1
|
Lee Y, Gjerdevik M, Jugessur A, Gjessing HK, Corfield E, Havdahl A, Harris JR, Magnus MC, Håberg SE, Magnus P. Parent-of-Origin Effects in Childhood Asthma at Seven Years of Age. Genet Epidemiol 2025; 49:e70007. [PMID: 40133993 PMCID: PMC11937430 DOI: 10.1002/gepi.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 01/06/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
Childhood asthma is more common among children whose mothers have asthma than among those whose fathers have asthma. The reasons for this are unknown, and we hypothesize that genomic imprinting may partly explain this observation. Our aim is to assess parent-of-origin (PoO) effects on childhood asthma by analyzing SNP array genotype data from a large population-based cohort. To estimate PoO effects in parent-reported childhood asthma at 7 years of age, we fit a log-linear model implemented in the HAPLIN R package to SNP array genotype data from 915 mother-father-child case triads, 603 mother-child case dyads, and 113 father-child case dyads participating in the Norwegian Mother, Father, and Child Cohort Study (MoBa). We found that alleles at two SNPs-rs3003214 and rs3003211-near the adenylosuccinate synthase 2 gene (ADSS2 on chromosome 1q44) showed significant PoO effects at a false positive rate ≤ 0.05. The ratio of the effect of the maternally and paternally inherited G-allele at rs3003214 was 1.68 (95% CI: 1.41-2.03, p value = 1.13E-08). Our results suggest PoO effects at the ADSS2 gene, particularly the maternally inherited G-allele at rs3003214, may contribute to the maternal effect in childhood asthma.
Collapse
Affiliation(s)
- Yunsung Lee
- Centre for Fertility and HealthNorwegian Institute of Public HealthOsloNorway
| | - Miriam Gjerdevik
- Centre for Fertility and HealthNorwegian Institute of Public HealthOsloNorway
- Department of Computer Science, Electrical Engineering and Mathematical SciencesWestern Norway University of Applied SciencesBergenNorway
| | - Astanand Jugessur
- Centre for Fertility and HealthNorwegian Institute of Public HealthOsloNorway
- Department of Global Public Health and Primary CareUniversity of BergenBergenNorway
| | - Håkon Kristian Gjessing
- Centre for Fertility and HealthNorwegian Institute of Public HealthOsloNorway
- Department of Global Public Health and Primary CareUniversity of BergenBergenNorway
| | - Elizabeth Corfield
- PsychGen Centre for Genetic Epidemiology and Mental HealthNorwegian Institute of Public HealthOsloNorway
- Nic Waals InstituteLovisenberg Diaconal HospitalOsloNorway
| | - Alexandra Havdahl
- PsychGen Centre for Genetic Epidemiology and Mental HealthNorwegian Institute of Public HealthOsloNorway
- Nic Waals InstituteLovisenberg Diaconal HospitalOsloNorway
| | | | | | - Siri Eldevik Håberg
- Centre for Fertility and HealthNorwegian Institute of Public HealthOsloNorway
| | - Per Magnus
- Centre for Fertility and HealthNorwegian Institute of Public HealthOsloNorway
| |
Collapse
|
2
|
Willmer T, Mabasa L, Sharma J, Muller CJF, Johnson R. Blood-Based DNA Methylation Biomarkers to Identify Risk and Progression of Cardiovascular Disease. Int J Mol Sci 2025; 26:2355. [PMID: 40076974 PMCID: PMC11900213 DOI: 10.3390/ijms26052355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
Non-communicable diseases (NCDs) are the leading cause of death worldwide, with cardiovascular disease (CVD) accounting for half of all NCD-related deaths. The biological onset of CVD may occur long before the development of clinical symptoms, hence the urgent need to understand the molecular alterations underpinning CVD, which would facilitate intervention strategies to prevent or delay the onset of the disease. There is evidence to suggest that CVD develops through a complex interplay between genetic, lifestyle, and environmental factors. Epigenetic modifications, including DNA methylation, serve as proxies linking genetics and the environment to phenotypes and diseases. In the past decade, a growing list of studies has implicated DNA methylation in the early events of CVD pathogenesis. In this regard, screening for these epigenetic marks in asymptomatic individuals may assist in the early detection of CVD and serve to predict the response to therapeutic interventions. This review discusses the current literature on the relationship between blood-based DNA methylation alterations and CVD in humans. We highlight a set of differentially methylated genes that show promise as candidates for diagnostic and prognostic CVD biomarkers, which should be prioritized and replicated in future studies across additional populations. Finally, we discuss key limitations in DNA methylation studies, including genetic diversity, interpatient variability, cellular heterogeneity, study confounders, different methodological approaches used to isolate and measure DNA methylation, sample sizes, and cross-sectional study design.
Collapse
Affiliation(s)
- Tarryn Willmer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Jyoti Sharma
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
| | - Christo J. F. Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa 3886, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (L.M.); (J.S.); (C.J.F.M.); (R.J.)
- Centre for Cardio-metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| |
Collapse
|
3
|
Lunar P, Meglič H, Vehar M, Ugovšek S, Rehberger Likozar A, Šebeštjen M, Zupan J. Effect of PCSK9 Inhibitors on Regulators of Lipoprotein Homeostasis, Inflammation and Coagulation. Biomedicines 2025; 13:294. [PMID: 40002707 PMCID: PMC11852752 DOI: 10.3390/biomedicines13020294] [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: 12/23/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND PCSK9 inhibitors (PCSK9i) represent a newer form of atherosclerosis treatment. Inflammation and haemostasis are key processes in the development of atherosclerosis. In this study, we investigated the influence of therapy with PCSK9i in patients with coronary artery disease (CAD) on regulators for lipoprotein homeostasis, inflammation and coagulation. METHODS Using quantitative polymerase chain reaction (qPCR), we measured the expression of the genes involved in lipoprotein homeostasis, namely for sterol regulatory element-binding protein 1 (SREBP1), SREBP2, low-density lipoprotein receptor (LDLR), hepatic lipase type C (LIPC), LDLR-related protein 8 (LRP8), and the genes associated with inflammation and coagulation, such as cluster of differentiation (CD) 36 (CD36), CD63, and CD14 in 96 patients with CAD and 25 healthy subjects. RESULTS Significant differences in the expression of the investigated genes between patients and healthy controls were found. Treatment with PCSK9i also resulted in significant changes in the expression of all studied genes. CONCLUSIONS We established that PCSK9i may have a significant effect on the gene expression of lipid regulators, inflammatory markers, and coagulation parameters, independent of their lipolytic effect.
Collapse
Affiliation(s)
- Patricija Lunar
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (P.L.); (H.M.); (S.U.); (M.Š.)
| | - Hana Meglič
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (P.L.); (H.M.); (S.U.); (M.Š.)
| | - Mateja Vehar
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia;
| | - Sabina Ugovšek
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (P.L.); (H.M.); (S.U.); (M.Š.)
- Department of Cardiology, University Medical Centre Ljubljana, Zaloška cesta 7, 1000 Ljubljana, Slovenia
| | - Andreja Rehberger Likozar
- Department of Vascular Diseases, University Medical Centre Ljubljana, Zaloška cesta 7, 1000 Ljubljana, Slovenia;
| | - Miran Šebeštjen
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (P.L.); (H.M.); (S.U.); (M.Š.)
- Department of Cardiology, University Medical Centre Ljubljana, Zaloška cesta 7, 1000 Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, Zaloška cesta 7, 1000 Ljubljana, Slovenia;
| | - Janja Zupan
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia;
| |
Collapse
|
4
|
Turkson V, Haller A, Jaeschke A, Hui DY. ApoE Receptor-2 R952Q Variant in Macrophages Elevates Soluble LRP1 to Potentiate Hyperlipidemia and Accelerate Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2025; 45:37-48. [PMID: 39508104 DOI: 10.1161/atvbaha.124.321748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND apoER2 (apolipoprotein E receptor-2) is a transmembrane receptor in the low-density lipoprotein receptor (LDLR) family with unique tissue expression. A single-nucleotide polymorphism that encodes the R952Q sequence variant has been associated with elevated plasma cholesterol levels and increased myocardial infarction risk in humans. The objective of this study was to delineate the mechanism underlying the association between the apoER2 variant with arginine-to-glutamine substitution at residue 952 (R952Q) and increased atherosclerosis risk. METHODS An apoER2 R952Q mouse model was generated and intercrossed with LDLR knockout mice, followed by feeding a Western-type high-fat high-cholesterol diet for 16 weeks. Atherosclerosis was investigated by immunohistology. Plasma lipids and lipid distributions among the various lipoprotein classes were analyzed by colorimetric assay. Tissue-specific effects of the R952Q sequence variant on atherosclerosis were analyzed by bone marrow transplant studies. sLRP1 (soluble low-density lipoprotein receptor-related protein 1) was measured in plasma and conditioned media from bone marrow-derived macrophages by ELISA and GST-RAP (glutathione S-transferase-receptor-associated protein) pull-down, respectively. P38 MAPK (mitogen-activated protein kinase) phosphorylation in VLDL (very-low-density lipoprotein)-treated macrophages was determined by Western blot analysis. RESULTS Consistent with observations in humans with this sequence variant, the apoER2 R952Q mutation exacerbated diet-induced hypercholesterolemia, via impediment of plasma triglyceride-rich lipoprotein clearance, to accelerate atherosclerosis in Western diet-fed LDLR knockout mice. Reciprocal bone marrow transplant experiments revealed that the apoER2 R952Q mutation in bone marrow-derived cells instead of non-bone marrow-derived cells was responsible for the increase in hypercholesterolemia and atherosclerosis. Additional data showed that the apoER2 R952Q mutation in macrophages promotes VLDL-induced LRP1 (low-density lipoprotein receptor-related protein 1) shedding in a p38 MAPK-dependent manner. CONCLUSIONS The apoER2 R952Q mouse model recapitulates characteristics observed in human disease. The underlying mechanism is that the apoER2 R952Q mutation in macrophages exacerbates VLDL-stimulated sLRP1 production in a p38 MAPK-dependent manner, resulting in its competition with cell surface LRP1 to impede triglyceride-rich lipoprotein clearance, thereby resulting in increased hypercholesterolemia and accelerated atherosclerosis.
Collapse
Affiliation(s)
- Vanessa Turkson
- Department of Pharmacology and Systems Physiology (V.T.), University of Cincinnati College of Medicine, OH
| | - April Haller
- Department of Pathology and Laboratory Medicine (A.H., A.J., D.Y.H.), University of Cincinnati College of Medicine, OH
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine (A.H., A.J., D.Y.H.), University of Cincinnati College of Medicine, OH
| | - David Y Hui
- Department of Pathology and Laboratory Medicine (A.H., A.J., D.Y.H.), University of Cincinnati College of Medicine, OH
| |
Collapse
|
5
|
Strickland DK, Cooper JM. Tale of 2 Receptors. Arterioscler Thromb Vasc Biol 2025; 45:49-52. [PMID: 39588647 PMCID: PMC11932498 DOI: 10.1161/atvbaha.124.322058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Affiliation(s)
- Dudley K. Strickland
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Joanna M. Cooper
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
6
|
Wang Q, Ni S, Ling L, Wang S, Xie H, Ren Z. Ginkgolide B Blocks Vascular Remodeling after Vascular Injury via Regulating Tgf β1/Smad Signaling Pathway. Cardiovasc Ther 2023; 2023:8848808. [PMID: 38125702 PMCID: PMC10732976 DOI: 10.1155/2023/8848808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/27/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Coronary artery disease (CAD) is the most prevalent cardiovascular disease worldwide, resulting in myocardial infarction (MI) and even sudden death. Following percutaneous coronary intervention (PCI), restenosis caused by vascular remodeling is always formed at the stent implantation site. Here, we show that Ginkgolide B (GB), a naturally occurring terpene lactone, effectively suppresses vascular remodeling and subsequent restenosis in wild-type mice following left carotid artery (LCA) injury. Additional experiments reveal that GB exerts a protective effect on vascular remodeling and further restenosis through modulation of the Tgfβ1/Smad signaling pathway in vivo and in human vascular smooth muscle cells (HVSMAs) but not in human umbilical vein endothelial cells (HUVECs) in vitro. Moreover, the beneficial effect of GB is abolished after incubated with pirfenidone (PFD, a drug for idiopathic pulmonary fibrosis, IPF), which can inhibit Tgfβ1. In Tgfβ1-/- mice, treatment with pirfenidone capsules and Yinxingneizhi Zhusheye (including Ginkgolide B) fails to improve vascular remodeling and restenosis. In conclusion, our data identify that GB could be a potential novel therapeutic agent to block vessel injury-associated vascular remodeling and further restenosis and show significant repression of Tgfβ1/Smad signaling pathway.
Collapse
Affiliation(s)
- Quan Wang
- Hubei University of Science and Technology, Xianning 437100, China
| | - Shuai Ni
- German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Li Ling
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Siqi Wang
- Hubei University of Science and Technology, Xianning 437100, China
| | - Hanbin Xie
- Collections Conservation Research Center, Shanghai Natural History Museum (Branch of Shanghai Science and Technology Museum), Shanghai 200041, China
| | - Zhanhong Ren
- Hubei University of Science and Technology, Xianning 437100, China
| |
Collapse
|
7
|
Wolfkiel PR, Haller AM, Kirby J, Jaeschke A, Hui DY. Different sensitivity to diet-induced hyperinsulinemia and hyperglycemia between mice with global or bone marrow-specific apoE receptor-2 deficiency. Am J Physiol Regul Integr Comp Physiol 2023; 325:R55-R68. [PMID: 37212552 PMCID: PMC10281787 DOI: 10.1152/ajpregu.00007.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 05/23/2023]
Abstract
This study explored the role of apoE receptor-2 (apoER2), a unique member of the LDL receptor family proteins with a restricted tissue expression profile, in modulating diet-induced obesity and diabetes. Unlike wild-type mice and humans in which chronic feeding of a high-fat Western-type diet leads to obesity and the prediabetic state of hyperinsulinemia before hyperglycemia onset, the Lrp8-/- mice with global apoER2 deficiency displayed lower body weight and adiposity, slower development of hyperinsulinemia, but the accelerated onset of hyperglycemia. Despite their lower adiposity, adipose tissues in Western diet-fed Lrp8-/- mice were more inflamed compared with wild-type mice. Additional experiments revealed that the hyperglycemia observed in Western diet-fed Lrp8-/- mice was due to impaired glucose-induced insulin secretion, ultimately leading to hyperglycemia, adipocyte dysfunction, and inflammation upon chronic feeding of the Western diet. Interestingly, bone marrow-specific apoER2-deficient mice were not defective in insulin secretion, exhibiting increased adiposity and hyperinsulinemia compared with wild-type mice. Analysis of bone marrow-derived macrophages revealed that apoER2 deficiency impeded inflammation resolution with lower secretion of IFN-β and IL-10 in response to LPS stimulation of IL-4 primed cells. The apoER2-deficient macrophages also showed an increased level of disabled-2 (Dab2) as well as increased cell surface TLR4, suggesting that apoER2 participates in Dab2 regulation of TLR4 signaling. Taken together, these results showed that apoER2 deficiency in macrophages sustains diet-induced tissue inflammation and accelerates obesity and diabetes onset while apoER2 deficiency in other cell types contributes to hyperglycemia and inflammation via defective insulin secretion.
Collapse
Affiliation(s)
- Patrick R Wolfkiel
- Molecular Genetics, Biochemistry, and Microbiology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - April M Haller
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Jillian Kirby
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| |
Collapse
|
8
|
miR-1322 protects against the myocardial ischemia via LRP8/PI3K/AKT pathway. Biochem Biophys Res Commun 2023; 638:120-126. [PMID: 36446154 DOI: 10.1016/j.bbrc.2022.10.101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/12/2022] [Accepted: 10/29/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Myocardial infarction is a fatal disease that causes millions of deaths worldwide every year. The damage and recovery of cardiomyocytes are closely related to changes in gene expression. miRNA may be a new therapeutic target of myocardial ischemia-reperfusion. METHODS The differential expression genes were analyzed based on GSE83500, GSE60993 and GSE154733. miRNA expression profile data and clinical data were downloaded from GSE76591. Bioinformatics analysis including limma package, cluster analysis, WGCNA analysis were performed. H9c2 cell hypoxia model and mouse myocardial ischemia model were established. Q-PCR, Western blot and luciferase assay were carried out. RESULTS miR-1322 was identified as a significantly differentially expressed miRNA in myocardial ischemi. Yin Yang 1(YY1) was significantly highly expressed in cells with hypoxia treatment (P < 0.05), and myocardial ischemia mice (P < 0.01), which was identified as the transcription factor of miR-1322. The protein expression of LRP8 was lower in cells with hypoxia treatment and myocardial ischemia mice (P < 0.05) and LRP8 was the target gene of miR-1322. The overexpression of LRP8 could significantly increase the expression of p-PI3K, p-AKT, and P70 S6K (P < 0.05). LRP8 regulated PI3K/AKT/P70 S6K signaling pathway, eventually resulting in cell apoptosis. CONCLUSION Our results suggested that miR-1322 can protect against the myocardial ischemia via LRP8/PI3K/AKT pathway.
Collapse
|
9
|
Calvier L, Herz J, Hansmann G. Interplay of Low-Density Lipoprotein Receptors, LRPs, and Lipoproteins in Pulmonary Hypertension. JACC Basic Transl Sci 2022; 7:164-180. [PMID: 35257044 PMCID: PMC8897182 DOI: 10.1016/j.jacbts.2021.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022]
Abstract
The low-density lipoprotein receptor (LDLR) gene family includes LDLR, very LDLR, and LDL receptor-related proteins (LRPs) such as LRP1, LRP1b (aka LRP-DIT), LRP2 (aka megalin), LRP4, and LRP5/6, and LRP8 (aka ApoER2). LDLR family members constitute a class of closely related multifunctional, transmembrane receptors, with diverse functions, from embryonic development to cancer, lipid metabolism, and cardiovascular homeostasis. While LDLR family members have been studied extensively in the systemic circulation in the context of atherosclerosis, their roles in pulmonary arterial hypertension (PAH) are understudied and largely unknown. Endothelial dysfunction, tissue infiltration of monocytes, and proliferation of pulmonary artery smooth muscle cells are hallmarks of PAH, leading to vascular remodeling, obliteration, increased pulmonary vascular resistance, heart failure, and death. LDLR family members are entangled with the aforementioned detrimental processes by controlling many pathways that are dysregulated in PAH; these include lipid metabolism and oxidation, but also platelet-derived growth factor, transforming growth factor β1, Wnt, apolipoprotein E, bone morpohogenetic proteins, and peroxisome proliferator-activated receptor gamma. In this paper, we discuss the current knowledge on LDLR family members in PAH. We also review mechanisms and drugs discovered in biological contexts and diseases other than PAH that are likely very relevant in the hypertensive pulmonary vasculature and the future care of patients with PAH or other chronic, progressive, debilitating cardiovascular diseases.
Collapse
Key Words
- ApoE, apolipoprotein E
- Apoer2
- BMP
- BMPR, bone morphogenetic protein receptor
- BMPR2
- COPD, chronic obstructive pulmonary disease
- CTGF, connective tissue growth factor
- HDL, high-density lipoprotein
- KO, knockout
- LDL receptor related protein
- LDL, low-density lipoprotein
- LDLR
- LDLR, low-density lipoprotein receptor
- LRP
- LRP, low-density lipoprotein receptor–related protein
- LRP1
- LRP1B
- LRP2
- LRP4
- LRP5
- LRP6
- LRP8
- MEgf7
- Mesd, mesoderm development
- PAH
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary artery smooth muscle cell
- PDGF
- PDGFR-β, platelet-derived growth factor receptor-β
- PH, pulmonary hypertension
- PPARγ
- PPARγ, peroxisome proliferator-activated receptor gamma
- PVD
- RV, right ventricle/ventricular
- RVHF
- RVSP, right ventricular systolic pressure
- TGF-β1
- TGF-β1, transforming growth factor β1
- TGFBR, transforming growth factor β1 receptor
- TNF, tumor necrosis factor receptor
- VLDLR
- VLDLR, very low density lipoprotein receptor
- VSMC, vascular smooth muscle cell
- Wnt
- apolipoprotein E receptor 2
- endothelial cell
- gp330
- low-density lipoprotein receptor
- mRNA, messenger RNA
- megalin
- monocyte
- multiple epidermal growth factor-like domains 7
- pulmonary arterial hypertension
- pulmonary vascular disease
- right ventricle heart failure
- smooth muscle cell
- very low density lipoprotein receptor
- β-catenin
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
- Pulmonary Vascular Research Center, Hannover Medical School, Hannover, Germany
| |
Collapse
|
10
|
Rowland MJ, Garry P, Ezra M, Corkill R, Baker I, Jezzard P, Westbrook J, Douaud G, Pattinson KTS. Early brain injury and cognitive impairment after aneurysmal subarachnoid haemorrhage. Sci Rep 2021; 11:23245. [PMID: 34853362 PMCID: PMC8636506 DOI: 10.1038/s41598-021-02539-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/29/2021] [Indexed: 12/21/2022] Open
Abstract
The first 72 h following aneurysm rupture play a key role in determining clinical and cognitive outcomes after subarachnoid haemorrhage (SAH). Yet, very little is known about the impact of so called "early brain injury" on patents with clinically good grade SAH (as defined as World Federation of Neurosurgeons Grade 1 and 2). 27 patients with good grade SAH underwent MRI scanning were prospectively recruited at three time-points after SAH: within the first 72 h (acute phase), at 5-10 days and at 3 months. Patients underwent additional, comprehensive cognitive assessment 3 months post-SAH. 27 paired healthy controls were also recruited for comparison. In the first 72 h post-SAH, patients had significantly higher global and regional brain volume than controls. This change was accompanied by restricted water diffusion in patients. Persisting abnormalities in the volume of the posterior cerebellum at 3 months post-SAH were present to those patients with worse cognitive outcome. When using this residual abnormal brain area as a region of interest in the acute-phase scans, we could predict with an accuracy of 84% (sensitivity 82%, specificity 86%) which patients would develop cognitive impairment 3 months later, despite initially appearing clinically indistinguishable from those making full recovery. In an exploratory sample of good clinical grade SAH patients compared to healthy controls, we identified a region of the posterior cerebellum for which acute changes on MRI were associated with cognitive impairment. Whilst further investigation will be required to confirm causality, use of this finding as a risk stratification biomarker is promising.
Collapse
Affiliation(s)
- Matthew J Rowland
- Nuffield Division of Anaesthetics, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK.
- Neurosciences Intensive Care Unit, Oxford University Hospitals NHS Trust, Oxford, UK.
| | - Payashi Garry
- Nuffield Division of Anaesthetics, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Neurosciences Intensive Care Unit, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Martyn Ezra
- Nuffield Division of Anaesthetics, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Neurosciences Intensive Care Unit, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Rufus Corkill
- Neurosciences Intensive Care Unit, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Ian Baker
- Department of Psychological Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 9DU, UK
| | - Peter Jezzard
- FMRIB, Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jon Westbrook
- Nuffield Division of Anaesthetics, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Neurosciences Intensive Care Unit, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Gwenaëlle Douaud
- FMRIB, Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Kyle T S Pattinson
- Nuffield Division of Anaesthetics, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Neurosciences Intensive Care Unit, Oxford University Hospitals NHS Trust, Oxford, UK
| |
Collapse
|
11
|
Guo Y, Tang Z, Yan B, Yin H, Tai S, Peng J, Cui Y, Gui Y, Belke D, Zhou S, Zheng XL. PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) Triggers Vascular Smooth Muscle Cell Senescence and Apoptosis: Implication of Its Direct Role in Degenerative Vascular Disease. Arterioscler Thromb Vasc Biol 2021; 42:67-86. [PMID: 34809446 DOI: 10.1161/atvbaha.121.316902] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE PCSK9 (proprotein convertase subtilisin/kexin type 9) plays a critical role in cholesterol metabolism via the PCSK9-LDLR (low-density lipoprotein receptor) axis in the liver; however, evidence indicates that PCSK9 directly contributes to the pathogenesis of various diseases through mechanisms independent of its LDL-cholesterol regulation. The objective of this study was to determine how PCSK9 directly acts on vascular smooth muscle cells (SMCs), contributing to degenerative vascular disease. Approach and Results: We first examined the effects of PCSK9 on cultured human aortic SMCs. Overexpression of PCSK9 downregulated the expression of ApoER2 (apolipoprotein E receptor 2), a known target of PCSK9. Treatment with soluble recombinant human ApoER2 or the DNA synthesis inhibitor, hydroxyurea, inhibited PCSK9-induced polyploidization and other cellular responses of human SMCs. Treatment with antibodies against ApoER2 resulted in similar effects to those observed with PCSK9 overexpression. Inducible, SMC-specific knockout of Pcsk9 accelerated neointima formation in mouse carotid arteries and reduced age-related arterial stiffness. PCSK9 was expressed in SMCs of human atherosclerotic lesions and abundant in the "shoulder" regions of vulnerable atherosclerotic plaques. PCSK9 was also expressed in SMCs of abdominal aortic aneurysm, which was inversely related to the expression of smooth muscle α-actin. CONCLUSIONS Our findings demonstrate that PCSK9 inhibits proliferation and induces polyploidization, senescence, and apoptosis, which may be relevant to various degenerative vascular diseases.
Collapse
Affiliation(s)
- Yanan Guo
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (Y. Guo, S.T., S.Z.)
| | - Zhihan Tang
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Binjie Yan
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Hao Yin
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Now with Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada (H.Y.)
| | - Shi Tai
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (Y. Guo, S.T., S.Z.)
| | - Juan Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Yuting Cui
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Yu Gui
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng)
| | - Darrell Belke
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng)
| | - Shenghua Zhou
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (Y. Guo, S.T., S.Z.)
| | - Xi-Long Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng)
| |
Collapse
|
12
|
Luo C, Wang D, Huang W, Song Y, Ge L, Zhang X, Yang L, Lu J, Tu X, Chen Q, Yang J, Xu C, Wang Q. Feedback regulation of coronary artery disease susceptibility gene ADTRP and LDL receptors LDLR/CD36/LOX-1 in endothelia cell functions involved in atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166130. [PMID: 33746034 DOI: 10.1016/j.bbadis.2021.166130] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/11/2022]
Abstract
A high level of low-density lipoprotein cholesterol (LDL) is one of the most important risk factors for coronary artery disease (CAD), the leading cause of death worldwide. However, a low concentration of LDL may be protective. Genome-wide association studies revealed that variation in ADTRP gene increased the risk of CAD. In this study, we found that a low concentration of oxidized-LDL induced the expression of ADTRP. Further analyses showed that knockdown of the expression of LDL receptor genes LDLR, CD36, or LOX-1 significantly downregulated ADTRP expression, whereas overexpression of LDLR/CD36/LOX-1 markedly increased ADTRP expression through the NF-κB pathway. Like ADTRP, LDLR, CD36 and LOX-1 were all involved in endothelial cell (EC) functions relevant to the initiation of atherosclerosis. Downregulation of LDLR/CD36/LOX-1 promoted monocyte adhesion to ECs and transendothelial migration of monocytes by increasing expression of ICAM-1, VCAM-1, E-selectin and P-selectin, decreased EC proliferation and migration, and increased EC apoptosis, thereby promoting the initiation of atherosclerosis. Opposite effects were observed with the overexpression of ADTRP and LDLR/CD36/LOX-1 in ECs. Interestingly, through the NF-κB and AKT pathways, overexpression of ADTRP significantly upregulated the expression of LDLR, CD36, and LOX-1, and knockdown of ADTRP expression significantly downregulated the expression of LDLR, CD36, and LOX-1. These data suggest that ADTRP and LDL receptors LDLR/CD36/LOX-1 positively regulate each other, and form a positive regulatory loop that regulates endothelial cell functions, thereby providing a potential protective mechanism against atherosclerosis. Our findings provide a new molecular mechanism by which deregulation of ADTRP and LDLR/CD36/LOX-1 promote the development of atherosclerosis and CAD.
Collapse
Affiliation(s)
- Chunyan Luo
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang 443002, Hubei, PR China; The Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, Hubei, PR China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China
| | - Decheng Wang
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang 443002, Hubei, PR China; The Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, Hubei, PR China
| | - Weifeng Huang
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang 443002, Hubei, PR China; The Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, Hubei, PR China
| | - Yinhong Song
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang 443002, Hubei, PR China; The Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, Hubei, PR China
| | - Lisha Ge
- The Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, Hubei, PR China
| | - Xinyue Zhang
- The Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, Hubei, PR China
| | - Lixue Yang
- The Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, Hubei, PR China
| | - Jiao Lu
- The Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, Hubei, PR China
| | - Xiancong Tu
- The Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, Hubei, PR China
| | - Qiuyun Chen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Jian Yang
- Department of Cardiology, the People's Hospital of China Three Gorges University, Yichang 443000, Hubei, PR China.
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China.
| | - Qing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, Hubei, PR China.
| |
Collapse
|
13
|
Mineo C. Lipoprotein receptor signalling in atherosclerosis. Cardiovasc Res 2021; 116:1254-1274. [PMID: 31834409 DOI: 10.1093/cvr/cvz338] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/01/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
The founding member of the lipoprotein receptor family, low-density lipoprotein receptor (LDLR) plays a major role in the atherogenesis through the receptor-mediated endocytosis of LDL particles and regulation of cholesterol homeostasis. Since the discovery of the LDLR, many other structurally and functionally related receptors have been identified, which include low-density lipoprotein receptor-related protein (LRP)1, LRP5, LRP6, very low-density lipoprotein receptor, and apolipoprotein E receptor 2. The scavenger receptor family members, on the other hand, constitute a family of pattern recognition proteins that are structurally diverse and recognize a wide array of ligands, including oxidized LDL. Among these are cluster of differentiation 36, scavenger receptor class B type I and lectin-like oxidized low-density lipoprotein receptor-1. In addition to the initially assigned role as a mediator of the uptake of macromolecules into the cell, a large number of studies in cultured cells and in in vivo animal models have revealed that these lipoprotein receptors participate in signal transduction to modulate cellular functions. This review highlights the signalling pathways by which these receptors influence the process of atherosclerosis development, focusing on their roles in the vascular cells, such as macrophages, endothelial cells, smooth muscle cells, and platelets. Human genetics of the receptors is also discussed to further provide the relevance to cardiovascular disease risks in humans. Further knowledge of the vascular biology of the lipoprotein receptors and their ligands will potentially enhance our ability to harness the mechanism to develop novel prophylactic and therapeutic strategies against cardiovascular diseases.
Collapse
Affiliation(s)
- Chieko Mineo
- Department of Pediatrics and Cell Biology, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| |
Collapse
|
14
|
Luo C, Pook E, Wang F, Archacki SR, Tang B, Zhang W, Hu JS, Yang J, Leineweber K, Bechem M, Huang W, Song Y, Cheung SH, Laux V, Ke T, Ren X, Tu X, Chen Q, Wang QK, Xu C. ADTRP regulates TFPI expression via transcription factor POU1F1 involved in coronary artery disease. Gene 2020; 753:144805. [PMID: 32445923 DOI: 10.1016/j.gene.2020.144805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022]
Abstract
Genomic variants in both ADTRP and TFPI genes are associated with risk of coronary artery disease (CAD). ADTRP regulates TFPI expression and endothelial cell functions involved in the initiation of atherosclerotic CAD. ADTRP also specifies primitive myelopoiesis and definitive hematopoiesis by upregulating TFPI expression. However, the underlying molecular mechanism is unknown. Here we show that transcription factor POU1F1 is the key by which ADTRP regulates TFPI expression. Luciferase reporter assays, chromatin-immunoprecipitation (ChIP) and electrophoretic mobility shift assay (EMSA) in combination with analysis of large and small deletions of the TFPI promoter/regulatory region were used to identify the molecular mechanism by which ADTRP regulates TFPI expression. Genetic association was assessed using case-control association analysis and phenome-wide association analysis (PhenGWA). ADTRP regulates TFPI expression at the transcription level in a dose-dependent manner. The ADTRP-response element was localized to a 50 bp region between -806 bp and -756 bp upstream of TFPI transcription start site, which contains a binding site for POU1F1. Deletion of POU1F1-binding site or knockdown of POU1F1 expression abolished ADTRP-mediated transcription of TFPI. ChIP and EMSA demonstrated that POU1F1 binds to the ADTRP response element. Genetic analysis identified significant association between POU1F1 variants and risk of CAD. PhenGWA identified other phenotypic traits associated with the ADTRP-POU1F1-TFPI axis such as lymphocyte count (ADTRP), waist circumference (TFPI), and standing height (POU1F1). These data identify POU1F1 as a transcription factor that regulates TFPI transcription in response to ADTRP, and link POU1F1 variants to risk of CAD for the first time.
Collapse
Affiliation(s)
- Chunyan Luo
- The Institute of Infection and Inflammation, Department of Microbiology and Immunology, Medical College, Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, China Three Gorges University, Yichang, Hubei 443002, PR China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | | | - Fan Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Stephen R Archacki
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Bo Tang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Weiyi Zhang
- Bayer Healthcare Co Ltd, Innovation Center China, Beijing, PR China
| | - Jing-Shan Hu
- Bayer Healthcare Co Ltd, Innovation Center China, Beijing, PR China
| | - Jian Yang
- The Institute of Infection and Inflammation, Department of Microbiology and Immunology, Medical College, Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, China Three Gorges University, Yichang, Hubei 443002, PR China
| | | | | | - Weifeng Huang
- The Institute of Infection and Inflammation, Department of Microbiology and Immunology, Medical College, Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, China Three Gorges University, Yichang, Hubei 443002, PR China
| | - Yinhong Song
- The Institute of Infection and Inflammation, Department of Microbiology and Immunology, Medical College, Key Laboratory of Ischemic Cardiovascular and Cerebrovascular Disease Translational Medicine, China Three Gorges University, Yichang, Hubei 443002, PR China
| | - Shing-Hu Cheung
- Bayer Healthcare Co Ltd, Innovation Center China, Beijing, PR China
| | - Volker Laux
- BayerAG, Drug Discovery, 42096 Wuppertal, Germany
| | - Tie Ke
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Xiang Ren
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Xin Tu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Qiuyun Chen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44195, USA.
| | - Qing Kenneth Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44195, USA.
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China.
| |
Collapse
|
15
|
From Anti-SARS-CoV-2 Immune Responses to COVID-19 via Molecular Mimicry. Antibodies (Basel) 2020; 9:antib9030033. [PMID: 32708525 PMCID: PMC7551747 DOI: 10.3390/antib9030033] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022] Open
Abstract
Aim: To define the autoimmune potential of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection. Methods: Experimentally validated epitopes cataloged at the Immune Epitope DataBase (IEDB) and present in SARS-CoV-2 were analyzed for peptide sharing with the human proteome. Results: Immunoreactive epitopes present in SARS-CoV-2 were mostly composed of peptide sequences present in human proteins that—when altered, mutated, deficient or, however, improperly functioning—may associate with a wide range of disorders, from respiratory distress to multiple organ failure. Conclusions: This study represents a starting point or hint for future scientific–clinical investigations and suggests a range of possible protein targets of autoimmunity in SARS-CoV-2 infection. From an experimental perspective, the results warrant the testing of patients’ sera for autoantibodies against these protein targets. Clinically, the results warrant a stringent surveillance on the future pathologic sequelae of the current SARS-CoV-2 pandemic.
Collapse
|
16
|
Angiotensin II Infusion Leads to Aortic Dissection in LRP8 Deficient Mice. Int J Mol Sci 2020; 21:ijms21144916. [PMID: 32664652 PMCID: PMC7404218 DOI: 10.3390/ijms21144916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 01/27/2023] Open
Abstract
Myeloid cells are crucial for the development of vascular inflammation. Low-density lipoprotein receptor-related protein 8 (LRP8) or Apolipoprotein E receptor 2 (ApoER2), is expressed by macrophages, endothelial cells and platelets and has been implicated in the development of cardiovascular diseases. Our aim was to evaluate the role of LRP8, in particular from immune cells, in the development of vascular inflammation. Methods. LRP8+/+ and LRP8−/− mice (on B6;129S background) were infused with angiotensin II (AngII, 1 mg/kg/day for 7 to 28 day) using osmotic minipumps. Blood pressure was recorded using tail cuff measurements. Vascular reactivity was assessed in isolated aortic segments. Leukocyte activation and infiltration were assessed by flow cytometry of aortic tissue and intravital videomicroscopy imaging. Histological analysis of aortic sections was conducted using sirius red staining. Results. AngII infusion worsened endothelial-dependent vascular relaxation and immune cells rolling and adherence to the carotid artery in both LRP8+/+ as well as LRP8−/− mice. However, only LRP8−/− mice demonstrated a drastically increased mortality rate in response to AngII due to aortic dissection. Bone marrow transplantation revealed that chimeras with LRP8 deficient myeloid cells phenocopied LRP8−/− mice. Conclusion. AngII-infused LRP8 deficient mice could be a useful animal model to study aortic dissection reflecting the lethality of this disease in humans.
Collapse
|
17
|
Cho H, Li Y, Archacki S, Wang F, Yu G, Chakrabarti S, Guo Y, Chen Q, Wang QK. Splice variants of lncRNA RNA ANRIL exert opposing effects on endothelial cell activities associated with coronary artery disease. RNA Biol 2020; 17:1391-1401. [PMID: 32602777 DOI: 10.1080/15476286.2020.1771519] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Each gene typically has multiple alternatively spliced transcripts. Different transcripts are assumed to play a similar biological role; however, some transcripts may simply lose their function due to loss of important functional domains. Here, we show that two different transcripts of lncRNA gene ANRIL associated with coronary artery disease (CAD) play antagonizing roles against each other. We previously reported that DQ485454, the short transcript, is downregulated in coronary arteries from CAD patients, and reduces monocyte adhesion to endothelial cells (ECs) and transendothelial monocyte migration (TEM). Interestingly, the longest transcript NR_003529 is significantly upregulated in coronary arteries from CAD patients. Overexpression of ANRIL transcript NR_003529 increases monocyte adhesion to ECs and TEM, whereas knockdown of NR_003529 expression reduces monocyte adhesion to ECs and TEM. Much more dramatic effects were observed for the combination of overexpression of NR_003529 and knockdown of DQ485454 or the combination of knockdown of NR_003529 and overexpression of DQ485454. The antagonizing effects of ANRIL transcripts NR_003529 and DQ485454 were associated with their opposite effects on expression of downstream target genes EZR, CXCL11 or TMEM106B. Our results demonstrate that different transcripts of lncRNA can exert antagonizing effects on biological functions, thereby providing important insights into the biology of lncRNA. The data further support the hypothesis that ANRIL is the causative gene at the 9p21 CAD susceptibility locus.
Collapse
Affiliation(s)
- Hyosuk Cho
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine , Cleveland, OH, USA.,Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic , Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, OH, USA
| | - Yabo Li
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic , Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, OH, USA
| | - Stephen Archacki
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic , Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, OH, USA
| | - Fan Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic , Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, OH, USA
| | - Gang Yu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic , Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, OH, USA.,Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology , Wuhan, P. R. China
| | - Susmita Chakrabarti
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic , Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, OH, USA
| | - Yang Guo
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic , Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, OH, USA
| | - Qiuyun Chen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic , Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, OH, USA
| | - Qing Kenneth Wang
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine , Cleveland, OH, USA.,Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic , Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, OH, USA
| |
Collapse
|
18
|
Komaravolu RK, Waltmann MD, Konaniah E, Jaeschke A, Hui DY. ApoER2 (Apolipoprotein E Receptor-2) Deficiency Accelerates Smooth Muscle Cell Senescence via Cytokinesis Impairment and Promotes Fibrotic Neointima After Vascular Injury. Arterioscler Thromb Vasc Biol 2019; 39:2132-2144. [PMID: 31412739 PMCID: PMC6761011 DOI: 10.1161/atvbaha.119.313194] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Genome-wide studies showed that mutation in apoER2 (apolipoprotein E receptor-2) is additive with ε4 polymorphism in the APOE gene on cardiovascular disease risk in humans. ApoE or apoER2 deficiency also accelerates atherosclerosis lesion necrosis in hypercholesterolemic mice and promotes neointima formation after vascular injury. This study tests the hypothesis that apoE and apoER2 modulate vascular occlusive diseases through distinct mechanisms. Approach and Results: Carotid endothelial denudation induced robust neointima formation in both apoE-/- and apoER2-deficient Lrp8-/- mice. The intima in apoE-/- mice was rich in smooth muscle cells, but the intima in Lrp8-/- mice was cell-poor and rich in extracellular matrix. Vascular smooth muscle cells isolated from apoE-/- mice were hyperplastic whereas Lrp8-/- smooth muscle cells showed reduced proliferation but responded robustly to TGF (transforming growth factor)-β-induced fibronectin synthesis indicative of a senescence-associated secretory phenotype, which was confirmed by increased β-galactosidase activity, p16INK4a immunofluorescence, and number of multinucleated cells. Western blot analysis of cell cycle-associated proteins showed that apoER2 deficiency promotes cell cycle arrest at the metaphase/anaphase. Coimmunoprecipitation experiments revealed that apoER2 interacts with the catalytic subunit of protein phosphatase 2A. In the absence of apoER2, PP2A-C (protein phosphatase 2A catalytic subunit) failed to interact with CDC20 (cell-division cycle protein 20) thus resulting in inactive anaphase-promoting complex and impaired cell cycle exit. CONCLUSIONS This study showed that apoER2 participates in APC (anaphase-promoting complex)/CDC20 complex formation during mitosis, and its absence impedes cytokinesis abscission thereby accelerating premature cell senescence and vascular disease. This mechanism is distinct from apoE deficiency, which causes smooth muscle cell hyperplasia to accelerate vascular disease.
Collapse
Affiliation(s)
- Ravi K. Komaravolu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Meaghan D. Waltmann
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Eddy Konaniah
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| | - David Y. Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, OH 45237
| |
Collapse
|
19
|
Barboza-Cerda MC, Barboza-Quintana O, Martínez-Aldape G, Garza-Guajardo R, Déctor MA. Phenotypic severity in a family with MEND syndrome is directly associated with the accumulation of potentially functional variants of cholesterol homeostasis genes. Mol Genet Genomic Med 2019; 7:e931. [PMID: 31397093 PMCID: PMC6732292 DOI: 10.1002/mgg3.931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 07/23/2019] [Indexed: 11/19/2022] Open
Abstract
Background Male EBP disorder with neurologic defects (MEND) syndrome is an X‐linked disease caused by hypomorphic mutations in the EBP (emopamil‐binding protein) gene. Modifier genes may explain the clinical variability among individuals who share a primary mutation. Methods We studied four males (Patient 1 to Patient 4) exhibiting a descending degree of phenotypic severity from a family with MEND syndrome. To identify candidate modifier genes that explain the phenotypic variability, variants of homeostasis cholesterol genes identified by whole‐exome sequencing (WES) were ranked according to the predicted magnitude of their effect through an in‐house scoring system. Results Twenty‐seven from 105 missense variants found in 45 genes of the four exomes were considered significant (−5 to −9 scores). We found a direct genotype–phenotype association based on the differential accumulation of potentially functional gene variants among males. Patient 1 exhibited 17 variants, both Patients 2 and 3 exhibited nine variants, and Patient 4 exhibited only five variants. Conclusion We conclude that APOA5 (rs3135506), ABCA1 (rs9282541), and APOB (rs679899 and rs12714225) are the most relevant candidate modifier genes in this family. Relative accumulation of the deficiencies associated with variants of these genes along with other lesser deficiencies in other genes appears to explain the variable expressivity in MEND syndrome.
Collapse
Affiliation(s)
- María Carmen Barboza-Cerda
- Facultad de Medicina y Hospital Universitario "Dr. José E. González", Servicio de Anatomía Patológica y Citopatología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico.,Facultad de Medicina y Hospital Universitario "Dr. José E. González", Departamento de Bioquímica y Medicina Molecular, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Oralia Barboza-Quintana
- Facultad de Medicina y Hospital Universitario "Dr. José E. González", Servicio de Anatomía Patológica y Citopatología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Gerardo Martínez-Aldape
- Facultad de Medicina y Hospital Universitario "Dr. José E. González", Servicio de Anatomía Patológica y Citopatología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Raquel Garza-Guajardo
- Facultad de Medicina y Hospital Universitario "Dr. José E. González", Servicio de Anatomía Patológica y Citopatología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Miguel Angel Déctor
- Facultad de Medicina y Hospital Universitario "Dr. José E. González", Servicio de Anatomía Patológica y Citopatología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico.,Facultad de Medicina y Hospital Universitario "Dr. José E. González", Departamento de Bioquímica y Medicina Molecular, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| |
Collapse
|
20
|
Yang Q, Xiong H, Xu C, Huang Y, Tu X, Wu G, Fu F, Wang Z, Wang L, Zhao Y, Li S, Huang Y, Wang C, Wang D, Yao Y, Wang F, Wang Y, Xue Y, Wang P, Chen Q, Pu J, Wang QK. Identification of rare variants in cardiac sodium channel β4-subunit gene SCN4B associated with ventricular tachycardia. Mol Genet Genomics 2019; 294:1059-1071. [PMID: 31020414 DOI: 10.1007/s00438-019-01567-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 04/10/2019] [Indexed: 12/19/2022]
Abstract
Ventricular tachycardia (VT) causes sudden cardiac death, however, the majority of risk genes for VT remain unknown. SCN4B encodes a β-subunit, Navβ4, for the voltage-gated cardiac sodium channel complex involved in generation and conduction of the cardiac action potential. We hypothesized that genomic variants in SCN4B increase the risk of VT. We used high-resolution melt analysis followed by Sanger sequencing to screen 199 VT patients to identify nonsynonymous variants in SCN4B. Two nonsynonymous heterozygous variants in SCN4B were identified in VT patients, including p.Gly8Ser in four VT patients and p.Ala145Ser in one VT patient. Case-control association studies were used to assess the association between variant p.Gly8Ser and VT in two independent populations for VT (299 VT cases vs. 981 controls in population 1 and 270 VT patients vs. 639 controls in population 2). Significant association was identified between p.Gly8Ser and VT in population 1 (P = 1.21 × 10-4, odds ratio or OR = 11.04), and the finding was confirmed in population 2 (P = 0.03, OR = 3.62). The association remained highly significant in the combined population (P = 3.09 × 10-5, OR = 6.17). Significant association was also identified between p.Gly8Ser and idiopathic VT (P = 1.89 × 10-5, OR = 7.27). Functional analysis with Western blotting showed that both p.Gly8Ser and p.Ala145Ser variants significantly reduced the expression level of Navβ4. Based on 2015 ACMG Standards and Guidelines, p.Gly8Ser and p.Ala145Ser can be classified as the pathogenic and likely pathogenic variant, respectively. Our data suggest that SCN4B is a susceptibility gene for common VT and idiopathic VT and link rare SCN4B variants with large effects (OR = 6.17-7.27) to common VT.
Collapse
Affiliation(s)
- Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Hongbo Xiong
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Yuan Huang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Xin Tu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Gang Wu
- Renmin Hospital, Wuhan University, Wuhan, People's Republic of China
| | - Fenfen Fu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Zhijie Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Longfei Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Yuanyuan Zhao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Sisi Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Yufeng Huang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Chuchu Wang
- School of Life Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, People's Republic of China
| | - Dan Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Fan Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Yongbo Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Yu Xue
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Pengyun Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Qiuyun Chen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA.
| | - Jielin Pu
- Department of Cardiology, East Hospital, Tongji University, Shanghai, Beijing, 100037, People's Republic of China. .,Tongji University Affiliated East Hospital, Beijing, 100037, People's Republic of China.
| | - Qing K Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China. .,Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA. .,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
21
|
Wang F, Wang IZ, Ellis S, Archacki S, Barnard J, Hubbard C, Topol EJ, Chen Q, Wang QK. Analysis of causal effect of APOA5 variants on premature coronary artery disease. Ann Hum Genet 2018; 82:437-447. [PMID: 30024021 DOI: 10.1111/ahg.12273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 05/17/2018] [Accepted: 06/18/2018] [Indexed: 11/30/2022]
Abstract
Apolipoprotein A5 (APOA5) regulates the metabolisms of triglyceride and HDL. APOA5 variants have been linked to coronary artery disease (CAD), but their causal roles are not well studied yet. This study aims to identify the causal effects of APOA5 variants on premature CAD. Sequencing analysis of APOA5 in 128 premature, familiar CAD patients from GeneQuest identified 11 genomic variants, including p.S19W (rs3135506). SKAT analysis showed that all sequenced variants, in aggregate, significantly increased the risk of premature CAD (P-skat = 0.037). Individually, the p.S19W variant was significantly associated with risk of premature CAD (OR = 2.30, P = 0.008) in an independent set of 342 premature CAD patients and 537 controls after adjusting for covariates of sex, age, hypertension, body mass index, triglycerides (TGs), and total, LDL-, and HDL-cholesterol levels. Meanwhile, p.S19W significantly correlated with HDL-C levels (P = 0.048) and TG levels (P = 0.025). Mediation analysis yielded a mediation effect of p.S19W on risk of premature CAD through HDL-C (OR = 0.98, P = 0.040) and TG (OR = 0.98, P = 0.042), suggesting a causal relationship between p.S19W and premature CAD partially through its effects on HDL-C and TG levels. These results suggest that APOA5 variation regulates TG and HDL levels, thus displaying a causal role in the development of CAD.
Collapse
Affiliation(s)
- Fan Wang
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Department of Genetics and Genome Science, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Isabel Z Wang
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Shaker Heights High School, Shaker Heights, OH, USA
| | - Stephen Ellis
- Department of Cardiovascular Medicine, Sydell & Arnold Miller Family Heart & Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Stephen Archacki
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Department of Genetics and Genome Science, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - John Barnard
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Carlos Hubbard
- Department of Cardiovascular Medicine, Sydell & Arnold Miller Family Heart & Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric J Topol
- Scripps Translational Science Institute, Scripps Research Institute, Scripps Clinic, La Jolla, CA, USA
| | - Qiuyun Chen
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Department of Genetics and Genome Science, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Qing K Wang
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Department of Genetics and Genome Science, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.,Department of Cardiovascular Medicine, Sydell & Arnold Miller Family Heart & Vascular Institute, Cleveland Clinic, Cleveland, OH, USA.,The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| |
Collapse
|
22
|
Asif M, Bhat S, Nizamuddin S, Mustak MS. TG haplotype in the LRP8 is associated with myocardial infarction in south Indian population. Gene 2017; 642:225-229. [PMID: 29032149 DOI: 10.1016/j.gene.2017.10.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/15/2017] [Accepted: 10/11/2017] [Indexed: 01/02/2023]
Abstract
Myocardial infarction (MI) is a complex multifactorial cardiovascular disease. India experiences a much greater burden of MI, also suggesting an experimental increase of this burden in the future. The absolute reasons for MI are context dependent and differ with different geographical settings. Several reports indicate that SNPs that are associated with certain diseases in other populations may not be associated with Indian population. It is, therefore, important to validate the association of SNPs. Low density lipoprotein receptor related protein 8 (LRP8) gene plays central role in human lipoprotein metabolism as it facilitates the clearance of bad cholesterol LDL, VLDL from plasma and is reported to be associated with MI in the western population. However, this gene has not been studied in the South Indian population. We aim to test the role of the LRP8 gene variants correlating with the lipid profile in MI patients in South Indian population. We sequenced regions of SNPs rs10788952, rs7546246, rs2297660 and rs5174 of LRP8 in 100 MI patients and 100 age-matched controls. Our result revealed a total of 4 variations. None of the SNPs were significantly associated with MI (p>0.973). Interestingly, haplotype based association analysis showed TG and CG of rs10788952 and rs7546246 significantly associated with MI (p<0.01 and p<0.00005) and in particular, haplotype TG was positively correlated with the risk of MI, as this increased the LDL and total cholesterol level in MI patients in south Indians. Our results suggest that haplotype TG is a risk factor for MI in South Indian population.
Collapse
Affiliation(s)
- Muhammed Asif
- Department of Anatomy, Yenepoya Medical College and Hospital, Mangalore 575018, Karnataka, India
| | - Shivarama Bhat
- Department of Anatomy, Yenepoya Medical College and Hospital, Mangalore 575018, Karnataka, India
| | - Sheikh Nizamuddin
- Centre for Cellular and Molecular Biology, Hyderabad, Telangana, India
| | - Mohammed S Mustak
- Department of Applied Zoology, Mangalore University, Mangalagangothri, 574199 Mangalore, India.
| |
Collapse
|
23
|
Naji DH, Tan C, Han F, Zhao Y, Wang J, Wang D, Fa J, Li S, Chen S, Chen Q, Xu C, Wang QK. Significant genetic association of a functional TFPI variant with circulating fibrinogen levels and coronary artery disease. Mol Genet Genomics 2017; 293:119-128. [PMID: 28894953 DOI: 10.1007/s00438-017-1365-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 08/29/2017] [Indexed: 01/17/2023]
Abstract
The tissue factor pathway inhibitor (TFPI) gene encodes a protease inhibitor with a critical role in regulation of blood coagulation. Some genomic variants in TFPI were previously associated with plasma TFPI levels, however, it remains to be further determined whether TFPI variants are associated with other coagulation factors. In this study, we carried out a large population-based study with 2313 study subjects for blood coagulation data, including fibrinogen levels, prothrombin time (PT), activated partial thromboplastin time (APTT), and thrombin time (TT). We identified significant association of TFPI variant rs10931292 (a functional promoter variant with reduced transactivation) with increased plasma fibrinogen levels (P = 0.017 under a recessive model), but not with PT, APTT or TT (P > 0.05). Using a large case-control association study population with 4479 CAD patients and 3628 controls, we identified significant association between rs10931292 and CAD under a recessive model (OR 1.23, P = 0.005). For the first time, we show that a TFPI variant is significantly associated with fibrinogen levels and risk of CAD. Our finding contributes significantly to the elucidation of the genetic basis and biological pathways responsible for fibrinogen levels and development of CAD.
Collapse
Affiliation(s)
- Duraid Hamid Naji
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Chengcheng Tan
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Fabin Han
- The Institute for Translational Medicine, The Second Affiliated Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yuanyuan Zhao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Junhan Wang
- University Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Dan Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jingjing Fa
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Sisi Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Shanshan Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qiuyun Chen
- Department of Molecular Cardiology, Center for Cardiovascular Genetics, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine/CCLCM, Case Western Reserve University, Cleveland, OH, 44195, USA.
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| | - Qing K Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China. .,Department of Molecular Cardiology, Center for Cardiovascular Genetics, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine/CCLCM, Case Western Reserve University, Cleveland, OH, 44195, USA. .,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44195, USA.
| |
Collapse
|
24
|
Genome-Wide Linkage Analysis of Large Multiple Multigenerational Families Identifies Novel Genetic Loci for Coronary Artery Disease. Sci Rep 2017; 7:5472. [PMID: 28710368 PMCID: PMC5511258 DOI: 10.1038/s41598-017-05381-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 05/30/2017] [Indexed: 01/10/2023] Open
Abstract
Coronary artery disease (CAD) is the leading cause of death, and genetic factors contribute significantly to risk of CAD. This study aims to identify new CAD genetic loci through a large-scale linkage analysis of 24 large and multigenerational families with 433 family members (GeneQuest II). All family members were genotyped with markers spaced by every 10 cM and a model-free nonparametric linkage (NPL-all) analysis was carried out. Two highly significant CAD loci were identified on chromosome 17q21.2 (NPL score of 6.20) and 7p22.2 (NPL score of 5.19). We also identified four loci with significant NPL scores between 4.09 and 4.99 on 2q33.3, 3q29, 5q13.2 and 9q22.33. Similar analyses in individual families confirmed the six significant CAD loci and identified seven new highly significant linkages on 9p24.2, 9q34.2, 12q13.13, 15q26.1, 17q22, 20p12.3, and 22q12.1, and two significant loci on 2q11.2 and 11q14.1. Two loci on 3q29 and 9q22.33 were also successfully replicated in our previous linkage analysis of 428 nuclear families. Moreover, two published risk variants, SNP rs46522 in UBE2Z and SNP rs6725887 in WDR12 by GWAS, were found within the 17q21.2 and 2q33.3 loci. These studies lay a foundation for future identification of causative variants and genes for CAD.
Collapse
|
25
|
Sacharidou A, Shaul PW, Mineo C. New Insights in the Pathophysiology of Antiphospholipid Syndrome. Semin Thromb Hemost 2017; 44:475-482. [PMID: 28129662 DOI: 10.1055/s-0036-1597286] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The antiphospholipid syndrome (APS) is an autoimmune disorder characterized by an elevated risk for arterial and venous thrombosis and pregnancy-related morbidity. Since the discovery of the disease in 1980s, numerous studies in cell culture systems, in animal models, and in patient populations have been reported, leading to a deeper understanding of the pathogenesis of APS. These studies have determined that circulating autoantibodies, collectively called antiphospholipid antibodies (aPL), the majority of which recognize cell surface proteins attached to the plasma membrane phospholipids, play a causal role in the development of the disease. The binding of aPL to the cell surface antigens triggers interaction of the complex with transmembrane receptors to initiate intracellular signaling in critical cell types, including platelets, monocytes, endothelial cells, and trophoblasts. Subsequent alteration of various cell functions results in inflammation, thrombus formation, and pregnancy complications. Apolipoprotein E receptor 2 (apoER2), a lipoprotein receptor family member, has been implicated as a mediator for aPL actions in platelets and endothelial cells. Nitric oxide (NO) is a signaling molecule known to exert potent antithrombotic, anti-inflammatory, and anti-atherogenic effects. NO insufficiency and oxidative stress have been linked to APS pathogenesis. This review will focus on the recent findings on how apoER2 and dysregulation of NO production contribute to aPL-mediated pathologies in APS.
Collapse
Affiliation(s)
- Anastasia Sacharidou
- Department of Pediatrics, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philip W Shaul
- Department of Pediatrics, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chieko Mineo
- Department of Pediatrics, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
26
|
Integrative mutation, haplotype and G × G interaction evidence connects ABGL4, LRP8 and PCSK9 genes to cardiometabolic risk. Sci Rep 2016; 6:37375. [PMID: 27853278 PMCID: PMC5112603 DOI: 10.1038/srep37375] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/28/2016] [Indexed: 12/19/2022] Open
Abstract
This study is expected to investigate the association of ATP/GTP binding protein-like 4 (AGBL4), LDL receptor related protein 8 (LRP8) and proprotein convertase subtilisin/kexin type 9 (PCSK9) gene single nucleotide variants (SNVs) with lipid metabolism in 2,552 individuals (Jing, 1,272 and Han, 1,280). We identified 12 mutations in this motif. The genotype and allele frequencies of these variants were different between the two populations. Multiple-locus linkage disequilibrium (LD) elucidated the detected sites are not statistically independent. Possible integrative haplotypes and gene-by-gene (G × G) interactions, comprising mutations of the AGBL4, LRP8 and PCSK9 associated with total cholesterol (TC, AGBL4 G-G-A, PCSK9 C-G-A-A and G-G-A-A-C-A-T-T-T-G-G-A), triglyceride (TG, AGBL4 G-G-A, LRP8 G-A-G-C-C, PCSK9 C-A-A-G, A-A-G-G-A-G-C-C-C-A-A-G and A-A-G-G-A-G-C-C-C-G-A-A), HDL cholesterol (HDL-C, AGBL4 A-A-G and A-A-G-A-A-G-T-C-C-A-A-G) and the apolipoprotein(Apo)A1/ApoB ratio (A1/B, PCSK9 C-A-A-G) in Jing minority. However, in the Hans, with TG (AGBL4 G-G-A, LRP8 G-A-G-C-C, PCSK9 C-A-A-G, A-A-G-G-A-G-C-C-C-A-A-G and A-A-G-G-A-G-C-C-C-G-A-A), HDL-C (LRP8 A-A-G-T-C), LDL-C (LRP8 A-A-G-T-C and A-A-G-A-A-G-T-C-C-A-A-G) and A1/B (LRP8 A-C-A-T-T and PCSK9 C-A-A-G). Association analysis based on haplotype clusters and G × G interactions probably increased power over single-locus tests especially for TG.
Collapse
|
27
|
Okoro EU, Zhang H, Guo Z, Yang F, Smith C, Yang H. A Subregion of Reelin Suppresses Lipoprotein-Induced Cholesterol Accumulation in Macrophages. PLoS One 2015; 10:e0136895. [PMID: 26317415 PMCID: PMC4552883 DOI: 10.1371/journal.pone.0136895] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/10/2015] [Indexed: 11/18/2022] Open
Abstract
Activation of apolipoprotein E receptor-2 (apoER2) and very low density lipoprotein receptor (VLDLR) inhibits foam cell formation. Reelin is a ligand of these receptors. Here we generated two reelin subregions containing the receptor binding domain with or without its C-terminal region (R5-6C and R5-6, respectively) and studied the impact of these peptides on macrophage cholesterol metabolism. We found that both R5-6C and R5-6 can be secreted by cells. Purified R5-6 protein can bind apoER2 and VLDLR. Overexpression of apoER2 in macrophages increased the amount of R5-6 bound to the cell surface. Treatment of macrophages with 0.2 μg/ml R5-6 elevated ATP binding cassette A1 (ABCA1) protein level by ~72% and apoAI-mediated cholesterol efflux by ~39%. In addition, the medium harvested from cells overexpressing R5-6 or R5-6C (R5-6- and R5-6C-conditioned media, respectively) also up-regulated ABCA1 protein expression, which was associated with accelerated cholesterol efflux and enhanced phosphorylation of phosphatidylinositol 3 kinase (PI3K) and specificity protein-1 (Sp1) in macrophages. The increased ABCA1 expression and cholesterol efflux by R5-6- and R5-6C-conditioned media were diminished by Sp1 or PI3K inhibitors mithramycin A and LY294002. Further, the cholesterol accumulation induced by apoB-containing, apoE-free lipoproteins was significantly less in macrophages incubated with R5-6- or R5-6C-conditioned medium than in those incubated with control conditioned medium. Knockdown of apoER2 or VLDLR attenuated the inhibitory role of R5-6-conditioned medium against lipoprotein-induced cholesterol accumulation. These results suggest that the reelin subregion R5-6 can serve as a tool for studying the role of apoER2 and VLDLR in atherogenesis.
Collapse
Affiliation(s)
- Emmanuel U. Okoro
- Department of Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Hongfeng Zhang
- Department of Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
- Department of Pathology, Central Hospital of Wuhan, Wuhan City, People’s Republic of China
| | - Zhongmao Guo
- Department of Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Fang Yang
- Department of Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
- Wuhan University School of Basic Medical Science, Wuhan City, People’s Republic of China
| | - Carlie Smith
- Department of Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
| | - Hong Yang
- Department of Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
28
|
Suppression of cell division-associated genes by Helicobacter pylori attenuates proliferation of RAW264.7 monocytic macrophage cells. Sci Rep 2015; 5:11046. [PMID: 26078204 PMCID: PMC4468580 DOI: 10.1038/srep11046] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 05/07/2015] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori at multiplicity of infection (MOI ≥ 50) have been shown to cause apoptosis in RAW264.7 monocytic macrophage cells. Because chronic gastric infection by H. pylori results in the persistence of macrophages in the host's gut, it is likely that H. pylori is present at low to moderate, rather than high numbers in the infected host. At present, the effect of low-MOI H. pylori infection on macrophage has not been fully elucidated. In this study, we investigated the genome-wide transcriptional regulation of H. pylori-infected RAW264.7 cells at MOI 1, 5 and 10 in the absence of cellular apoptosis. Microarray data revealed up- and down-regulation of 1341 and 1591 genes, respectively. The expression of genes encoding for DNA replication and cell cycle-associated molecules, including Aurora-B kinase (AurkB) were down-regulated. Immunoblot analysis verified the decreased expression of AurkB and downstream phosphorylation of Cdk1 caused by H. pylori infection. Consistently, we observed that H. pylori infection inhibited cell proliferation and progression through the G1/S and G2/M checkpoints. In summary, we suggest that H. pylori disrupts expression of cell cycle-associated genes, thereby impeding proliferation of RAW264.7 cells, and such disruption may be an immunoevasive strategy utilized by H. pylori.
Collapse
|
29
|
Hussain S, Iqbal T, Javed Q. TNF-alpha-308G>A polymorphism and the risk of familial CAD in a Pakistani population. Hum Immunol 2014; 76:13-8. [PMID: 25500256 DOI: 10.1016/j.humimm.2014.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 07/17/2014] [Accepted: 12/03/2014] [Indexed: 01/10/2023]
Abstract
A case-control and trio-families study was performed to establish a potential association between TNF-alpha gene promoter SNPs at -308 and -238, and occurrence of CAD in a Pakistani population. In the first phase, 150 patients and 150 controls were enrolled in the case-control association study. In the second phase, heritability of susceptible alleles was investigated from 88 trio-families with CAD affected offspring. Biochemical analysis of lipids and hs-CRP was carried out spectrophotometrically, while serum TNF-alpha concentrations were determined by enzyme-linked immunosorbent assay. Genotyping of the TNF-alpha SNPs were determined by PCR-RFLP method. Elevated serum TNF-alpha and hs-CRP were observed from CAD vs. controls (P<0.0001; for both). The evaluation of TNF-alpha-308G>A polymorphism in case-control study revealed that the said SNP was significantly associated with the increased risk of CAD. The findings demonstrated a significant link between the TNF-alpha variant allele A at -308 and CAD (P=0.0035), whereas the -238 SNP was not associated with the disease. Haplotype A-G of the TNF-alpha gene at -308G>A and -238G>A showed higher frequency in the patient group compared with controls (P<0.05). Moreover, data showed preferential transmission of the disease susceptible allele A at TNF-alpha-308 from parent to affected offspring in a trio-family study (P<0.0001). The current research leads to conclusion that the TNF-alpha-308G>A polymorphism is associated with CAD in the study population. Furthermore, for the first time, we showed that the TNF-alpha-308A allele was significantly associated with the familial CAD in our high risk population.
Collapse
Affiliation(s)
- Sabir Hussain
- Department of Biosciences, COMSATS Institute of Information Technology, Park Road, Chak Shazad, Islamabad 44000, Pakistan.
| | - Tahir Iqbal
- Department of Internal Medicine, Shifa College of Medicine, Shifa International Hospital, H-8/4, Islamabad 44000, Pakistan
| | - Qamar Javed
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|
30
|
Gao H, Li L, Rao S, Shen G, Xi Q, Chen S, Zhang Z, Wang K, Ellis SG, Chen Q, Topol EJ, Wang QK. Genome-wide linkage scan identifies two novel genetic loci for coronary artery disease: in GeneQuest families. PLoS One 2014; 9:e113935. [PMID: 25485937 PMCID: PMC4259362 DOI: 10.1371/journal.pone.0113935] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/30/2014] [Indexed: 11/18/2022] Open
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Recent genome-wide association studies (GWAS) identified >50 common variants associated with CAD or its complication myocardial infarction (MI), but collectively they account for <20% of heritability, generating a phenomena of “missing heritability”. Rare variants with large effects may account for a large portion of missing heritability. Genome-wide linkage studies of large families and follow-up fine mapping and deep sequencing are particularly effective in identifying rare variants with large effects. Here we show results from a genome-wide linkage scan for CAD in multiplex GeneQuest families with early onset CAD and MI. Whole genome genotyping was carried out with 408 markers that span the human genome by every 10 cM and linkage analyses were performed using the affected relative pair analysis implemented in GENEHUNTER. Affected only nonparametric linkage (NPL) analysis identified two novel CAD loci with highly significant evidence of linkage on chromosome 3p25.1 (peak NPL = 5.49) and 3q29 (NPL = 6.84). We also identified four loci with suggestive linkage on 9q22.33, 9q34.11, 17p12, and 21q22.3 (NPL = 3.18–4.07). These results identify novel loci for CAD and provide a framework for fine mapping and deep sequencing to identify new susceptibility genes and novel variants associated with risk of CAD.
Collapse
Affiliation(s)
- Hanxiang Gao
- Heart Center, the First Affiliated Hospital, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
| | - Lin Li
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
| | - Shaoqi Rao
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
- Institute of Medical Systems Biology and School of Public Health, Guangdong Medical College, Dongguan, Guangdong, 523808, P. R. China
| | - Gongqing Shen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
| | - Quansheng Xi
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
| | - Shenghan Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
| | - Zheng Zhang
- Heart Center, the First Affiliated Hospital, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Kai Wang
- Center for Cardiovascular Genetics, Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
| | - Stephen G. Ellis
- Center for Cardiovascular Genetics, Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
| | - Qiuyun Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
| | - Eric J. Topol
- Scripps Translational Science Institute, Scripps Research Institute, Scripps Clinic, La Jolla, California, 92037, United States of America
- * E-mail: (EJT); (QKW)
| | - Qing K. Wang
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
- Center for Cardiovascular Genetics, Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart and Vascular Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
- Center for Sleep Medicine, Neurological Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, Ohio, United States of America
- Department of Genetics and Genome Sciences, Case Western Reserve University, 9500 Euclid Ave., Cleveland, Ohio, 44195, United States of America
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P.R. China
- * E-mail: (EJT); (QKW)
| |
Collapse
|
31
|
Genetic variants of ApoE and ApoER2 differentially modulate endothelial function. Proc Natl Acad Sci U S A 2014; 111:13493-8. [PMID: 25197062 DOI: 10.1073/pnas.1402106111] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
It is poorly understood why there is greater cardiovascular disease risk associated with the apolipoprotein E4 (apoE) allele vs. apoE3, and also greater risk with the LRP8/apolipoprotein E receptor 2 (ApoER2) variant ApoER2-R952Q. Little is known about the function of the apoE-ApoER2 tandem outside of the central nervous system. We now report that in endothelial cells apoE3 binding to ApoER2 stimulates endothelial NO synthase (eNOS) and endothelial cell migration, and it also attenuates monocyte-endothelial cell adhesion. However, apoE4 does not stimulate eNOS or endothelial cell migration or dampen cell adhesion, and alternatively it selectively antagonizes apoE3/ApoER2 actions. The contrasting endothelial actions of apoE4 vs. apoE3 require the N-terminal to C-terminal interaction in apoE4 that distinguishes it structurally from apoE3. Reconstitution experiments further reveal that ApoER2-R952Q is a loss-of-function variant of the receptor in endothelium. Carotid artery reendothelialization is decreased in ApoER2(-/-) mice, and whereas adenoviral-driven apoE3 expression in wild-type mice has no effect, apoE4 impairs reendothelialization. Moreover, in a model of neointima formation invoked by carotid artery endothelial denudation, ApoER2(-/-) mice display exaggerated neointima development. Thus, the apoE3/ApoER2 tandem promotes endothelial NO production, endothelial repair, and endothelial anti-inflammatory properties, and it prevents neointima formation. In contrast, apoE4 and ApoER2-R952Q display dominant-negative action and loss of function, respectively. Thus, genetic variants of apoE and ApoER2 impact cardiovascular health by differentially modulating endothelial function.
Collapse
|
32
|
Shen GQ, Girelli D, Li L, Rao S, Archacki S, Olivieri O, Martinelli N, Park JE, Chen Q, Topol EJ, Wang QK. A novel molecular diagnostic marker for familial and early-onset coronary artery disease and myocardial infarction in the LRP8 gene. ACTA ACUST UNITED AC 2014; 7:514-20. [PMID: 24867879 DOI: 10.1161/circgenetics.113.000321] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Many single-nucleotide polymorphisms have been associated with coronary artery disease (CAD)/myocardial infarction (MI) by genome-wide association studies, but the diagnostic value of these variants is limited. Functional single-nucleotide polymorphism R952Q in LRP8 is associated with familial and early-onset CAD/MI. The objective of this study is to test whether fine mapping and haplotype analysis for single-nucleotide polymorphisms flanking R952Q may identify a haplotype that may serve as a molecular diagnostic marker for familial and early-onset CAD/MI. METHODS AND RESULTS Five single-nucleotide polymorphisms (rs7546246, rs2297660, rs3737983, R952Q, and rs5177) were genotyped and analyzed in GeneQuest (381 patients with familial, early-onset CAD and 183 patients with MI versus 560 controls) and the Italian population (248 patients with familial MI versus 308 controls). One novel risk haplotype, TACGC, was found only in patients with CAD and MI but not in controls. It was significantly associated with CAD (P=7.4×10(-7)) and MI (P=2.2×10(-9)) in GeneQuest. The finding was replicated in the Italian cohort (P=0.041). Sib-transmission disequilibrium test analysis showed a significant association between haplotype TACGC and CAD in GeneQuest II (P=0.039). Haplotype TACGC was not present in a South Korean population of 611 patients with CAD and 294 normal controls. TACGC/TACGC homozygotes tended to develop CAD/MI earlier and showed higher low-density lipoprotein cholesterol levels than heterozygotes (P<0.05). CONCLUSIONS The rare haplotype TACGC in LRP8 confers a significant risk of familial, early-onset CAD/MI. Because the risk haplotype exists only in patients with familial and early-onset CAD/MI, we propose that it may be a molecular diagnostic marker for diagnosis of familial, early-onset CAD/MI in some white populations.
Collapse
Affiliation(s)
- Gong-Qing Shen
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Domenico Girelli
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Lin Li
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Shaoqi Rao
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Stephen Archacki
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Oliviero Olivieri
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Nicola Martinelli
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Jeong Euy Park
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Qiuyun Chen
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Eric J Topol
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.)
| | - Qing K Wang
- From the Department of Molecular Cardiology, Center for Cardiovascular Genetics, Lerner Research Institute, Cleveland Clinic, OH (G.-Q.S., L.L., S.R., S.A., Q.C., Q.K.W.); Department of Medicine, University of Verona, Verona, Italy (D.G., O.O., N.M.); Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea (J.E.P.); The Scripps Research Institute, Scripps Clinic, La Jolla, CA (E.J.T.); and Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China (Q.K.W.).
| |
Collapse
|
33
|
Waltmann MD, Basford JE, Konaniah ES, Weintraub NL, Hui DY. Apolipoprotein E receptor-2 deficiency enhances macrophage susceptibility to lipid accumulation and cell death to augment atherosclerotic plaque progression and necrosis. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1395-405. [PMID: 24840660 DOI: 10.1016/j.bbadis.2014.05.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 01/29/2023]
Abstract
Genome-wide association studies have linked LRP8 polymorphisms to premature coronary artery disease and myocardial infarction in humans. However, the mechanisms by which dysfunctions of apolipoprotein E receptor-2 (apoER2), the protein encoded by LRP8 gene, influence atherosclerosis have not been elucidated completely. The current study focused on the role of apoER2 in macrophages, a cell type that plays an important role in atherosclerosis. Results showed that apoER2-deficient mouse macrophages accumulated more lipids and were more susceptible to oxidized LDL (oxLDL)-induced death compared to control cells. Consistent with these findings, apoER2 deficient macrophages also displayed defective serum-induced Akt activation and higher levels of the pro-apoptotic protein phosphorylated p53. Furthermore, the expression and activation of peroxisome proliferator-activated receptor γ (PPARγ) were increased in apoER2-deficient macrophages. Deficiency of apoER2 in hypercholesterolemic LDL receptor-null mice (Lrp8(-/-)Ldlr(-/-) mice) also resulted in accelerated atherosclerosis with more complex lesions and extensive lesion necrosis compared to Lrp8(+/+)Ldlr(-/-) mice. The atherosclerotic plaques of Lrp8(-/-)Ldlr(-/-) mice displayed significantly higher levels of p53-positive macrophages, indicating that the apoER2-deficient macrophages contribute to the accelerated atherosclerotic lesion necrosis observed in these animals. Taken together, this study indicates that apoER2 in macrophages limits PPARγ expression and protects against oxLDL-induced cell death. Thus, abnormal apoER2 functions in macrophages may at least in part contribute to the premature coronary artery disease and myocardial infarction in humans with LRP8 polymorphisms. Moreover, the elevated PPARγ expression in apoER2-deficient macrophages suggests that LRP8 polymorphism may be a genetic modifier of cardiovascular risk with PPARγ therapy.
Collapse
Affiliation(s)
- Meaghan D Waltmann
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Joshua E Basford
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Eddy S Konaniah
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Neal L Weintraub
- Department of Internal Medicine, Division of Cardiovascular Disease, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA.
| |
Collapse
|
34
|
de Gonzalo-Calvo D, Revuelta-López E, Llorente-Cortés V. [Basic mechanisms. Regulation and clearance of lipoproteins that contain apolipoprotein B]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2013; 25:194-200. [PMID: 23768652 DOI: 10.1016/j.arteri.2013.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 05/17/2013] [Indexed: 06/02/2023]
Affiliation(s)
- David de Gonzalo-Calvo
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Barcelona, España
| | | | | |
Collapse
|
35
|
Chen K, Chen YP, Song W, Huang R, Zhao B, Cao B, Yang Y, Satake W, Toda T, Shang HF. Association analysis of LRP8 SNP rs3820198 and rs5174 with Parkinson's disease in Han Chinese population. Neurol Res 2013; 34:725-9. [PMID: 22889673 DOI: 10.1179/1743132812y.0000000075] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES The single-nucleotide polymorphism (SNP) rs5174 of the low-density lipoprotein receptor-related protein 8 (LRP8) gene has been linked to decreased risk for Parkinson's disease (PD) on Caucasian populations. However, this association has not been proven in Han Chinese populations. METHODS A total of 378 unrelated Han patients with PD from the Department of Neurology, West China Hospital of Sichuan University and 274 unrelated Han healthy controls (HCs) from the same region were included in this study. SNPs rs5174 and rs3820198 were genotyped using the Sequenom iPLEX Assay technology. RESULTS No significant difference was found in the genotype and minor allele frequencies (MAFs) of SNPs rs5174 and rs3820198 between the PD and HC groups, the early-onset PD and HC groups, the late-onset PD and HC groups, as well as the early-onset PD and late-onset PD groups. CONCLUSION This report is the first one on the lack of association of the LRP8 SNPs rs5174 and rs3820198 with PD in Han Chinese population. Together with a Japanese study, the results indicate that the variants within the LRP8 gene do not contribute to the risk of developing PD in Asian populations.
Collapse
Affiliation(s)
- Ke Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Shen GQ, Girelli D, Li L, Olivieri O, Martinelli N, Chen Q, Topol EJ, Wang QK. Multi-allelic haplotype association identifies novel information different from single-SNP analysis: a new protective haplotype in the LRP8 gene is against familial and early-onset CAD and MI. Gene 2013; 521:78-81. [PMID: 23524007 DOI: 10.1016/j.gene.2013.03.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/07/2013] [Indexed: 12/13/2022]
Abstract
Our previous studies identified a functional SNP, R952Q in the LRP8 gene, that was associated with increased platelet activation and familial and early-onset coronary artery disease (CAD) and myocardial infarction (MI) in American and Italian Caucasian populations. In this study, we analyzed four additional SNPs near R952Q (rs7546246, rs2297660, rs3737983, rs5177) to identify a specific LRP8 SNP haplotype that is associated with familial and early-onset CAD and MI. We employed a case-control association design involving 381 premature CAD and MI probands and 560 controls in GeneQuest, 441 individuals from 22 large pedigrees in GeneQuest II, and 248 MI patients with family history and 308 controls in an Italian cohort. Like R952Q, LRP8 SNPs rs7546246, rs2297660, rs3737983, and rs5177 were significantly associated with early-onset CAD/MI in both population-based and family-based association studies in GeneQuest. The results were replicated in the GeneQuest II family-based population and the Italian population. We then carried out a haplotype analysis for all five SNPs including R952Q. One common haplotype (TCCGC) was significantly associated with CAD (P=4.0×10(-11)) and MI (P=6.5×10(-12)) in GeneQuest with odds ratios of 0.53 and 0.42, respectively. The results were replicated in the Italian cohort (P=0.004, OR=0.71). The sib-TDT analysis also showed significant association between the TCCGC haplotype and CAD in GeneQuest II (P=0.001). These results suggest that a common LRP8 haplotype TCCGC confers a significant protective effect on the development of familial, early-onset CAD and/or MI.
Collapse
Affiliation(s)
- Gong-Qing Shen
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Baardman ME, Kerstjens-Frederikse WS, Berger RM, Bakker MK, Hofstra RM, Plösch T. The Role of Maternal-Fetal Cholesterol Transport in Early Fetal Life: Current Insights1. Biol Reprod 2013; 88:24. [DOI: 10.1095/biolreprod.112.102442] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
38
|
Hu S, Liu H, Pan Z, Ding F, Kou J, Li L, Wang J. The cloning, characterization, and expression profiling of the LRP8 gene in duck (Anas platyrhynchos). Mol Cell Biochem 2012; 375:139-49. [PMID: 23224277 DOI: 10.1007/s11010-012-1536-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 11/23/2012] [Indexed: 01/22/2023]
Abstract
Low-density lipoprotein receptor-related protein 8 (LRP8) is a member of the low-density lipoprotein receptor gene family that functions in body lipoprotein homeostasis. In this study, reverse transcription-polymerase chain reaction, rapid amplification of cDNA ends, and real-time PCR were performed to characterize the duck LRP8 gene. The cDNA of duck LRP8 contained a 14-bp 5' UTR, a 2754-bp open reading frame, and a 189-bp 3' UTR. The duck LRP8 encoded a protein of 917 amino acid residues composed of five functional domains and resembling other members of the LDLR family, and it displayed high nucleotide and amino acid homology to the LRP8 sequences in other avian species. The mRNA expression level of LRP8 was greater in duck extra-hepatic adipose tissue than in the liver. The peak expression values of LRP8 in both liver and adipose tissues occurred at week 1 and were significantly higher than the values observed during any other week (p < 0.05). Differences in the expression patterns of LRP8 mRNA from weeks 2 to 8 of growth were observed in different organs. A consistent low expression was observed in the liver, and fluctuating expression was observed in the subcutaneous adipose tissue (up- and then down-regulated) and abdominal adipose tissue (down-, then up-, then down-regulated). These findings suggest that LRP8 might play more important roles in regulating lipid metabolism in extra-hepatic adipose tissues than in the liver during early growth after hatching in the duck.
Collapse
Affiliation(s)
- Shenqiang Hu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Ya'an 625014, Sichuan, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
39
|
Gupta R, Ratan A, Rajesh C, Chen R, Kim HL, Burhans R, Miller W, Santhosh S, Davuluri RV, Butte AJ, Schuster SC, Seshagiri S, Thomas G. Sequencing and analysis of a South Asian-Indian personal genome. BMC Genomics 2012; 13:440. [PMID: 22938532 PMCID: PMC3534380 DOI: 10.1186/1471-2164-13-440] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 08/18/2012] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND With over 1.3 billion people, India is estimated to contain three times more genetic diversity than does Europe. Next-generation sequencing technologies have facilitated the understanding of diversity by enabling whole genome sequencing at greater speed and lower cost. While genomes from people of European and Asian descent have been sequenced, only recently has a single male genome from the Indian subcontinent been published at sufficient depth and coverage. In this study we have sequenced and analyzed the genome of a South Asian Indian female (SAIF) from the Indian state of Kerala. RESULTS We identified over 3.4 million SNPs in this genome including over 89,873 private variations. Comparison of the SAIF genome with several published personal genomes revealed that this individual shared ~50% of the SNPs with each of these genomes. Analysis of the SAIF mitochondrial genome showed that it was closely related to the U1 haplogroup which has been previously observed in Kerala. We assessed the SAIF genome for SNPs with health and disease consequences and found that the individual was at a higher risk for multiple sclerosis and a few other diseases. In analyzing SNPs that modulate drug response, we found a variation that predicts a favorable response to metformin, a drug used to treat diabetes. SNPs predictive of adverse reaction to warfarin indicated that the SAIF individual is not at risk for bleeding if treated with typical doses of warfarin. In addition, we report the presence of several additional SNPs of medical relevance. CONCLUSIONS This is the first study to report the complete whole genome sequence of a female from the state of Kerala in India. The availability of this complete genome and variants will further aid studies aimed at understanding genetic diversity, identifying clinically relevant changes and assessing disease burden in the Indian population.
Collapse
Affiliation(s)
- Ravi Gupta
- SciGenom Labs Pvt Ltd., Plot 43A, SDF 3rd Floor CSEZ, Kakkanad, Cochin, Kerala, 682037, India
| | - Aakrosh Ratan
- Center for Comparative Genomics and Bioinformatics, Pennsylvania State University, 310 Wartik Lab, University Park, , Pennsylvania, 16802, USA
| | - Changanamkandath Rajesh
- SciGenom Labs Pvt Ltd., Plot 43A, SDF 3rd Floor CSEZ, Kakkanad, Cochin, Kerala, 682037, India
| | - Rong Chen
- , , Personalis, 1350 Willow Road, Suite 202, Menlo Park, CA, 94025, USA
| | - Hie Lim Kim
- Center for Comparative Genomics and Bioinformatics, Pennsylvania State University, 310 Wartik Lab, University Park, , Pennsylvania, 16802, USA
| | - Richard Burhans
- Center for Comparative Genomics and Bioinformatics, Pennsylvania State University, 310 Wartik Lab, University Park, , Pennsylvania, 16802, USA
| | - Webb Miller
- Center for Comparative Genomics and Bioinformatics, Pennsylvania State University, 310 Wartik Lab, University Park, , Pennsylvania, 16802, USA
| | - Sam Santhosh
- SciGenom Labs Pvt Ltd., Plot 43A, SDF 3rd Floor CSEZ, Kakkanad, Cochin, Kerala, 682037, India
| | - Ramana V Davuluri
- Center for Systems The Wistar Institute,, , Philadelphia, PA, 19104, USA
| | - Atul J Butte
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephan C Schuster
- Center for Comparative Genomics and Bioinformatics, Pennsylvania State University, 310 Wartik Lab, University Park, , Pennsylvania, 16802, USA
- Singapore Centre on Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, SBS-01N-27, Singapore, Singapore , 637551
| | - Somasekar Seshagiri
- Department of Molecular Biology, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - George Thomas
- SciGenom Labs Pvt Ltd., Plot 43A, SDF 3rd Floor CSEZ, Kakkanad, Cochin, Kerala, 682037, India
| |
Collapse
|
40
|
Schunkert H, König IR, Erdmann J. Molecular Signatures of Cardiovascular Disease Risk. Mol Diagn Ther 2012; 12:281-7. [DOI: 10.1007/bf03256293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
41
|
Shen GQ, Li L, Wang QK. Genetic variant R952Q in LRP8 is associated with increased plasma triglyceride levels in patients with early-onset CAD and MI. Ann Hum Genet 2012; 76:193-9. [PMID: 22404453 DOI: 10.1111/j.1469-1809.2012.00705.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We previously identified a novel locus for plasma triglyceride (TG) levels on chromosome 1p31-32 by genome-wide linkage analysis in the GeneQuest population with familial and premature coronary artery disease (CAD). Here we tested a hypothesis that variants in LRP8, a gene that is under the 1p31-32 linkage peak and associated with risk of familial and premature CAD and increased platelet activation, are associated with TG levels. Seven tagSNPs that cover the entire LRP8 gene were characterized in 358 GeneQuest Caucasian probands. Only SNP R952Q (rs5174) was associated with TG levels (P-adj = 0.0016), and this finding was replicated in one other independent population of 134 patients with early-onset myocardial infarction (males <45; females <55; P-adj = 0.0098). TG levels were higher in the group with higher body mass index (BMI ≥ 25) than in the group with lower BMI (BMI < 25). The association was significant in the overweight group (P-adj = 0.0029) or in the smoking group (P-adj = 0.0004), but not in the group with normal BMI or without smoking history. These results suggest that genetic variant R952Q of LRP8 is associated with increased plasma TG levels in patients who are overweight and have premature CAD/MI and history of smoking.
Collapse
Affiliation(s)
- Gong-Qing Shen
- Center for Cardiovascular Genetics, Cleveland Clinic, OH, USA
| | | | | |
Collapse
|
42
|
Chen X, Guo Z, Okoro EU, Zhang H, Zhou L, Lin X, Rollins AT, Yang H. Up-regulation of ATP binding cassette transporter A1 expression by very low density lipoprotein receptor and apolipoprotein E receptor 2. J Biol Chem 2011; 287:3751-9. [PMID: 22170052 DOI: 10.1074/jbc.m111.310888] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of very low density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (apoER2) results in either pro- or anti-atherogenic effects depending on the ligand. Using reelin and apoE as ligands, we studied the impact of VLDLR- and apoER2-mediated signaling on the expression of ATP binding cassette transporter A1 (ABCA1) and cholesterol efflux using RAW264.7 cells. Treatment of these mouse macrophages with reelin or human apoE3 significantly increased ABCA1 mRNA and protein levels, and apoAI-mediated cholesterol efflux. In addition, both reelin and apoE3 significantly increased phosphorylated disabled-1 (Dab1), phosphatidylinositol 3-kinase (PI3K), protein kinase Cζ (PKCζ), and specificity protein 1 (Sp1). This reelin- or apoER2-mediated up-regulation of ABCA1 expression was suppressed by 1) knockdown of Dab1, VLDLR, and apoER2 with small interfering RNAs (siRNAs), 2) inhibition of PI3K and PKC with kinase inhibitors, 3) overexpression of kinase-dead PKCζ, and 4) inhibition of Sp1 DNA binding with mithramycin A. Activation of the Dab1-PI3K signaling pathway has been implicated in VLDLR- and apoER2-mediated cellular functions, whereas the PI3K-PKCζ-Sp1 signaling cascade has been implicated in the regulation of ABCA1 expression induced by apoE/apoB-carrying lipoproteins. Taken together, these data support a model in which activation of VLDLR and apoER2 by reelin and apoE induces ABCA1 expression and cholesterol efflux via a Dab1-PI3K-PKCζ-Sp1 signaling cascade.
Collapse
Affiliation(s)
- Xinping Chen
- Department of Physiology, Meharry Medical College, Nashville, Tennessee 37208, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Rawal L, Ali S, Ali S. Molecular mining of GGAA tagged transcripts and their expression in water buffalo Bubalus bubalis. Gene 2011; 492:290-5. [PMID: 22037482 DOI: 10.1016/j.gene.2011.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/15/2011] [Accepted: 10/01/2011] [Indexed: 12/27/2022]
Abstract
Repeat sequences are involved in regulation of gene expression both at the transcriptional and translational level. In the mammalian genomes, tri- and tetranucleotide repeats like ATA, AATA, GGAA and GAAA have been associated with diseases. In silico analysis of (GGAA)5 distribution across the species showed maximum number of this repeat in the mouse transcriptome compared to that in other species. Following this, we conducted minisatellite associated sequence amplification (MASA) to explore the buffalo's transcriptome using cDNA from different tissues and an oligo based on (GGAA)5 repeats. MASA uncovered twenty six mRNA transcripts showing homology to known genes in the database. qPCR studies showed the highest expression of twelve transcripts in the spleen. A transcript, pLRC107 with its partial sequence of 203 nucleotides showed sequence variation at several positions in spleen as compared to other four tissues examined. Transcript pLRC100 was found to represent the partial coding sequence of Bos taurus HECT {(homologous to E6-associated protein (UBE3A) carboxyl-terminus domain) and RCC1 (CHC1)-like domain (RLD) 1}, mRNA. We ascertained full length coding sequence of HECT gene and localized the same on buffalo chromosome 10 employing FISH. This gene was found to be conserved across the species. Another gene LRP8 uncovered in the process showed copy number variation between buffalo males (4-9) and females (34-54). The MASA approach enabled us to identify several genes in Bubalus bubalis without screening an entire cDNA library. The highest expression of 12 mRNA transcripts in spleen suggests their likely involvement with immuno transaction. A comprehensive knowledge of the repeat tagged transcriptomes is envisaged to help in understanding their significance in genome organization and evolution forming rich basis of functional and comparative genomics.
Collapse
Affiliation(s)
- Leena Rawal
- Molecular Genetics Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi-110067, India.
| | | | | |
Collapse
|
44
|
Sivapalaratnam S, Motazacker MM, Maiwald S, Hovingh GK, Kastelein JJP, Levi M, Trip MD, Dallinga-Thie GM. Genome-wide association studies in atherosclerosis. Curr Atheroscler Rep 2011; 13:225-32. [PMID: 21369780 PMCID: PMC3085784 DOI: 10.1007/s11883-011-0173-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease remains the major cause of worldwide morbidity and mortality. Its pathophysiology is complex and multifactorial. Because the phenotype of cardiovascular disease often shows a marked heritable pattern, it is likely that genetic factors play an important role. In recent years, large genome-wide association studies have been conducted to decipher the molecular mechanisms underlying this heritable and prevalent phenotype. The emphasis of this review is on the recently identified 17 susceptibility loci for coronary artery disease. Implications of their discovery for biology and clinical medicine are discussed. A description of the landscape of human genetics in the near future in the context of next-generation sequence technologies is provided at the conclusion of this review.
Collapse
Affiliation(s)
- S Sivapalaratnam
- Department of Vascular Medicine, Academic Medical Center Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Yang Q, Li L, Chen Q, Foldvary-Schaefer N, Ondo WG, Wang QK. Association studies of variants in MEIS1, BTBD9, and MAP2K5/SKOR1 with restless legs syndrome in a US population. Sleep Med 2011; 12:800-4. [PMID: 21925394 PMCID: PMC3514407 DOI: 10.1016/j.sleep.2011.06.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Revised: 06/17/2011] [Accepted: 06/17/2011] [Indexed: 01/10/2023]
Abstract
BACKGROUND A genome-wide association study (GWAS) identified significant association between variants in MEIS1, BTBD9, and MAP2K5/SKOR1 and restless legs syndrome (RLS). However, many independent replication studies are needed to unequivocally establish a valid genotype-phenotype association across various populations. To further validate the GWAS findings, we investigated three variants, rs2300478 in MEIS1, rs9357271 in BTBD9, and rs1026732 in MAP2K5/SKOR1 in 38 RLS families and 189 RLS patients/560 controls from the US for their association with RLS. METHOD Both family-based and population-based case-control association studies were carried out. RESULTS The family-based study showed that SNP rs1026732 in MAP2K5/SKOR1 was significantly associated with RLS (P=0.01). Case-control association studies showed significant association between all three variants and RLS (P=0.0001/OR=1.65, P=0.0021/OR=1.59, and P=0.0011/OR=1.55 for rs2300478, rs9357271, and rs1026732, respectively). CONCLUSION Variants in MEIS1, BTBD9, and MAP2K5/SKOR1 confer a significant risk of RLS in a US population.
Collapse
Affiliation(s)
- Qinbo Yang
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, OH, USA
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
- Department of Molecular Medicine, CCLCM of Case Western Reserve University, Cleveland, OH, USA
| | - Lin Li
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, OH, USA
- Department of Molecular Medicine, CCLCM of Case Western Reserve University, Cleveland, OH, USA
| | - Qiuyun Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, OH, USA
- Department of Molecular Medicine, CCLCM of Case Western Reserve University, Cleveland, OH, USA
| | - Nancy Foldvary-Schaefer
- Center for Sleep Medicine, Neurological Institute, Cleveland Clinic, OH, USA
- Department of Molecular Medicine, CCLCM of Case Western Reserve University, Cleveland, OH, USA
| | - William G. Ondo
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Qing Kenneth Wang
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, OH, USA
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P. R. China
- Center for Sleep Medicine, Neurological Institute, Cleveland Clinic, OH, USA
- Department of Molecular Medicine, CCLCM of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
46
|
Platelet CD36 surface expression levels affect functional responses to oxidized LDL and are associated with inheritance of specific genetic polymorphisms. Blood 2011; 117:6355-66. [PMID: 21478428 DOI: 10.1182/blood-2011-02-338582] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CD36 modulates platelet function via binding to oxidized LDL (oxLDL), cell-derived microparticles, and thrombospondin-1. We hypothesized that the level of platelet CD36 expression may be associated with inheritance of specific genetic polymorphisms and that this would determine platelet reactivity to oxLDL. Analysis of more than 500 subjects revealed that CD36 expression levels were consistent in individual donors over time but varied widely among donors (200-14,000 molecules per platelet). Platelet aggregometry and flow cytometry in a subset of subjects with various CD36 expression levels revealed a high level of correlation (r² = 0.87) between platelet activation responses to oxLDL and level of CD36 expression. A genome-wide association study of 374 white subjects from the Cleveland Clinic ASCLOGEN study showed strong associations of single nucleotide polymorphisms in CD36 with platelet surface CD36 expression. Most of these findings were replicated in a smaller subset of 25 black subjects. An innovative gene-based genome-wide scan provided further evidence that single nucleotide polymorphisms in CD36 were strongly associated with CD36 expression. These studies show that CD36 expression on platelets varies widely, correlates with functional responses to oxLDL, and is associated with inheritance of specific CD36 genetic polymorphisms, and suggest that inheritance of specific CD36 polymorphisms could affect thrombotic risk.
Collapse
|
47
|
Wang AZ, Li L, Zhang B, Shen GQ, Wang QK. Association of SNP rs17465637 on chromosome 1q41 and rs599839 on 1p13.3 with myocardial infarction in an American caucasian population. Ann Hum Genet 2011; 75:475-82. [PMID: 21463265 DOI: 10.1111/j.1469-1809.2011.00646.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Recent genome-wide single nucleotide polymorphism (SNP) association studies (GWAS) have identified a number of SNPs that were significantly associated with coronary artery disease and myocardial infarction (MI). However, many independent replication studies in other populations are needed to unequivocally confirm the GWAS association. To assess GWAS association, we have established a case-control cohort consisting of 1231 well-characterised MI patients and 560 controls without detectable coronary stenosis, all selected from the Cleveland Genebank population. The Genebank cohort has sufficient power to detect the association between MI and four GWAS SNPs, including rs17465637 within the MIA3 gene, rs2943634 (intergenic), rs6922269 in MTHFD1L, and rs599839 near SORT1. SNPs were genotyped by TaqMan assays and follow-up multivariate logistic regression analysis with incorporation of significant covariates showed significant association with MI for MIA3 SNP rs17465637 (P-adj= 0.0034) and SORT1 SNP rs599839 (P-adj= 0.009). The minor allele G of rs599839 was also associated with a decreased LDL-C level of 5-9 mg/dL per allele, but not with HDL-C or triglyceride levels. No association for MI or lipid levels was found for SNPs rs2943634 and rs6922269 (P-adj > 0.05). Our results establish two SNPs, rs17465637 in MIA3 and rs599839 near SORT1 as significant risk factors for MI in the American Genebank Caucasian population.
Collapse
Affiliation(s)
- Annabel Z Wang
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | | | | | | | |
Collapse
|
48
|
Genome-wide association identifies a susceptibility locus for coronary artery disease in the Chinese Han population. Nat Genet 2011; 43:345-9. [PMID: 21378986 DOI: 10.1038/ng.783] [Citation(s) in RCA: 213] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 02/10/2011] [Indexed: 12/13/2022]
Abstract
Coronary artery disease (CAD) causes more than 700,000 deaths each year in China. Previous genome-wide association studies (GWAS) in populations of European ancestry identified several genetic loci for CAD, but no such study has yet been reported in the Chinese population. Here we report a three-stage GWAS in the Chinese Han population. We identified a new association between rs6903956 in a putative gene denoted as C6orf105 on chromosome 6p24.1 and CAD (P = 5.00 × 10⁻³, stage 2 validation; P = 3.00 × 10⁻³, P = 1.19 × 10⁻⁸ and P = 4.00 × 10⁻³ in three independent stage 3 replication populations; P = 4.87 × 10⁻¹², odds ratio = 1.51 in the combined population). The minor risk allele A of rs6903956 is associated with decreased C6orf105 mRNA expression. We report the first GWAS for CAD in the Chinese Han population and identify a SNP, rs6903956, in C6orf105 associated with susceptibility to CAD in this population.
Collapse
|
49
|
Yang Q, Li L, Yang R, Shen GQ, Chen Q, Foldvary-Schaefer N, Ondo WG, Wang QK. Family-based and population-based association studies validate PTPRD as a risk factor for restless legs syndrome. Mov Disord 2011; 26:516-9. [PMID: 21264940 DOI: 10.1002/mds.23459] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 08/12/2010] [Accepted: 09/07/2010] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE We previously mapped a genetic locus for restless legs syndrome (RLS) to chromosome 9p22-24 (RLS3) and a later genome-wide association study (GWAS) implicated the PTPRD gene at the RLS3 locus as a susceptibility gene for RLS. However, from the standpoint of genetics, the GWAS association needs to be validated by independent studies. In this study, we used both family-based and population-based association studies to assess the association between PTPRD and RLS in an American Caucasian population. METHODS We genotyped two intronic SNPs rs1975197 and rs4626664 in PTPRD in 144 family members from 15 families and a case control cohort of 189 patients and 560 controls. Direct DNA sequence analysis was used to screen coding exons and exon-intron boundaries of PTPRD for rare mutations. RESULTS A family-based sibling transmission disequilibrium test showed association of RLS with SNP rs1975197 (P = 0.015), but not with rs4626664 (P = 0.622). The association with rs1975197 was significantly replicated by a population-based case control association study (allelic P = 0.0004, odds ratio = 1.68; genotypic P = 0.0013 and 0.0003 for an additive and dominant model, respectively). One rare p.E1639D variant was identified in exon 39 in kindred RLS40005. The rare D1639 allele did not co-segregate with RLS in the family, suggesting that p.E1639D variant is not a causative mutation. CONCLUSIONS This represents the first independent study to validate the association between PTPRD variants and RLS. Both family-based and population-based association studies suggest that PTPRD variant rs1975197 confers risk of RLS.
Collapse
Affiliation(s)
- Qinbo Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Apolipoprotein E receptor 2 is involved in the thrombotic complications in a murine model of the antiphospholipid syndrome. Blood 2010; 117:1408-14. [PMID: 21119114 DOI: 10.1182/blood-2010-07-299099] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Antiphospholipid (aPL)/anti-β(2) glycoprotein I (anti-β(2)GPI) antibodies stimulates tissue factor (TF) expression within vasculature and in blood cells, thereby leading to increased thrombosis. Several cellular receptors have been proposed to mediate these effects, but no convincing evidence for the involvement of a specific one has been provided. We investigated the role of Apolipoprotein E receptor 2 (ApoER2') on the pathogenic effects of a patient-derived polyclonal aPL IgG preparation (IgG-APS), a murine anti-β(2)GPI monoclonal antibody (E7) and of a constructed dimeric β(2)GPI I (dimer), which in vitro mimics β(2)GPI-antibody immune complexes, using an animal model of thrombosis, and ApoER2-deficient (-/-) mice. In wild type mice, IgG-APS, E7 and the dimer increased thrombus formation, carotid artery TF activity as well as peritoneal macrophage TF activity/expression. Those pathogenic effects were significantly reduced in ApoER2 (-/-) mice. In addition, those effects induced by the IgG-APS, by E7 and by the dimer were inhibited by treatment of wild-type mice with soluble binding domain 1 of ApoER2 (sBD1). Altogether these data show that ApoER2 is involved in pathogenesis of antiphospholipids antibodies.
Collapse
|