and Biological Science and Technology, China University of Science and Technology, Taipei, Taiwan
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan.
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan.
- Institute of Biomedical Engineering, National Cheng-Kung University, Tainan, Taiwan.
- College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
17
|
Olsen T, Vinknes KJ, Svingen GFT, Pedersen ER, Dhar I, Tell GS, Blomhoff R, Ueland PM, Midttun Ø, Refsum H, Nygård OK. The risk association of plasma total homocysteine with acute myocardial infarction is modified by serum vitamin A. Eur J Prev Cardiol 2018; 25:1612-1620. [DOI: 10.1177/2047487318788587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Plasma total homocysteine (tHcy) has been implicated in the development of cardiovascular disease, but the mechanisms remain unclear. Vitamin A (Vit-A) is involved in homocysteine metabolism and we therefore explored the potential interaction between plasma tHcy and serum Vit-A in relation to incident acute myocardial infarction. Methods Cox proportional hazards models were used to assess the prospective relationships between tHcy and acute myocardial infarction in 2205 patients from Western Norway undergoing elective coronary angiography for suspected stable angina pectoris. Results are reported as hazard ratio per standard deviation increase in log-transformed tHcy. An interaction term for tHcy × Vit-A was added to multivariate models including age, sex, smoking, apolipoprotein B fasting, statin and aspirin prescription and estimated glomerular filtration rate. Results Geometric mean (geometric standard deviation) age of the participants (64.3% men) was 62.3 (1.24) years. Plasma tHcy was higher among participants in the upper versus lower Vit-A tertile. During 7 (2.4) years of follow-up, 15.1% suffered an AMI. A significant association of plasma tHcy with AMI in the total study population was observed. When we stratified the population according to Vit-A tertiles, plasma tHcy was associated with acute myocardial infarction only in the upper Vit-A tertile (hazard ratio per SD: 1.25, 95% confidence interval: 1.04–1.53, pinteraction = 0.03). Conclusions The risk relationship between plasma tHcy and acute myocardial infarction was modified by serum concentrations of Vit-A in patients with suspected stable angina pectoris. This finding may clarify the relationship between tHcy and cardiovascular disease.
Collapse
Affiliation(s)
- Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway
| | - Gard FT Svingen
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Eva R Pedersen
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
- KG Jebsen Centre for Diabetes Research, University of Bergen, Norway
| | - Indu Dhar
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Grethe S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Norway
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Bergen, Norway
| | - Rune Blomhoff
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway
- Department of Clinical Service, Division of Cancer Medicine, Oslo University Hospital, Norway
| | - Per M Ueland
- Laboratory of Clinical Biochemistry, Haukeland University Hospital, Bergen, Norway
- Bevital AS, Bergen, Norway
| | | | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway
| | - Ottar K Nygård
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
- KG Jebsen Centre for Diabetes Research, University of Bergen, Norway
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| |
Collapse
|
18
|
Gregory EK, Webb A, Vercammen JM, Kelly ME, Akar B, van Lith R, Bahnson EM, Jiang W, Ameer GA, Kibbe MR. Inhibiting intimal hyperplasia in prosthetic vascular grafts via immobilized all-trans retinoic acid. J Control Release 2018; 274:69-80. [PMID: 29391231 PMCID: PMC5847482 DOI: 10.1016/j.jconrel.2018.01.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/15/2017] [Accepted: 01/22/2018] [Indexed: 12/24/2022]
Abstract
Peripheral arterial disease is a leading cause of morbidity and mortality. The most commonly utilized prosthetic material for peripheral bypass grafting is expanded polytetrafluoroethylene (ePTFE) yet it continues to exhibit poor performance from restenosis due to neointimal hyperplasia, especially in femoral distal bypass procedures. Recently, we demonstrated that periadventitial delivery of all-trans retinoic acid (atRA) immobilized throughout porous poly(1,8 octamethylene citrate) (POC) membranes inhibited neointimal formation in a rat arterial injury model. Thus, the objective of this study was to investigate whether atRA immobilized throughout the lumen of ePTFE vascular grafts would inhibit intimal formation following arterial bypass grafting. Utilizing standard ePTFE, two types of atRA-containing ePTFE vascular grafts were fabricated and evaluated: grafts whereby all-trans retinoic acid was directly immobilized on ePTFE (atRA-ePTFE) and grafts where all-trans retinoic acid was immobilized onto ePTFE grafts coated with POC (atRA-POC-ePTFE). All grafts were characterized by SEM, HPLC, and FTIR and physical characteristics were evaluated in vitro. Modification of these grafts, did not significantly alter their physical characteristics or biocompatibility, and resulted in inhibition of intimal formation in a rat aortic bypass model, with atRA-POC-ePTFE inhibiting intimal formation at both the proximal and distal graft sections. In addition, treatment with atRA-POC-ePTFE resulted in increased graft endothelialization and decreased inflammation when compared to the other treatment groups. This work further confirms the biocompatibility and efficacy of locally delivered atRA to inhibit intimal formation in a bypass setting. Thus, atRA-POC-ePTFE grafts have the potential to improve patency rates in small diameter bypass grafts and warrant further investigation.
Collapse
Affiliation(s)
- Elaine K Gregory
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - Antonio Webb
- The University of Florida, Gainesville, FL 32611, United States
| | - Janet M Vercammen
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - Megan E Kelly
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - Banu Akar
- Biomedical Engineering Department, McCormick School of Engineering, Northwestern University, Evanston, IL 60201, United States
| | - Robert van Lith
- Biomedical Engineering Department, McCormick School of Engineering, Northwestern University, Evanston, IL 60201, United States; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - Edward M Bahnson
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Wulin Jiang
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - Guillermo A Ameer
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Biomedical Engineering Department, McCormick School of Engineering, Northwestern University, Evanston, IL 60201, United States; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States
| | - Melina R Kibbe
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, United States; Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
19
|
Prevention of intimal hyperplasia by immobilized all-trans retinoic acid. J Control Release 2018. [DOI: 10.1016/j.jconrel.2018.02.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
20
|
Tao L, Nie Y, Wang G, Ding Y, Ding J, Xiong F, Tang S, Wang Y, Zhou B, Zhu H. All‑trans retinoic acid reduces endothelin‑1 expression and increases endothelial nitric oxide synthase phosphorylation in rabbits with atherosclerosis. Mol Med Rep 2017; 17:2619-2625. [PMID: 29207193 DOI: 10.3892/mmr.2017.8156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 10/24/2017] [Indexed: 11/06/2022] Open
Abstract
All-trans retinoic acid (ATRA) is a natural derivative of vitamin A that ameliorates atherosclerosis (AS) by regulating inflammatory factors. However, studies concerning the role of retinoic acid in artery endothelial function are rare. Therefore, the present study investigated its role in regulating the production of endothelin‑1 (ET‑1) and nitric oxide (NO) in rabbits with AS. The rabbits were randomly divided into 3 groups: The control group was administered an ordinary diet, while the high fat group and the ATRA drug intervention group were administered a high fat diet. After 12 weeks, the blood lipid levels of rabbits, the morphological structure of the arterial wall, the arterial intimal permeability, the activity of blood endothelial nitric oxide synthase (eNOS) and the level of plasma NO were investigated. Western blot analysis was used to detect the levels of ET‑1, eNOS and eNOS phosphorylation at Ser‑1177 (p‑eNOS), and a radioimmunoassay was performed to detect the level of ET‑1 in the plasma. It was identified that plaque formation was alleviated in the ATRA group compared with the high fat group, as revealed by hematoxylin and eosin and oil red O staining, and a similar trend was reflected in the immunofluorescence results for endothelial permeability. Western blotting demonstrated significantly decreased ET‑1 expression levels in the arterial tissue of rabbits in the ATRA group compared with the high fat group, together with increased p‑eNOS level (P<0.05), however, no difference was observed in the expression of eNOS (P>0.05). The trends observed for ET‑1 and the activity of eNOS in plasma were similar to those for arterial tissue. Therefore, the present study demonstrated that ATRA may regulate the grade of AS by the reduction of ET‑1 secretion and increased NO formation via increased phosphorylation of eNOS. ATRA provides a potential novel method for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Linlin Tao
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yumei Nie
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Ganxian Wang
- Laboratory of Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yanhui Ding
- Laboratory of Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Junli Ding
- Laboratory of Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Fangyuan Xiong
- Laboratory of Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Songtao Tang
- Laboratory of Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuan Wang
- Laboratory of Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Birong Zhou
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Huaqing Zhu
- Laboratory of Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
21
|
Nordlohne J, von Vietinghoff S. Interleukin 17A in atherosclerosis - Regulation and pathophysiologic effector function. Cytokine 2017; 122:154089. [PMID: 28663097 DOI: 10.1016/j.cyto.2017.06.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/01/2017] [Accepted: 06/21/2017] [Indexed: 12/20/2022]
Abstract
This review summarizes the current data on the interleukin (IL)-17A pathway in experimental atherosclerosis and clinical data. IL-17A is a prominent cytokine for early T cell response produced by both innate and adaptive leukocytes. In atherosclerosis, increased total IL-17A levels and expression in CD4+ T helper and γδ T cells have been demonstrated. Cytokines including IL-6 and TGFβ that increase IL-17A expression are elevated. Many other factors such as lipids, glucose and sodium chloride concentrations as well as vitamins and arylhydrocarbon receptor agonists that promote IL-17A expression are closely associated with cardiovascular risk in the human population. In acute inflammation models, IL-17A mediates innate leukocyte recruitment of both neutrophils and monocytes. In atherosclerosis, IL-17A increased aortic macrophage and T cell infiltration in most models. Secondary recruitment effects via the endothelium and according to recent data also pericytes have been demonstrated. IL-17 receptor A is highly expressed on monocytes and direct effects have been reported as well. Beyond leukocyte accumulation, IL-17A may affect other factors of plaque formation such as endothelial function, and according to some reports, fibrous cap formation and vascular relaxation with an increase in blood pressure. Anti-IL-17A agents are now available for clinical use. Cardiovascular side effect profiles are benign at this point. IL-17A appears to be a differential regulator of atherosclerosis and its effects in mouse models suggest that its modulation may have contradictory effects on plaque size and possibly stability in different patient populations.
Collapse
Affiliation(s)
- Johannes Nordlohne
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Sibylle von Vietinghoff
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
22
|
Albuquerque MNDL, Diniz ADS, Arruda IKGD. Elevated Serum Retinol and Low Beta-Carotene but not Alpha-Tocopherol Concentrations Are Associated with Dyslipidemia in Brazilian Adolescents. J Nutr Sci Vitaminol (Tokyo) 2017; 62:73-80. [PMID: 27264090 DOI: 10.3177/jnsv.62.73] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The purpose of this study was to investigate the status of retinol, beta-carotene, and alpha-tocopherol serum concentrations in adolescents with dyslipidemia. A case series dyslipidemia study was conducted, with an attached control group, including 104 adolescents of public schools in Recife during the months of March/April 2013. Retinol, beta-carotene and alpha-tocopherol serum concentrations were analysed by high efficiency liquid chromatography. Sociodemographic, anthropometric, clinical and biochemical variables were analysed. Dyslipidemic adolescents had high serum concentrations of both retinol (p=0.007) and beta-carotene/apolipoprotein A-I ratio (p=0.034); they also had low concentrations of beta-carotene/total cholesterol (p<0.0001) and beta-carotene/apolipoprotein B ratios (p=0.033) when compared to the controls. The alpha-tocopherol serum status was not associated with dyslipidemia. Overweight, abdominal obesity, lipid profile markers, and systolic and diastolic blood pressures were more prevalent in dyslipidemic adolescents. The findings show an association between vitamin A and dyslipidemia in adolescents. However, additional investigations of this risk group are necessary to clarify the mechanisms of action of this nutrient in the pathogenesis of this syndrome, aiming at reducing cardiometabolic risks as of earlier ages.
Collapse
|
23
|
MicroRNA-10a is crucial for endothelial response to different flow patterns via interaction of retinoid acid receptors and histone deacetylases. Proc Natl Acad Sci U S A 2017; 114:2072-2077. [PMID: 28167758 DOI: 10.1073/pnas.1621425114] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Histone deacetylases (HDACs) and microRNAs (miRs) have emerged as two important epigenetic factors in the regulation of vascular physiology. This study aimed to elucidate the relationship between HDACs and miRs in the hemodynamic modulation of endothelial cell (EC) dysfunction. We found that miR-10a has the lowest expression among all examined shear-responsive miRs in ECs under oscillatory shear stress (OS), and a relatively high expression under pulsatile shear stress (PS). PS and OS alter EC miR-10a expression to regulate the expression of its direct target GATA6 and downstream vascular cell adhesion molecule (VCAM)-1. PS induces the expression, nuclear accumulation, and association of retinoid acid receptor-α (RARα) and retinoid X receptor-α (RXRα). RARα and RXRα serve as a "director" and an "enhancer," respectively, to enhance RARα binding to RA-responsive element (RARE) and hence miR-10a expression, thus down-regulating GATA6/VCAM-1 signaling in ECs. In contrast, OS induces associations of "repressors" HDAC-3/5/7 with RARα to inhibit the RARα-directed miR-10a signaling. The flow-mediated miR-10a expression is regulated by Krüppel-like factor 2 through modulation in RARα-RARE binding, with the consequent regulation in GATA6/VCAM-1 in ECs. These results are confirmed in vivo by en face staining on the aortic arch vs. the straight thoracic aorta of rats. Our findings identify a mechanism by which HDACs and RXRα modulate the hormone receptor RARα to switch miR-10a expression and hence the proinflammatory vs. anti-inflammatory responses of vascular endothelium under different hemodynamic forces.
Collapse
|
24
|
Photodecomposition, photomutagenicity and photocytotoxicity of retinyl palmitate under He-Ne laser photoirradiation and its effects on photodynamic therapy of cancer cells in vitro. Photodiagnosis Photodyn Ther 2015; 13:316-322. [PMID: 26365099 DOI: 10.1016/j.pdpdt.2015.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 08/06/2015] [Accepted: 09/08/2015] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Our aim was to study photodecomposition, photomutagenicity and cytotoxicity of retinyl palmitate (RP), a principal storage form of vitamin A in humans and animals, under He-Ne laser photoirradiation. Moreover, the effect of different concentrations and timing protocol of antioxidants on photodynamic therapy (PDT) is contradictory, so the effect of RP (as antioxidant) on the PDT cytotoxicity was studied. METHODS Photomutagenicity was tested by Ames test. Photodecomposition was studied by UV-vis spectroscopy. Cytotoxicity was measured with MTT-assay. Moreover, the effect of PDT, using hematoporphyrin derivatives (HpD) as photosensitizer under He-Ne laser irradiation (10 J/cm(2)), was studied on HeLa cells either with or without RP (1-100 μM) which incubated with the cells for short or long incubation period (1 h or 24 h) prior to PDT. RESULTS No photodecomposition of RP alone was obseved whereas there is a little photodecomposition of RP only in presence of HpD under irradiation with He-Ne laser. Moreover, no photomutagenicity was observed in Salmonella typhimurium strains under laser irradiation in presence or absence of HpD. RP alone (1-100 μM) significantly decrease the viability of HeLa cells. Laser irradiation of HeLa cells pre-incubated with RP alone for 24 h showed further significant decrease in viability of the cells. While RP incubations for 1 h before PDT had slight effect on the cells, 24 h incubation before PDT enhanced the cytotoxicity of PDT on HeLa cells. CONCLUSIONS RP can be used 24 h before PDT to enhance its effects. RP is not mutagenic under irradiation with He-Ne laser.
Collapse
|
25
|
Lee JY, Moon YJ, Lee HO, Park AK, Choi SA, Wang KC, Han JW, Joung JG, Kang HS, Kim JE, Phi JH, Park WY, Kim SK. Deregulation of Retinaldehyde Dehydrogenase 2 Leads to Defective Angiogenic Function of Endothelial Colony-Forming Cells in Pediatric Moyamoya Disease. Arterioscler Thromb Vasc Biol 2015; 35:1670-7. [PMID: 26023078 DOI: 10.1161/atvbaha.115.305363] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 05/06/2015] [Indexed: 01/07/2023]
Abstract
OBJECTIVE-- Moyamoya disease (MMD) is a common cause of childhood stroke, in which the abnormal function of the endothelial colony-forming cell (ECFC) plays a key role in the pathogenesis of the disease. This study was designed to identify genes involved in MMD pathogenesis using gene expression profiling and to understand the defective function of MMD ECFCs. APPROACH AND RESULTS-- We compared gene expression profiles of ECFCs isolated from patients with MMD and normal controls. Among the differentially expressed genes, we selected a gene with the most downregulated expression, retinaldehyde dehydrogenase 2 (RALDH2). The activity of RALDH2 in MMD ECFCs was assessed by in vitro tube formation assay and in vivo Matrigel plug assay in the presence of all-trans retinoic acid. The transcriptional control of RALDH2 was tested using ChIP assays on acetyl-histone H3. In the results, MMD ECFCs inefficiently formed capillary tubes in vitro and capillaries in vivo, a defect restored by all-trans retinoic acid treatment. Knockdown of RALDH2 mRNA in normal ECFCs also induced decreased activity of capillary formation in vitro. The decreased level of RALDH2 mRNA in MMD ECFCs was attributed to defective acetyl-histone H3 binding to the promoter region. CONCLUSIONS-- From these results, we conclude that the expression of RALDH2 was epigenetically suppressed in ECFCs from patients with MMD, which may play a key role in their functional impairment.
Collapse
Affiliation(s)
- Ji Yeoun Lee
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Youn Joo Moon
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Hae-Ock Lee
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Ae-Kyung Park
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Seung-Ah Choi
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Kyu-Chang Wang
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Jung Woo Han
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Je-Gun Joung
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Hyun Seung Kang
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Jeong Eun Kim
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Ji Hoon Phi
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.)
| | - Woong-Yang Park
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.).
| | - Seung-Ki Kim
- From the Department of Anatomy (J.Y.L.), Division of Pediatric Neurosurgery, Seoul National University Children's Hospital (J.Y.L., Y.J.M., S.-A.C., K.-C.W., J.W.H., J.H.P., S.-K.K.), and Department of Neurosurgery (H.S.K., J.E.K.), Seoul National University College of Medicine, Seoul, Korea; Samsung Genome Institute, Samsung Medical Center, Seoul, Korea (H.-O.L., J.-G.J., W.-Y.P.); College of Pharmacy, Sunchon National University, Jeonnam, Korea (A.-K.P.); and Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea (H.-O.L., W.-Y.P.).
| |
Collapse
|
26
|
Role of Retinoic Acid-Metabolizing Cytochrome P450s, CYP26, in Inflammation and Cancer. ADVANCES IN PHARMACOLOGY 2015; 74:373-412. [PMID: 26233912 DOI: 10.1016/bs.apha.2015.04.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vitamin A (retinol) and its active metabolite, all-trans-retinoic acid (atRA), play critical roles in regulating the differentiation, growth, and migration of immune cells. Similarly, as critical signaling molecules in the regulation of the cell cycle, retinoids are important in cancers. Concentrations of atRA are tightly regulated in tissues, predominantly by the availability of retinol, synthesis of atRA by ALDH1A enzymes and metabolism and clearance of atRA by CYP26 enzymes. The ALDH1A and CYP26 enzymes are expressed in several cell types in the immune system and in cancer cells. In the immune system, the ALDH1A and CYP26 enzymes appear to modulate RA concentrations. Consequently, alterations in the activity of ALDH1A and CYP26 enzymes are expected to change disease outcomes in inflammation. There is increasing evidence from various disease models of intestinal and skin inflammation that treatment with atRA has a positive effect on disease markers. However, whether aberrant atRA concentrations or atRA synthesis and metabolism play a role in inflammatory disease development and progression is not well understood. In cancers, especially in acute promyelocytic leukemia and neuroblastoma, increasing intracellular concentrations of atRA appears to provide clinical benefit. Inhibition of the CYP26 enzymes to increase atRA concentrations and combat therapy resistance has been pursued as a drug target in these cancers. This chapter covers the current knowledge of how atRA and retinol regulate the immune system and inflammation, how retinol and atRA metabolism is altered in inflammation and cancer, and what roles atRA-metabolizing enzymes have in immune responses and cancers.
Collapse
|
27
|
Zhao W, Smith JA, Mao G, Fornage M, Peyser PA, Sun YV, Turner ST, Kardia SLR. The cis and trans effects of the risk variants of coronary artery disease in the Chr9p21 region. BMC Med Genomics 2015; 8:21. [PMID: 25958224 PMCID: PMC4432789 DOI: 10.1186/s12920-015-0094-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/24/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Recent genome-wide association studies (GWAS) have shown that single nucleotide polymorphisms (SNPs) in the Chr9p21 region are associated with coronary artery disease (CAD). Most of the SNPs identified in this region are non-coding SNPs, suggesting that they may influence gene expression by cis or trans mechanisms to affect disease susceptibility. Since all cells from an individual have the same DNA sequence variations, levels of gene expression in immortalized cell lines can reflect the functional effects of DNA sequence variations that influence or regulate gene expression. The objective of this study is to evaluate the functional consequences of the risk variants in the Chr9p21 region on gene expression. METHODS We examined the association between the variants in the Chr9p21 region and the transcript-level mRNA expression of the adjacent genes (cis) as well as all other genes across the whole genome (trans) from transformed beta-lymphocytes in 801 non-Hispanic white participants from The Genetic Epidemiology Network of Arteriopathy (GENOA) study. RESULTS We found that the CAD risk variants in the Chr9p21 region were significantly associated with the mRNA expression of the ANRIL transcript ENST00000428597 (p = 8.58e-06). Importantly, a few distant transcripts were also found to be associated with the variants in this region, including the well-known CAD risk gene ABCA1 (p = 1.01e-05). Gene enrichment testing suggests that retinol metabolism, N-Glycan biosynthesis, and TGF signaling pathways may be involved. CONCLUSION These results suggest that the effect of risk variants in the Chr9p21 region on susceptibility to CAD is likely to be mediated through both cis and trans mechanisms.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Guangmei Mao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Myriam Fornage
- Institute of Molecular Medicine and Human Genetics Center, University of Texas Health Science Center, Houston, TX, USA.
| | - Patricia A Peyser
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Kraus BJ, Sartoretto JL, Polak P, Hosooka T, Shiroto T, Eskurza I, Lee SA, Jiang H, Michel T, Kahn BB. Novel role for retinol-binding protein 4 in the regulation of blood pressure. FASEB J 2015; 29:3133-40. [PMID: 25911613 DOI: 10.1096/fj.14-266064] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/31/2015] [Indexed: 12/23/2022]
Abstract
Elevated levels of serum retinol-binding protein 4 (RBP4) contribute to insulin resistance and correlate with increased prevalence of hypertension and myocardial infarction. We sought to determine whether lowering RBP4 would improve blood pressure (BP) and protect against obesity- or angiotensin (Ang)-II-induced hypertension. Systolic and diastolic BP were lower in the RBP4-knockout (RBP4-KO) mice and higher in the RBP4-overexpressing (RBP4-Tg) mice compared with BP in the wild-type (WT) littermates. Carbachol-induced vasodilatation was increased in arteries from the RBP4-KO compared with the WT mice and was impaired in the RBP4-Tg mice. Aortic eNOS(Ser1177) phosphorylation was enhanced ∼50% in the RBP4-KO mice, with no change in total eNOS protein. Feeding a high-fat diet increased BP in the RBP4-KO mice only to the level in the WT mice fed chow and had no effect on aortic eNOS(Ser1177) phosphorylation. Ang-II infusion resulted in 22 mmHg lower systolic BP in the RBP4-KO than in the WT mice, although the relative BP increase over saline infusion was ∼30% in both. Ang-II treatment decreased aortic eNOS(Ser1177) phosphorylation in the WT and RBP4-KO mice, but phosphorylation remained higher in the RBP4-KO mice. Cardiac hypertrophy with Ang-II treatment was diminished by 56% in the RBP4-KO mice. Thus, elevated serum RBP4 raises BP and lack of RBP4 reduces it, with commensurate changes in aortic eNOS(Ser1177) phosphorylation. Lowering RBP4 may reduce BP through enhanced eNOS-mediated vasodilatation and may be a novel therapeutic approach for hypertension.
Collapse
Affiliation(s)
- Bettina J Kraus
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Juliano L Sartoretto
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Pazit Polak
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Tetsuya Hosooka
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Takashi Shiroto
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Iratxe Eskurza
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Seung-Ah Lee
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Hongfeng Jiang
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Thomas Michel
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Barbara B Kahn
- *Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Cardiovascular Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
29
|
Vitamin A-deficient diet accelerated atherogenesis in apolipoprotein E(-/-) mice and dietary β-carotene prevents this consequence. BIOMED RESEARCH INTERNATIONAL 2015; 2015:758723. [PMID: 25802864 PMCID: PMC4352738 DOI: 10.1155/2015/758723] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 11/25/2014] [Accepted: 12/03/2014] [Indexed: 12/26/2022]
Abstract
Vitamin A is involved in regulation of glucose concentrations, lipid metabolism, and inflammation, which are major risk factors for atherogenesis. However, the effect of vitamin A deficiency on atherogenesis has not been investigated. Therefore, the objective of the current study was to examine whether vitamin A deficiency accelerates atherogenesis in apolipoprotein E-deficient mice (apoE−/−). ApoE−/− mice were allocated into the following groups: control, fed vitamin A-containing chow diet; BC, fed chow diet fortified with Dunaliella powder containing βc isomers; VAD, fed vitamin A-deficient diet; and VAD-BC group, fed vitamin A-deficient diet fortified with a Dunaliella powder. Following 15 weeks of treatment, liver retinol concentration had decreased significantly in the VAD group to about 30% that of control group. Vitamin A-deficient diet significantly increased both plasma cholesterol concentrations and the atherosclerotic lesion area at the aortic sinus (+61%) compared to the control group. Dietary βc fortification inhibited the elevation in plasma cholesterol and retarded atherogenesis in mice fed the vitamin A-deficient diet. The results imply that dietary vitamin A deficiency should be examined as a risk factor for atherosclerosis and that dietary βc, as a sole source of retinoids, can compensate for vitamin A deficiency.
Collapse
|
30
|
The inhibition of macrophage foam cell formation by 9-cis β-carotene is driven by BCMO1 activity. PLoS One 2015; 10:e0115272. [PMID: 25629601 PMCID: PMC4309590 DOI: 10.1371/journal.pone.0115272] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/23/2014] [Indexed: 01/04/2023] Open
Abstract
Atherosclerosis is a major cause of morbidity and mortality in developed societies, and begins when activated endothelial cells recruit monocytes and T-cells from the bloodstream into the arterial wall. Macrophages that accumulate cholesterol and other fatty materials are transformed into foam cells. Several epidemiological studies have demonstrated that a diet rich in carotenoids is associated with a reduced risk of heart disease; while previous work in our laboratory has shown that the 9-cis β-carotene rich alga Dunaliella inhibits atherogenesis in mice. The effect of 9-cis β-carotene on macrophage foam cell formation has not yet been investigated. In the present work, we sought to study whether the 9-cis β-carotene isomer, isolated from the alga Dunaliella, can inhibit macrophage foam cell formation upon its conversion to retinoids. The 9-cis β-carotene and Dunaliella lipid extract inhibited foam cell formation in the RAW264.7 cell line, similar to 9-cis retinoic acid. Furthermore, dietary enrichment with the algal powder in mice resulted in carotenoid accumulation in the peritoneal macrophages and in the inhibition of foam cell formation ex-vivo and in-vivo. We also found that the β-carotene cleavage enzyme β-carotene 15,15’-monooxygenase (BCMO1) is expressed and active in macrophages. Finally, 9-cis β-carotene, as well as the Dunaliella extract, activated the nuclear receptor RXR in hepa1-6 cells. These results indicate that dietary carotenoids, such as 9-cis β-carotene, accumulate in macrophages and can be locally cleaved by endogenous BCMO1 to form 9-cis retinoic acid and other retinoids. Subsequently, these retinoids activate the nuclear receptor RXR that, along with additional nuclear receptors, can affect various metabolic pathways, including those involved in foam cell formation and atherosclerosis.
Collapse
|
31
|
Schooling CM, Jones HE. Could child vitamin A supplementation have long-term health effects? Int J Epidemiol 2015; 44:365-6. [DOI: 10.1093/ije/dyu278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
32
|
Mottaghi A, Ebrahimof S, Angoorani P, Saboor-Yaraghi AA. Vitamin A Supplementation Reduces IL-17 and RORc Gene Expression in Atherosclerotic Patients. Scand J Immunol 2014; 80:151-7. [DOI: 10.1111/sji.12190] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 04/24/2014] [Indexed: 01/12/2023]
Affiliation(s)
- A. Mottaghi
- Obesity Research Center; Nutrition and Endocrine Research Center; Research Institute of Endocrine Sciences; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - S. Ebrahimof
- Students' Research Committee; Faculty of Nutrition and Food Technology; National Nutrition and Food Technology Research Institute; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - P. Angoorani
- Faculty of Nutrition and Food Technology; National Nutrition and Food Technology Research Institute; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - A.-A. Saboor-Yaraghi
- Department of Cellular and Molecular Nutrition; School of Nutrition and Dietetics; Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
33
|
Prevention of atherosclerosis progression by 9-cis-β-carotene rich alga Dunaliella in apoE-deficient mice. BIOMED RESEARCH INTERNATIONAL 2013; 2013:169517. [PMID: 24175283 PMCID: PMC3794645 DOI: 10.1155/2013/169517] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 08/23/2013] [Indexed: 12/31/2022]
Abstract
Introduction. β-Carotene-rich diet has been shown to be inversely associated with the risk of coronary heart disease. However, clinical trials using synthetic all-trans-β-carotene failed to demonstrate a beneficial effect. We therefore sought to study the effect of natural source of β-carotene, the alga Dunaliella, containing both all-trans and 9-cis-β-carotene on atherosclerosis. In a previous study we showed that 9-cis-β-carotene-rich powder of the alga Dunaliella inhibits early atherogenesis in low-density lipoprotein receptor knockout mice. Aims. The aims of the current work were to study whether diet enriched with Dunaliella powder would inhibit the progression of established atherosclerosis in old male apoE-deficient mice and to compare the effect of Dunaliella on lipid profile and atherosclerosis in a low-versus high-fat diet fed mice. Methods. In the first experiment, young mice (12 weeks old) were allocated into 3 groups: (1) low-fat diet; (2) low-fat diet + Dunaliella powder (8%); (3) low-fat diet + β-carotene-deficient Dunaliella. In the second experiment, old mice (7 months old) with established atherosclerotic lesions were allocated into 4 groups: (1) low-fat diet; (2) low-fat diet + Dunaliella; (3) high fat-diet; (4) high-fat diet + Dunaliella. Results. In young mice fed a low-fat diet, a trend toward lower atherosclerotic lesion area in the aortic sinus was found in the Dunaliella group compared with the control group. In old mice with established atherosclerotic lesion, Dunaliella inhibited significantly plasma cholesterol elevation and atherosclerosis progression in mice fed a high-fat diet. Conclusion. The results of this study suggest that a diet containing natural carotenoids, rich in 9-cis-β-carotene, has the potential to inhibit atherosclerosis progression, particularly in high-fat diet regime.
Collapse
|
34
|
Kurakula K, Hamers AAJ, de Waard V, de Vries CJM. Nuclear Receptors in atherosclerosis: a superfamily with many 'Goodfellas'. Mol Cell Endocrinol 2013; 368:71-84. [PMID: 22664910 DOI: 10.1016/j.mce.2012.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 05/23/2012] [Accepted: 05/25/2012] [Indexed: 01/07/2023]
Abstract
Nuclear Receptors form a superfamily of 48 transcription factors that exhibit a plethora of functions in steroid hormone signaling, regulation of metabolism, circadian rhythm and cellular differentiation. In this review, we describe our current knowledge on the role of Nuclear Receptors in atherosclerosis, which is a multifactorial disease of the vessel wall. Various cell types are involved in this chronic inflammatory pathology in which multiple cellular processes and numerous genes are dysregulated. Systemic risk factors for atherosclerosis are among others adverse blood lipid profiles, enhanced circulating cytokine levels, as well as increased blood pressure. Since many Nuclear Receptors modulate lipid profiles or regulate blood pressure they indirectly affect atherosclerosis. In the present review, we focus on the functional involvement of Nuclear Receptors within the atherosclerotic vessel wall, more specifically on their modulation of cellular functions in endothelial cells, smooth muscle cells and macrophages. Collectively, this overview shows that most of the Nuclear Receptors are athero-protective in atherosclerotic lesions.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
35
|
Alberts-Grill N, Denning TL, Rezvan A, Jo H. The role of the vascular dendritic cell network in atherosclerosis. Am J Physiol Cell Physiol 2013; 305:C1-21. [PMID: 23552284 DOI: 10.1152/ajpcell.00017.2013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A complex role has been described for dendritic cells (DCs) in the potentiation and control of vascular inflammation and atherosclerosis. Resident vascular DCs are found in the intima of atherosclerosis-prone vascular regions exposed to disturbed blood flow patterns. Several phenotypically and functionally distinct vascular DC subsets have been described. The functional heterogeneity of these cells and their contributions to vascular homeostasis, inflammation, and atherosclerosis are only recently beginning to emerge. Here, we review the available literature, characterizing the origin and function of known vascular DC subsets and their important role contributing to the balance of immune activation and immune tolerance governing vascular homeostasis under healthy conditions. We then discuss how homeostatic DC functions are disrupted during atherogenesis, leading to atherosclerosis. The effectiveness of DC-based "atherosclerosis vaccine" therapies in the treatment of atherosclerosis is also reviewed. We further provide suggestions for distinguishing DCs from macrophages and discuss important future directions for the field.
Collapse
Affiliation(s)
- Noah Alberts-Grill
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | | | | | |
Collapse
|
36
|
Mottaghi A, Salehi E, Keshvarz A, Sezavar H, Saboor-Yaraghi AA. The influence of vitamin A supplementation on Foxp3 and TGF-β gene expression in atherosclerotic patients. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2013; 5:314-26. [PMID: 23363776 DOI: 10.1159/000341916] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/17/2012] [Indexed: 12/17/2022]
Abstract
The aim of this study was to investigate the role of vitamin A in Foxp3 and TGF-β gene expression in atherosclerotic patients. Patients and healthy controls in the vitamin A group received 25,000 IU retinyl palmitate per day, while patients in the placebo group took one capsule of placebo per day for 4 months. Gene expressions of regulatory T cells were studied by real-time PCR. The levels of Foxp3 expression in phytohemagglutinin-activated cells were much higher in the patients who received vitamin A than in placebo-treated patients and healthy controls, while Foxp3 gene expression in oxidized low-density lipoprotein-activated cells showed no significant differences between all groups (p=0.357). A significant difference in the expression level of TGF-β gene in fresh cells was observed between patients and healthy controls (p=0.009). TGF-β gene expression in oxidized low-density lipoprotein-activated cells increased in all groups; however, these changes were not statistically significant (p=0.65); the changes obtained were 2.8-, 2.2- and 3.9-fold in the vitamin A, placebo, and control groups, respectively. Based on suppressing actions of regulatory T cells on effector T cells and findings that show that vitamin A has the effect of increasing expression of regulatory T cells, it can be concluded that supplementation with vitamin A in atherosclerotic patients may be effective in slowing disease progression.
Collapse
Affiliation(s)
- Azadeh Mottaghi
- Department of Cellular and Molecular Nutrition, School of Nutrition and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | |
Collapse
|
37
|
Huk DJ, Hammond HL, Kegechika H, Lincoln J. Increased dietary intake of vitamin A promotes aortic valve calcification in vivo. Arterioscler Thromb Vasc Biol 2012. [PMID: 23202364 DOI: 10.1161/atvbaha.112.300388] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Calcific aortic valve disease (CAVD) is a major public health problem with no effective treatment available other than surgery. We previously showed that mature heart valves calcify in response to retinoic acid (RA) treatment through downregulation of the SRY transcription factor Sox9. In this study, we investigated the effects of excess vitamin A and its metabolite RA on heart valve structure and function in vivo and examined the molecular mechanisms of RA signaling during the calcification process in vitro. METHODS AND RESULTS Using a combination of approaches, we defined calcific aortic valve disease pathogenesis in mice fed 200 IU/g and 20 IU/g of retinyl palmitate for 12 months at molecular, cellular, and functional levels. We show that mice fed excess vitamin A develop aortic valve stenosis and leaflet calcification associated with increased expression of osteogenic genes and decreased expression of cartilaginous markers. Using a pharmacological approach, we show that RA-mediated Sox9 repression and calcification is regulated by classical RA signaling and requires both RA and retinoid X receptors. CONCLUSIONS Our studies demonstrate that excess vitamin A dietary intake promotes heart valve calcification in vivo. Therefore suggesting that hypervitaminosis A could serve as a new risk factor of calcific aortic valve disease in the human population.
Collapse
Affiliation(s)
- Danielle J Huk
- Center for Cardiovascular and Pulmonary Research, Columbus, OH, USA
| | | | | | | |
Collapse
|
38
|
Rydén M, Garvin P, Kristenson M, Leanderson P, Ernerudh J, Jonasson L. Provitamin A carotenoids are independently associated with matrix metalloproteinase-9 in plasma samples from a general population. J Intern Med 2012; 272:371-84. [PMID: 22486775 DOI: 10.1111/j.1365-2796.2012.2534.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIM Carotenoids in plasma are inversely associated with cardiovascular risk. Low levels can be explained by low dietary intake but also by a number of other factors including inflammatory activity. Given that matrix metalloproteinase (MMP)-9 has an important role in inflammation and cardiovascular disease, we hypothesized that circulating MMP-9 levels would be inversely related to total or single carotenoids in a general population cohort. METHODS A well-characterized population-based cohort of 285 Swedish men and women (45-69 years) was used for the present study. The intake of carotenoid-rich fruits and vegetables was estimated from a food frequency questionnaire. Levels of MMP-9, C-reactive protein (CRP), interleukin (IL)-6 and six major carotenoids [β-cryptoxanthine, α-carotene, β-carotene, lutein (+zeaxanthin) and lycopene] were determined in plasma. RESULTS Lower plasma levels of total and single carotenoids were associated with lower dietary intake of carotenoids, older age, male sex, lower physical activity, higher alcohol consumption, higher body mass index (BMI), higher systolic and diastolic blood pressures, lower levels of total cholesterol and HDL cholesterol and higher levels of CRP, IL-6 and MMP-9. After multivariate adjustments, plasma levels of total carotenoids and provitamin A carotenoids (β-cryptoxanthine, α-carotene and β-carotene) remained independently associated with sex, dietary intake of carotenoids, BMI, HDL cholesterol and MMP-9, whilst associations with CRP and IL-6 were not maintained. Neither dietary intake of carotenoid-rich fruits and vegetables, nor vitamin supplement use was associated with MMP-9, CRP or IL-6 levels. CONCLUSION Plasma carotenoids were associated with a variety of factors including age, sex, dietary intake and metabolic variables. A new finding was the independent relationship in plasma between low provitamin A carotenoids and high MMP-9, suggesting a link between these carotenoids, matrix turnover and arterial remodelling.
Collapse
Affiliation(s)
- M Rydén
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | | | | | | | | | | |
Collapse
|
39
|
Elmabsout AA, Kumawat A, Saenz-Méndez P, Krivospitskaya O, Sävenstrand H, Olofsson PS, Eriksson LA, Strid A, Valen G, Törmä H, Sirsjö A. Cloning and functional studies of a splice variant of CYP26B1 expressed in vascular cells. PLoS One 2012; 7:e36839. [PMID: 22666329 PMCID: PMC3362586 DOI: 10.1371/journal.pone.0036839] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 04/09/2012] [Indexed: 11/18/2022] Open
Abstract
Background All-trans retinoic acid (atRA) plays an essential role in the regulation of gene expression, cell growth and differentiation and is also important for normal cardiovascular development but may in turn be involved in cardiovascular diseases, i.e. atherosclerosis and restenosis. The cellular atRA levels are under strict control involving several cytochromes P450 isoforms (CYPs). CYP26 may be the most important regulator of atRA catabolism in vascular cells. The present study describes the molecular cloning, characterization and function of atRA-induced expression of a spliced variant of the CYP26B1 gene. Methodology/Principal Findings The coding region of the spliced CYP26B1 lacking exon 2 was amplified from cDNA synthesized from atRA-treated human aortic smooth muscle cells and sequenced. Both the spliced variant and full length CYP26B1 was found to be expressed in cultured human endothelial and smooth muscle cells, and in normal and atherosclerotic vessel. atRA induced both variants of CYP26B1 in cultured vascular cells. Furthermore, the levels of spliced mRNA transcript were 4.5 times higher in the atherosclerotic lesion compared to normal arteries and the expression in the lesions was increased 20-fold upon atRA treatment. The spliced CYP26B1 still has the capability to degrade atRA, but at an initial rate one-third that of the corresponding full length enzyme. Transfection of COS-1 and THP-1 cells with the CYP26B1 spliced variant indicated either an increase or a decrease in the catabolism of atRA, probably depending on the expression of other atRA catabolizing enzymes in the cells. Conclusions/Significance Vascular cells express the spliced variant of CYP26B1 lacking exon 2 and it is also increased in atherosclerotic lesions. The spliced variant displays a slower and reduced degradation of atRA as compared to the full-length enzyme. Further studies are needed, however, to clarify the substrate specificity and role of the CYP26B1 splice variant in health and disease.
Collapse
Affiliation(s)
- Ali Ateia Elmabsout
- Department of Clinical Medicine, School of Health Sciences, Örebro University, Örebro, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|