1
|
Pan BY, Chen CS, Chen FY, Shen MY. Multifaceted Role of Apolipoprotein C3 in Cardiovascular Disease Risk and Metabolic Disorder in Diabetes. Int J Mol Sci 2024; 25:12759. [PMID: 39684468 PMCID: PMC11641554 DOI: 10.3390/ijms252312759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Apolipoprotein C3 (APOC3) plays a critical role in regulating triglyceride levels and serves as a key predictor of cardiovascular disease (CVD) risk, particularly in patients with diabetes. While APOC3 is known to inhibit lipoprotein lipase, recent findings reveal its broader influence across lipoprotein metabolism, where it modulates the structure and function of various lipoproteins. Therefore, this review examines the complex metabolic cycle of APOC3, emphasizing the impact of APOC3-containing lipoproteins on human metabolism, particularly in patients with diabetes. Notably, APOC3 affects triglyceride-rich lipoproteins and causes structural changes in high-, very low-, intermediate-, and low-density lipoproteins, thereby increasing CVD risk. Evidence suggests that elevated APOC3 levels-above the proposed safe range of 10-15 mg/dL-correlate with clinically significant CVD outcomes. Recognizing APOC3 as a promising biomarker for CVD, this review underscores the urgent need for high-throughput, clinically feasible methods to further investigate its role in lipoprotein physiology in both animal models and human studies. Additionally, we analyze the relationship between APOC3-related genes and lipoproteins, reinforcing the value of large-population studies to understand the impact of APOC3 on metabolic diseases. Ultimately, this review supports the development of therapeutic strategies targeting APOC3 reduction as a preventive approach for diabetes-related CVD.
Collapse
Affiliation(s)
- Bo-Yi Pan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (B.-Y.P.); (F.-Y.C.)
| | - Chen-Sheng Chen
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 40402, Taiwan;
| | - Fang-Yu Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (B.-Y.P.); (F.-Y.C.)
| | - Ming-Yi Shen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (B.-Y.P.); (F.-Y.C.)
- Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan
- Department of Nursing, Asia University, Taichung 413305, Taiwan
| |
Collapse
|
2
|
Fukase T, Dohi T, Nishio R, Takeuchi M, Takahashi N, Chikata Y, Endo H, Doi S, Nishiyama H, Okai I, Iwata H, Koga S, Okazaki S, Miyauchi K, Daida H, Minamino T. Association between apolipoprotein C-III levels and coronary calcification detected by intravascular ultrasound in patients who underwent percutaneous coronary intervention. Front Cardiovasc Med 2024; 11:1430203. [PMID: 39234605 PMCID: PMC11371589 DOI: 10.3389/fcvm.2024.1430203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
There are few reports on the association between apolipoprotein C-III (ApoC-III) and coronary calcification using intravascular modalities. This study aimed to investigate the impacts of ApoC-III levels on coronary calcification using grayscale intravascular ultrasound (IVUS). Consecutive 263 culprit lesions for 202 patients who underwent percutaneous coronary intervention using grayscale IVUS were included in this study and divided into four groups based on quartile ApoC-III values. This study assessed plaque characteristics, including severe calcification (>180° arc) at the minimum lumen area site and presence of calcified nodules within the culprit lesion using grayscale IVUS, and evaluated whether ApoC-III levels were associated with coronary calcified plaques. The highest ApoC-III quartile [Quartile 4 (Q4)] had a higher proportion of complex lesions, calcified plaques, severe calcification, calcified nodules, plaque burden, and total atheroma volume than the lowest ApoC-III quartile [Quartile 1 (Q1)]. Additionally, multivariable logistic regression analysis showed that Q4 was significantly associated with severe calcification and calcified nodules, with Q1 as the reference (odds ratio [OR]: 2.70, 95% confidence intervals [CIs]: 1.04-7.00, p = 0.042; and OR: 3.72, 95% CIs 1.26-11.0, p = 0.017, respectively). Furthermore, ApoC-III level (1-mg/dl increase) was a strong significant predictor of severe calcification (OR: 1.07, 95% CIs: 1.00-1.15, p = 0.040) and calcified nodules (OR: 1.09, 95% CIs: 1.01-1.19, p = 0.034) according to the multivariable logistic regression analysis. This study is the first to verify that elevated ApoC-III levels are associated with the development of severe calcification and progression to calcified nodules as detected by grayscale IVUS.
Collapse
Affiliation(s)
- Tatsuya Fukase
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Tomotaka Dohi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Ryota Nishio
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Mitsuhiro Takeuchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Norihito Takahashi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Yuichi Chikata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Hirohisa Endo
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Shinichiro Doi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Hiroki Nishiyama
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Iwao Okai
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Hiroshi Iwata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Seiji Koga
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Shinya Okazaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Katsumi Miyauchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
- Department of Radiological Technology, Faculty of Health Science, Juntendo University Graduate School, Bunkyo-ku, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Chiyoda-ku, Japan
| |
Collapse
|
3
|
Kim DH, Lee S, Noh SG, Lee J, Chung HY. FoxO6-mediated ApoC3 upregulation promotes hepatic steatosis and hyperlipidemia in aged rats fed a high-fat diet. Aging (Albany NY) 2024; 16:4095-4115. [PMID: 38441531 PMCID: PMC10968681 DOI: 10.18632/aging.205610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/24/2024] [Indexed: 03/22/2024]
Abstract
FoxO6, an identified factor, induces hyperlipidemia and hepatic steatosis during aging by activating hepatic lipoprotein secretion and lipogenesis leading to increased ApoC3 concentrations in the bloodstream. However, the intricate mechanisms underlying hepatic steatosis induced by elevated FoxO6 under hyperglycemic conditions remain intricate and require further elucidation. In order to delineate the regulatory pathway involving ApoC3 controlled by FoxO6 and its resultant functional impacts, we employed a spectrum of models including liver cell cultures, aged rats subjected to HFD, transgenic mice overexpressing FoxO6 (FoxO6-Tg), and FoxO6 knockout mice (FoxO6-KO). Our findings indicate that FoxO6 triggered ApoC3-driven lipid accumulation in the livers of aged rats on an HFD and in FoxO6-Tg, consequently leading to hepatic steatosis and hyperglycemia. Conversely, the absence of FoxO6 attenuated the expression of genes involved in lipogenesis, resulting in diminished hepatic lipid accumulation and mitigated hyperlipidemia in murine models. Additionally, the upregulation of FoxO6 due to elevated glucose levels led to increased ApoC3 expression, consequently instigating cellular triglyceride mediated lipid accumulation. The transcriptional activation of FoxO6 induced by both the HFD and high glucose levels resulted in hepatic steatosis by upregulating ApoC3 and genes associated with gluconeogenesis in aged rats and liver cell cultures. Our conclusions indicate that the upregulation of ApoC3 by FoxO6 promotes the development of hyperlipidemia, hyperglycemia, and hepatic steatosis in vivo, and in vitro. Taken together, our findings underscore the significance of FoxO6 in driving hyperlipidemia and hepatic steatosis specifically under hyperglycemic states by enhancing the expression of ApoC3 in aged rats.
Collapse
Affiliation(s)
- Dae Hyun Kim
- Department of Food Science and Technology, College of Natural Resources and Life Science, Pusan National University, Miryang-si, Gyeongsangnam-do 50463, Republic of Korea
| | - Seulah Lee
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Sang Gyun Noh
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Hae Young Chung
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| |
Collapse
|
4
|
Liu Y, Dai S, Qin S, Zhou J, Wang Z, Yin G. The pathogenic mutations of APOA5 in Chinese patients with hyperlipidemic acute pancreatitis. Lipids Health Dis 2024; 23:44. [PMID: 38331899 PMCID: PMC10851583 DOI: 10.1186/s12944-024-02011-5] [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: 10/31/2023] [Accepted: 01/09/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND AND AIMS To study the role of gene mutations in the development of severe hypertriglyceridemia (HTG) in patients with hyperlipidemic acute pancreatitis (HLAP), especially different apolipoprotein A5 (APOA5) mutations. METHODS Whole-exome sequencing was performed on 163 patients with HLAP and 30 patients with biliary acute pancreatitis (BAP). The pathogenicity of mutations was then assessed by combining clinical information, predictions of bioinformatics programs, information from multiple gene databases, and residue location and conservation. The pathogenic mutations of APOA5 were visualized using the software. RESULTS 1. Compared with BAP patients, pathogenic mutations of APOA5 were frequent in HLAP patients; among them, the heterozygous mutation of p.G185C was the most common. 2. All six pathogenic mutations of APOA5 identified in this study (p.S35N, p.D167V, p.G185C, p.K188I, p.R223C, and p.H182fs) were positively correlated with severe HTG; they were all in the important domains of apolipoprotein A-V (apoA-V). Residue 223 is strictly conserved in multiple mammals and is located in the lipoprotein lipase (LPL)-binding domain (Pro215-Phe261). When Arg 223 is mutated to Cys 223, the positive charge of this residue is reduced, which is potentially destructive to the binding function of apoA-V to LPL. 3. Four new APOA5 mutations were identified, namely c.563A > T, c.667C > T, c.788G > A, and c.544_545 insGGTGC. CONCLUSIONS The pathogenic mutations of APOA5 were specific to the patients with HLAP and severe HTG in China, and identifying such mutations had clinical significance in elucidating the etiology and subsequent treatment.
Collapse
Affiliation(s)
- Yuxin Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, District, No.1055, San-Xiang Road, Gu-Su, Suzhou, 215000, Jiangsu Province, China
| | - Si Dai
- Department of Gastroenterology, Songtao Miao Autonomous County People's Hospital, Tongren, 554199, Guizhou Province, China
| | - Shuqi Qin
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, District, No.1055, San-Xiang Road, Gu-Su, Suzhou, 215000, Jiangsu Province, China
| | - Jing Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, District, No.1055, San-Xiang Road, Gu-Su, Suzhou, 215000, Jiangsu Province, China
| | - Zhendan Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, District, No.1055, San-Xiang Road, Gu-Su, Suzhou, 215000, Jiangsu Province, China
| | - Guojian Yin
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, District, No.1055, San-Xiang Road, Gu-Su, Suzhou, 215000, Jiangsu Province, China.
| |
Collapse
|
5
|
Zhu D, Zhu Y, Liu L, He X, Fu S. Metabolomic analysis of vascular cognitive impairment due to hepatocellular carcinoma. Front Neurol 2023; 13:1109019. [PMID: 37008043 PMCID: PMC10062391 DOI: 10.3389/fneur.2022.1109019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/26/2022] [Indexed: 03/18/2023] Open
Abstract
IntroductionScreening for metabolically relevant differentially expressed genes (DEGs) shared by hepatocellular carcinoma (HCC) and vascular cognitive impairment (VCI) to explore the possible mechanisms of HCC-induced VCI.MethodsBased on metabolomic and gene expression data for HCC and VCI, 14 genes were identified as being associated with changes in HCC metabolites, and 71 genes were associated with changes in VCI metabolites. Multi-omics analysis was used to screen 360 DEGs associated with HCC metabolism and 63 DEGs associated with VCI metabolism.ResultsAccording to the Cancer Genome Atlas (TCGA) database, 882 HCC-associated DEGs were identified and 343 VCI-associated DEGs were identified. Eight genes were found at the intersection of these two gene sets: NNMT, PHGDH, NR1I2, CYP2J2, PON1, APOC2, CCL2, and SOCS3. The HCC metabolomics prognostic model was constructed and proved to have a good prognostic effect. The HCC metabolomics prognostic model was constructed and proved to have a good prognostic effect. Following principal component analyses (PCA), functional enrichment analyses, immune function analyses, and TMB analyses, these eight DEGs were identified as possibly affecting HCC-induced VCI and the immune microenvironment. As well as gene expression and gene set enrichment analyses (GSEA), a potential drug screen was conducted to investigate the possible mechanisms involved in HCC-induced VCI. The drug screening revealed the potential clinical efficacy of A-443654, A-770041, AP-24534, BI-2536, BMS- 509744, CGP-60474, and CGP-082996.ConclusionHCC-associated metabolic DEGs may influence the development of VCI in HCC patients.
Collapse
Affiliation(s)
- Dan Zhu
- Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yamei Zhu
- Deptartment of Infectious Diseases, Wuhua Ward, 920th Hospital of Joint Logistics Support Force of Chinese PLA, Kunming, Yunnan, China
| | - Lin Liu
- Dalian Hunter Information Consulting Co. LTD, Dalian, China
| | - Xiaoxue He
- Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shizhong Fu
- Deptartment of Infectious Diseases, Wuhua Ward, 920th Hospital of Joint Logistics Support Force of Chinese PLA, Kunming, Yunnan, China
- *Correspondence: Shizhong Fu ;
| |
Collapse
|
6
|
ENO1 Binds to ApoC3 and Impairs the Proliferation of T Cells via IL-8/STAT3 Pathway in OSCC. Int J Mol Sci 2022; 23:ijms232112777. [DOI: 10.3390/ijms232112777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Lymph node metastasis is associated with poor prognosis of oral squamous cell carcinoma (OSCC), and few studies have explored the relevance of postoperative lymphatic drainage (PLD) in metastatic OSCC. Alpha-enolase (ENO1) is a metabolic enzyme, which is related to lymphatic metastasis of OSCC. However, the role of ENO1 in PLD in metastatic OSCC has not been elucidated. Herein, we collected lymphatic drainage after lymphadenectomy between metastatic and non-metastatic lymph nodes in OSCC patients to investigate the relationship between ENO1 expression and metastasis, and to identify the proteins which interacted with ENO1 in PLD of patients with metastatic OSCC by MS/GST pulldown assay. Results revealed that the metabolic protein apolipoprotein C-III (ApoC3) was a novel partner of ENO1. The ENO1 bound to ApoC3 in OSCC cells and elicited the production of interleukin (IL)-8, as demonstrated through a cytokine antibody assay. We also studied the function of IL-8 on Jurkat T cells co-cultured with OSCC cells in vitro. Western blot analysis was applied to quantitate STAT3 (signal transducer and activator of transcription 3) and p-STAT3 levels. Mechanistically, OSCC cells activated the STAT3 signaling pathway on Jurkat T cells through IL-8 secretion, promoted apoptosis, and inhibited the proliferation of Jurkat T cells. Collectively, these findings illuminate the molecular mechanisms underlying the function of ENO1 in metastasis OSCC and provide new strategies for targeting ENO1 for OSCC treatment.
Collapse
|
7
|
The Role of RNA-Targeted Therapeutics to Reduce ASCVD Risk: What Have We Learned Recently? Curr Atheroscler Rep 2021; 23:40. [PMID: 34146170 DOI: 10.1007/s11883-021-00936-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW To discuss advances on the RNA-targeted therapies to treat dyslipidemia with the aim of reducing atherosclerotic cardiovascular disease (ASCVD). RECENT FINDINGS Genetic studies have paved the way for therapies that reduce translation of proteins that play causal roles in dyslipidemia and atherosclerosis like proprotein convertase subtilisin/kexin type 9 (PCSK9), apolipoprotein B-100 (apoB), apolipoprotein(a) [apo(a)], apolipoprotein C3 (apoC3), and angiopoietin-like 3 (ANGPTL3). Either antisense oligonucleotide (ASO) therapies and small interfering RNA (siRNA) molecules inhibit protein synthesis and consequently improve dyslipidemia. Most of these molecules contain N-acetylgalactosamine (GalNAc) moieties that have high specificity for hepatocytes and therefore reduce concentration in other tissues. Inclisiran, an siRNA for PCSK9, has shown robust LDL-C reductions, with good tolerability, in severe forms of hypercholesterolemia as well as in high cardiovascular disease patients with injections every 3 to 6 months. Pelacarsen is an ASO against apolipoprotein(a) that reduces Lp(a) up to 80% with good tolerability. Either inclisiran or pelacarsen is being tested to show it can prevent ASCVD. AMG 890, an siRNA compound aimed at reducing apo(a) synthesis, is also under investigation. Volanesorsen is an ASO against apoC3 that reduces triglyceride levels up to 70% and is being tested in severe hypertriglyceridemic patients. Vupanorsen is an ASO against ANGPTL3 that reduced triglyceride levels 36-53% among moderate hypertriglyceridemic individuals. Interestingly, it also reduces ApoC3 and non-HDL cholesterol and apoB; however, it lowers HDL cholesterol. RNA-targeted therapies are being extensively tested for dyslipidemia treatment with promising results.
Collapse
|
8
|
D'Erasmo L, Di Costanzo A, Gallo A, Bruckert E, Arca M. ApoCIII: A multifaceted protein in cardiometabolic disease. Metabolism 2020; 113:154395. [PMID: 33058850 DOI: 10.1016/j.metabol.2020.154395] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/20/2020] [Accepted: 09/26/2020] [Indexed: 01/15/2023]
Abstract
ApoCIII has a well-recognized role in triglyceride-rich lipoproteins metabolism. A considerable amount of data has clearly highlighted that high levels of ApoCIII lead to hypertriglyceridemia and, thereby, may influence the risk of cardiovascular disease. However, recent findings indicate that ApoCIII might also act beyond lipid metabolism. Indeed, ApoCIII has been implicated in other physiological processes such as glucose homeostasis, monocyte adhesion, activation of inflammatory pathways, and modulation of the coagulation cascade. As the inhibition of ApoCIII is emerging as a new promising therapeutic strategy, the complete understanding of multifaceted pathophysiological role of this apoprotein may be relevant. Therefore, the purpose of this work is to review available evidences not only related to genetics and biochemistry of ApoCIII, but also highlighting the role of this apoprotein in triglyceride and glucose metabolism, in the inflammatory process and coagulation cascade as well as in cardiovascular disease.
Collapse
Affiliation(s)
- Laura D'Erasmo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Italy; Department of Endocrinology and Cardiovascular Disease Prevention, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Sorbonne University Paris, France.
| | - Alessia Di Costanzo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Italy.
| | - Antonio Gallo
- Department of Endocrinology and Cardiovascular Disease Prevention, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Sorbonne University Paris, France
| | - Eric Bruckert
- Department of Endocrinology and Cardiovascular Disease Prevention, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Sorbonne University Paris, France
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| |
Collapse
|
9
|
Xu QY, Li H, Cao HX, Pan Q, Fan JG. APOC3 rs2070667 Associates with Serum Triglyceride Profile and Hepatic Inflammation in Nonalcoholic Fatty Liver Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8869674. [PMID: 33294458 PMCID: PMC7718051 DOI: 10.1155/2020/8869674] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
Single-nucleotide polymorphisms (SNPs) of apolipoprotein C3 (APOC3) play important role in lipid metabolism, and dyslipidemia underlies nonalcoholic fatty liver disease (NAFLD). But the correlation of serum lipidomics, APOC3 SNPs, and NAFLD remains limited understood. Enrolling thirty-four biopsy-proven NAFLD patients from Tianjin, Shanghai, Fujian, we investigated their APOC3 genotype and serum lipid profile by DNA sequencing and ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), respectively. Scoring of hepatocyte steatosis, ballooning, lobular inflammation, and liver fibrosis was then performed to reveal the role of lipidomics-affecting APOC3 SNPs in NAFLD-specific pathological alterations. Here, we reported that APOC3 SNPs (rs4225, rs4520, rs5128, rs2070666, and rs2070667) intimately correlated to serum lipidomics in NAFLD patients. A allele instead of G allele at rs2070667, which dominated the SNPs underlying lipidomic alteration, exhibited downregulatory effect on triacylglycerols (TGs: TG 54 : 7, TG 54 : 8, and TG 56 : 9) containing polyunsaturated fatty acid (PUFA). Moreover, subjects with low-level PUFA-containing TGs were predisposed to high-grade lobular inflammation (TG 54 : 7, rho = -0.454 and P = 0.007; TG 54 : 8, rho = -0.411 and P =0.016; TG 56 : 9, rho = -0.481 and P = 0.004). The significant correlation of APOC3 rs2070667 and inflammation grading [G/G vs. G/A+A/A: 0.00 (0.00 and 1.00) vs. 1.50 (0.75 and 2.00), P = 0.022] further confirmed its pathological action on the basis of lipidomics-impacting activity. These findings suggest an inhibitory effect of A allele at APOC3 rs2070667 on serum levels of PUFA-containing TGs, which are associated with high-grade lobular inflammation in NAFLD patients.
Collapse
Affiliation(s)
- Qing-Yang Xu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, China
| | - Han Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, China
| | - Hai-Xia Cao
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, China
| | - Qin Pan
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200092, China
- Department of Pediatric Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Shanghai Key Laboratory of Children's Digestion and Nutrition, Shanghai 200092, China
| |
Collapse
|
10
|
Wade H, Pan K, Su Q. CREBH: A Complex Array of Regulatory Mechanisms in Nutritional Signaling, Metabolic Inflammation, and Metabolic Disease. Mol Nutr Food Res 2020; 65:e2000771. [DOI: 10.1002/mnfr.202000771] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Henry Wade
- Institute for Global Food Security School of Biological Sciences Queen's University Belfast Belfast BT9 5DL UK
| | - Kaichao Pan
- Institute for Global Food Security School of Biological Sciences Queen's University Belfast Belfast BT9 5DL UK
| | - Qiaozhu Su
- Institute for Global Food Security School of Biological Sciences Queen's University Belfast Belfast BT9 5DL UK
| |
Collapse
|
11
|
Dron JS, Dilliott AA, Lawson A, McIntyre AD, Davis BD, Wang J, Cao H, Movsesyan I, Malloy MJ, Pullinger CR, Kane JP, Hegele RA. Loss-of-Function
CREB3L3
Variants in Patients With Severe Hypertriglyceridemia. Arterioscler Thromb Vasc Biol 2020; 40:1935-1941. [DOI: 10.1161/atvbaha.120.314168] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Objective:
Genetic determinants of severe hypertriglyceridemia include both common variants with small effects (assessed using polygenic risk scores) plus heterozygous and homozygous rare variants in canonical genes directly affecting triglyceride metabolism. Here, we broadened our scope to detect associations with rare loss-of-function variants in genes affecting noncanonical pathways, including those known to affect triglyceride metabolism indirectly.
Approach and Results:
From targeted next-generation sequencing of 69 metabolism-related genes in 265 patients of European descent with severe hypertriglyceridemia (≥10 mmol/L or ≥885 mg/dL) and 477 normolipidemic controls, we focused on the association of rare heterozygous loss-of-function variants in individual genes. We observed that compared with controls, severe hypertriglyceridemia patients were 20.2× (95% CI, 1.11–366.1;
P
=0.03) more likely than controls to carry a rare loss-of-function variant in
CREB3L3
, which encodes a transcription factor that regulates several target genes with roles in triglyceride metabolism.
Conclusions:
Our findings indicate that rare variants in a noncanonical gene for triglyceride metabolism, namely
CREB3L3
, contribute significantly to severe hypertriglyceridemia. Secondary genes and pathways should be considered when evaluating the genetic architecture of this complex trait.
Collapse
Affiliation(s)
- Jacqueline S. Dron
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
- Department of Biochemistry (J.S.D., A.A.D., A.L., R.A.H.), Western University, London, ON, Canada
| | - Allison A. Dilliott
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
- Department of Biochemistry (J.S.D., A.A.D., A.L., R.A.H.), Western University, London, ON, Canada
| | - Arden Lawson
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
- Department of Biochemistry (J.S.D., A.A.D., A.L., R.A.H.), Western University, London, ON, Canada
| | - Adam D. McIntyre
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
| | - Brent D. Davis
- Schulich School of Medicine and Dentistry, and Department of Computer Science (B.D.D.), Western University, London, ON, Canada
| | - Jian Wang
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
| | - Henian Cao
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
| | - Irina Movsesyan
- Cardiovascular Research Institute, University of California, San Francisco (I.M., M.J.M., C.R.P., J.P.K.)
| | - Mary J. Malloy
- Cardiovascular Research Institute, University of California, San Francisco (I.M., M.J.M., C.R.P., J.P.K.)
| | - Clive R. Pullinger
- Cardiovascular Research Institute, University of California, San Francisco (I.M., M.J.M., C.R.P., J.P.K.)
| | - John P. Kane
- Cardiovascular Research Institute, University of California, San Francisco (I.M., M.J.M., C.R.P., J.P.K.)
| | - Robert A. Hegele
- From the Robarts Research Institute (J.S.D., A.A.D., A.L., A.D.M., J.W., H.C., R.A.H.), Western University, London, ON, Canada
- Department of Biochemistry (J.S.D., A.A.D., A.L., R.A.H.), Western University, London, ON, Canada
- Department of Medicine (R.A.H.), Western University, London, ON, Canada
| |
Collapse
|
12
|
Corbin LJ, Hughes DA, Chetwynd AJ, Taylor AE, Southam AD, Jankevics A, Weber RJM, Groom A, Dunn WB, Timpson NJ. Metabolic characterisation of disturbances in the APOC3/triglyceride-rich lipoprotein pathway through sample-based recall by genotype. Metabolomics 2020; 16:69. [PMID: 32494907 PMCID: PMC7270992 DOI: 10.1007/s11306-020-01689-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 05/15/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION High plasma triacylglyceride levels are known to be associated with increased risk of atherosclerotic cardiovascular disease. Apolipoprotein C-III (apoC-III) is a key regulator of plasma triacylglyceride levels and is associated with hypertriglyceridemia via a number of pathways. There is consistent evidence for an association of cardiovascular events with blood apoC-III level, with support from human genetic studies of APOC3 variants. As such, apoC-III has been recognised as a potential therapeutic target for patients with severe hypertriglyceridaemia with one of the most promising apoC-III-targeting drugs, volanesorsen, having recently progressed through Phase III trials. OBJECTIVES To exploit a rare loss of function variant in APOC3 (rs138326449) to characterise the potential long-term treatment effects of apoC-III targeting interventions on the metabolome. METHODS In a recall-by-genotype study, 115 plasma samples were analysed by UHPLC-MS to acquire non-targeted metabolomics data. The study included samples from 57 adolescents and 33 adults. Overall, 12 985 metabolic features were tested for an association with APOC3 genotype. RESULTS 161 uniquely annotated metabolites were found to be associated with rs138326449(APOC3). The highest proportion of associated metabolites belonged to the acyl-acyl glycerophospholipid and triacylglyceride metabolite classes. In addition to the anticipated (on-target) reduction of metabolites in the triacylglyceride and related classes, carriers of the rare variant exhibited previously unreported increases in levels of a number of metabolites from the acyl-alkyl glycerophospholipid class. CONCLUSION Overall, our results suggest that therapies targeting apoC-III may potentially achieve a broad shift in lipid profile that favours better metabolic health.
Collapse
Affiliation(s)
- Laura J Corbin
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - David A Hughes
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Andrew J Chetwynd
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Amy E Taylor
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
- NIHR Biomedical Research Centre at the University Hospitals Bristol NHS Foundation Trust and the University of Bristol, Bristol, BS8 2BN, UK
| | - Andrew D Southam
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Andris Jankevics
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Ralf J M Weber
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Alix Groom
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Warwick B Dunn
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, BS8 2BN, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK.
| |
Collapse
|
13
|
The Role of Overexpressed Apolipoprotein AV in Insulin-Resistant Hepatocytes. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3268505. [PMID: 32382544 PMCID: PMC7193279 DOI: 10.1155/2020/3268505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/16/2019] [Accepted: 08/16/2019] [Indexed: 11/17/2022]
Abstract
In this paper, we sought to explore the relationship between apolipoprotein AV (APOAV) overexpression and insulin resistance in hepatocytes. The insulin-resistant HepG2 cell model was constructed, and then, APOAV-overexpressed HepG2 cells (B-M) were induced by infecting with a recombinant adenovirus vector. Microarray data were developed from B-M samples compared with negative controls (A-con), and the microarray data were analyzed by bioinformatic methods. APOAV-overexpression induced 313 upregulated genes and 563 downregulated ones in B-M sample. The differentially expressed genes (DEGs) were significantly classified in fat digestion and absorption pathway. Protein-protein interaction network was constructed, and AGTR1 (angiotensin II receptor type 1) and P2RY2 (purinergic receptor P2Y, G-protein coupled 2) were found to be the significant nodes closely related with G-protein related signaling. Additionally, overexpression of APOAV could change the expression of Glut4 and release the insulin resistance of hepatic cells. Thus, APOAV overexpression may prevent the insulin resistance in liver cells by mediating the genes such as AGTR1 and P2RY2.
Collapse
|
14
|
Su X, Peng D. The exchangeable apolipoproteins in lipid metabolism and obesity. Clin Chim Acta 2020; 503:128-135. [PMID: 31981585 DOI: 10.1016/j.cca.2020.01.015] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/29/2022]
Abstract
Dyslipidemia, characterized by increased plasma levels of low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), triglyceride (TG), and reduced plasma levels of high-density lipoprotein cholesterol (HDL-C), is confirmed as a hallmark of obesity and cardiovascular diseases (CVD), posing serious risks to the future health of humans. Thus, it is important to understand the molecular metabolism of dyslipidemia, which could help reduce the morbidity and mortality of obesity and CVD. Currently, several exchangeable apolipoproteins, such as apolipoprotein A1 (ApoA1), apolipoprotein A5 (ApoA5), apolipoprotein E (ApoE), and apolipoprotein C3 (ApoC3), have been verified to exert vital effects on modulating lipid metabolism and homeostasis both in plasma and in cells, which consequently affect dyslipidemia. In the present review, we summarize the findings of the effect of exchangeable apolipoproteins on affecting lipid metabolism in adipocytes and hepatocytes. Furthermore, we also provide new insights into the mechanisms by which the exchangeable apolipoproteins influence the pathogenesis of dyslipidemia and its related cardio-metabolic disorders.
Collapse
Affiliation(s)
- Xin Su
- Department of Cardiovascular Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
15
|
Borén J, Packard CJ, Taskinen MR. The Roles of ApoC-III on the Metabolism of Triglyceride-Rich Lipoproteins in Humans. Front Endocrinol (Lausanne) 2020; 11:474. [PMID: 32849270 PMCID: PMC7399058 DOI: 10.3389/fendo.2020.00474] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. It is well-established based on evidence accrued during the last three decades that high plasma concentrations of cholesterol-rich atherogenic lipoproteins are causatively linked to CVD, and that lowering these reduces atherosclerotic cardiovascular events in humans (1-9). Historically, most attention has been on low-density lipoproteins (LDL) since these are the most abundant atherogenic lipoproteins in the circulation, and thus the main carrier of cholesterol into the artery wall. However, with the rise of obesity and insulin resistance in many populations, there is increasing interest in the role of triglyceride-rich lipoproteins (TRLs) and their metabolic remnants, with accumulating evidence showing they too are causatively linked to CVD. Plasma triglyceride, measured either in the fasting or non-fasting state, is a useful index of the abundance of TRLs and recent research into the biology and genetics of triglyceride heritability has provided new insight into the causal relationship of TRLs with CVD. Of the genetic factors known to influence plasma triglyceride levels variation in APOC3- the gene for apolipoprotein (apo) C-III - has emerged as being particularly important as a regulator of triglyceride transport and a novel therapeutic target to reduce dyslipidaemia and CVD risk (10).
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- *Correspondence: Jan Borén
| | - Chris J. Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Marja-Riitta Taskinen
- Research Programs Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Emerging evidences for the opposite role of apolipoprotein C3 and apolipoprotein A5 in lipid metabolism and coronary artery disease. Lipids Health Dis 2019; 18:220. [PMID: 31836003 PMCID: PMC6909560 DOI: 10.1186/s12944-019-1166-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 12/06/2019] [Indexed: 12/16/2022] Open
Abstract
Apolipoprotein C3 (apoC3) and apolipoprotein A5 (apoA5), encoded by APOA1/C3/A4/A5 gene cluster, are two critical regulators of plasma triglyceride (TG) metabolism. Deficiency of apoC3 or apoA5 led to significant decreased or increased plasma TG levels, respectively. Recent studies indicated apoC3 and apoA5 also played roles in plasma remnant cholesterol, high density lipoprotein (HDL) and hepatic TG metabolisms. Moreover, large scale population genetic studies indicated that loss of function mutations in APOC3 and APOA5 gene conferred decreased and increased risk of coronary artery disease (CAD), respectively. This manuscript mainly reviewed existing evidences suggesting the opposite role of apoC3 and apoA5 in lipid metabolism and CAD risk, and discussed the potential correlation between these two apolipoproteins.
Collapse
|
17
|
Treviño-Villarreal JH, Reynolds JS, Bartelt A, Langston PK, MacArthur MR, Arduini A, Tosti V, Veronese N, Bertozzi B, Brace LE, Mejia P, Trocha K, Kajitani GS, Longchamp A, Harputlugil E, Gathungu R, Bird SS, Bullock AD, Figenshau RS, Andriole GL, Thompson A, Heeren J, Ozaki CK, Kristal BS, Fontana L, Mitchell JR. Dietary protein restriction reduces circulating VLDL triglyceride levels via CREBH-APOA5-dependent and -independent mechanisms. JCI Insight 2018; 3:99470. [PMID: 30385734 DOI: 10.1172/jci.insight.99470] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022] Open
Abstract
Hypertriglyceridemia is an independent risk factor for cardiovascular disease. Dietary interventions based on protein restriction (PR) reduce circulating triglycerides (TGs), but underlying mechanisms and clinical relevance remain unclear. Here, we show that 1 week of a protein-free diet without enforced calorie restriction significantly lowered circulating TGs in both lean and diet-induced obese mice. Mechanistically, the TG-lowering effect of PR was due, in part, to changes in very low-density lipoprotein (VLDL) metabolism both in liver and peripheral tissues. In the periphery, PR stimulated VLDL-TG consumption by increasing VLDL-bound APOA5 expression and promoting VLDL-TG hydrolysis and clearance from circulation. The PR-mediated increase in Apoa5 expression was controlled by the transcription factor CREBH, which coordinately regulated hepatic expression of fatty acid oxidation-related genes, including Fgf21 and Ppara. The CREBH-APOA5 axis activation upon PR was intact in mice lacking the GCN2-dependent amino acid-sensing arm of the integrated stress response. However, constitutive hepatic activation of the amino acid-responsive kinase mTORC1 compromised CREBH activation, leading to blunted APOA5 expression and PR-recalcitrant hypertriglyceridemia. PR also contributed to hypotriglyceridemia by reducing the rate of VLDL-TG secretion, independently of activation of the CREBH-APOA5 axis. Finally, a randomized controlled clinical trial revealed that 4-6 weeks of reduced protein intake (7%-9% of calories) decreased VLDL particle number, increased VLDL-bound APOA5 expression, and lowered plasma TGs, consistent with mechanistic conservation of PR-mediated hypotriglyceridemia in humans with translational potential as a nutraceutical intervention for dyslipidemia.
Collapse
Affiliation(s)
| | - Justin S Reynolds
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Alexander Bartelt
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Sabri Ülker Center for Nutrient, Genetic, and Metabolic Research, Boston, Massachusetts, USA.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - P Kent Langston
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Michael R MacArthur
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Alessandro Arduini
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Sabri Ülker Center for Nutrient, Genetic, and Metabolic Research, Boston, Massachusetts, USA
| | - Valeria Tosti
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Nicola Veronese
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Beatrice Bertozzi
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Lear E Brace
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Pedro Mejia
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kaspar Trocha
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Division of Vascular and Endovascular Surgery, Department of Surgery, and
| | - Gustavo S Kajitani
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Alban Longchamp
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Division of Vascular and Endovascular Surgery, Department of Surgery, and
| | - Eylul Harputlugil
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Rose Gathungu
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Division of Sleep Medicine, Department of Medicine, Harvard Medical School (HMS), Boston, Massachusetts, USA
| | - Susan S Bird
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Surgery, HMS, Boston, Massachusetts, USA
| | - Arnold D Bullock
- Division of Urology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert S Figenshau
- Division of Urology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gerald L Andriole
- Division of Urology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrew Thompson
- Dana Farber Cancer Institute/HMS Rodent Histopathology Core Facility, HMS, Boston, Massachusetts, USA
| | - Jöerg Heeren
- Department for Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - C Keith Ozaki
- Division of Vascular and Endovascular Surgery, Department of Surgery, and
| | - Bruce S Kristal
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Division of Sleep Medicine, Department of Medicine, Harvard Medical School (HMS), Boston, Massachusetts, USA.,Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Surgery, HMS, Boston, Massachusetts, USA
| | - Luigi Fontana
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,School of Medicine and Charles Perkins Centre, University of Sydney, Sydney, Australia
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Umano GR, Martino M, Santoro N. The Association between Pediatric NAFLD and Common Genetic Variants. CHILDREN-BASEL 2017. [PMID: 28629152 PMCID: PMC5483624 DOI: 10.3390/children4060049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common complications of obesity. Several studies have shown that genetic predisposition probably plays an important role in its pathogenesis. In fact, in the last few years a large number of genetic studies have provided compelling evidence that some gene variants, especially those in genes encoding proteins regulating lipid metabolism, are associated with intra-hepatic fat accumulation. Here we provide a comprehensive review of the gene variants that have affected the natural history of the disease.
Collapse
Affiliation(s)
- Giuseppina Rosaria Umano
- Department of Pediatrics, Yale University, 06520, New Haven, CT, USA.
- Dipartimento della Donna, del Bambino, di Vhirurgia Generale e Specialistica, Universita' della Campania Luigi Vanvitelli, 80138, Napoli, Italy.
| | - Mariangela Martino
- Department of Pediatrics, Yale University, 06520, New Haven, CT, USA.
- Dipartimento di Medicina V. Tiberio, Universita' del Molise, 86100, Campobasso, Italy.
| | - Nicola Santoro
- Department of Pediatrics, Yale University, 06520, New Haven, CT, USA.
- Dipartimento di Medicina V. Tiberio, Universita' del Molise, 86100, Campobasso, Italy.
| |
Collapse
|
19
|
Chan AWH, Wong GLH, Chan HY, Tong JHM, Yu YH, Choi PCL, Chan HLY, To KF, Wong VWS. Concurrent fatty liver increases risk of hepatocellular carcinoma among patients with chronic hepatitis B. J Gastroenterol Hepatol 2017; 32:667-676. [PMID: 27547913 DOI: 10.1111/jgh.13536] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/14/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Concurrent fatty liver in hepatitis B virus (HBV)-infected patients without significant alcohol intake is a frequent and increasingly alarming problem because of the non-alcoholic fatty liver disease pandemic. The risk of HBV-related hepatocellular carcinoma (HCC) development was increased by concomitant obesity and diabetes. Direct evidence of the hepatocarcinogenic effect of fatty liver in chronic HBV remains elusive. We aimed to evaluate the risk of concurrent histologically proven fatty liver in HBV hepatocarcinogenesis. METHODS We conducted a retrospective cohort study on a liver biopsy cohort of HBV-infected patients without significant alcohol intake to evaluate the prevalence of concurrent histologically proven fatty liver and its association with subsequent HCC development. We also examined nine polymorphisms on six non-alcoholic fatty liver disease-related candidate genes (ADIPOQ, APOC3, GCKR, LEPR, PNPLA3, and PPARG). RESULTS Among 270 HBV-infected patients, concurrent fatty liver was found in 107 patients (39.6%) and was associated with metabolic risks, cirrhosis (P = 0.016) and PNPLA3 rs738409 CG/GG genotype (P = 0.002). At a median follow-up of 79.9 months, 11 patients (4.1%) developed HCC, and nine of them had concurrent fatty liver. By multivariable Cox analysis, concurrent fatty liver (HR 7.27, 95% confidence interval: 1.52-34.76; P = 0.013), age, cirrhosis, and APOC3 rs2854116 TC/CC genotype (HR 3.93, 95% confidence interval: 1.30-11.84; P = 0.013) were independent factors predicting HCC development. CONCLUSIONS Concurrent fatty liver is common in HBV-infected patients and an independent risk factor potentiating HBV-associated HCC development by 7.3-fold. The risk of HBV-related HCC is increased by APOC3 gene polymorphism, and further characterization is required by its role.
Collapse
Affiliation(s)
- Anthony W H Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong.,Li Ka Shing Institute of Health Science, Sir Y. K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong
| | - Grace L H Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong.,State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong
| | - Hoi-Yun Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong.,State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong
| | - Joanna H M Tong
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong.,Li Ka Shing Institute of Health Science, Sir Y. K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong
| | - Yau-Hei Yu
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong.,Li Ka Shing Institute of Health Science, Sir Y. K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong
| | - Paul C L Choi
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong
| | - Henry L Y Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong.,State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong.,Li Ka Shing Institute of Health Science, Sir Y. K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong.,State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong
| | - Vincent W S Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong.,State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
20
|
Lack of evidence for a liver or intestinal miRNA regulation involved in the hypertriglyceridemic effect of APOC3 3'UTR variant SstI. Atherosclerosis 2016; 255:6-10. [PMID: 27794214 DOI: 10.1016/j.atherosclerosis.2016.10.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/19/2016] [Accepted: 10/12/2016] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS APOC3 is a major regulator of triglycerides metabolism. Several APOC3 variants are associated with hypertriglyceridemia (HTG). Our aim was to establish the potential regulation of APOC3 3'UTR variants associated with HTG by liver or intestinal miRNAs. METHODS We sequenced APOC3 3'UTR in 100 type 2 diabetic (TD2) patients with severe HTG (TG > 15 mmol/L) (HTG group) compared to 100 normotriglyceridemic patients (NTG group). We performed in silico studies to identify potential loss of miRNA binding induced by APOC3 3'UTR variants. We also performed in vitro studies to test the functionality of miRNA/APOC3 variants interactions: APOC3 3'UTR plasmids coupled with a firefly luciferase reporter were transfected in HepG2, HuH-7 and Caco-2 cells. RESULTS We identified only two variants: SstI (rs5128) and BbvI (rs5225) in APOC3 3'UTR in the 2 groups of patients. Only the SstI-S2 rare allele was significantly associated with HTG (allele frequency 19,5% in HTG group vs. 9,5% in NTG group, p = 0.0045). In silico studies predicted a potential loss in the binding of 5 miRNAs induced by the S2 variant. These 5 miRNAs are all endogenously expressed in human liver and intestine, as well as in the cell models studied. However, in vitro, the S2 variant did not modulate APOC3 3'UTR reporter gene expression in HepG2, HuH-7 and Caco-2 cells. CONCLUSIONS Our results do not confirm the hypothesis of a direct regulation of the APOC3 SstI variant by hepatic or intestinal miRNAs.
Collapse
|
21
|
Druley TE, Wang L, Lin SJ, Lee JH, Zhang Q, Daw EW, Abel HJ, Chasnoff SE, Ramos EI, Levinson BT, Thyagarajan B, Newman AB, Christensen K, Mayeux R, Province MA. Candidate gene resequencing to identify rare, pedigree-specific variants influencing healthy aging phenotypes in the long life family study. BMC Geriatr 2016; 16:80. [PMID: 27060904 PMCID: PMC4826550 DOI: 10.1186/s12877-016-0253-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 04/04/2016] [Indexed: 11/22/2022] Open
Abstract
Background The Long Life Family Study (LLFS) is an international study to identify the genetic components of various healthy aging phenotypes. We hypothesized that pedigree-specific rare variants at longevity-associated genes could have a similar functional impact on healthy phenotypes. Methods We performed custom hybridization capture sequencing to identify the functional variants in 464 candidate genes for longevity or the major diseases of aging in 615 pedigrees (4,953 individuals) from the LLFS, using a multiplexed, custom hybridization capture. Variants were analyzed individually or as a group across an entire gene for association to aging phenotypes using family based tests. Results We found significant associations to three genes and nine single variants. Most notably, we found a novel variant significantly associated with exceptional survival in the 3’ UTR OBFC1 in 13 individuals from six pedigrees. OBFC1 (chromosome 10) is involved in telomere maintenance, and falls within a linkage peak recently reported from an analysis of telomere length in LLFS families. Two different algorithms for single gene associations identified three genes with an enrichment of variation that was significantly associated with three phenotypes (GSK3B with the Healthy Aging Index, NOTCH1 with diastolic blood pressure and TP53 with serum HDL). Conclusions Sequencing analysis of family-based associations for age-related phenotypes can identify rare or novel variants. Electronic supplementary material The online version of this article (doi:10.1186/s12877-016-0253-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Todd E Druley
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA
| | - Lihua Wang
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Shiow J Lin
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Joseph H Lee
- Sergievsky Center, College of Physicians and Surgeons, Columbia University New York, New York, NY, USA.,Taub Institute, College of Physicians and Surgeons, Columbia University New York, New York, NY, USA.,Department of Epidemiology, School of Public Health, Columbia University New York, New York, NY, USA
| | - Qunyuan Zhang
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - E Warwick Daw
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Haley J Abel
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Sara E Chasnoff
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA
| | - Enrique I Ramos
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA
| | - Benjamin T Levinson
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Kaare Christensen
- The Danish Aging Research Center, Epidemiology, University of Southern Denmark, Odense, Denmark
| | - Richard Mayeux
- Gertrude H. Sergievsky Center and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York City, NY, USA
| | - Michael A Province
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA. .,Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
22
|
Serum Levels of ApoA1 and ApoA2 Are Associated with Cognitive Status in Older Men. BIOMED RESEARCH INTERNATIONAL 2015; 2015:481621. [PMID: 26682220 PMCID: PMC4670907 DOI: 10.1155/2015/481621] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/04/2015] [Accepted: 11/08/2015] [Indexed: 01/02/2023]
Abstract
Background. Advancing age, chronic inflammation, oxidative damage, and disorders of lipid metabolism are positively linked to the late-life cognitive impairment. Serum biomarkers may be associated with the cognitive status in older men. Methods. 440 old male subjects with different cognitive functions were recruited to investigate probable serum markers. Pearson Chi-Squared test, univariate analysis, and multivariate logistic regression analysis were performed to evaluate biomarkers which may be associated with cognitive status. Results. Levels of fundus atherosclerosis (AS) (P < 0.001), age (P < 0.001), serum biomarkers peroxidase (POD) (P = 0.026) and interleukin-6 (IL-6) (P = 0.001), serum levels of high-density lipoprotein cholesterol (HDL-C) (P < 0.001), apolipoprotein A2 (ApoA2) (P = 0.001), and ApoC2 (P = 0.005) showed significant differences. Compared to group 3, ApoA1 in group 1 (OR = 1.30, 95% CI 1.01–1.67) and group 2 (OR = 1.47, 95% CI 1.11–1.94) were higher, while ApoA2 were lower (group 1: OR = 0.43, 95% CI 0.18–1.02; group 2: OR = 0.21, 95% CI 0.08–0.54) after adjusting for control variables. Conclusion. The results demonstrated that age, AS levels, POD, IL-6, HDL-C, ApoA2, and ApoC2 were significantly related to cognitive status. Moreover, ApoA1 and ApoA2 were independently associated with cognitive impairment and late-life dementia.
Collapse
|
23
|
Patel SB. Mendel, Molecular Biology, and Apolipoprotein C-III: A Heady Combination. Metab Syndr Relat Disord 2015; 13:55-6. [DOI: 10.1089/met.2014.1506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Shailendra B. Patel
- Clement J. Zablocki VAMC and Division of Endocrinology, Diabetes, and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
24
|
Almeda-Valdes P, Cuevas-Ramos D, Mehta R, Muñoz-Hernandez L, Cruz-Bautista I, Perez-Mendez O, Tusie-Luna MT, Gomez-Perez FJ, Pajukanta P, Matikainen N, Taskinen MR, Aguilar-Salinas CA. Factors associated with postprandial lipemia and apolipoprotein A-V levels in individuals with familial combined hyperlipidemia. BMC Endocr Disord 2014; 14:90. [PMID: 25425215 PMCID: PMC4253986 DOI: 10.1186/1472-6823-14-90] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 11/17/2014] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Alterations in postprandial metabolism have been described in familial combined hyperlipidemia (FCH); however, their underlying mechanisms are not well characterized. We aimed to identify factors related to the magnitude of postprandial lipemia and apolipoprotein (apo) A-V levels in subjects with FCH. METHODS FCH cases (n = 99) were studied using a standardized meal test. Abdominal obesity was assessed using the waist to hip ratio (WHR). A linear regression model was performed to investigate the variables associated with the triglycerides incremental area under the curve (iAUC). Independent associations between metabolic variables and apo A-V iAUC were also investigated in a randomly selected subgroup (n = 44). The study sample was classified according to the presence of fasting hypertriglyceridemia (≥150 mg/dL) and abdominal obesity (WHR ≥0.92 in men and ≥0.85 in women) to explore differences in parameters. RESULTS The fasting apo B-48 levels (r = 0.404), and the WHR (r = 0.359) were independent factors contributing to the triglycerides iAUC (r2 = 0.29, P < 0.001). The triglycerides iAUC was independently associated with the apo A-V iAUC (r2 = 0.54, P < 0.01). Patients with both hypertriglyceridemia and abdominal obesity showed the most robust triglycerides and apo A-V postprandial responses. CONCLUSIONS In patients with FCH the fasting apo B-48 level is the main factor associated with postprandial lipemia. Abdominal obesity also contributes to the magnitude of the postprandial response.The triglycerides postprandial increment is the principal factor associated with the apo A-V postprandial response.
Collapse
Affiliation(s)
- Paloma Almeda-Valdes
- />Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Daniel Cuevas-Ramos
- />Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Roopa Mehta
- />Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Liliana Muñoz-Hernandez
- />Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Ivette Cruz-Bautista
- />Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Oscar Perez-Mendez
- />Department of Molecular Biology, Instituto Nacional de Cardiologia Ignacio A. Chavez, Mexico City, Mexico
| | - Maria Teresa Tusie-Luna
- />Molecular Biology and Genomic Medicine Units. Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran. Biomedical Investigation Institute, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Francisco J Gomez-Perez
- />Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Päivi Pajukanta
- />Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Niina Matikainen
- />Diabetes and Obesity Units, Heart and Lung Center, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Marja-Riitta Taskinen
- />Diabetes and Obesity Units, Heart and Lung Center, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Carlos A Aguilar-Salinas
- />Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| |
Collapse
|
25
|
Emilian O, Gheorghe-Emilian O. Apolipoprotein C-III's role in cardiovascular diseases, a short review. SCIENCEOPEN RESEARCH 2014. [DOI: 10.14293/s2199-1006.1.sor-med.afafp0.v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In this short review we show the important role played by apoC-III in the lipid dysregulation present in the majority of cardiovascular diseases. With emphasis on the mutations present in a minority of individuals that confer protection. With this in mind we state that apoC-III should be considered a valid target for pharmaceutical intervention and cardiovascular disease control and progression.
Collapse
|
26
|
Shou W, Wang Y, Xie F, Wang B, Yang L, Wu H, Wang Y, Wang Z, Shi J, Huang W. A functional polymorphism affecting the APOA5 gene expression is causally associated with plasma triglyceride levels conferring coronary atherosclerosis risk in Han Chinese Population. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2147-54. [DOI: 10.1016/j.bbadis.2014.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/29/2014] [Accepted: 08/13/2014] [Indexed: 01/21/2023]
|
27
|
Li S, Hu B, Wang Y, Wu D, Jin L, Wang X. Influences of APOA5 variants on plasma triglyceride levels in Uyghur population. PLoS One 2014; 9:e110258. [PMID: 25313938 PMCID: PMC4196964 DOI: 10.1371/journal.pone.0110258] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 09/11/2014] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Single nucleotide polymorphisms (SNPs) in apolipoprotein A5 (APOA5) gene are associated with triglyceride (TG) levels. However, the minor allele frequencies and linkage disequilibriums (LDs) of the SNPs in addition to their effects on TG levels vary greatly between Caucasians and East Asians. The distributions of the SNPs/haplotypes and their associations with TG levels in Uyghur population, an admixture population of Caucasians and East Asians, have not been reported to date. Here, we performed a cross-sectional study to address these. METHODS Genotyping of four SNPs in APOA5 (rs662799, rs3135506, rs2075291, and rs2266788) was performed in 1174 unrelated Uyghur subjects. SNP/haplotype and TG association analyses were conducted. RESULTS The frequencies of the SNPs in Uyghurs were in between those in Caucasians and East Asians. The LD between rs662799 and rs2266788 in Uyghurs was stronger than that in East Asians but weaker than that in Caucasians, and the four SNPs resulted in four haplotypes (TGGT, CGGC, TCGT, and CGTT arranged in the order of rs662799, rs3135506, rs2075291, and rs2266788) representing 99.2% of the population. All the four SNPs were significantly associated with TG levels. Compared with non-carriers, carriers of rs662799-C, rs3135506-C, rs2075291-T, and rs2266788-C alleles had 16.0%, 15.1%, 17.1%, and 12.4% higher TG levels, respectively. When haplotype TGGT was defined as the reference, the haplotypes CGGC, TCGT, and CGTT resulted in 16.1%, 19.0%, and 19.8% higher TG levels, respectively. The proportions of variance in TG explained by APOA5 locus were 2.5%, 0.3%, 0.4%, and 1.9% for single SNP rs662799, rs3135506, rs2075291, and rs2266788, respectively, and 3.0% for the haplotypes constructed by them. CONCLUSIONS The association profiles between the SNPs and haplotypes at APOA5 locus and TG levels in this admixture population differed from those in Caucasians and East Asians. The functions of these SNPs and haplotypes need to be elucidated comprehensively.
Collapse
Affiliation(s)
- Shuyuan Li
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China; Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Bin Hu
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China; Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Yi Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China; Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Di Wu
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Li Jin
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China; Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Xiaofeng Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China; Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| |
Collapse
|
28
|
Wu CL, Zhao SP, Yu BL. Intracellular role of exchangeable apolipoproteins in energy homeostasis, obesity and non-alcoholic fatty liver disease. Biol Rev Camb Philos Soc 2014; 90:367-76. [PMID: 24834836 DOI: 10.1111/brv.12116] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 04/10/2014] [Accepted: 04/17/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Chen-Lu Wu
- Department of Cardiology; The Second Xiangya Hospital, Central South University; Changsha Hunan 410011 China
| | - Shui-Ping Zhao
- Department of Cardiology; The Second Xiangya Hospital, Central South University; Changsha Hunan 410011 China
| | - Bi-Lian Yu
- Department of Cardiology; The Second Xiangya Hospital, Central South University; Changsha Hunan 410011 China
| |
Collapse
|
29
|
Abstract
Despite the critical importance of plasma lipoproteins in the development of atherosclerosis, varying degrees of evidence surround the causal associations of lipoproteins with coronary artery disease (CAD). These causal contributions can be assessed by employing genetic variants as unbiased proxies for lipid levels. A relatively large number of low-density lipoprotein cholesterol (LDL-C) variants strongly associate with CAD, confirming the causal impact of this lipoprotein on atherosclerosis. Although not as firmly established, genetic evidence supporting a causal role of triglycerides (TG) in CAD is growing. Conversely, high-density lipoprotein cholesterol (HDL-C) variants not associated with LDL-C or TG have not yet been shown to be convincingly associated with CAD, raising questions about the causality of HDL-C in atherosclerosis. Finally, genetic variants at the LPA locus associated with lipoprotein(a) [Lp(a)] are decisively linked to CAD, indicating a causal role for Lp(a). Translational investigation of CAD-associated lipid variants may identify novel regulatory pathways with therapeutic potential to alter CAD risk.
Collapse
|
30
|
Sehgal A, Vaishnaw A, Fitzgerald K. Liver as a target for oligonucleotide therapeutics. J Hepatol 2013; 59:1354-9. [PMID: 23770039 DOI: 10.1016/j.jhep.2013.05.045] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 05/17/2013] [Accepted: 05/29/2013] [Indexed: 12/18/2022]
Abstract
Oligonucleotide-based therapeutics are an emerging class of drugs that hold the promise for silencing "un-druggable" targets,thus creating unique opportunities for innovative medicines. As opposed to gene therapy, oligonucleotides are considered to be more akin to small molecule therapeutics because they are small,completely synthetic in origin, do not integrate into the host genome,and have a defined duration of therapeutic activity after which effects recover to baseline. They offer a high degree of specificity at the genetic level, thereby reducing off-target effects.At the same time, they provide a strategy for targeting any gene in the genome, including transcripts that produce mutated proteins.Oligonucleotide-based therapeutics include short interfering RNA (siRNA), that degrade target mRNA through RISC mediated RNAi; anti-miRs, that target miRNAs; miRNA mimics, that regulate target mRNA; antisense oligonucleotides, that may be working through RNAseH mediated mRNA decay; mRNA upregulation,by targeting long non-coding RNAs; and oligonucleotides induced alternative splicing [1]. All these approaches require some minimal degree of homology at the nucleic acid sequence level for them to be functional. The different mechanisms of action and their relevant activity are outlined in Fig. 1. Besides homology,RNA secondary structure has also been exploited in the case of ribozymes and aptamers, which act by binding to nucleic acids or proteins, respectively. While there have been many reports of gene knockdown and gene modulation in cell lines and mice with all these methods, very few have advanced to clinical stages.The main obstacle to date has been the safe and effective intracellular delivery of these compounds in higher species, including humans. Indeed, their action requires direct interaction with DNA/RNA within the target cell so even when one solves the issues of tissue and cellular access, intracellular/intranuclear location represents yet another barrier to overcome. To date,hepatic delivery of oligonucleotides has been the area with greatest progress, and thus we have focused on liver-targeted therapeutics that have shown promise at the preclinical and/or clinical level.The liver is the largest internal organ in the body, playing a central role in metabolism, detoxification, synthesis, and secretion of major plasma proteins (carrier proteins, coagulation factors,complement components, hormones, and apolipoproteins),and iron homeostasis. It is therefore not surprising that a large number of disease targets reside in the liver where they are susceptible to modulation by oligonucleotide therapies.
Collapse
Affiliation(s)
- Alfica Sehgal
- Alnylam Pharmaceuticals Inc., Cambridge, MA 02142, USA.
| | | | | |
Collapse
|
31
|
Abdulreda MH, Berggren PO. Islet inflammation in plain sight. Diabetes Obes Metab 2013; 15 Suppl 3:105-16. [PMID: 24003927 PMCID: PMC3777660 DOI: 10.1111/dom.12160] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 04/10/2013] [Indexed: 01/09/2023]
Abstract
Although, diabetes is reaching pandemic proportions, the exact aetiology of either type 1 (T1D) or type 2 diabetes (T2D) remains to be determined. Mounting evidence, however, suggests that islet inflammation is a likely common denominator during early development of either type of the disease. In this review, we highlight some of the inflammatory mechanisms that appear to be shared between T1D and T2D, and we explore the utility of intravital imaging in the study of islet inflammation. Intravital imaging has emerged as an indispensable tool in biomedical research and a variety of in vivo imaging approaches have been developed to study pancreatic islet physiology and pathophysiology in the native environment in health and disease. However, given the scattered distribution of the islets of Langerhans within the 'sea' of the exocrine pancreas located deep within the body and the fact that the islets only constitute 1-2% of the total volume of pancreatic tissue, studying the pancreatic islet in situ has been challenging. Here, we focus on a new experimental approach that enables studying local islet inflammation with single-cell resolution in the relevant context of the in vivo environment non-invasively and longitudinally and, thereby improving our understanding of diabetes pathogenesis.
Collapse
Affiliation(s)
- Midhat H. Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, Stockholm, Sweden
- Department of Surgery, University of Miami Miller School of Medicine, Stockholm, Sweden
| | - Per-Olof Berggren
- Diabetes Research Institute, University of Miami Miller School of Medicine, Stockholm, Sweden
- Department of Surgery, University of Miami Miller School of Medicine, Stockholm, Sweden
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
- Correspondence details: The Rolf Luft Research Center for Diabetes and Endocrinology Karolinska Institutet, Karolinska University Hospital L1 SE-171 76 Stockholm, Sweden Phone +46 8 517 757 31, +46 70 729 5731 Fax +46 8 517 717 81
| |
Collapse
|
32
|
Zampino R, Coppola N, Cirillo G, Boemio A, Pisaturo M, Marrone A, Macera M, Sagnelli E, Perrone L, Adinolfi LE, Miraglia del Giudice E. Abdominal fat interacts with PNPLA3 I148M, but not with the APOC3 variant in the pathogenesis of liver steatosis in chronic hepatitis C. J Viral Hepat 2013; 20:517-23. [PMID: 23808989 DOI: 10.1111/jvh.12053] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 12/02/2012] [Indexed: 12/12/2022]
Abstract
The patatin-like phospholipase domain-containing 3 gene (PNPLA3) and the apolipoprotein C3 gene (APOC3) have been studied in relation to liver steatosis and liver disease outcome. The aim of this study was to evaluate the influence of PNPLA3 p.I148M and APOC3 rs2854116 and rs2854117 polymorphisms on the clinical and histological presentation of chronic hepatitis C in an Italian population and their relationship with viral and anthropometric parameters. Patients with hepatitis C (n = 166) entered the study receiving a clinical, histological, virological and biochemical evaluation. APOC3 (rs2854116 and rs2854117) and PNPLA3 (p.I148M) variants were genotyped. PNPLA3 polymorphisms were associated with liver steatosis, which was significantly higher in patients with p.148I/M (P = 0.034) and p.148M/M (P = 0.004) variants than those homozygous for the PNPLA3 wild type. Excluding patients with HCV genotype 3, the association with liver steatosis and PNPLA3 variants was more marked (p.148I/I genotype vs p.148I/M, P = 0.02, and vs p.148M/M, P = 0.005). The APOC3 polymorphism was not associated with any of the evaluated parameters. Among the interacting factors, BMI and waist circumference correlated with liver steatosis (P = 0.008 and 0.004, respectively). Relationship between waist circumference and liver steatosis was analysed for the different PNPLA3 genotypes. Homozygous 148M patients showed a stronger correlation between waist circumference and steatosis than those carrying the other genotypes (P = 0.0047). In our hepatitis C-infected population, the PNPLA3 polymorphism influenced the development of liver steatosis, but not fibrosis progression. APOC3 polymorphisms had no effect on the development of steatosis and no influence on the PNPLA3 polymorphism. The amount of abdominal fat can increase the association of PNPLA3 p.I148M with liver steatosis.
Collapse
Affiliation(s)
- R Zampino
- Internal Medicine and Hepatology, Second University of Naples, Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Asselbergs F, Guo Y, van Iperen E, Sivapalaratnam S, Tragante V, Lanktree M, Lange L, Almoguera B, Appelman Y, Barnard J, Baumert J, Beitelshees A, Bhangale T, Chen YD, Gaunt T, Gong Y, Hopewell J, Johnson T, Kleber M, Langaee T, Li M, Li Y, Liu K, McDonough C, Meijs M, Middelberg R, Musunuru K, Nelson C, O’Connell J, Padmanabhan S, Pankow J, Pankratz N, Rafelt S, Rajagopalan R, Romaine S, Schork N, Shaffer J, Shen H, Smith E, Tischfield S, van der Most P, van Vliet-Ostaptchouk J, Verweij N, Volcik K, Zhang L, Bailey K, Bailey K, Bauer F, Boer J, Braund P, Burt A, Burton P, Buxbaum S, Chen W, Cooper-DeHoff R, Cupples L, deJong J, Delles C, Duggan D, Fornage M, Furlong C, Glazer N, Gums J, Hastie C, Holmes M, Illig T, Kirkland S, Kivimaki M, Klein R, Klein B, Kooperberg C, Kottke-Marchant K, Kumari M, LaCroix A, Mallela L, Murugesan G, Ordovas J, Ouwehand W, Post W, Saxena R, Scharnagl H, Schreiner P, Shah T, Shields D, Shimbo D, Srinivasan S, Stolk R, Swerdlow D, Taylor H, Topol E, Toskala E, van Pelt J, van Setten J, Yusuf S, Whittaker J, Zwinderman A, Anand S, Balmforth A, Berenson G, Bezzina C, et alAsselbergs F, Guo Y, van Iperen E, Sivapalaratnam S, Tragante V, Lanktree M, Lange L, Almoguera B, Appelman Y, Barnard J, Baumert J, Beitelshees A, Bhangale T, Chen YD, Gaunt T, Gong Y, Hopewell J, Johnson T, Kleber M, Langaee T, Li M, Li Y, Liu K, McDonough C, Meijs M, Middelberg R, Musunuru K, Nelson C, O’Connell J, Padmanabhan S, Pankow J, Pankratz N, Rafelt S, Rajagopalan R, Romaine S, Schork N, Shaffer J, Shen H, Smith E, Tischfield S, van der Most P, van Vliet-Ostaptchouk J, Verweij N, Volcik K, Zhang L, Bailey K, Bailey K, Bauer F, Boer J, Braund P, Burt A, Burton P, Buxbaum S, Chen W, Cooper-DeHoff R, Cupples L, deJong J, Delles C, Duggan D, Fornage M, Furlong C, Glazer N, Gums J, Hastie C, Holmes M, Illig T, Kirkland S, Kivimaki M, Klein R, Klein B, Kooperberg C, Kottke-Marchant K, Kumari M, LaCroix A, Mallela L, Murugesan G, Ordovas J, Ouwehand W, Post W, Saxena R, Scharnagl H, Schreiner P, Shah T, Shields D, Shimbo D, Srinivasan S, Stolk R, Swerdlow D, Taylor H, Topol E, Toskala E, van Pelt J, van Setten J, Yusuf S, Whittaker J, Zwinderman A, Anand S, Balmforth A, Berenson G, Bezzina C, Boehm B, Boerwinkle E, Casas J, Caulfield M, Clarke R, Connell J, Cruickshanks K, Davidson K, Day I, de Bakker P, Doevendans P, Dominiczak A, Hall A, Hartman C, Hengstenberg C, Hillege H, Hofker M, Humphries S, Jarvik G, Johnson J, Kaess B, Kathiresan S, Koenig W, Lawlor D, März W, Melander O, Mitchell B, Montgomery G, Munroe P, Murray S, Newhouse S, Onland-Moret N, Poulter N, Psaty B, Redline S, Rich S, Rotter J, Schunkert H, Sever P, Shuldiner A, Silverstein R, Stanton A, Thorand B, Trip M, Tsai M, van der Harst P, van der Schoot E, van der Schouw Y, Verschuren W, Watkins H, Wilde A, Wolffenbuttel B, Whitfield J, Hovingh G, Ballantyne C, Wijmenga C, Reilly M, Martin N, Wilson J, Rader D, Samani N, Reiner A, Hegele R, Kastelein J, Hingorani A, Talmud P, Hakonarson H, Elbers C, Keating B, Drenos F. Large-scale gene-centric meta-analysis across 32 studies identifies multiple lipid loci. Am J Hum Genet 2012; 91:823-38. [PMID: 23063622 PMCID: PMC3487124 DOI: 10.1016/j.ajhg.2012.08.032] [Show More Authors] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Revised: 08/02/2012] [Accepted: 08/30/2012] [Indexed: 12/19/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified many SNPs underlying variations in plasma-lipid levels. We explore whether additional loci associated with plasma-lipid phenotypes, such as high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC), and triglycerides (TGs), can be identified by a dense gene-centric approach. Our meta-analysis of 32 studies in 66,240 individuals of European ancestry was based on the custom ∼50,000 SNP genotyping array (the ITMAT-Broad-CARe array) covering ∼2,000 candidate genes. SNP-lipid associations were replicated either in a cohort comprising an additional 24,736 samples or within the Global Lipid Genetic Consortium. We identified four, six, ten, and four unreported SNPs in established lipid genes for HDL-C, LDL-C, TC, and TGs, respectively. We also identified several lipid-related SNPs in previously unreported genes: DGAT2, HCAR2, GPIHBP1, PPARG, and FTO for HDL-C; SOCS3, APOH, SPTY2D1, BRCA2, and VLDLR for LDL-C; SOCS3, UGT1A1, BRCA2, UBE3B, FCGR2A, CHUK, and INSIG2 for TC; and SERPINF2, C4B, GCK, GATA4, INSR, and LPAL2 for TGs. The proportion of explained phenotypic variance in the subset of studies providing individual-level data was 9.9% for HDL-C, 9.5% for LDL-C, 10.3% for TC, and 8.0% for TGs. This large meta-analysis of lipid phenotypes with the use of a dense gene-centric approach identified multiple SNPs not previously described in established lipid genes and several previously unknown loci. The explained phenotypic variance from this approach was comparable to that from a meta-analysis of GWAS data, suggesting that a focused genotyping approach can further increase the understanding of heritability of plasma lipids.
Collapse
Affiliation(s)
- Folkert W. Asselbergs
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Department of Medical Genetics, Biomedical Genetics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Yiran Guo
- Center for Applied Genomics, Abramson Research Center, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Erik P.A. van Iperen
- Durrer Center for Cardiogenetic Research, 1105 AZ Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Suthesh Sivapalaratnam
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Vinicius Tragante
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Department of Medical Genetics, Biomedical Genetics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Matthew B. Lanktree
- Departments of Medicine and Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 3K6, Canada
| | - Leslie A. Lange
- Department of Genetics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Berta Almoguera
- Center for Applied Genomics, Abramson Research Center, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yolande E. Appelman
- Department of Cardiology, VU University Medical Center, 1081 HV Amsterdam, the Netherlands
| | - John Barnard
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jens Baumert
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Amber L. Beitelshees
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Tushar R. Bhangale
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, CA 94080, USA
| | - Yii-Der Ida Chen
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tom R. Gaunt
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL 32603, USA
| | - Jemma C. Hopewell
- Clinical Trial Service Unit, University of Oxford, Oxford OX3 7BN, UK
| | - Toby Johnson
- Clinical Pharmacology and Barts and the London Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Marcus E. Kleber
- LURIC Study, Platz der alten Synagoge 1, 79098 Freiburg, Germany
- Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty of Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 7-11, 68167 Mannheim, Germany
| | - Taimour Y. Langaee
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL 32603, USA
| | - Mingyao Li
- Cardiovascular Institute, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yun R. Li
- Center for Applied Genomics, Abramson Research Center, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kiang Liu
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Caitrin W. McDonough
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL 32603, USA
| | - Matthijs F.L. Meijs
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Rita P.S. Middelberg
- Genetic Epidemiology, Queensland Institute of Medical Research, Locked Bag 2000, Royal Brisbane Hospital, Queensland 4029, Australia
- Department of Medicine, Prince Charles Hospital, Rode Road, Chermside, Queensland 4032, Australia
| | - Kiran Musunuru
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Christopher P. Nelson
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Jeffery R. O’Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sandosh Padmanabhan
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - James S. Pankow
- Division of Epidemiology & Community Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Suzanne Rafelt
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Ramakrishnan Rajagopalan
- Division of Medical Genetics, Department of Medicine and the Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Simon P.R. Romaine
- Leeds Institute of Genetics, Health & Therapeutics, University of Leeds, Leeds LS2 9JT, UK
| | - Nicholas J. Schork
- The Scripps Translational Science Institute and The Scripps Research Institute, 3344 N. Torrey Pines Court, Suite 300, La Jolla, CA 92037, USA
| | - Jonathan Shaffer
- Center for Behavioral Cardiovascular Health, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Haiqing Shen
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Erin N. Smith
- Department of Pediatrics and Rady’s Children’s Hospital, University of California at San Diego School of Medicine, La Jolla, CA 9293, USA
| | - Sam E. Tischfield
- Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Peter J. van der Most
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Jana V. van Vliet-Ostaptchouk
- Molecular Genetics Laboratory, Medical Biology Division, Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen University, 9700 RB Groningen, The Netherlands
- Department of Endocrinology, University Medical Center Groningen, Groningen University, 9700 RB Groningen, The Netherlands
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9700 RB, The Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Kelly A. Volcik
- Division of Epidemiology, Human Genetics and Environmental Sciences, Human Genetics Center, School of Public Health, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Li Zhang
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kent R. Bailey
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Kristian M. Bailey
- Leeds Institute of Genetics, Health & Therapeutics, University of Leeds, Leeds LS2 9JT, UK
| | - Florianne Bauer
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Complex Genetics Section, Department of Medical Genetics, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Jolanda M.A. Boer
- National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Peter S. Braund
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Amber Burt
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Paul R. Burton
- Department of Health Sciences, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Sarah G. Buxbaum
- Jackson Heart Study and School of Health Sciences, Department of Epidemiology and Biostatistics, Jackson State University, Jackson, MS 39217, USA
| | - Wei Chen
- Department of Epidemiology, Tulane University, 1440 Canal Street, Suite 1829, New Orleans, LA 70118, USA
| | - Rhonda M. Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL 32603, USA
| | - L. Adrienne Cupples
- Boston University, Boston, MA 02215, USA
- The National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA 01702, USA
| | - Jonas S. deJong
- Heart Failure Research Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - Christian Delles
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - David Duggan
- Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Myriam Fornage
- The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Clement E. Furlong
- Division of Medical Genetics, Department of Medicine and the Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | | | - John G. Gums
- Departments of Pharmacotherapy and Translational Research and Community Health and Family Medicine, University of Florida, Gainesville, FL 32607, USA
| | - Claire Hastie
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Michael V. Holmes
- Research Department of Epidemiology & Public Health, University College London Institute of Epidemiology & Health Care, University College London, 1-19 Torrington Place, London WC1E 6BT, UK
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, 30625 Hannover, Germany
| | - Susan A. Kirkland
- Department of Community Health and Epidemiology and Department of Medicine, Dalhousie University, Halifax NS B3H 1V7, Canada
| | - Mika Kivimaki
- Research Department of Epidemiology & Public Health, University College London Institute of Epidemiology & Health Care, University College London, 1-19 Torrington Place, London WC1E 6BT, UK
| | - Ronald Klein
- Department of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Barbara E. Klein
- Department of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Meena Kumari
- Research Department of Epidemiology & Public Health, University College London Institute of Epidemiology & Health Care, University College London, 1-19 Torrington Place, London WC1E 6BT, UK
| | - Andrea Z. LaCroix
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Laya Mallela
- Center for Applied Genomics, Abramson Research Center, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Gurunathan Murugesan
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jose Ordovas
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge & Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Richa Saxena
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria
| | - Pamela J. Schreiner
- School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tina Shah
- Research Department of Epidemiology & Public Health, University College London Institute of Epidemiology & Health Care, University College London, 1-19 Torrington Place, London WC1E 6BT, UK
| | - Denis C. Shields
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Daichi Shimbo
- Center for Behavioral Cardiovascular Health, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Sathanur R. Srinivasan
- Department of Epidemiology, Tulane University, 1440 Canal Street, Suite 2000, New Orleans, LA 70118 USA
| | - Ronald P. Stolk
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Daniel I. Swerdlow
- Genetic Epidemiology Group, Research Department of Epidemiology and Public Health, University College London Institute of Epidemiology and Health Care, University College London, London WC1E 6BT, UK
| | - Herman A. Taylor
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39217, USA
| | - Eric J. Topol
- The Scripps Translational Science Institute and The Scripps Research Institute, 3344 N. Torrey Pines Court, Suite 300, La Jolla, CA 92037, USA
| | - Elina Toskala
- Center for Applied Genomics, Abramson Research Center, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Joost L. van Pelt
- Laboratory Center, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Jessica van Setten
- Department of Medical Genetics, Biomedical Genetics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Salim Yusuf
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
| | - John C. Whittaker
- GlaxoSmithKline, Stevenage SG1 2NY, UK
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - A.H. Zwinderman
- Department of Clinical Epidemiology, Biostatistics, and Bioinformatics, Academic Medical Center, 1100DE Amsterdam, The Netherlands
| | - LifeLines Cohort Study
- LifeLines Cohort Study, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Sonia S. Anand
- Population Health Research Institute, Hamilton Health Sciences, McMaster University, Hamilton, ON L8L 2X2, Canada
| | - Anthony J. Balmforth
- Leeds Institute of Genetics, Health & Therapeutics, University of Leeds, Leeds LS2 9JT, UK
| | - Gerald S. Berenson
- Department of Epidemiology, Tulane University, 1440 Canal Street, Suite 1829, New Orleans, LA 70118, USA
| | - Connie R. Bezzina
- Heart Failure Research Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
- Molecular and Experimental Cardiology Group, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Bernhard O. Boehm
- Department of Internal Medicine, Department of Medicine, University Hospital, 89069 Ulm, Germany
| | - Eric Boerwinkle
- The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Juan P. Casas
- Research Department of Epidemiology & Public Health, University College London Institute of Epidemiology & Health Care, University College London, 1-19 Torrington Place, London WC1E 6BT, UK
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Mark J. Caulfield
- Clinical Pharmacology and Barts and the London Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Robert Clarke
- Clinical Trial Service Unit, University of Oxford, Oxford OX3 7BN, UK
| | - John M. Connell
- University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - Karen J. Cruickshanks
- Department of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Karina W. Davidson
- Departments of Medicine and Psychiatry, Columbia University, New York, NY 10027, USA
| | - Ian N.M. Day
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Paul I.W. de Bakker
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Medical Genetics, Biomedical Genetics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Pieter A. Doevendans
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Anna F. Dominiczak
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Alistair S. Hall
- Leeds Institute of Genetics, Health & Therapeutics, University of Leeds, Leeds LS2 9JT, UK
| | - Catharina A. Hartman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, 9700 AR Groningen, The Netherlands
| | | | - Hans L. Hillege
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9700 RB, The Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Marten H. Hofker
- Molecular Genetics, Medical Biology Section, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Steve E. Humphries
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, 5 University Street, London WC1E 6JF, UK
| | - Gail P. Jarvik
- Division of Medical Genetics, Department of Medicine and the Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL 32603, USA
| | - Bernhard M. Kaess
- Klinik und Poliklinik für Innere Medizin II, University of Regensburg, 93042 Germany
| | - Sekar Kathiresan
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Wolfgang Koenig
- Department of Internal Medicine II–Cardiology, University of Ulm Medical Centre, 89081 Ulm, Germany
| | - Debbie A. Lawlor
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Winfried März
- Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty of Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 7-11, 68167 Mannheim, Germany
- Synlab Academy, Gottlieb-Daimler-Straße 25, 68165 Mannheim, Germany
| | - Olle Melander
- Clinical Research Center, Malmö University Hospital, Malmö SE-205 02, Sweden
| | - Braxton D. Mitchell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Grant W. Montgomery
- Molecular Epidemiology, Queensland Institute of Medical Research, Locked Bag 2000, Royal Brisbane Hospital, Queensland 4029, Australia
| | - Patricia B. Munroe
- Clinical Pharmacology and Barts and the London Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Sarah S. Murray
- The Scripps Translational Science Institute and The Scripps Research Institute, 3344 N. Torrey Pines Court, Suite 300, La Jolla, CA 92037, USA
| | - Stephen J. Newhouse
- Clinical Pharmacology and Barts and the London Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - N. Charlotte Onland-Moret
- Complex Genetics Section, Department of Medical Genetics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Neil Poulter
- International Centre for Circulatory Health, Imperial College London, London W2 1PG, UK
| | - Bruce Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA 98195, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, WA 98109, USA
| | - Susan Redline
- University Hospitals, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | - Jerome I. Rotter
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - Peter Sever
- International Centre for Circulatory Health, Imperial College London, London W2 1PG, UK
| | - Alan R. Shuldiner
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Geriatric Research and Education Clinical Center, Veterans Administration Medical Center, Baltimore, MD 21201, USA
| | - Roy L. Silverstein
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Case Western Reserve University, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Alice Stanton
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Barbara Thorand
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Mieke D. Trip
- Department of Cardiology, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen 9700 RB, The Netherlands
| | - Ellen van der Schoot
- Department of Experimental Immunology, Sanquin, 1105 AZ Amsterdam, The Netherlands
| | - Yvonne T. van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - W.M. Monique Verschuren
- National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands
| | - Hugh Watkins
- Department of Cardiovascular Medicine, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Arthur A.M. Wilde
- Heart Failure Research Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - Bruce H.R. Wolffenbuttel
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - John B. Whitfield
- Genetic Epidemiology, Queensland Institute of Medical Research, Locked Bag 2000, Royal Brisbane Hospital, Queensland 4029, Australia
| | - G. Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | | | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, the Netherlands
| | - Muredach P. Reilly
- Cardiovascular Institute, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas G. Martin
- Genetic Epidemiology, Queensland Institute of Medical Research, Locked Bag 2000, Royal Brisbane Hospital, Queensland 4029, Australia
| | - James G. Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Daniel J. Rader
- Cardiovascular Institute, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester LE3 9QP, UK
- National Institute of Health Research Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Alex P. Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Robert A. Hegele
- Robarts Research Institute, University of Western Ontario, London, ON N6A5K8, Canada
| | - John J.P. Kastelein
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Aroon D. Hingorani
- Research Department of Epidemiology & Public Health, University College London Institute of Epidemiology & Health Care, University College London, 1-19 Torrington Place, London WC1E 6BT, UK
| | - Philippa J. Talmud
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, 5 University Street, London WC1E 6JF, UK
| | - Hakon Hakonarson
- Center for Applied Genomics, Abramson Research Center, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Clara C. Elbers
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Complex Genetics Section, Department of Medical Genetics, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Brendan J. Keating
- Center for Applied Genomics, Abramson Research Center, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Fotios Drenos
- Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, 5 University Street, London WC1E 6JF, UK
| |
Collapse
|
34
|
Guardiola M, Alvaro A, Vallvé JC, Rosales R, Solà R, Girona J, Serra N, Duran P, Esteve E, Masana L, Ribalta J. APOA5 gene expression in the human intestinal tissue and its response to in vitro exposure to fatty acid and fibrate. Nutr Metab Cardiovasc Dis 2012; 22:756-762. [PMID: 21489765 DOI: 10.1016/j.numecd.2010.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 10/05/2010] [Accepted: 12/11/2010] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND AIMS APOA5, a key gene regulating triglyceride (TG) levels, is reported to be expressed exclusively in the liver where it may regulate TG-rich particle synthesis and secretion. Since the same lipoprotein processing occurs in the intestine, we have postulated that this organ would also express APOA5. METHODS AND RESULTS We have detected the APOA5 gene expression in C57BL/6J mouse and in human small intestine samples. In humans, it is expressed mainly in the duodenum and colon, with messenger RNA (mRNA) levels four orders of magnitude lower than in the liver, and the protein product being one-sixth of the liver equivalent. Subsequently, we carried out in vitro experiments in TC-7/CaCo(2) human intestinal cells to analyse the expression of APOA5, APOC3, APOB and MTP genes after the incubation with long- and short-chain fatty acids, and a peroxisome proliferator-activated receptor alpha (PPARα) agonist (Wy 14643, a fibrate therapeutic agent). In the TC-7 cell line, APOA5 expression was significantly upregulated by saturated fatty acids. The short-chain fatty acid butyrate increased APOA5 expression almost fourfold while APOB was downregulated by increasing butyrate concentrations. When TC-7 cells were incubated with PPARα agonist, the APOA5 expression was increased by 60%, while the expression of APOB, MTP and APOC3 was decreased by 50%, 30% and 45%, respectively. CONCLUSION Our results demonstrate that APOA5 is expressed in the intestine, albeit at a much lower concentration than in the liver. While it remains to be determined whether intestinal apo A-V is functional, our in vitro experiments show that its expression is modifiable by dietary and pharmacological stimuli.
Collapse
Affiliation(s)
- M Guardiola
- Unitat de Recerca en Lípids i Arteriosclerosi, Institut d'Investigacions Sanitàries Pere, Virgili, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas, Universitat Rovira i Virgili, Reus, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Liu CF, Yang QF, Chen XL, Liu CY. Apolipoprotein a5 gene polymorphism and risk for metabolic syndrome: a meta-analysis. Genet Test Mol Biomarkers 2012; 16:1241-5. [PMID: 22905904 DOI: 10.1089/gtmb.2012.0183] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
BACKGROUND Many studies have focused on the association between the apolipoprotein A5 (ApoA5) polymorphism and the risk of metabolic syndrome (MetS). However, these studies drew inconsistent conclusions. The aim of this study was to evaluate the exact association between the ApoA5 polymorphism and MetS in a large-scale meta-analysis. METHODS The PubMed, Embase, and Science Citation Index (ISI Web of Science) databases were searched to collect all publications on the association between the ApoA5 polymorphism and MetS. Two common variants of ApoA5 (namely -1131T>C in the promoter region and c.56C>G in the coding region) with the risk of MetS were analyzed. The overall odd ratios (ORs) and 95% confidence intervals (CIs) for -1131T>C (CC+TC) versus TT genotype and c.C56G (GG+GC) versus CC were assessed between the MetS and control group. Subgroup analysis was further performed by ethnicity. The meta-analysis was performed by Stata11.0. RESULTS Twelve studies from 10 publications were chosen in our meta-analysis. The combined results showed that C allele carriers (CC+TC) of -1131T>C had a significantly higher risk of MetS for the overall (OR=1.32; 95% CI: 1.14-1.53; p=0.000) with moderate heterogeneity (I2=54.9%, p=0.014). Subgroup analysis was further performed according to ethnicity, and the association was still significant in Asians (OR=1.42; 95% CI: 1.25-1.62; p=0.000), but not in white populations (OR=1.25; 95% CI: 0.97-1.61; p=0.087). When analyzing the association between c.C56G and MetS, the G allele carrier (GG+GC) genotype significantly increased the risk of MetS (OR=1.32; 95% CI: 1.15-1.50; p=0.000) in white populations. No significant publication bias was observed in either -1131T>C or c.C56G. CONCLUSIONS Our study suggested that the ApoA5 -1131T>C polymorphism was significantly associated with the risk of MetS in Asians, but not in white populations. However, the c.C56G polymorphism was significantly associated with MetS in white populations.
Collapse
Affiliation(s)
- Cun-Fei Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | | | | | | |
Collapse
|
36
|
Lopez MF, Sarracino DA, Prakash A, Athanas M, Krastins B, Rezai T, Sutton JN, Peterman S, Gvozdyak O, Chou S, Lo E, Buonanno F, Ning M. Discrimination of ischemic and hemorrhagic strokes using a multiplexed, mass spectrometry-based assay for serum apolipoproteins coupled to multi-marker ROC algorithm. Proteomics Clin Appl 2012; 6:190-200. [PMID: 22532455 DOI: 10.1002/prca.201100041] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
PURPOSE Typically, apolipoproteins are individually measured in blood by immunoassay. In this report, we describe the development of a multiplexed selected reaction monitoring (SRM) based assay for a panel of apolipoproteins and its application to a clinical cohort of samples derived from acute stroke patients. EXPERIMENTAL DESIGN An SRM assay for a panel of nine apolipoproteins was developed on a triple quadrupole mass spectrometer. Quantitative data for each apolipoprotein were analyzed to determine expression ratio and receiver operating characteristic (ROC) values for ischemic versus hemorrhagic stroke. RESULTS The optimized SRM assay was used to interrogate a small cohort of well-characterized plasma samples obtained from patients with acute ischemic and hemorrhagic strokes. The ROC analyses demonstrated good classification power for several single apolipoproteins, most notably apoC-III and apoC-I. When a novel multi-marker ROC algorithm was applied, the ischemic versus hemorrhagic groups were best differentiated by a combination of apoC-III and apoA-I with an area under the curve (AUC) value of 0.92. CONCLUSIONS AND CLINICAL RELEVANCE This proof-of-concept study provides interesting and provocative data for distinguishing ischemic versus hemorrhage within first week of symptom onset. However, the observations are based on one cohort of patient samples and further confirmation will be required.
Collapse
Affiliation(s)
- Mary F Lopez
- ThermoFisher Scientific BRIMS, Cambridge, MA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Song F, Poljak A, Crawford J, Kochan NA, Wen W, Cameron B, Lux O, Brodaty H, Mather K, Smythe GA, Sachdev PS. Plasma apolipoprotein levels are associated with cognitive status and decline in a community cohort of older individuals. PLoS One 2012; 7:e34078. [PMID: 22701550 PMCID: PMC3372509 DOI: 10.1371/journal.pone.0034078] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 02/21/2012] [Indexed: 11/29/2022] Open
Abstract
Objectives Apolipoproteins have recently been implicated in the etiology of Alzheimer’s disease (AD). In particular, Apolipoprotein J (ApoJ or clusterin) has been proposed as a biomarker of the disease at the pre-dementia stage. We examined a group of apolipoproteins, including ApoA1, ApoA2, ApoB, ApoC3, ApoE, ApoH and ApoJ, in the plasma of a longitudinal community based cohort. Methods 664 subjects (257 with Mild Cognitive Impairment [MCI] and 407 with normal cognition), mean age 78 years, from the Sydney Memory and Aging Study (MAS) were followed up over two years. Plasma apolipoprotein levels at baseline (Wave 1) were measured using a multiplex bead fluorescence immunoassay technique. Results At Wave 1, MCI subjects had lower levels of ApoA1, ApoA2 and ApoH, and higher levels of ApoE and ApoJ, and a higher ApoB/ApoA1 ratio. Carriers of the apolipoprotein E ε4 allele had significantly lower levels of plasma ApoE, ApoC3 and ApoH and a significantly higher level of ApoB. Global cognitive scores were correlated positively with ApoH and negatively with ApoJ levels. ApoJ and ApoE levels were correlated negatively with grey matter volume and positively with cerebrospinal fluid (CSF) volume on MRI. Lower ApoA1, ApoA2 and ApoH levels, and higher ApoB/ApoA1 ratio, increased the risk of cognitive decline over two years in cognitively normal individuals. ApoA1 was the most significant predictor of decline. These associations remained after statistically controlling for lipid profile. Higher ApoJ levels predicted white matter atrophy over two years. Conclusions Elderly individuals with MCI have abnormal apolipoprotein levels, which are related to cognitive function and volumetric MRI measures cross-sectionally and are predictive of cognitive impairment in cognitively normal subjects. ApoA1, ApoH and ApoJ are potential plasma biomarkers of cognitive decline in non-demented elderly individuals.
Collapse
Affiliation(s)
- Fei Song
- Brain and Aging Research Program, University of New South Wales, Sydney, Australia
- School of Psychiatry, University of New South Wales, Sydney, Australia
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, Australia
| | - Anne Poljak
- Brain and Aging Research Program, University of New South Wales, Sydney, Australia
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - John Crawford
- Brain and Aging Research Program, University of New South Wales, Sydney, Australia
- School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Nicole A. Kochan
- Brain and Aging Research Program, University of New South Wales, Sydney, Australia
- School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Wei Wen
- Brain and Aging Research Program, University of New South Wales, Sydney, Australia
- School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Barbara Cameron
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Ora Lux
- Brain and Aging Research Program, University of New South Wales, Sydney, Australia
| | - Henry Brodaty
- Brain and Aging Research Program, University of New South Wales, Sydney, Australia
- School of Psychiatry, University of New South Wales, Sydney, Australia
- Dementia Collaborative Research Centre, University of New South Wales, Sydney, Australia
| | - Karen Mather
- Brain and Aging Research Program, University of New South Wales, Sydney, Australia
- School of Psychiatry, University of New South Wales, Sydney, Australia
| | - George A. Smythe
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Perminder S. Sachdev
- Brain and Aging Research Program, University of New South Wales, Sydney, Australia
- School of Psychiatry, University of New South Wales, Sydney, Australia
- * E-mail:
| |
Collapse
|
38
|
Holleboom AG, Karlsson H, Lin RS, Beres TM, Sierts JA, Herman DS, Stroes ES, Aerts JM, Kastelein JJ, Motazacker MM, Dallinga-Thie GM, Levels JH, Zwinderman AH, Seidman JG, Seidman CE, Ljunggren S, Lefeber DJ, Morava E, Wevers RA, Fritz TA, Tabak LA, Lindahl M, Hovingh GK, Kuivenhoven JA. Heterozygosity for a loss-of-function mutation in GALNT2 improves plasma triglyceride clearance in man. Cell Metab 2011; 14:811-8. [PMID: 22152306 PMCID: PMC3523677 DOI: 10.1016/j.cmet.2011.11.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 08/15/2011] [Accepted: 11/13/2011] [Indexed: 01/31/2023]
Abstract
Genome-wide association studies have identified GALNT2 as a candidate gene in lipid metabolism, but it is not known how the encoded enzyme ppGalNAc-T2, which contributes to the initiation of mucin-type O-linked glycosylation, mediates this effect. In two probands with elevated plasma high-density lipoprotein cholesterol and reduced triglycerides, we identified a mutation in GALNT2. It is shown that carriers have improved postprandial triglyceride clearance, which is likely attributable to attenuated glycosylation of apolipoprotein (apo) C-III, as observed in their plasma. This protein inhibits lipoprotein lipase (LPL), which hydrolyses plasma triglycerides. We show that an apoC-III-based peptide is a substrate for ppGalNAc-T2 while its glycosylation by the mutant enzyme is impaired. In addition, neuraminidase treatment of apoC-III which removes the sialic acids from its glycan chain decreases its potential to inhibit LPL. Combined, these data suggest that ppGalNAc-T2 can affect lipid metabolism through apoC-III glycosylation, thereby establishing GALNT2 as a lipid-modifying gene.
Collapse
Affiliation(s)
- Adriaan G. Holleboom
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | - Helen Karlsson
- Center of Occupational and Environmental Medicine, County Council of Östergötland, Linköping S-581 85, Sweden
- Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping S-581 85, Sweden
| | - Ruei-Shiuan Lin
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas M. Beres
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeroen A. Sierts
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | - Daniel S. Herman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | - Johannes M. Aerts
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | - John J.P. Kastelein
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | - Mohammad M. Motazacker
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | - Geesje M. Dallinga-Thie
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | - Johannes H.M. Levels
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | - Aeilko H. Zwinderman
- Department of Clinical Epidemiology, Biostatistics, and Bioinformatics, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | | | | | - Stefan Ljunggren
- Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping S-581 85, Sweden
| | - Dirk J. Lefeber
- Department of Neurology, Radboud University Nijmegen Medical Center, Nijmegen 6525GA, The Netherlands
- Department of Laboratory Medicine, Radboud University Nijmegen Medical Center, Nijmegen 6525GA, The Netherlands
- Institute for Genetic and Metabolic Disease, Radboud University Nijmegen Medical Center, Nijmegen 6525GA, The Netherlands
| | - Eva Morava
- Institute for Genetic and Metabolic Disease, Radboud University Nijmegen Medical Center, Nijmegen 6525GA, The Netherlands
- Department of Pediatrics, Radboud University Nijmegen Medical Center, Nijmegen 6525GA, The Netherlands
| | - Ron A. Wevers
- Department of Laboratory Medicine, Radboud University Nijmegen Medical Center, Nijmegen 6525GA, The Netherlands
- Institute for Genetic and Metabolic Disease, Radboud University Nijmegen Medical Center, Nijmegen 6525GA, The Netherlands
| | | | - Lawrence A. Tabak
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mats Lindahl
- Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping S-581 85, Sweden
| | - G. Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
| | - Jan Albert Kuivenhoven
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam 1105AZ, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen 9713AV, The Netherlands
| |
Collapse
|
39
|
Apolipoprotein A-V; a potent triglyceride reducer. Atherosclerosis 2011; 219:15-21. [DOI: 10.1016/j.atherosclerosis.2011.07.019] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 07/03/2011] [Accepted: 07/04/2011] [Indexed: 11/21/2022]
|
40
|
Johansen CT, Hegele RA. Allelic and phenotypic spectrum of plasma triglycerides. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:833-42. [PMID: 22033228 DOI: 10.1016/j.bbalip.2011.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 10/04/2011] [Indexed: 01/10/2023]
Abstract
The genetic underpinnings of both normal and pathological variation in plasma triglyceride (TG) concentration are relatively well understood compared to many other complex metabolic traits. For instance, genome-wide association studies (GWAS) have revealed 32 common variants that are associated with plasma TG concentrations in healthy epidemiologic populations. Furthermore, GWAS in clinically ascertained hypertriglyceridemia (HTG) patients have shown that almost all of the same TG-raising alleles from epidemiologic samples are also associated with HTG disease status, and that greater accumulation of these alleles reflects the severity of the HTG phenotype. Finally, comprehensive resequencing studies show a burden of rare variants in some of these same genes - namely in LPL, GCKR, APOB and APOA5 - in HTG patients compared to normolipidemic controls. A more complete understanding of the genes and genetic variants associated with plasma TG concentration will enrich our understanding of the molecular pathways that modulate plasma TG metabolism, which may translate into clinical benefit. This article is part of a Special Issue entitled Triglyceride Metabolism and Disease.
Collapse
|
41
|
Bandgar BP, Sarangdhar RJ, Khan F, Mookkan J, Shetty P, Singh G. Synthesis and biological evaluation of orally active hypolipidemic agents. J Med Chem 2011; 54:5915-26. [PMID: 21770455 DOI: 10.1021/jm200704f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A series of novel fenofibric acid ester prodrugs 1c-1h were synthesized and evaluated with the aim of obtaining potent hypolipidemic agents. Prodrugs 1c and 1d exhibited potent hypochlolesterolemic activity, lowering the mice plasma triglyceride level up to 47% in Swiss albino mice after oral administration of 50 mg/kg/day for 8 days. Fenofibric acid ester prodrugs 1c-1h were found lipophilic like fenofibrate (1b), indicated by partition coefficients measured in octanol-buffer system at pH 7.4. On the basis of in vivo studies, prodrugs 1c and 1d emerged as potent hypolipidemic agents.
Collapse
Affiliation(s)
- Babasaheb P. Bandgar
- Medicinal Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur-413 255, India
| | - Rajendra J. Sarangdhar
- Medicinal Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur-413 255, India
| | - Fruthous Khan
- Medicinal Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur-413 255, India
| | - Jeyamurugan Mookkan
- Medicinal Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur-413 255, India
| | - Pranesha Shetty
- Medicinal Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur-413 255, India
| | - Gajendra Singh
- Medicinal Chemistry Research Laboratory, School of Chemical Sciences, Solapur University, Solapur-413 255, India
| |
Collapse
|
42
|
Zhang Z, Peng B, Gong R, Gao L, Du J, Fang D, Song Y, Li Y, Ou G. Apolipoprotein A5 polymorphisms and risk of coronary artery disease: A meta-analysis. Biosci Trends 2011; 5:165-72. [PMID: 21914952 DOI: 10.5582/bst.2011.v5.4.165] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Zhen Zhang
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, and State Key Laboratory of Oral Diseases, Sichuan University
| | - Bo Peng
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University
| | - Renrong Gong
- Department of Thoracic and Cardiovascular Surgery, West China Hospital, Sichuan University
| | - Linbo Gao
- Laboratory of Molecular and Translational Medicine, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University
| | - Juan Du
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, and State Key Laboratory of Oral Diseases, Sichuan University
| | - Dingzhi Fang
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, and State Key Laboratory of Oral Diseases, Sichuan University
| | - Yongyan Song
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, and State Key Laboratory of Oral Diseases, Sichuan University
| | - Yuanhao Li
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, and State Key Laboratory of Oral Diseases, Sichuan University
| | - Guojing Ou
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, and State Key Laboratory of Oral Diseases, Sichuan University
| |
Collapse
|
43
|
Presence of Apolipoprotein C-III Attenuates Apolipoprotein E-Mediated Cellular Uptake of Cholesterol-Containing Lipid Particles by HepG2 Cells. Lipids 2010; 46:323-32. [DOI: 10.1007/s11745-010-3498-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 10/26/2010] [Indexed: 11/28/2022]
|
44
|
Johansen CT, Kathiresan S, Hegele RA. Genetic determinants of plasma triglycerides. J Lipid Res 2010; 52:189-206. [PMID: 21041806 DOI: 10.1194/jlr.r009720] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Plasma triglyceride (TG) concentration is reemerging as an important cardiovascular disease risk factor. More complete understanding of the genes and variants that modulate plasma TG should enable development of markers for risk prediction, diagnosis, prognosis, and response to therapies and might help specify new directions for therapeutic interventions. Recent genome-wide association studies (GWAS) have identified both known and novel loci associated with plasma TG concentration. However, genetic variation at these loci explains only ∼10% of overall TG variation within the population. As the GWAS approach may be reaching its limit for discovering genetic determinants of TG, alternative genetic strategies, such as rare variant sequencing studies and evaluation of animal models, may provide complementary information to flesh out knowledge of clinically and biologically important pathways in TG metabolism. Herein, we review genes recently implicated in TG metabolism and describe how some of these genes likely modulate plasma TG concentration. We also discuss lessons regarding plasma TG metabolism learned from various genomic and genetic experimental approaches. Treatment of patients with moderate to severe hypertriglyceridemia with existing therapies is often challenging; thus, gene products and pathways found in recent genetic research studies provide hope for development of more effective clinical strategies.
Collapse
Affiliation(s)
- Christopher T Johansen
- Department of Biochemistry, Robarts Research Institute, University of Western Ontario, London, Ontario N6A 5K8, Canada
| | | | | |
Collapse
|
45
|
Clemente-Postigo M, Queipo-Ortuño M, Valdivielso P, Tinahones F, Cardona F. Effect of apolipoprotein C3 and apolipoprotein A1 polymorphisms on postprandial response to a fat overload in metabolic syndrome patients. Clin Biochem 2010; 43:1300-4. [DOI: 10.1016/j.clinbiochem.2010.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 07/14/2010] [Accepted: 08/08/2010] [Indexed: 10/19/2022]
|
46
|
|
47
|
Hernandez C, Molusky M, Li Y, Li S, Lin JD. Regulation of hepatic ApoC3 expression by PGC-1β mediates hypolipidemic effect of nicotinic acid. Cell Metab 2010; 12:411-419. [PMID: 20889132 PMCID: PMC2950832 DOI: 10.1016/j.cmet.2010.09.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 06/24/2010] [Accepted: 07/21/2010] [Indexed: 10/19/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR) γ coactivator-1β (PGC-1β) is a transcriptional coactivator that induces hypertriglyceridemia in response to dietary fats through activating hepatic lipogenesis and lipoprotein secretion. The expression of PGC-1β is regulated by free fatty acids. Here we show that PGC-1β regulates plasma triglyceride metabolism through stimulating apolipoprotein C3 (APOC3) expression and elevating APOC3 levels in circulation. Remarkably, liver-specific knockdown of APOC3 significantly ameliorates PGC-1β-induced hypertriglyceridemia in mice. Hepatic expression of PGC-1β and APOC3 is reduced in response to acute and chronic treatments with nicotinic acid, a widely prescribed drug for lowering plasma triglycerides. Adenoviral-mediated knockdown of PGC-1β or APOC3 in the liver recapitulates the hypolipidemic effect of nicotinic acid. Proteomic analysis of hepatic PGC-1β transcriptional complex indicates that it stimulates APOC3 expression through coactivating orphan nuclear receptor ERRα and recruiting chromatin-remodeling cofactors. Together, these studies identify PGC-1β as an important regulator of the APOC3 gene cluster and reveal a mechanism through which nicotinic acid achieves its therapeutic effects.
Collapse
Affiliation(s)
- Carlos Hernandez
- Life Sciences Institute, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Matthew Molusky
- Life Sciences Institute, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yaqiang Li
- Life Sciences Institute, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Siming Li
- Life Sciences Institute, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jiandie D Lin
- Life Sciences Institute, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
48
|
Heeren J, Merkel M. Hypertriglyceridemia in obese subjects: Caused by reduced apolipoprotein A5 plasma levels? Atherosclerosis 2010; 212:386-7. [DOI: 10.1016/j.atherosclerosis.2010.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 06/09/2010] [Indexed: 10/19/2022]
|
49
|
Sundaram M, Yao Z. Recent progress in understanding protein and lipid factors affecting hepatic VLDL assembly and secretion. Nutr Metab (Lond) 2010; 7:35. [PMID: 20423497 PMCID: PMC2873297 DOI: 10.1186/1743-7075-7-35] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 04/27/2010] [Indexed: 02/06/2023] Open
Abstract
Excess lipid induced metabolic disorders are one of the major existing challenges for the society. Among many different causes of lipid disorders, overproduction and compromised catabolism of triacylglycerol-rich very low density lipoproteins (VLDL) have become increasingly prevalent leading to hyperlipidemia worldwide. This review provides the latest understanding in different aspects of VLDL assembly process, including structure-function relationships within apoB, mutations in APOB causing hypobetalipoproteinemia, significance of modulating microsomal triglyceride-transfer protein activity in VLDL assembly, alterations of VLDL assembly by different fatty acid species, and hepatic proteins involved in vesicular trafficking, and cytosolic lipid droplet metabolism that contribute to VLDL assembly. The role of lipoprotein receptors and exchangeable apolipoproteins that promote or diminish VLDL assembly and secretion is discussed. New understanding on dysregulated insulin signaling as a consequence of excessive triacylglycerol-rich VLDL in the plasma is also presented. It is hoped that a comprehensive view of protein and lipid factors that contribute to molecular and cellular events associated with VLDL assembly and secretion will assist in the identification of pharmaceutical targets to reduce disease complications related to hyperlipidemia.
Collapse
Affiliation(s)
- Meenakshi Sundaram
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Zemin Yao
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Department of Pathology and Laboratory Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
50
|
Ken-Dror G, Goldbourt U, Dankner R. Different effects of apolipoprotein A5 SNPs and haplotypes on triglyceride concentration in three ethnic origins. J Hum Genet 2010; 55:300-7. [PMID: 20395964 DOI: 10.1038/jhg.2010.27] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Several polymorphisms in the ApoA5 gene emerged as important candidate genes in triglyceride metabolism. The aim of this study was to determine the associations between ApoA5 polymorphisms, plasma triglyceride concentrations and the presence of cardiovascular disease (CVD) in three ethnic origins. Genotypes for 15 single nucleotide polymorphisms (SNPs) were determined in 659 older adults (mean age 71+/-7 years) who immigrated to Israel or whose ancestors originated from East Europe (Ashkenazi), North Africa, Asia (Sephardic) or Yemen (Yemenite). The minor alleles of the four common SNPs (rs662799, rs651821, rs2072560 and rs2266788) are associated with an increase of 27-38% in triglyceride concentration among Ashkenazi and Yemenite Jews compared with the major alleles, but not among those of Sephardic origin. Conversely, among the Sephardic group, the presence of the minor allele in SNP rs3135506 compared with the major allele was associated with an increase of 34% in triglyceride concentration. The four SNPs were in significant linkage disequilibrium (D'=0.96-0.99), resulting in three haplotypes H1, H2 and H3, representing 98-99% of the population. Haplotype H2 was significantly associated with triglyceride concentration among Ashkenazi and Yemenite but not among Sephardic Jews. Conversely, haplotype H3 was associated with triglyceride concentration in Sephardic but not in Ashkenazi and Yemenite Jews. Ashkenazi carriers of H2 haplotype had a CVD odds ratio of 2.19 (95% CI: 1.05-4.58) compared with H1 (the most frequent), after adjustment for all other risk factors. These results suggest that different SNPs in ApoA5 polymorphisms may be associated with triglyceride concentration and CVD in each of these ethnic origins.
Collapse
Affiliation(s)
- Gie Ken-Dror
- Division of Epidemiology and Preventive Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | | | | |
Collapse
|