1
|
Lin S, Wang L, Jia Y, Sun Y, Qiao P, Quan Y, Liu J, Hu H, Yang B, Zhou H. Lipin-1 deficiency deteriorates defect of fatty acid β-oxidation and lipid-related kidney damage in diabetic kidney disease. Transl Res 2024; 266:1-15. [PMID: 37433392 DOI: 10.1016/j.trsl.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/13/2023]
Abstract
Diabetic lipo-toxicity is a fundamental pathophysiologic mechanism in DM and is now increasingly recognized a key determinant of DKD. Targeting lipid metabolic disorders is an important therapeutic strategy for the treatment of DM and its complications, including DKD. This study aimed to explore the molecular mechanism of lipid metabolic regulation in kidney, especially renal PTECs, and elucidate the role of lipid metabolic related molecule lipin-1 in diabetic lipid-related kidney damage. In this study, lipin-1-deficient db/db mouse model and STZ/HFD-induced T2DM mouse model were used to determine the effect of lipin-1 on DKD development. Then RPTCs and LPIN1 knockdown or overexpressed HK-2 cells induced by PA were used to investigate the mechanism. We found that the expression of lipin-1 increased early and then decreased in kidney during the progression of DKD. Glucose and lipid metabolic disorders and renal insufficiency were found in these 2 types of diabetic mouse models. Interestingly, lipin-1 deficiency might be a pathogenic driver of DKD-to-CKD transition, which could further accelerate the imbalance of renal lipid homeostasis, the dysfunction of mitochondrial and energy metabolism in PTECs. Mechanistically, lipin-1 deficiency resulted in aggravated PTECs injury to tubulointerstitial fibrosis in DKD by downregulating FAO via inhibiting PGC-1α/PPARα mediated Cpt1α/HNF4α signaling and upregulating SREBPs to promote fat synthesis. This study provided new insights into the role of lipin-1 as a regulator for maintaining lipid homeostasis in the kidney, especially PTECs, and its deficiency led to the progression of DKD.
Collapse
Affiliation(s)
- Simei Lin
- Department of Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Liang Wang
- Department of Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yingli Jia
- Department of Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Panshuang Qiao
- Department of Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yazhu Quan
- Department of Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jihan Liu
- Department of Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Huihui Hu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China
| | - Baoxue Yang
- Department of Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Hong Zhou
- Department of Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.
| |
Collapse
|
2
|
Thakuri LS, Park CM, Kim HA, Kim HJ, Park JW, Park JC, Rhyu DY. Gracilaria chorda subcritical water ameliorates hepatic lipid accumulation and regulates glucose homeostasis in a hepatic steatosis cell model and obese C57BL/6J mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117395. [PMID: 37952731 DOI: 10.1016/j.jep.2023.117395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/03/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Red seaweed, known as Rhodophyta, has a long history of use in traditional Asian medicine, including Traditional Chinese Medicine and Ayurveda. It is believed to have cooling and detoxification properties. Red seaweed species, such as Gracilaria, have been used in traditional remedies to address various conditions, such as inflammation, thyroid disorders, and digestive issues. AIM OF THE STUDY Obesity is a risk factor of hepatic steatosis, a hallmark of non-alcoholic fatty liver disease (NAFLD) that affects nearly 25% of the worldwide population. Gracilaria chorda (GC) contains bioactive peptides that may be applicable in the prevention of metabolic syndrome diseases. This study investigated the effects of GC subcritical water extract at 210 °C (GCSW210) on preventing liver injury and lipid and glucose dysregulation in an oleic acid (OA)-induced hepatic steatosis cell model (HepG2) and high-fat diet (HFD)-induced obese animal model (C57BL/6J mice). MATERIALS AND METHODS Human hepatoma HepG2 cells were exposed to 0.1 mM OA for 24 h to induce hepatic steatosis and C57BL/6J mice were fed a HFD for 13 weeks. For lipid accumulation, triglyceride (TG) content was measured in both models, along with free fatty acid (FFA), plasma glucose, and insulin levels in HFD-fed mice. Protein expression of master regulators of adipogenesis and lipogenesis, as well as cholesterol and mitochondrial biosynthesis, was studied via western blotting in hepatic steatosis-induced in vitro and in vivo models. In addition, protein expression of the insulin signaling cascade in skeletal muscle tissues of HFD-fed mice was studied. RESULTS GCSW210 significantly decreased lipid accumulation in HepG2 cells exposed to OA and suppressed the expression of lipogenic factors, such as sterol regulatory element-binding protein (SREBP)-1c and fatty acid synthase. In addition, GCSW210 abrogated transcription factors related to cholesterol biosynthesis, such as SREBP-2 and low-density lipoprotein receptor. Similarly, FFA, TG, serum glutamic acid, aspartate transaminase, alanine transferase, plasma glucose, and insulin levels were also significantly reduced in GCSW210-treated HFD-fed mice, which were comparable to the positive control mice treated with Garcinia cambogia extract. Additionally, GCSW210 enhanced the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase in the hepatic tissues of HFD-fed mice. Moreover, GCSW210 treatment improved insulin signal transduction by reducing insulin receptor substrate 1 Ser307 phosphorylation and elevated phosphatidylinositol 3-kinase/protein kinase B and glucose transporter type 4 protein expression in muscle tissue. 5-Hdroxymethylfufural (5-HMF) was confirmed to be active substances isolated from GCSW210 through LC-PDA and LC-MS. CONCLUSIONS GCSW210 significantly regulated glucose metabolism, alleviated insulin resistance (IR) induced by high fatty acid synthesis and lipid accumulation, and elevated de novo lipogenesis by activating AMPK phosphorylation in both the liver and muscle tissues of HFD-fed mice. GCSW210 may be a potential functional food for preventing HFD-induced metabolic diseases, such as IR, NAFLD, and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Laxmi Sen Thakuri
- Department of Nutraceutical Resources, Mokpo National University, Jeonnam 58554, Korea; Department of Biomedicine, Health & Life Convergence Sciences, BK21 FOUR, Mokpo National University, Jeonnam 58554, Korea
| | - Chul Min Park
- Department of Nutraceutical Resources, Mokpo National University, Jeonnam 58554, Korea; Division of Practical Research, Honam National Institute of Biological Resources, Jeonam 58762, Korea
| | - Hyeon-A Kim
- Department of Food and Nutrition, Mokpo National University, Jeonnam 58554, Korea
| | - Hyung Jung Kim
- Department of Pharmacy, Mokpo National University, Jeonnam 58554, Korea
| | - Jin Woo Park
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 FOUR, Mokpo National University, Jeonnam 58554, Korea; Department of Pharmacy, Mokpo National University, Jeonnam 58554, Korea
| | - Jong Cheol Park
- Department of Oriental Medicine Resources, Sunchon National University, Jeonnam 57922, Korea; Chunsu Mountain Medicinal Herb Research Association, Gyeonggi-do 13637, Korea
| | - Dong Young Rhyu
- Department of Nutraceutical Resources, Mokpo National University, Jeonnam 58554, Korea; Department of Biomedicine, Health & Life Convergence Sciences, BK21 FOUR, Mokpo National University, Jeonnam 58554, Korea.
| |
Collapse
|
3
|
Beccacece L, Abondio P, Giorgetti A, Bini C, Pelletti G, Luiselli D, Pelotti S. A Genome-Wide Analysis of a Sudden Cardiac Death Cohort: Identifying Novel Target Variants in the Era of Molecular Autopsy. Genes (Basel) 2023; 14:1265. [PMID: 37372445 DOI: 10.3390/genes14061265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Sudden cardiac death (SCD) is an unexpected natural death due to cardiac causes, usually happening within one hour of symptom manifestation or in individuals in good health up to 24 h before the event. Genomic screening has been increasingly applied as a useful approach to detecting the genetic variants that potentially contribute to SCD and helping the evaluation of SCD cases in the post-mortem setting. Our aim was to identify the genetic markers associated with SCD, which might enable its target screening and prevention. In this scope, a case-control analysis through the post-mortem genome-wide screening of 30 autopsy cases was performed. We identified a high number of novel genetic variants associated with SCD, of which 25 polymorphisms were consistent with a previous link to cardiovascular diseases. We ascertained that many genes have been already linked to cardiovascular system functioning and diseases and that the metabolisms most implicated in SCD are the lipid, cholesterol, arachidonic acid, and drug metabolisms, suggesting their roles as potential risk factors. Overall, the genetic variants pinpointed herein might be useful markers of SCD, but the novelty of these results requires further investigations.
Collapse
Affiliation(s)
- Livia Beccacece
- Computational Genomics Lab, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Paolo Abondio
- aDNA Lab, Department of Cultural Heritage, University of Bologna, Ravenna Campus, 48121 Ravenna, Italy
| | - Arianna Giorgetti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Carla Bini
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Guido Pelletti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Donata Luiselli
- aDNA Lab, Department of Cultural Heritage, University of Bologna, Ravenna Campus, 48121 Ravenna, Italy
| | - Susi Pelotti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
4
|
Lara-Hernandez F, Alvarez L, Chaves J, Garcia-Garcia AB. Molecular Research on Genes Involved in Metabolic Diseases. Biomedicines 2023; 11:1671. [PMID: 37371766 DOI: 10.3390/biomedicines11061671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Numerous genes involved in different metabolic diseases have been identified, and this number is increasing [...].
Collapse
Affiliation(s)
| | - Luis Alvarez
- Genomic and Diabetes Unit, INCLIVA Biomedical Research Institute, 46021 Valencia, Spain
| | - Javier Chaves
- Genomic and Diabetes Unit, INCLIVA Biomedical Research Institute, 46021 Valencia, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), ISCIII, 28029 Madrid, Spain
| | - Ana-Barbara Garcia-Garcia
- Genomic and Diabetes Unit, INCLIVA Biomedical Research Institute, 46021 Valencia, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), ISCIII, 28029 Madrid, Spain
| |
Collapse
|
5
|
Lai MW, Chow N, Checco A, Kunar B, Redmond D, Rafii S, Rabbany SY. Systems Biology Analysis of Temporal Dynamics That Govern Endothelial Response to Cyclic Stretch. Biomolecules 2022; 12:1837. [PMID: 36551265 PMCID: PMC9775567 DOI: 10.3390/biom12121837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Endothelial cells in vivo are subjected to a wide array of mechanical stimuli, such as cyclic stretch. Notably, a 10% stretch is associated with an atheroprotective endothelial phenotype, while a 20% stretch is associated with an atheroprone endothelial phenotype. Here, a systems biology-based approach is used to present a comprehensive overview of the functional responses and molecular regulatory networks that characterize the transition from an atheroprotective to an atheroprone phenotype in response to cyclic stretch. Using primary human umbilical vein endothelial cells (HUVECs), we determined the role of the equibiaxial cyclic stretch in vitro, with changes to the radius of the magnitudes of 10% and 20%, which are representative of physiological and pathological strain, respectively. Following the transcriptome analysis of next-generation sequencing data, we identified four key endothelial responses to pathological cyclic stretch: cell cycle regulation, inflammatory response, fatty acid metabolism, and mTOR signaling, driven by a regulatory network of eight transcription factors. Our study highlights the dynamic regulation of several key stretch-sensitive endothelial functions relevant to the induction of an atheroprone versus an atheroprotective phenotype and lays the foundation for further investigation into the mechanisms governing vascular pathology. This study has significant implications for the development of treatment modalities for vascular disease.
Collapse
Affiliation(s)
- Michael W. Lai
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, New York, NY 11549, USA
| | - Nathan Chow
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, New York, NY 11549, USA
| | - Antonio Checco
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, New York, NY 11549, USA
| | - Balvir Kunar
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine (WCM), New York, NY 10065, USA
| | - David Redmond
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine (WCM), New York, NY 10065, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine (WCM), New York, NY 10065, USA
| | - Sina Y. Rabbany
- Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, New York, NY 11549, USA
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine (WCM), New York, NY 10065, USA
| |
Collapse
|
6
|
García-García AB, Martínez-Hervás S, Vernia S, Ivorra C, Pulido I, Martín-Escudero JC, Casado M, Carretero J, Real JT, Chaves FJ. A Very Rare Variant in SREBF2, a Possible Cause of Hypercholesterolemia and Increased Glycemic Levels. Biomedicines 2022; 10:biomedicines10051178. [PMID: 35625914 PMCID: PMC9138625 DOI: 10.3390/biomedicines10051178] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 02/01/2023] Open
Abstract
Patients with high cholesterol and glucose levels are at high risk for cardiovascular disease. The Sterol Regulatory Element Binding Protein (SREBP) system regulates genes involved in lipid, cholesterol and glucose pathways. Autosomal Dominant Hypercholesterolemias (ADHs) are a group of diseases with increased cholesterol levels. They affect 1 out of every 500 individuals. About 20–30% of patients do not present any mutation in the known genes (LDLR, APOB and PCSK9). ADHs constitute a good model to identify the genes involved in the alteration of lipid levels or possible therapeutic targets. In this paper, we studied whether a mutation in the SREBP system could be responsible for ADH and other metabolic alterations present in these patients. Forty-one ADH patients without mutations in the main responsible genes were screened by direct sequencing of SREBP system genes. A luciferase reporter assay of the found mutation and an oral glucose tolerance test in carriers and non-carriers were performed. We found a novel mutation in the SREBF2 gene that increases transcription levels and cosegregates with hypercholesterolemia, and we found increased glucose levels in one family. SREBP2 is known to be involved in cholesterol synthesis, plasma levels and glucose metabolism in humans. The found mutation may involve the SREBF2 gene in hypercholesterolemia combined with hyperglycemia.
Collapse
Affiliation(s)
- Ana-Bárbara García-García
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain; (A.-B.G.-G.); (S.M.-H.); (J.T.R.)
- Genomic and Diabetes Unit, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Sergio Martínez-Hervás
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain; (A.-B.G.-G.); (S.M.-H.); (J.T.R.)
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
- Endocrinology Service, University Clinical Hospital of Valencia, 46010 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Santiago Vernia
- Biomedical Institute of Valencia (IBV-CSIC), 46010 Valencia, Spain; (S.V.); (M.C.)
| | - Carmen Ivorra
- Genomic and Diabetes Unit, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Inés Pulido
- University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois Chicago, Chicago, IL 60612, USA;
- Department of Physiology, University of Valencia, 46010 Valencia, Spain;
| | | | - Marta Casado
- Biomedical Institute of Valencia (IBV-CSIC), 46010 Valencia, Spain; (S.V.); (M.C.)
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
| | - Julián Carretero
- Department of Physiology, University of Valencia, 46010 Valencia, Spain;
| | - José T. Real
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain; (A.-B.G.-G.); (S.M.-H.); (J.T.R.)
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
- Endocrinology Service, University Clinical Hospital of Valencia, 46010 Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Felipe Javier Chaves
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), 28029 Madrid, Spain; (A.-B.G.-G.); (S.M.-H.); (J.T.R.)
- Genomic and Diabetes Unit, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
- Correspondence: ; Tel.: +34-96-38-64100 (ext. 51905)
| |
Collapse
|
7
|
Kang H, Yu H, Fan J, Cao G. Rotigotine protects against oxidized low-density lipoprotein(ox-LDL)-induced damages in human umbilical vein endothelial cells(HUVECs). Bioengineered 2021; 12:10568-10579. [PMID: 34860135 PMCID: PMC8810014 DOI: 10.1080/21655979.2021.2000224] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Rotigotine is a non-ergoline dopamine agonist that has been licensed for the treatment of Parkinson’s disease. Cardiovascular diseases are the world’s leading cause of death. Ox-LDL- induced endothelial damages are involved in the initiation and progression of cardiovascular diseases. In this study, we assessed the beneficial properties of Rotigotine on ox-LDL-induced insults to HUVECs to highlight its potential use in the treatment of cardiovascular diseases. Our findings show that Rotigotine suppresses the expressions of low-density lipoprotein receptor (LDL-R), proprotein convertase subtilisin/kexin type 9 (PCSK-9), and sterol regulatory element-binding protein (SREBP-2). It also inhibits ox-LDL-induced cholesterol accumulation in endothelial cells (ECs), improves U937 monocytes adhesion, and decreases the representation of NADPH oxidase (NOX-4) and generation of reactive oxygen species (ROS) in endothelial cells (ECs). Furthermore, Rotigotine inhibited the expressions of both vascular cellular adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) in HUVECs and had anti-inflammatory efficacy in ox-LDL-induced cells by inhibiting the expressions of pro-inflammatory cytokines. Notably, Rotigotine inhibits the activation of nuclear factor-kappaB (NF-κB) by preventing nuclear translocation of NF-κB p65 and reducing the luciferase activity of NF-κB reporter. We, therefore, conclude that these effects of Rotigotine on HUVECs suggest that it may play a therapeutic role in cardiovascular diseases.
Collapse
Affiliation(s)
- Hui Kang
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Hui Yu
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jingxiu Fan
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ge Cao
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Espinoza C, Fuenzalida B, Leiva A. Increased Fetal Cardiovascular Disease Risk: Potential Synergy Between Gestational Diabetes Mellitus and Maternal Hypercholesterolemia. Curr Vasc Pharmacol 2021; 19:601-623. [PMID: 33902412 DOI: 10.2174/1570161119666210423085407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVD) remain a major cause of death worldwide. Evidence suggests that the risk for CVD can increase at the fetal stages due to maternal metabolic diseases, such as gestational diabetes mellitus (GDM) and maternal supraphysiological hypercholesterolemia (MSPH). GDM is a hyperglycemic, inflammatory, and insulin-resistant state that increases plasma levels of free fatty acids and triglycerides, impairs endothelial vascular tone regulation, and due to the increased nutrient transport, exposes the fetus to the altered metabolic conditions of the mother. MSPH involves increased levels of cholesterol (mainly as low-density lipoprotein cholesterol) which also causes endothelial dysfunction and alters nutrient transport to the fetus. Despite that an association has already been established between MSPH and increased CVD risk, however, little is known about the cellular processes underlying this relationship. Our knowledge is further obscured when the simultaneous presentation of MSPH and GDM takes place. In this context, GDM and MSPH may substantially increase fetal CVD risk due to synergistic impairment of placental nutrient transport and endothelial dysfunction. More studies on the separate and/or cumulative role of both processes are warranted to suggest specific treatment options.
Collapse
Affiliation(s)
- Cristian Espinoza
- Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Andrea Leiva
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Providencia 7510157, Chile
| |
Collapse
|
9
|
Li H, Li J, Zhang X, Li J, Xi C, Wang W, Lu Y, Xuan L. Euphornin L promotes lipid clearance by dual regulation of LDLR and PCSK9. Exp Ther Med 2021; 22:1381. [PMID: 34650629 DOI: 10.3892/etm.2021.10817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 03/24/2021] [Indexed: 11/05/2022] Open
Abstract
Our previous study identified euphornin L as an active lipid-lowering compound in high-fat diet-fed Golden Syrian hamsters. The aim of the present study was to investigate the mechanisms underlying the lipid-lowering effects of euphornin L. Euphornin L in HepG2 cells was assessed via DiI-LDL update assays and found to increase LDL-update and LDLR protein levels. RNA interference assays demonstrated that its LDL-update effects were LDLR-dependent. Dual luciferase reporter and mRNA stability assays revealed that euphornin L had little effect on LDLR mRNA transcription but lengthened the half-life of LDLR mRNA by activating ERK protein in cells. Euphornin L decreased the secretion of PCSK9 protein and alleviated PCSK9-mediated LDLR protein degradation. In vivo experiments in hamsters, which were treated with euphornin L (30 mg/kg/day) for 3 weeks, confirmed these findings. LDLR protein levels in liver tissue were upregulated, while PCSK9 protein levels in serum were downregulated. Altogether, the present study demonstrated that euphornin L increased LDLR protein levels by dual regulation of LDLR mRNA and PCSK9 protein, and represented an active compound for lipid-lowering drug development.
Collapse
Affiliation(s)
- Huihui Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Jun Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xianjing Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Jiaomeng Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Cong Xi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Wenqiong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Youli Lu
- Central Laboratory, Shanghai Xuhui Central Hospital/Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200031, P.R. China.,Shanghai Clinical Research Center, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Lijiang Xuan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China.,University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
10
|
Zou H, Yang N, Zhang X, Chen HW. RORγ is a context-specific master regulator of cholesterol biosynthesis and an emerging therapeutic target in cancer and autoimmune diseases. Biochem Pharmacol 2021; 196:114725. [PMID: 34384758 DOI: 10.1016/j.bcp.2021.114725] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 01/04/2023]
Abstract
Aberrant cholesterol metabolism and homeostasis in the form of elevated cholesterol biosynthesis and dysregulated efflux and metabolism is well recognized as a major feature of metabolic reprogramming in solid tumors. Recent studies have emphasized on major drivers and regulators such as Myc, mutant p53, SREBP2, LXRs and oncogenic signaling pathways that play crucial roles in tumor cholesterol metabolic reprogramming. Therapeutics such as statins targeting the mevalonate pathway were tried at the clinic without showing consistent benefits to cancer patients. Nuclear receptors are prominent regulators of mammalian metabolism. Their de-regulation often drives tumorigenesis. RORγ and its immune cell-specific isoform RORγt play important functions in control of mammalian metabolism, circadian rhythm and immune responses. Although RORγ, together with its closely related members RORα and RORβ were identified initially as orphan receptors, recent studies strongly support the conclusion that specific intermediates and metabolites of cholesterol pathways serve as endogenous ligands of RORγ. More recent studies also reveal a critical role of RORγ in tumorigenesis through major oncogenic pathways including acting a new master-like regulator of tumor cholesterol biosynthesis program. Importantly, an increasing number of RORγ orthosteric and allosteric ligands are being identified that display potent activities in blocking tumor growth and autoimmune disorders in preclinical models. This review summarizes the recent preclinical and clinical progress on RORγ with emphasis on its role in reprogramming tumor cholesterol metabolism and its regulation. It will also discuss RORγ functional mechanisms, context-specificity and its value as a therapeutic target for effective cancer treatment.
Collapse
Affiliation(s)
- Hongye Zou
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Nianxin Yang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Xiong Zhang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, USA; UC Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, California, USA; VA Northern California Health Care System, Mather, California, USA.
| |
Collapse
|
11
|
Rutin and Quercetin Decrease Cholesterol in HepG2 Cells but Not Plasma Cholesterol in Hamsters by Oral Administration. Molecules 2021; 26:molecules26123766. [PMID: 34205604 PMCID: PMC8234066 DOI: 10.3390/molecules26123766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 11/18/2022] Open
Abstract
Rutin (R) and quercetin (Q) are two widespread dietary flavonoids. Previous studies regarding the plasma cholesterol-lowering activity of R and Q generated inconsistent results. The present study was therefore carried out to investigate the effects of R and Q on cholesterol metabolism in both HepG2 cells and hypercholesterolemia hamsters. Results from HepG2 cell experiments demonstrate that both R and Q decreased cholesterol at doses of 5 and 10 µM. R and Q up-regulated both the mRNA and protein expression of sterol regulatory element binding protein 2 (SREBP2), low-density lipoprotein receptor (LDLR), and liver X receptor alpha (LXRα). The immunofluorescence study revealed that R and Q increased the LDLR expression, while only Q improved LDL-C uptake in HepG2 cells. Results from hypercholesterolemia hamsters fed diets containing R (5.5 g/kg diet) and Q (2.5 g/kg diet) for 8 weeks demonstrate that both R and Q had no effect on plasma total cholesterol. In the liver, only Q reduced cholesterol significantly. The discrepancy between the in vitro and in vivo studies was probably due to a poor bioavailability of flavonoids in the intestine. It was therefore concluded that R and Q were effective in reducing cholesterol in HepG2 cells in vitro, whereas in vivo, the oral administration of the two flavonoids had little effect on plasma cholesterol in hamsters.
Collapse
|
12
|
Luquero A, Badimon L, Borrell-Pages M. PCSK9 Functions in Atherosclerosis Are Not Limited to Plasmatic LDL-Cholesterol Regulation. Front Cardiovasc Med 2021; 8:639727. [PMID: 33834043 PMCID: PMC8021767 DOI: 10.3389/fcvm.2021.639727] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/01/2021] [Indexed: 12/31/2022] Open
Abstract
The relevance of PCSK9 in atherosclerosis progression is demonstrated by the benefits observed in patients that have followed PCSK9-targeted therapies. The impact of these therapies is attributed to the plasma lipid-lowering effect induced when LDLR hepatic expression levels are recovered after the suppression of soluble PCSK9. Different studies show that PCSK9 is involved in other mechanisms that take place at different stages during atherosclerosis development. Indeed, PCSK9 regulates the expression of key receptors expressed in macrophages that contribute to lipid-loading, foam cell formation and atherosclerotic plaque formation. PCSK9 is also a regulator of vascular inflammation and its expression correlates with pro-inflammatory cytokines release, inflammatory cell recruitment and plaque destabilization. Furthermore, anti-PCSK9 approaches have demonstrated that by inhibiting PCSK9 activity, the progression of atherosclerotic disease is diminished. PCSK9 also modulates thrombosis by modifying platelets steady-state, leukocyte recruitment and clot formation. In this review we evaluate recent findings on PCSK9 functions in cardiovascular diseases beyond LDL-cholesterol plasma levels regulation.
Collapse
Affiliation(s)
- Aureli Luquero
- Cardiovascular Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación en Red- Área Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Centro de Investigación en Red- Área Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
13
|
Abstract
The main strategy for lowering blood cholesterol levels is through the inhibition of the NADPH-dependent HMG-CoA reductase (3-hydroxy-3-methyl-glutaryl-CoA reductase). The enzyme catalyses the reduction of HMG-CoA to mevalonate and this process is inhibited by statins that form the bulk of the therapeutic agents to treat high cholesterol since the 1970s. Newer drugs that are safer than statins are constantly being developed. The inhibition of candidate drugs to HMG-CoA reductase remains the mainstay of drug development research. The determination of the enzyme activity is important for the correct assessment of potency of the enzyme as well as determining the inhibition of potential therapeutic agents from the plant and microbial extracts. Also, this chapter covers the use of the popular four-parameter logistics model that can yield accurate estimation of the IC50 values of therapeutic agents and their 95% confidence intervals.
Collapse
Affiliation(s)
- Baskaran Gunasekaran
- Faculty of Biotechnology and Biomolecular Sciences, Department of Biochemistry, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Mohd Yunus Shukor
- Faculty of Biotechnology and Biomolecular Sciences, Department of Biochemistry, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|
14
|
Wang R, You YM, Liu X. Effect of Zanthoxylum alkylamides on lipid metabolism and its mechanism in rats fed with a high-fat diet. J Food Biochem 2020; 45:e13548. [PMID: 33270233 DOI: 10.1111/jfbc.13548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/10/2020] [Accepted: 10/15/2020] [Indexed: 01/14/2023]
Abstract
This research aimed at exploring the effect of Zanthoxylum alkylamides on lipid metabolism and its potential mechanisms using high-fat diet rat model. Treatment with Zanthoxylum alkylamides for 6 weeks, food efficiency and atherogenic index of the low, medium, and high doses of Zanthoxylum alkylamides-treated groups were significantly reduced. Meanwhile, the histopathological structure of the livers showed that hepatic steatosis in the groups treated with Zanthoxylum alkylamides was reduced, particularly the HD group. Moreover, the related genes were studied, such as, liver X receptor (LXR), cholesterol 7 alpha-hydroxylase (CYP7A1), hepatic 3-hydroxyl-2-methylglutaryl CoA (HMG-CoA) reductase, sterol regulatory element-binding protein 2 (SREBP-2), ileal bile acid-binding protein (IBABP), sodium-dependent bile acid transporter (ASBT), and transient receptor potential vanilloid subtype1 (TRPV1). These results demonstrated that Zanthoxylum alkylamides could ameliorate abnormal lipid metabolism in rats fed with a high-fat diet. The underlying mechanism may be the downregulation of the expression levels of cholesterol synthesis and bile acid reabsorption-related genes, reduction of endogenous cholesterol synthesis, and increase in bile acid and neutral sterol excretion. PRACTICAL APPLICATIONS: High-energy diet is a potential risk of lipid metabolic disorder. Many studies have shown that hyperlipidemia can lead to atherosclerosis and even hemangioma, cerebral thrombosis, coronary heart disease, and other diseases, which seriously threaten human health. Therefore, seeking an effective and safe way to prevent the obesity-related disease is necessary. This research found that Zanthoxylum alkylamide could ameliorate abnormal lipid metabolism in rats fed with a high-fat diet. The underlying mechanism may be the downregulation of the expression levels of cholesterol synthesis and ileal absorption of bile acid genes, reduction of endogenous cholesterol synthesis, and increase in bile acid and neutral sterol excretion. Therefore, Zanthoxylum alkylamide has the potential for preventing or alleviating high-energy intake-related obesity.
Collapse
Affiliation(s)
- Rui Wang
- College of Food Science, Southwest University, Chongqing, China.,Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing, China
| | - Yu-Ming You
- College of Forestry and Life Sciences, Chongqing University of Arts and Sciences, Chongqing, China
| | - Xiong Liu
- College of Food Science, Southwest University, Chongqing, China
| |
Collapse
|
15
|
Wang D, Yang X, Chen Y, Gong K, Yu M, Gao Y, Wu X, Hu H, Liao C, Han J, Duan Y. Ascorbic acid enhances low-density lipoprotein receptor expression by suppressing proprotein convertase subtilisin/kexin 9 expression. J Biol Chem 2020; 295:15870-15882. [PMID: 32913121 DOI: 10.1074/jbc.ra120.015623] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/04/2020] [Indexed: 01/30/2023] Open
Abstract
Ascorbic acid, a water-soluble antioxidant, regulates various biological processes and is thought to influence cholesterol. However, little is known about the mechanisms underpinning ascorbic acid-mediated cholesterol metabolism. Here, we determined if ascorbic acid can regulate expression of proprotein convertase subtilisin/kexin 9 (PCSK9), which binds low-density lipoprotein receptor (LDLR) leading to its intracellular degradation, to influence low-density lipoprotein (LDL) metabolism. At cellular levels, ascorbic acid inhibited PCSK9 expression in HepG2 and Huh7 cell lines. Consequently, LDLR expression and cellular LDL uptake were enhanced. Similar effects of ascorbic acid on PCSK9 and LDLR expression were observed in mouse primary hepatocytes. Mechanistically, ascorbic acid suppressed PCSK9 expression in a forkhead box O3-dependent manner. In addition, ascorbic acid increased LDLR transcription by regulating sterol regulatory element-binding protein 2. In vivo, administration of ascorbic acid reduced serum PCSK9 levels and enhanced liver LDLR expression in C57BL/6J mice. Reciprocally, lack of ascorbic acid supplementation in L-gulono-γ-lactone oxidase deficient (Gulo -/-) mice increased circulating PCSK9 and LDL levels, and decreased liver LDLR expression, whereas ascorbic acid supplementation decreased PCSK9 and increased LDLR expression, ameliorating LDL levels in Gulo -/- mice fed a high fat diet. Moreover, ascorbic acid levels were negatively correlated to PCSK9, total and LDL levels in human serum samples. Taken together, these findings suggest that ascorbic acid reduces PCSK9 expression, leading to increased LDLR expression and cellular LDL uptake. Thus, supplementation of ascorbic acid may ameliorate lipid profiles in ascorbic acid-deficient species.
Collapse
Affiliation(s)
- Dandan Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Maoyun Yu
- School of Biological and Pharmaceutical Engineering, West Anhui University, Lu'An, China
| | - Yongyao Gao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ximei Wu
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Huaqing Hu
- First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China; College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China.
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
16
|
Chen X, Huang K, Hu S, Lan G, Gan X, Gao S, Deng Y, Hu J, Li L, Hu B, He H, Liu H, Xia L, Wang J. FASN-Mediated Lipid Metabolism Regulates Goose Granulosa Cells Apoptosis and Steroidogenesis. Front Physiol 2020; 11:600. [PMID: 32676035 PMCID: PMC7333536 DOI: 10.3389/fphys.2020.00600] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/14/2020] [Indexed: 12/27/2022] Open
Abstract
Lipid metabolism participates in regulating the functions of granulosa cells (GCs), which is important for follicular development. In this experiment, goose GCs from pre-hierarchical follicles and hierarchical follicles were selected to be the model for studying the putative regulatory role of lipid metabolism in apoptosis and steroidogenesis, through overexpression and interference with fatty acid synthase (FASN). When FASN was overexpressed, the lipid accumulation was increased in hierarchical GCs (hGCs) and it was increased in the two categorized GCs when FASN was interfered. In addition, the apoptosis of the two categorized GCs was increased when FASN was overexpressed, and their progesterone production was decreased when FASN was interfered. The results of qRT-PCR showed that, when FASN was overexpressed, the expression level of CYP11A1 was decreased in pre-hierarchical GCs (phGCs), while the expression levels of SCD1, DGAT2, APOB, and StAR were increased in hGCs. When FASN was interfered, the expression levels of CPT-1, DGAT2, and StAR were decreased whereas the expression level of CYP11A1 was increased in phGCs, and the expression levels of CPT-1, SCD1, and StAR were decreased in hGCs. These results not only identify the different effects of manipulated FASN expression on lipid metabolism of goose phGCs and hGCs but also demonstrate that FASN-mediated lipid metabolism plays an important role in regulating apoptosis and steroidogenesis of in vitro cultured goose GCs.
Collapse
Affiliation(s)
- Xi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Kailiang Huang
- Key Laboratory of Agricultural Information Engineering of Sichuan Province, College of Information Engineering, Sichuan Agricultural University, Ya'an, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Gang Lan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xiang Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shanyan Gao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yan Deng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Lu Xia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
17
|
Pangestika I, Oksal E, Tengku Muhammad TS, Amir H, Syamsumir DF, Wahid MEA, Andriani Y. Inhibitory effects of tangeretin and trans-ethyl caffeate on the HMG-CoA reductase activity: Potential agents for reducing cholesterol levels. Saudi J Biol Sci 2020; 27:1947-1960. [PMID: 32714018 PMCID: PMC7376234 DOI: 10.1016/j.sjbs.2020.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 06/06/2020] [Accepted: 06/07/2020] [Indexed: 12/19/2022] Open
Abstract
One of the pathways to reduce cholesterol production in the liver is through the inhibition of HMG-Coa reductase (HMGCR) by current drugs, statins. However, these have side effects if consumed in prolonged periods. Tangeretin and trans-ethyl caffeate as alternative drugs in reducing hypercholesterolemia and preventing atherosclerosis have never been reported. Their effects on inhibiting HMGCR activity were investigated through enzymatic method (in vitro and in vivo). The toxicity property was analyzed on the Serum Glutamate Oxalate Transaminase (SGOT)/Serum Glutamate Piruvate Transaminase (SGPT) levels and rat liver histology. The results showed that both compounds inhibited HMGCR activity significantly compare to the control simvastatin (p < 0.05). Tangeretin which showed very good activity in inhibiting HMGCR (83.8 of % inhibition, equal to simvastatin) was selected and used for anti-hypercholesterolemia in vivo assessment. Furthermore, tangeretin was shown to effectively reduced Total Cholesterol (TC) and Low Density Lipoprotein (LDL), and increased High Density Lipoprotein (HDL) levels significantly compared to the simvastatin group (p < 0.05). Tangeretin group was also proven to inhibit HMGCR rat liver activity significantly compare to the control simvastatin (p < 0.05). The toxicity study on the SGOT/SGPT levels and liver histology revealed that there were no side effects after administration by tangeretin. Results found that both tangeretin and trans-ethyl caffeate are potent candidates as anti-hypercholesterolemia agent in vitro. In addition, tangeretin was also shown to be safe and suitable as an alternative treatment for controlling hypercholesterolemia in vivo as well as have potency for preventing atherosclerosis.
Collapse
Affiliation(s)
- Inten Pangestika
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Efriyana Oksal
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Tengku Sifzizul Tengku Muhammad
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot, 21030 Kuala Nerus, Terengganu, Malaysia.,Research Management Center, Universiti Malaysia Terengganu, Mengabang Telipot, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Hermansyah Amir
- Educational Chemistry Program, Faculty of Teacher Training and Education, Bengkulu University, Bengkulu 38371, Indonesia
| | - Desy Fitrya Syamsumir
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Mohd Effendy Abdul Wahid
- School of Fisheries and Aquaculture Sciences, Universiti Malaysia Terengganu, Mengabang Telipot, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Yosie Andriani
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Mengabang Telipot, 21030 Kuala Nerus, Terengganu, Malaysia
| |
Collapse
|
18
|
Song Z, Lv S, Wu H, Qin L, Cao H, Zhang B, Ren S. Identification of foam cell biomarkers by microarray analysis. BMC Cardiovasc Disord 2020; 20:211. [PMID: 32375652 PMCID: PMC7201525 DOI: 10.1186/s12872-020-01495-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/23/2020] [Indexed: 01/12/2023] Open
Abstract
Background Lipid infiltration and inflammatory response run through the occurrence of atherosclerosis. Differentiation into macrophages and foam cell formation are the key steps of AS. Aim of this study was that the differential gene expression between foam cells and macrophages was analyzed to search the key links of foam cell generation, so as to explore the pathogenesis of atherosclerosis and provide targets for the early screening and prevention of coronary artery disease (CAD). Methods The gene expression profiles of GSE9874 were downloaded from Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE9874) on GPL96 [HG-U133A] Affymetrix Human Genome U133. A total of 22,383 genes were analyzed for differentially expression genes (DEGs) by Bayes package. GO enrichment analysis and KEGG pathway analysis for DEGs were performed using KOBAS 3.0 software (Peking University, Beijing, China). STRING software (STRING 10.0; European Molecular Biology Laboratory, Heidelberg, Germany) was used to analyze the protein-protein interaction (PPI) of DEGs. Results A total of 167 DEGs between macrophages and foam cells were identified. Compared with macrophages, 102 genes were significantly upregulated and 65 genes were significantly downregulated (P < 0.01, fold-change > 1) in foam cells. DEGs were mainly enrich in ‘sterol biosynthetic and metabolic process’, ‘cholesterol metabolic and biosynthetic process’ by GO enrichment analysis. The results of KEGG pathway analysis showed all differential genes are involved in biological processes through 143 KEGG pathways. A PPI network of the DEGs was constructed and 10 outstanding genes of the PPI network was identified by using Cytoscape, which include HMGCR, SREBF2, LDLR, HMGCS1, FDFT1, LPL, DHCR24, SQLE, ABCA1 and FDPS. Conclusion: Lipid metabolism related genes and molecular pathways were the key to the transformation of macrophages into foam cells. Therefore, lipid metabolism disorder is the key to turn macrophages into foam cells, which plays a major role in CAD.
Collapse
Affiliation(s)
- Zikai Song
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Shijie Lv
- Department of Orthopedics, Jilin Province FAW General Hospital, Changchun, Jilin Province, China
| | - Haidi Wu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ling Qin
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Hongyan Cao
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Bo Zhang
- Department of Pediatric Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Shuping Ren
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
19
|
Albuquerque A, Óvilo C, Núñez Y, Benítez R, López-Garcia A, García F, Félix MDR, Laranjo M, Charneca R, Martins JM. Comparative Transcriptomic Analysis of Subcutaneous Adipose Tissue from Local Pig Breeds. Genes (Basel) 2020; 11:E422. [PMID: 32326415 PMCID: PMC7231169 DOI: 10.3390/genes11040422] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/07/2020] [Accepted: 04/11/2020] [Indexed: 12/12/2022] Open
Abstract
When compared to modern lean-type breeds, Portuguese local Alentejano (AL) and Bísaro (BI) pig breeds present a high potential for subcutaneous and intramuscular fat (IMF) deposition which contributes for better meat quality. The aim of this work was to explore the genome function to better understand the underlying physiological mechanisms associated with body fat accretion. Dorsal subcutaneous fat samples were collected at slaughter from adult animals (n = 4 for each breed) with ~150 kg body weight. Total RNA was obtained and sequenced for transcriptome analysis using DESeq2. A total of 458 differentially expressed (DE) genes (q-value < 0.05) were identified, with 263 overexpressed in AL and 195 in BI. Key genes involved in de novo fatty acid biosynthesis, elongation and desaturation were upregulated in AL such as ACLY, FASN, ME1, ELOVL6 and SCD. A functional enrichment analysis of the DE genes was performed using Ingenuity Pathway Analysis. Cholesterol synthesis is suggested to be higher in AL via SREBF2, SCAP and PPARG, while lipolytic activity may be more active in BI through GH and AMPK signalling. Increased signalling of CD40 together with the predicted activation of INSIG1 and INSIG2 in BI suggests that this breed is more sensitive to insulin whereas the AL is less sensitive like the Iberian breed.
Collapse
Affiliation(s)
- André Albuquerque
- MED-Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Formação Avançada & Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal;
| | - Cristina Óvilo
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (C.Ó.); (Y.N.); (R.B.); (A.L.-G.); (F.G.)
| | - Yolanda Núñez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (C.Ó.); (Y.N.); (R.B.); (A.L.-G.); (F.G.)
| | - Rita Benítez
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (C.Ó.); (Y.N.); (R.B.); (A.L.-G.); (F.G.)
| | - Adrián López-Garcia
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (C.Ó.); (Y.N.); (R.B.); (A.L.-G.); (F.G.)
| | - Fabián García
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28040 Madrid, Spain; (C.Ó.); (Y.N.); (R.B.); (A.L.-G.); (F.G.)
| | - Maria do Rosário Félix
- MED & Departamento de Fitotecnia, Escola de Ciências e Tecnologia, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal;
| | - Marta Laranjo
- MED-Mediterranean Institute for Agriculture, Environment and Development, Instituto de Investigação e Formação Avançada & Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal;
| | - Rui Charneca
- MED & Departamento de Medicina Veterinária, Escola de Ciências e Tecnologia, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal;
| | - José Manuel Martins
- MED & Departamento de Zootecnia, Escola de Ciências e Tecnologia, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal
| |
Collapse
|
20
|
Pengrattanachot N, Cherngwelling R, Jaikumkao K, Pongchaidecha A, Thongnak L, Swe MT, Chatsudthipong V, Lungkaphin A. Atorvastatin attenuates obese-induced kidney injury and impaired renal organic anion transporter 3 function through inhibition of oxidative stress and inflammation. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165741. [PMID: 32101757 DOI: 10.1016/j.bbadis.2020.165741] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/17/2020] [Accepted: 02/18/2020] [Indexed: 12/15/2022]
Abstract
An excessive consumption of high-fat diet can lead to the alterations of glucose and lipid metabolism, impaired insulin signaling and increased ectopic lipid accumulation resulting in renal lipotoxicity and subsequent renal dysfunction. Atorvastatin is a lipid-lowering drug in clinical treatment. Several studies have reported that atorvastatin has several significant pleiotropic effects including anti-inflammatory, antioxidant, and anti-apoptotic effects. However, the effects of atorvastatin on metabolic disturbance and renal lipotoxicity in obesity are not fully understood. In this study, obesity in rat was developed by high-fat diet (HFD) feeding for 16 weeks. After that, the HFD-fed rats were received either a vehicle (HF), atorvastatin (HFA) or vildagliptin (HFVIL), by oral gavage for 4 weeks. We found that HF rats showed insulin resistance, visceral fat expansion and renal lipid accumulation. Impaired renal function and renal organic anion transporter 3 (Oat3) function and expression were also observed in HF rats. The marked increases in MDA level, renal injury and NF-κB, TGF-β, NOX-4, PKC-α expression were demonstrated in HF rats. Atorvastatin or vildagliptin treatment attenuated insulin resistance and renal lipid accumulation-induced lipotoxicity in HFA and HFVIL rats. Moreover, the proteins involved in renal inflammation, fibrosis, oxidative stress and apoptosis were attenuated leading to improved renal Oat3 function and renal function in the treated groups. Interestingly, atorvastatin showed higher efficacy than vildagliptin in improving insulin resistance, renal lipid accumulation and in exerting renoprotective effects in obesity-induced renal injury and impaired renal Oat3 function.
Collapse
Affiliation(s)
| | - Rada Cherngwelling
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Krit Jaikumkao
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Anchalee Pongchaidecha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Laongdao Thongnak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Myat Theingi Swe
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Physiology, University of Medicine, Yangon, Myanmar
| | - Varanuj Chatsudthipong
- Research Center of Transport Protein for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Functional Food Research Center for Well-being, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
21
|
Thongnak L, Pongchaidecha A, Lungkaphin A. Renal Lipid Metabolism and Lipotoxicity in Diabetes. Am J Med Sci 2019; 359:84-99. [PMID: 32039770 DOI: 10.1016/j.amjms.2019.11.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/13/2019] [Accepted: 11/20/2019] [Indexed: 12/27/2022]
Abstract
The pathogenesis of diabetic kidney disease is a complex process caused by both glucotoxicity and lipotoxicity due to lipid accumulation. In cases of diabetic animals, lipid deposition is found in both tubular and glomerular portions of the kidneys, which are the major sites of diabetic nephropathy lesions. The aim of this review was to provide insights into the mechanisms that lead to the development of renal lipid accumulation and the effects of renal lipotoxicity in the diabetic condition. An increased number of lipogenic genes and a decreased number of lipid oxidation genes are also detected in diabetic kidneys, both of which lead to lipid accumulation. The induction of oxidative stress, inflammation, fibrosis and apoptosis caused by lipid accumulation and lipid metabolites is called lipotoxicity. Renal lipotoxicity due to derangement in lipid metabolism may be a pathogenic mechanism leading to diabetic nephropathy and renal dysfunction.
Collapse
Affiliation(s)
- Laongdao Thongnak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anchalee Pongchaidecha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Functional Food Research Center for Well-Being, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
22
|
Taghizadeh E, Taheri F, Gheibi Hayat SM, Montecucco F, Carbone F, Rostami D, Montazeri A, Sahebkar A. The atherogenic role of immune cells in familial hypercholesterolemia. IUBMB Life 2019; 72:782-789. [PMID: 31633867 DOI: 10.1002/iub.2179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/16/2019] [Indexed: 12/19/2022]
Abstract
Familial hypercholesterolemia (FH) is an autosomal dominant disorder of lipoprotein metabolism that mainly occurs due to mutations in the low-density lipoprotein receptor gene and is characterized by increased levels of low-density lipoprotein cholesterol, leading to accelerated atherogenesis and premature coronary heart disease. Both innate and adaptive immune responses, which mainly include monocytes, macrophages, neutrophils, T lymphocytes, and B lymphocytes, have been shown to play a key role for the initiation and progression of atherogenesis in the general population. In FH patients, these immune cells have been suggested to play specific pro-atherosclerotic activities, from the initial leukocyte recruitment to plaque rupture. In fact, the accumulation of cholesterol crystals and oxLDL in the vessels in FH patients is particularly high, with consequent abnormal mobilization of immune cells and secretion of various pro-inflammatory and chemokines. In addition, cholesterol accumulation in immune cells is exaggerated with chronic exposure to relevant pro-atherosclerotic triggers. The topics considered in this review may provide a more specific focus on the immune system alterations in FH and open new insights toward immune cells as potential therapeutic targets in FH.
Collapse
Affiliation(s)
- Eskandar Taghizadeh
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy.,First Clinic of Internal Medicine, IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Daryoush Rostami
- Department of School Allied, Zabol University of Medical Sciences, Zabol, Iran
| | - Ardalan Montazeri
- Department of Biology, Faculty of Science, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Vargas-Alarcon G, Gonzalez-Pacheco H, Perez-Mendez O, Posadas-Sanchez R, Cardoso-Saldaña G, Ramirez-Bello J, Escobedo G, Nieto-Lima B, Fragoso JM. SREBF1c and SREBF2 gene polymorphisms are associated with acute coronary syndrome and blood lipid levels in Mexican population. PLoS One 2019; 14:e0222017. [PMID: 31490983 PMCID: PMC6730878 DOI: 10.1371/journal.pone.0222017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/20/2019] [Indexed: 12/20/2022] Open
Abstract
Aim It has recently been reported that the sterol regulatory element-binding transcription factors (SREBF-1c, and -2) contribute to the variation in the plasma lipids levels, which have an important role in the atherosclerotic plaque development. The aim of the present study was to evaluate whether the SREBF1c and SREBF2 gene single nucleotide polymorphisms (SNPs) are associated with plasma lipids levels and ACS susceptibility in a case-control association study. Material and methods A case-control study was carried out in 625 patients with ACS (82% men and 18% women, with a mean age of 57.97 ± 10.5 years) and 700 healthy controls (66% men and 34% women, with a mean age of 54.37 ± 7.65 years). The sample size was calculated for a statistical power of 80%. We genotyped three SREBF1c (rs2297508, rs11656665 and rs11868035) and three SREBF2 (rs2267439, rs2267443, and rs2228314) gene polymorphisms by 5’ exonuclease TaqMan assays. The associations were evaluated by logistic regression under the co-dominant, dominant, recessive, over-dominant and additive inheritance models. The contribution of the genotypes on the plasma lipids levels was evaluated by Student’s t-test. Results Under different models, the SREBF1c rs2297508 (OR = 1.50, pCRes = 0.03), SREBF1c rs11656665 (OR = 1.35, pCDom = 0.02 and OR = 1.26, pCAdd = 0.02) and SREBF2 rs2228314 (OR = 1.78, pCRes = 0.03, OR = 1.27, pCAdd = 0.04) SNPs were associated with higher risk of ACS. On the other hand, the SREBF1c rs11868035 SNP was associated with lower risk of ACS (OR = 0.49, pCCo-dom = 0.001, OR = 0.66, pCDom = 0.003, OR = 0.57, PRes = 0.003 and OR = 0.71, pCAdd = 0.001). There was a statistically significant association of both SREBF1c rs11656665 and rs11868035 polymorphisms with plasma triglyceride levels. Conclusions In summary, our data suggest the association of the SREBF1c and SREBF2 SNPs with risk of developing ACS and with triglyceride levels in a Mexican population.
Collapse
Affiliation(s)
- Gilberto Vargas-Alarcon
- Department of Molecular Biology, Instituto Nacional de Cardiologia Ignacio Chavez, Mexico City, Mexico
- Atherosclerosis Study Group, Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| | - Hector Gonzalez-Pacheco
- Atherosclerosis Study Group, Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
- Coronary Unit, Instituto Nacional de Cardiologia Ignacio Chavez, Mexico City, Mexico
| | - Oscar Perez-Mendez
- Department of Molecular Biology, Instituto Nacional de Cardiologia Ignacio Chavez, Mexico City, Mexico
- Atherosclerosis Study Group, Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| | | | | | - Julian Ramirez-Bello
- Research Unit on Endocrine and Metabolic Diseases, Hospital Juarez de Mexico, Mexico City, Mexico
| | - Galileo Escobedo
- Unit of the Experimental Medicine, Hospital General de Mexico, Dr. Eduardo Liceaga, Mexico City, Mexico
| | - Betzabe Nieto-Lima
- Department of Molecular Biology, Instituto Nacional de Cardiologia Ignacio Chavez, Mexico City, Mexico
| | - Jose Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiologia Ignacio Chavez, Mexico City, Mexico
- Atherosclerosis Study Group, Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
- * E-mail:
| |
Collapse
|
24
|
Dolezelova E, Sa ICI, Prasnicka A, Hroch M, Hyspler R, Ticha A, Lastuvkova H, Cermanova J, Pericacho M, Visek J, Lasticova M, Micuda S, Nachtigal P. High soluble endoglin levels regulate cholesterol homeostasis and bile acids turnover in the liver of transgenic mice. Life Sci 2019; 232:116643. [PMID: 31299237 DOI: 10.1016/j.lfs.2019.116643] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/01/2019] [Accepted: 07/09/2019] [Indexed: 02/07/2023]
Abstract
AIMS Increased plasma soluble endoglin concentrations (sEng) are frequently detected in metabolic disorders accompanied with hypercholesterolemia in serum, but effect of sEng on the cholesterol biochemistry is unknown. Cholesterol and bile acids (BA) are important products of liver metabolism with numerous functions within the organism. Turnover of these substances requires precise regulation due to potential toxicities during their cumulation. In this study, we hypothesized that high sEng levels affect cholesterol homeostasis and BA turnover in mice liver. MAIN METHODS Nine-month-old transgenic male mice overexpressing human sEng and wild-type mice underwent plasma, bile, stool, and organ samples analysis by analytical, qRT-PCT and Western blot methods. KEY FINDINGS sEng mice demonstrated decreased plasma total and LDL cholesterol concentrations due to upregulation of hepatic Sr-b1 and Ldlr receptors, increased liver cholesterol content, and increased Abcg8-mediated cholesterol efflux into bile. sEng also increased conversion of cholesterol into bile acids (BA) via upregulation of Cyp7a1 and increased Mdr1 expression. Plasma concentrations of BA were increased in sEng mice due to their enhanced reabsorption via ileum. Increased hepatic disposition of BA led to their increased biliary excretion coupled with choleretic activity. SIGNIFICANCE For the first time, we have shown that high sEng plasma levels affect cholesterol and BA homeostasis on the basis of complex liver and intestinal effects. The significance of these findings for pathophysiology of diseases associated with increased sEng concentrations remains to be elucidated in prospective studies.
Collapse
Affiliation(s)
- Eva Dolezelova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Ivone Cristina Igreja Sa
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Alena Prasnicka
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Milos Hroch
- Department of Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Czech Republic
| | - Radomir Hyspler
- Centrum for Research and Development, University Hospital, Hradec Kralove, Czech Republic
| | - Alena Ticha
- Centrum for Research and Development, University Hospital, Hradec Kralove, Czech Republic
| | - Hana Lastuvkova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Czech Republic
| | - Jolana Cermanova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Czech Republic
| | - Miguel Pericacho
- Biomedical Research Institute of Salamanca and Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain
| | - Jakub Visek
- 3rd Department of Internal Medicine, Metabolism and Gerontology, University Hospital, Hradec Kralove, Czech Republic
| | - Martina Lasticova
- 3rd Department of Internal Medicine, Metabolism and Gerontology, University Hospital, Hradec Kralove, Czech Republic
| | - Stanislav Micuda
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Czech Republic.
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic.
| |
Collapse
|
25
|
Dumolt JH, Rideout TC. The Lipid-lowering Effects and Associated Mechanisms of Dietary Phytosterol Supplementation. Curr Pharm Des 2019; 23:5077-5085. [PMID: 28745211 DOI: 10.2174/1381612823666170725142337] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 07/01/2017] [Accepted: 07/21/2017] [Indexed: 12/12/2022]
Abstract
Phytosterols (PS) are plant-based structural analogous of mammalian cholesterol that have been shown to lower blood cholesterol concentrations by ~10%, although inter-individual response to PS supplementation due to subject-specific metabolic and genetic factors is evident. Recent work further suggests that PS may act as effective triglyceride (TG)-lowering agents with maximal TG reductions observed in hypertriglyceridemic subjects. Although PS have been demonstrated to interfere with cholesterol and perhaps TG absorption within the intestine, they also have the capacity to modulate the expression of lipid regulatory genes through liver X receptor (LXR) activation. Identification of single-nucleotide polymorphisms (SNP) in key cholesterol and TG regulating genes, in particular adenosine triphosphate binding cassette G8 (ABCG8) and apolipoprotein E (apoE) have provided insight into the potential of utilizing genomic identifiers as an indicator of PS responsiveness. While PS supplementation is deemed safe, expanding research into the atherogenic potential of oxidized phytosterols (oxyphytosterols) has emerged with their identification in arterial lesions. This review will highlight the lipid-lowering utility and associated mechanisms of PS and discuss novel applications and future research priorities for PS pertaining to in utero PS exposure for long-term cardiovascular disease risk protection and combination therapies with lipidlowering drugs.
Collapse
Affiliation(s)
- Jerad H Dumolt
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, 14214, United States
| | - Todd C Rideout
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, 14214, United States
| |
Collapse
|
26
|
Sulfated polysaccharides from the edible marine algae Padina tetrastromatica protects heart by ameliorating hyperlipidemia, endothelial dysfunction and inflammation in isoproterenol induced experimental myocardial infarction. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
27
|
Evidence of association of circulating epigenetic-sensitive biomarkers with suspected coronary heart disease evaluated by Cardiac Computed Tomography. PLoS One 2019; 14:e0210909. [PMID: 30673762 PMCID: PMC6343931 DOI: 10.1371/journal.pone.0210909] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/03/2019] [Indexed: 12/22/2022] Open
Abstract
Circulating biomarkers available in clinical practice do not allow to stratify patients with coronary heart disease (CHD) prior the onset of a clinically relevant event. We evaluated the methylation status of specific genomic segments and gene expression in peripheral blood of patients undergoing Cardiac Computed Tomography (CCT) for CHD (n = 95). We choose to investigate cholesterol metabolism. Methylation and gene expression of low density lipoprotein receptor (LDLR), sterol regulatory element-binding factor 2 (SREBF2) and ATP-binding cassette transporter 1 (ABCA1) were evaluated by qRT-PCR. Calcium score (CACS), stenosis degree, total plaque volume (TPV), calcified plaque volume (CPV), non-calcified plaque volume (NCPV) and plaque burden (PB) were assessed in all CHD patients (n = 65). The percentage of methylation at the specific analyzed segment of LDLR promoter was higher in CHD patients vs healthy subjects (HS) (n = 30) (p = 0.001). LDLR, SREBF2 and ABCA1 mRNAs were up-regulated in CHD patients vs HS (p = 0.02; p = 0.019; p = 0.008). SREBF2 was overexpressed in patients with coronary stenosis ≥50% vs subjects with stenosis <50% (p = 0.036). After adjustment for risk factors and clinical features, ABCA1 (p = 0.005) and SREBF2 (p = 0.010) gene expression were identified as independent predictors of CHD and severity. ROC curve analysis revealed a good performance of ABCA1 on predicting CHD (AUC = 0.768; p<0.001) and of SREBF2 for the prediction of disease severity (AUC = 0.815; p<0.001). Moreover, adjusted multivariate analysis demonstrated SREBF2 as independent predictor of CPV, NCPV and TPV (p = 0.022; p = 0.002 and p = 0.006) and ABCA1 as independent predictor of NCPV and TPV (p = 0.002 and p = 0.013). CHD presence and characteristics are related to selected circulating transcriptional and epigenetic-sensitive biomarkers linked to cholesterol pathway. More extensive analysis of CHD phenotypes and circulating biomarkers might improve and personalize cardiovascular risk stratification in the clinical settings.
Collapse
|
28
|
He Z, Lei L, Kwek E, Zhao Y, Liu J, Hao W, Zhu H, Liang N, Ma KY, Ho HM, He WS, Chen ZY. Ginger attenuates trimethylamine-N-oxide (TMAO)-exacerbated disturbance in cholesterol metabolism and vascular inflammation. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.10.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
29
|
Torres-Adorno AM, Vitrac H, Qi Y, Tan L, Levental KR, Fan YY, Yang P, Chapkin RS, Eckhardt BL, Ueno NT. Eicosapentaenoic acid in combination with EPHA2 inhibition shows efficacy in preclinical models of triple-negative breast cancer by disrupting cellular cholesterol efflux. Oncogene 2018; 38:2135-2150. [PMID: 30459358 PMCID: PMC6430703 DOI: 10.1038/s41388-018-0569-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/26/2018] [Accepted: 10/17/2018] [Indexed: 01/06/2023]
Abstract
Triple-negative breast cancer (TNBC), the most aggressive breast cancer subtype, currently lacks effective targeted therapy options. Eicosapentaenoic acid (EPA), an omega-3 fatty acid and constituent of fish oil, is a common supplement with anti-inflammatory properties. Although it is not a mainstream treatment, several preclinical studies have demonstrated that EPA exerts anti-tumor activity in breast cancer. However, against solid tumors, EPA as a monotherapy is clinically ineffective; thus, we sought to develop a novel targeted drug combination to bolster its therapeutic action against TNBC. Using a high-throughput functional siRNA screen, we identified Ephrin type-A receptor 2 (EPHA2), an oncogenic cell-surface receptor tyrosine kinase, as a therapeutic target that sensitizes TNBC cells to EPA. EPHA2 expression was uniquely elevated in TNBC cell lines and patient tumors. In independent functional expression studies in TNBC models, EPHA2 gene-silencing combined with EPA significantly reduced cell growth and enhanced apoptosis compared with monotherapies, both in vitro and in vivo. EPHA2 specific inhibitors similarly enhanced the therapeutic action of EPA. Finally, we identified that therapy-mediated apoptosis was attributed to a lethal increase in cancer cell membrane polarity due to ABCA1 inhibition and subsequent dysregulation of cholesterol homeostasis. This study provides new molecular and pre-clinical evidence to support a clinical evaluation of EPA combined with EPHA2 inhibition in patients with TNBC.
Collapse
Affiliation(s)
- Angie M Torres-Adorno
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Section of Translational Breast Cancer Research and Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heidi Vitrac
- Department of Biochemistry and Molecular Biology, UTHealth McGovern Medical School, Houston, TX, USA
| | - Yuan Qi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kandice R Levental
- Department of Integrative Biology and Pharmacology, UTHealth McGovern Medical School, Houston, TX, USA
| | - Yang-Yi Fan
- Program in Integrative Nutrition & Complex Diseases, Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Peiying Yang
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition & Complex Diseases, Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Bedrich L Eckhardt
- Section of Translational Breast Cancer Research and Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research and Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
30
|
DIAgnosis and Management Of familial hypercholesterolemia in a Nationwide Design (DIAMOND-FH): Prevalence in Switzerland, clinical characteristics and the diagnostic value of clinical scores. Atherosclerosis 2018; 277:282-288. [DOI: 10.1016/j.atherosclerosis.2018.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/25/2018] [Accepted: 08/17/2018] [Indexed: 11/20/2022]
|
31
|
Choi HK, Hwang JT, Nam TG, Kim SH, Min DK, Park SW, Chung MY. Welsh onion extract inhibits PCSK9 expression contributing to the maintenance of the LDLR level under lipid depletion conditions of HepG2 cells. Food Funct 2018; 8:4582-4591. [PMID: 29130084 DOI: 10.1039/c7fo00562h] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Statins mediate the transactivation of PCSK9, which in turn limits their cholesterol-lowering effects via LDL receptor (LDLR) degradation. The objective of the present study was to investigate the mechanism of action by which Welsh onion (Allium fistulosum L. [family Amaryllidaceae]) extract (WOE) regulates LDLR and PCSK9. HepG2 cells were cultured under lipid depletion conditions using a medium supplemented with delipidated serum (DLPS). WOE (50, 100, 200, and 400 μg ml-1) significantly attenuated the DLPS-mediated increases in LDLR, PCSK9, and SREBP2 gene expression. While WOE treatment maintained the DLPS-mediated increases in LDLR protein expression, it dose-dependently and significantly attenuated the DLPS-mediated increases in the protein content of PCSK9. The suppression of PCSK9 was associated with the WOE-mediated reductions in SREBP2, but not HNF1α. WOE also dose-dependently reduced PCSK9 protein expression that was otherwise markedly induced by concomitant statin treatment. WOE-mediated PCSK9 inhibition contributed to LDLR lysosomal degradation suppression, and subsequent LDLR protein stabilization. HPLC analysis indicated that WOE contains kaempferol, quercetin, ferulic acid, and p-coumaric acid. Kaempferol and p-coumaric acid contributed to the maintenance of LDLR expression by inhibiting PCSK9 in lipid depleted HepG2 cells. Altogether, these findings suggest that WOE inhibits PCSK9 transcription and protein expression via the reduction of SREBP2, and decreased PCSK9 further contributes to LDLR degradation prevention and LDLR protein stabilization under conditions of lipoprotein deficiency. The PCSK9 inhibition-mediated mechanism of WOE was likely attributed to the action of kaempferol and p-coumaric acid present in WOE.
Collapse
Affiliation(s)
- Hyo-Kyoung Choi
- Korea Food Research Institute, Seongnam City, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|
32
|
Hernández Flores TDJ, González García JR, Colima Fausto AG, Vázquez Cárdenas NA, Sánchez López Y, Zarate Morales CA, Magaña Torres MT. Screening of LDLR and APOB gene mutations in Mexican patients with homozygous familial hypercholesterolemia. J Clin Lipidol 2018; 12:693-701. [PMID: 29576406 DOI: 10.1016/j.jacl.2018.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/20/2018] [Accepted: 02/24/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is an autosomal dominant disorder that causes accumulation of serum low-density lipoprotein cholesterol and premature cardiovascular disease. It is mainly related to mutations in the LDLR gene. Homozygous FH (HoFH) patients have the most severe form of the disease accounting for a worldwide prevalence of 1:1,000,000. In Mexico, at least 5 cases of HoFH have been reported. OBJECTIVE The aim of this study was to describe the clinical, biochemical, and molecular data observed in patients with HoFH phenotype. METHODS We included 13 patients, belonging to 11 families, with clinical and biochemical diagnoses suggestive of HoFH. Molecular analyses of the LDLR and APOB genes were performed by means of polymerase chain reaction followed by Sanger sequencing. RESULTS The causal mutation of HoFH was found in 8 of 11 unrelated patients. Excepting 1, all were true homozygotes. Six different variants in LDLR were identified: c.-139delCTCCCCCTGC, p.Glu140Lys, p.Asp360His, p.Asn405Lys, p.Ala755Glyfs*7, and p.Leu759Serfs*6. Of these, p.Asp360His and p.Asn405Lys were detected for the first time in Mexico; p.Leu759Serfs*6 showed to be the most frequent (43.7% of the alleles 7/16), and c.-139delCTCCCCCTGC is a new variant located in the promoter region. CONCLUSIONS This work increases knowledge of biochemical and genetic features in Mexican patients with HoFH. A novel mutation in the LDLR gene promoter was detected: c.-139delCTCCCCCTGC, which possibly inhibits its expression.
Collapse
Affiliation(s)
- Teresita De Jesús Hernández Flores
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México; Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Juan Ramón González García
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Ana Gabriela Colima Fausto
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México; Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | | | - Yoaly Sánchez López
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México; Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - César Augusto Zarate Morales
- Hospital "Presidente Juárez" del Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Oaxaca, Oax, México
| | - María Teresa Magaña Torres
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México; Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México.
| |
Collapse
|
33
|
Rafeeq MM, Habib HS, Murad HAS, Gari MA, Gazzaz ZJ. Effect of genetic polymorphisms in SREBF-SCAP pathway on therapeutic response to rosuvastatin in Saudi metabolic syndrome patients. Pharmacogenomics 2018; 19:185-196. [PMID: 29318930 DOI: 10.2217/pgs-2017-0181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Genetic variants contribute to statins' therapeutic variability. SREBF-SCAP pathway is a key player in lipid homeostasis. Hence, effect of SREBF-SCAP polymorphisms on therapeutic response was studied. PATIENTS & METHODS Metabolic syndrome patients of either sex were prescribed rosuvastatin 10 mg for 24 weeks. Clinical, anthropometric and lipid measurements were done before and after treatment. Genotyping was done by pyrosequencing. RESULTS & CONCLUSION No associations of SCAP and SREBF-1a genotypes with baseline lipids but significant associations with lipid reductions were observed. Significant effect of SCAP (GG; B = -8.16, p = 0.001); SREBF-1a (GG; B = -7.47, p = 0.001) and SREBF-1a (-delG; B = -7.42, p = 0.001) was observed on total cholesterol reduction. Additive trend was found between SCAP genotypes and lipid reductions. A total of 88% responders have SCAP 'G' allele (p = 0.001). Patients carrying SCAP (GG) and SREBF-1a (GG and -delG) have 9.5-, 8.6- and 14.6-times more likelihood of being responders (p < 0.05). 'G' allele in SCAP and SREBF-1a is significant predictor of rosuvastatin response.
Collapse
Affiliation(s)
- Misbahuddin Mohd Rafeeq
- Department of Pharmacology, Faculty of Medicine, Rabigh Campus, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hamed Said Habib
- Department of Paediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hussam Aly Sayed Murad
- Department of Pharmacology, Faculty of Medicine, Rabigh Campus, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Rabigh Campus, King Abdulaziz University, Jeddah, Saudi Arabia & Faculty of Medicine, Ainshams University, Cairo, Egypt
| | - Mamdouh Abdullah Gari
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zohair Jamil Gazzaz
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
34
|
Wang D, Luo Y, Myakala K, Orlicky DJ, Dobrinskikh E, Wang X, Levi M. Serelaxin improves cardiac and renal function in DOCA-salt hypertensive rats. Sci Rep 2017; 7:9793. [PMID: 28851937 PMCID: PMC5574886 DOI: 10.1038/s41598-017-09470-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/26/2017] [Indexed: 11/09/2022] Open
Abstract
Serelaxin, a recombinant form of the naturally occurring peptide hormone relaxin-2, is a pleiotropic vasodilating hormone that has been studied in patients with acute heart failure. In this study, the effects of serelaxin on cardiac and renal function, fibrosis, inflammation and lipid accumulation were studied in DOCA-salt treated rats. Uninephrectomized rats were assigned to two groups: controls provided with normal drinking water and DOCA provided with DOCA pellets and sodium chloride drinking water. After 4 weeks, the DOCA-salt rats were randomly selected and implanted with osmotic minipumps delivering vehicle or serelaxin for another 4 weeks. Treatment with serelaxin prevented cardiac and renal dysfunction in DOCA-salt rats. Serelaxin prevented cardiac and renal fibrosis, as determined by Picrosirius Red staining and Second Harmonic Generation (SHG) Microscopy. Treatment of DOCA-salt rats with serelaxin decreased renal inflammation, including the expression of TGF-β, NFκB, MCP-1, IL-1, IL-6, ICAM-1, VCAM-1 and CD68 macrophages. Serelaxin also decreased lipid accumulation in kidney in part by decreasing SREBP-1c, SREBP-2, ChREBP, FATP1, HMGCoAR, and LDL receptor, and increasing Acox1 and ABCA1. In summary, serelaxin reversed DOCA-salt induced cardiac and renal dysfunction.
Collapse
Affiliation(s)
- Dong Wang
- Renal Diseases and Hypertension, School of Medicine, University of Colorado ANSCHUTZ MEDICAL CAMPUS, Aurora, Colorado, 80045, USA
| | - Yuhuan Luo
- Renal Diseases and Hypertension, School of Medicine, University of Colorado ANSCHUTZ MEDICAL CAMPUS, Aurora, Colorado, 80045, USA
| | - Komuraiah Myakala
- Renal Diseases and Hypertension, School of Medicine, University of Colorado ANSCHUTZ MEDICAL CAMPUS, Aurora, Colorado, 80045, USA
| | - David J Orlicky
- Department of Pathology, School of Medicine, University of Colorado ANSCHUTZ MEDICAL CAMPUS, Aurora, Colorado, 80045, USA
| | - Evgenia Dobrinskikh
- Renal Diseases and Hypertension, School of Medicine, University of Colorado ANSCHUTZ MEDICAL CAMPUS, Aurora, Colorado, 80045, USA
| | - Xiaoxin Wang
- Renal Diseases and Hypertension, School of Medicine, University of Colorado ANSCHUTZ MEDICAL CAMPUS, Aurora, Colorado, 80045, USA
| | - Moshe Levi
- Renal Diseases and Hypertension, School of Medicine, University of Colorado ANSCHUTZ MEDICAL CAMPUS, Aurora, Colorado, 80045, USA.
| |
Collapse
|
35
|
Hypercholesterolemia: The role of PCSK9. Arch Biochem Biophys 2017; 625-626:39-53. [DOI: 10.1016/j.abb.2017.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/29/2017] [Accepted: 06/02/2017] [Indexed: 01/06/2023]
|
36
|
Jeyakumar SM, Sheril A, Vajreswari A. Chronic vitamin A-enriched diet feeding regulates hypercholesterolaemia through transcriptional regulation of reverse cholesterol transport pathway genes in obese rat model of WNIN/GR-Ob strain. Indian J Med Res 2016; 144:238-244. [PMID: 27934803 PMCID: PMC5206875 DOI: 10.4103/0971-5916.195038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background & objectives: Hepatic scavenger receptor class B1 (SR-B1), a high-density lipoprotein (HDL) receptor, is involved in the selective uptake of HDL-associated esterified cholesterol (EC), thereby regulates cholesterol homoeostasis and improves reverse cholesterol transport. Previously, we reported in euglycaemic obese rats (WNIN/Ob strain) that feeding of vitamin A-enriched diet normalized hypercholesterolaemia, possibly through hepatic SR-B1-mediated pathway. This study was aimed to test whether it would be possible to normalize hypercholesterolaemia in glucose-intolerant obese rat model (WNIN/GR/Ob) through similar mechanism by feeding identical vitamin A-enriched diet. Methods: In this study, 30 wk old male lean and obese rats of WNIN/GR-Ob strain were divided into two groups and received either stock diet or vitamin A-enriched diet (2.6 mg or 129 mg vitamin A/kg diet) for 14 wk. Blood and other tissues were collected for various biochemical analyses. Results: Chronic vitamin A-enriched diet feeding decreased hypercholesterolaemia and normalized abnormally elevated plasma HDL-cholesterol (HDL-C) levels in obese rats as compared to stock diet-fed obese groups. Further, decreased free cholesterol (FC) and increased esterified cholesterol (EC) contents of plasma cholesterol were observed, which were reflected in higher EC to FC ratio of vitamin A-enriched diet-fed obese rats. However, neither lecithin-cholesterol acyltransferase (LCAT) activity of plasma nor its expression (both gene and protein) in the liver were altered. On the contrary, hepatic cholesterol levels significantly increased in vitamin A-enriched diet fed obese rats. Hepatic SR-B1 expression (both mRNA and protein) remained unaltered among groups. Vitamin A-enriched diet fed obese rats showed a significant increase in hepatic low-density lipoprotein receptor mRNA levels, while the expression of genes involved in HDL synthesis, namely, ATP-binding cassette protein 1 (ABCA1) and apolipoprotein A-I, were downregulated. No such response was seen in vitamin A-supplemented lean rats as compared with their stock diet-fed lean counterparts. Interpretation & conclusions: Chronic vitamin A-enriched diet feeding decreased hypercholesterolaemia and normalized HDL-C levels, possibly by regulating pathways involved in HDL synthesis and degradation, independent of hepatic SR-B1 in this glucose-intolerant obese rat model.
Collapse
Affiliation(s)
| | - Alex Sheril
- Division of Lipid Biochemistry, National Institute of Nutrition, Hyderabad, India
| | | |
Collapse
|
37
|
Dietary high oleic canola oil supplemented with docosahexaenoic acid attenuates plasma proprotein convertase subtilisin kexin type 9 (PCSK9) levels in participants with cardiovascular disease risk: A randomized control trial. Vascul Pharmacol 2016; 87:60-65. [DOI: 10.1016/j.vph.2016.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/21/2016] [Accepted: 06/26/2016] [Indexed: 12/13/2022]
|
38
|
Hussain A, Yadav MK, Bose S, Wang JH, Lim D, Song YK, Ko SG, Kim H. Daesiho-Tang Is an Effective Herbal Formulation in Attenuation of Obesity in Mice through Alteration of Gene Expression and Modulation of Intestinal Microbiota. PLoS One 2016; 11:e0165483. [PMID: 27812119 PMCID: PMC5094769 DOI: 10.1371/journal.pone.0165483] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/12/2016] [Indexed: 12/16/2022] Open
Abstract
Background Obesity has become a major global health challenge due to its increasing prevalence, and the associated health risk. It is the main cause of various metabolic diseases including diabetes, hypertension, cardiovascular disease, stroke and certain forms of cancer. Methods and Results In the present study we evaluated the anti-obesity property of Daesiho-tang (DSHT), an herbal medicine, using high fat diet (HFD)-induced obese mice as a model. Our results showed that DSHT ameliorated body weight gain, decreased total body fat, regulated expression of leptin and adiponectin genes of adipose tissue and exerted an anti-diabetic effect by attenuating fasting glucose level and serum insulin level in HFD-fed animals. In addition, DSHT-treatment significantly reduced total cholesterol (TC), triglycerides (TG) and increased high density lipoprotein-cholesterol (HDL), glutamic pyruvic transaminase (GPT) and glutamic oxaloacetic transaminase (GOT) levels in serum and reduced deposition of fat droplets in liver. DSHT treatment resulted in significantly increased relative abundance of bacteria including Bacteroidetes, Bacteroidetes/Firmicutes ratio, Akkermansia Bifidobacterium., Lactobacillus, and decreased the level of Firmicutes. Using RT2 profiler PCR array, 39 (46%) genes were found to be differentially expressed in HFD-fed mice compared to normal control. However, normal gene expressions were restored in 36 (92%) genes of HFD-fed mice, when co-exposed to DSHT. Conclusion/Major Findings The results of this study demonstrated that DSHT is an effective herbal formulation in attenuation of obesity in HFD-fed mice through alteration of gene expressions and modulation of intestinal microbiota.
Collapse
Affiliation(s)
- Ahtesham Hussain
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, Goyang, Republic of Korea
| | - Mukesh Kumar Yadav
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
- Institute for Medical Device Clinical Trials, Korea University College of Medicine, Seoul, Republic of Korea
| | - Shambhunath Bose
- Applied Surface Technology Inc., 11th Floor, Bldg. A, Advance Institutes of Convergence Technology, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Jing-Hua Wang
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, Goyang, Republic of Korea
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Traditional Chinese Medicine, Meishan Road 103, Hefei, Anhui Province, People’s Republic of China
| | - Dongwoo Lim
- Department of pathology, College of Korean Medicine, Dongguk University, Goyang, Republic of Korea
| | - Yun-Kyung Song
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Gachon University, Incheon, Republic of Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyunghee University, Seoul, Republic of Korea
| | - Hojun Kim
- Department of Rehabilitation Medicine of Korean Medicine, Dongguk University, Goyang, Republic of Korea
- * E-mail:
| |
Collapse
|
39
|
Intracellular cholesterol transport proteins: roles in health and disease. Clin Sci (Lond) 2016; 130:1843-59. [DOI: 10.1042/cs20160339] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/12/2016] [Indexed: 12/13/2022]
Abstract
Effective cholesterol homoeostasis is essential in maintaining cellular function, and this is achieved by a network of lipid-responsive nuclear transcription factors, and enzymes, receptors and transporters subject to post-transcriptional and post-translational regulation, whereas loss of these elegant, tightly regulated homoeostatic responses is integral to disease pathologies. Recent data suggest that sterol-binding sensors, exchangers and transporters contribute to regulation of cellular cholesterol homoeostasis and that genetic overexpression or deletion, or mutations, in a number of these proteins are linked with diseases, including atherosclerosis, dyslipidaemia, diabetes, congenital lipoid adrenal hyperplasia, cancer, autosomal dominant hearing loss and male infertility. This review focuses on current evidence exploring the function of members of the ‘START’ (steroidogenic acute regulatory protein-related lipid transfer) and ‘ORP’ (oxysterol-binding protein-related proteins) families of sterol-binding proteins in sterol homoeostasis in eukaryotic cells, and the evidence that they represent valid therapeutic targets to alleviate human disease.
Collapse
|
40
|
Henderson R, O'Kane M, McGilligan V, Watterson S. The genetics and screening of familial hypercholesterolaemia. J Biomed Sci 2016; 23:39. [PMID: 27084339 PMCID: PMC4833930 DOI: 10.1186/s12929-016-0256-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/03/2016] [Indexed: 11/14/2022] Open
Abstract
Familial Hypercholesterolaemia is an autosomal, dominant genetic disorder that leads to elevated blood cholesterol and a dramatically increased risk of atherosclerosis. It is perceived as a rare condition. However it affects 1 in 250 of the population globally, making it an important public health concern. In communities with founder effects, higher disease prevalences are observed. We discuss the genetic basis of familial hypercholesterolaemia, examining the distribution of variants known to be associated with the condition across the exons of the genes LDLR, ApoB, PCSK9 and LDLRAP1. We also discuss screening programmes for familial hypercholesterolaemia and their cost-effectiveness. Diagnosis typically occurs using one of the Dutch Lipid Clinic Network (DCLN), Simon Broome Register (SBR) or Make Early Diagnosis to Prevent Early Death (MEDPED) criteria, each of which requires a different set of patient data. New cases can be identified by screening the family members of an index case that has been identified as a result of referral to a lipid clinic in a process called cascade screening. Alternatively, universal screening may be used whereby a population is systematically screened. It is currently significantly more cost effective to identify familial hypercholesterolaemia cases through cascade screening than universal screening. However, the cost of sequencing patient DNA has fallen dramatically in recent years and if the rate of progress continues, this may change.
Collapse
Affiliation(s)
- Raymond Henderson
- Northern Ireland Centre for Stratified Medicine, Ulster University, C-TRIC, Altnagelvin Hospital Campus, Derry, Co Londonderry, Northern Ireland, BT47 6SB, UK
| | - Maurice O'Kane
- Department of Clinical Chemistry, Altnagelvin Hospital, Western Health and Social Care Trust, Londonderry, Northern Ireland, BT47 6SB, UK
| | - Victoria McGilligan
- Northern Ireland Centre for Stratified Medicine, Ulster University, C-TRIC, Altnagelvin Hospital Campus, Derry, Co Londonderry, Northern Ireland, BT47 6SB, UK
| | - Steven Watterson
- Northern Ireland Centre for Stratified Medicine, Ulster University, C-TRIC, Altnagelvin Hospital Campus, Derry, Co Londonderry, Northern Ireland, BT47 6SB, UK.
| |
Collapse
|
41
|
Obeidat M, Fishbane N, Nie Y, Chen V, Hollander Z, Tebbutt SJ, Bossé Y, Ng RT, Miller BE, McManus B, Rennard S, Paré PD, Sin DD. The Effect of Statins on Blood Gene Expression in COPD. PLoS One 2015; 10:e0140022. [PMID: 26462087 PMCID: PMC4604084 DOI: 10.1371/journal.pone.0140022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/19/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND COPD is currently the fourth leading cause of death worldwide. Statins are lipid lowering agents with documented cardiovascular benefits. Observational studies have shown that statins may have a beneficial role in COPD. The impact of statins on blood gene expression from COPD patients is largely unknown. OBJECTIVE Identify blood gene signature associated with statin use in COPD patients, and the pathways underpinning this signature that could explain any potential benefits in COPD. METHODS Whole blood gene expression was measured on 168 statin users and 451 non-users from the ECLIPSE study using the Affymetrix Human Gene 1.1 ST microarray chips. Factor Analysis for Robust Microarray Summarization (FARMS) was used to process the expression data. Differential gene expression analysis was undertaken using the Linear Models for Microarray data (Limma) package adjusting for propensity score and surrogate variables. Similarity of the expression signal with published gene expression profiles was performed in ProfileChaser. RESULTS 25 genes were differentially expressed between statin users and non-users at an FDR of 10%, including LDLR, CXCR2, SC4MOL, FAM108A1, IFI35, FRYL, ABCG1, MYLIP, and DHCR24. The 25 genes were significantly enriched in cholesterol homeostasis and metabolism pathways. The resulting gene signature showed correlation with Huntington's disease, Parkinson's disease and acute myeloid leukemia gene signatures. CONCLUSION The blood gene signature of statins' use in COPD patients was enriched in cholesterol homeostasis pathways. Further studies are needed to delineate the role of these pathways in lung biology.
Collapse
Affiliation(s)
- Ma’en Obeidat
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
| | - Nick Fishbane
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
| | - Yunlong Nie
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
| | - Virginia Chen
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
| | - Zsuzsanna Hollander
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
| | - Scott J. Tebbutt
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
- Respiratory Division, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec, Department of Molecular Medicine, Laval University, Québec, Canada
| | - Raymond T. Ng
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
| | - Bruce E. Miller
- Respiratory Therapy Area Unit, GlaxoSmithKline R&D, King of Prussia, Pennsylvania, United States of America
| | - Bruce McManus
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
| | - Stephen Rennard
- Division of Pulmonary and Critical Care Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Peter D. Paré
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Respiratory Division, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Don D. Sin
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Respiratory Division, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
42
|
Barley sprout extract containing policosanols and polyphenols regulate AMPK, SREBP2 and ACAT2 activity and cholesterol and glucose metabolism in vitro and in vivo. Food Res Int 2015. [DOI: 10.1016/j.foodres.2015.03.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
43
|
Wang Y, Tong J, Chang B, Wang BF, Zhang D, Wang BY. Relationship of SREBP-2 rs2228314 G>C polymorphism with nonalcoholic fatty liver disease in a Han Chinese population. Genet Test Mol Biomarkers 2014; 18:653-7. [PMID: 24992162 DOI: 10.1089/gtmb.2014.0116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE This study aims at investigating the relationship of SREBP-2 rs2228314 G>C polymorphism with the risk of nonalcoholic fatty liver disease (NAFLD) in a Han Chinese population. METHOD This case-control study was conducted at the First Affiliated Hospital of China Medical University. Three-hundred subjects who met the diagnostic criteria of NAFLD and had typical clinical and ultrasonographic findings were placed in the case group. There were 160 matched healthy controls in the control group. A common single nucleotide polymorphism (SNP) (rs2228314 G>C) in the SREBP-2 gene was tested. Genetic analyses were performed using genomic DNA extracted from peripheral blood leukocytes. Polymerase chain reaction-restriction fragment length polymorphism was applied to detect SNP. RESULTS Our results indicated that the GG genotype and G carrier (CG+GG) of rs2228314 G>C polymorphism in the SREBP-2 gene were strongly associated with susceptibility to NAFLD (both p<0.001). However, there was no significant difference in the frequency of G allele between NAFLD patients and healthy controls (p=0.328). Multivariate logistic regression analysis revealed that GG genotype, G carrier, body mass index, high-density lipoprotein cholesterol, total cholesterol, alanine aminotransferase, and γ-glutamyl-transferase might be associated with an increased risk of NAFLD (all p<0.05). CONCLUSION The results of this study provide evidence that the GG genotype and G carrier (CG+GG) of rs2228314 G>C polymorphism in the SREBP-2 gene may increase the risk of NAFLD.
Collapse
Affiliation(s)
- Ying Wang
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University , Shenyang, People's Republic of China
| | | | | | | | | | | |
Collapse
|
44
|
Inhibition of proprotein convertase subtilisin/kexin type 9: A novel mechanism of berberine and 8-hydroxy dihydroberberine against hyperlipidemia. Chin J Integr Med 2014; 21:132-8. [DOI: 10.1007/s11655-014-1775-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Indexed: 10/25/2022]
|
45
|
Zhang X, Liu J, Su W, Wu J, Wang C, Kong X, Gustafsson JÅ, Ding J, Ma X, Guan Y. Liver X receptor activation increases hepatic fatty acid desaturation by the induction of SCD1 expression through an LXRα-SREBP1c-dependent mechanism. J Diabetes 2014; 6:212-20. [PMID: 23945440 DOI: 10.1111/1753-0407.12081] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 08/01/2013] [Accepted: 08/12/2013] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Liver X receptors (LXRs) including LXRα and LXRβ are members of the nuclear hormone receptor superfamily of ligand activated transcription factors, which serve as lipid sensors to regulate expression of genes controlling many aspects of cholesterol and fatty acid metabolism. The liver is the central organ in controlling lipid metabolism. In the present study, we aimed at elucidating the role of LXR activation in hepatic fatty acid homeostasis. METHODS We treated C57BL/6 mice with a synthetic non-selective LXR agonist TO901317. Fatty acid profile of lipid esters in the livers was analyzed by gas-liquid chromatography. Real-time polymerase chain reaction (PCR) and western blot were used to determine the expression of SREBP1c and SCD1 in TO901317-treated livers and HepG2 cells. RESULTS Oral administration of TO901317 resulted in increased fatty acid desaturation in the liver, with concomitant increase in hepatic stearoyl CoA desaturase-1 (SCD1) expression. TO901317-induced SCD1 expression was observed in LXRβ-/- mice, but not in LXRα-/- mice. Furthermore, TO901317 significantly increased expression of sterol regulatory element-binding protein 1c (SREBP1c), the deficiency of which almost completely abolished the induction of SCD1 by TO901317. This drug induced both SREBP1c and SCD1 expression in HepG2 cells. Overexpression of SREBP1c resulted in a significant increase in SCD1 promoter activity and expression. CONCLUSIONS Taken together, the present studies demonstrate that pan-LXR activation increases hepatic fatty acid desaturation via the induction of SCD1 expression in an LXRα-dependent and SREBP1c-mediated manner.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China; Shenzhen University Diabetes Center, Shenzhen University Health Science Center, Shenzhen, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Influence of SREBP-2 and SCAP Gene Polymorphisms on Lipid-Lowering Response to Atorvastatin in a Cohort of Chilean Subjects with Amerindian Background. Mol Diagn Ther 2014; 18:435-43. [DOI: 10.1007/s40291-014-0094-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Silva LSE, de Miranda AM, de Brito Magalhães CL, dos Santos RC, Pedrosa ML, Silva ME. Diet supplementation with beta-carotene improves the serum lipid profile in rats fed a cholesterol-enriched diet. J Physiol Biochem 2013; 69:811-20. [DOI: 10.1007/s13105-013-0257-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 04/22/2013] [Indexed: 01/19/2023]
|
48
|
de Souza MO, Souza E Silva L, de Brito Magalhães CL, de Figueiredo BB, Costa DC, Silva ME, Pedrosa ML. The hypocholesterolemic activity of açaí (Euterpe oleracea Mart.) is mediated by the enhanced expression of the ATP-binding cassette, subfamily G transporters 5 and 8 and low-density lipoprotein receptor genes in the rat. Nutr Res 2012; 32:976-84. [PMID: 23244543 DOI: 10.1016/j.nutres.2012.10.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 09/28/2012] [Accepted: 10/01/2012] [Indexed: 01/01/2023]
Abstract
Previous studies have demonstrated that the ingestion of açaí pulp can improve serum lipid profile in various animal models; therefore, we hypothesized that açaí pulp (Euterpe oleracea Mart.) may modulate the expression of the genes involved in cholesterol homeostasis in the liver and increase fecal excretion, thus reducing serum cholesterol. To test this hypothesis, we analyzed the expression of 7α-hydroxylase and ATP-binding cassette, subfamily G transporters (ABCG5 and ABCG8), which are genes involved with the secretion of cholesterol in the rat. We also evaluated the expression of sterol regulatory element-binding protein 2, 3-hydroxy-3-methylglutaryl CoA reductase, low-density lipoprotein receptor (LDL-R), and apolipoprotein B100, which are involved in cholesterol biosynthesis. Female Fischer rats were divided into 4 groups: the C group, which was fed a standard AIN-93 M diet; the CA group, which was fed a standard diet supplemented with 2% açaí pulp; the H group, which was fed a hypercholesterolemic diet (25% soy oil and 1% cholesterol); and the HA group, which was fed a hypercholesterolemic diet supplemented with 2% açaí pulp. At the end of the experimental period, the rats were euthanized, and their blood and livers were collected. The HA group exhibited a significant decrease in serum total cholesterol, low-density lipoprotein cholesterol, and atherogenic index and also had increased high-density lipoprotein cholesterol and cholesterol excretion in feces compared with the H group. In addition, the expression of the LDL-R, ABCG5, and ABCG8 genes was significantly increased by the presence of açaí pulp. These results suggest that açaí pulp promotes a hypocholesterolemic effect in a rat model of dietary-induced hypercholesterolemia through an increase in the expression of ATP-binding cassette, subfamily G transporters, and LDL-R genes.
Collapse
Affiliation(s)
- Melina Oliveira de Souza
- Research Center in Biological Sciences (NUPEB), Federal University of Ouro Preto, Minas Gerais, Brazil
| | | | | | | | | | | | | |
Collapse
|
49
|
Kostopoulou F, Gkretsi V, Malizos KN, Iliopoulos D, Oikonomou P, Poultsides L, Tsezou A. Central role of SREBP-2 in the pathogenesis of osteoarthritis. PLoS One 2012; 7:e35753. [PMID: 22662110 PMCID: PMC3360703 DOI: 10.1371/journal.pone.0035753] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 03/25/2012] [Indexed: 01/01/2023] Open
Abstract
Background Recent studies have implied that osteoarthritis (OA) is a metabolic disease linked to deregulation of genes involved in lipid metabolism and cholesterol efflux. Sterol Regulatory Element Binding Proteins (SREBPs) are transcription factors regulating lipid metabolism with so far no association with OA. Our aim was to test the hypothesis that SREBP-2, a gene that plays a key role in cholesterol homeostasis, is crucially involved in OA pathogenesis and to identify possible mechanisms of action. Methodology/Principal Findings We performed a genetic association analysis using a cohort of 1,410 Greek OA patients and healthy controls and found significant association between single nucleotide polymorphism (SNP) 1784G>C in SREBP-2 gene and OA development. Moreover, the above SNP was functionally active, as normal chondrocytes’ transfection with SREBP-2-G/C plasmid resulted in interleukin-1β and metalloproteinase-13 (MMP-13) upregulation. We also evaluated SREBP-2, its target gene 3-hydroxy-3-methylglutaryl-coenzymeA reductase (HMGCR), phospho-phosphoinositide3-kinase (PI3K), phospho-Akt, integrin-alphaV (ITGAV) and transforming growth factor-β (TGF-β) mRNA and protein expression levels in osteoarthritic and normal chondrocytes and found that they were all significantly elevated in OA chondrocytes. To test whether TGF-β alone can induce SREBP-2, we treated normal chondrocytes with TGF-β and found significant upregulation of SREBP-2, HMGCR, phospho-PI3K and MMP-13. We also showed that TGF-β activated aggrecan (ACAN) in chondrocytes only through Smad3, which interacts with SREBP-2. Finally, we examined the effect of an integrin inhibitor, cyclo-RGDFV peptide, on osteoarthritic chondrocytes, and found that it resulted in significant upregulation of ACAN and downregulation of SREBP-2, HMGCR, phospho-PI3K and MMP-13 expression levels. Conclusions/Significance We demonstrated, for the first time, the association of SREBP-2 with OA pathogenesis and provided evidence on the molecular mechanism involved. We suggest that TGF-β induces SREBP-2 pathway activation through ITGAV and PI3K playing a key role in OA and that integrin blockage may be a potential molecular target for OA treatment.
Collapse
Affiliation(s)
- Fotini Kostopoulou
- Department of Cytogenetics and Molecular Genetics, University of Thessaly School of Medicine, Larissa, Greece
- Institute of Biomedical Research and Technology, Center for Research and Technology-Thessaly, Larissa, Greece
| | - Vasiliki Gkretsi
- Institute of Biomedical Research and Technology, Center for Research and Technology-Thessaly, Larissa, Greece
| | - Konstantinos N. Malizos
- Institute of Biomedical Research and Technology, Center for Research and Technology-Thessaly, Larissa, Greece
- Department of Orthopaedics, University of Thessaly School of Medicine, Larissa, Greece
| | - Dimitrios Iliopoulos
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute and Department of Pathology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Pagona Oikonomou
- Institute of Biomedical Research and Technology, Center for Research and Technology-Thessaly, Larissa, Greece
| | - Lazaros Poultsides
- Department of Orthopaedics, University of Thessaly School of Medicine, Larissa, Greece
| | - Aspasia Tsezou
- Department of Cytogenetics and Molecular Genetics, University of Thessaly School of Medicine, Larissa, Greece
- Institute of Biomedical Research and Technology, Center for Research and Technology-Thessaly, Larissa, Greece
- Department of Biology, University of Thessaly School of Medicine, Larissa, Greece
- * E-mail:
| |
Collapse
|
50
|
Jin X, Zeng F, Zhang N, Huang T, Meng Q, Liu Y. Association of Sterol Regulatory Element-Binding Transcription Factor Gene Polymorphisms with Ischaemic Stroke. J Int Med Res 2012; 40:157-66. [PMID: 22429355 DOI: 10.1177/147323001204000116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE: To explore the association between polymorphisms of the sterol regulatory element-binding transcription factor ( SREBF) gene and ischaemic stroke. METHODS: The SREBF1c 54G>C and SREBPF2 1784G>C genotypes were assessed using restriction fragment length polymorphism analysis in 446 Han Chinese ischaemic stroke patients and 355 Han Chinese control subjects without cerebrovascular disease. RESULTS: The frequencies of the SREBF2 1784G>C CC genotype and the C allele were significantly higher in the ischaemic stroke group than in controls. Patients with ischaemic stroke who had the SREBF2 1784G>C CC genotype had significantly lower high-density lipoprotein (HDL) levels, compared with ischaemic stroke patients and control subjects with the GC or GG genotypes. Multivariate logistic regression analysis revealed a significant positive association between SREBF2 1784G>C and ischaemic stroke; an inverse association was observed between HDL level and risk of ischaemic stroke. CONCLUSIONS: The CC genotype of the SREBF2 1784G>C polymorphism was associated with an increased risk of ischaemic stroke, possibly through decreasing the HDL level, which was inversely associated with the risk of ischaemic stroke.
Collapse
Affiliation(s)
- X Jin
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - F Zeng
- Department of Emergency Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - N Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - T Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Q Meng
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Y Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|