1
|
Peng D, Yang W, Tang T, He A, Xu X, Jing T, Xia D. PLA2G7 promotes immune evasion of bladder cancer through the JAK-STAT-PDL1 axis. Cell Death Dis 2025; 16:234. [PMID: 40169540 PMCID: PMC11962123 DOI: 10.1038/s41419-025-07593-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
Targeting immune checkpoints such as Programmed death ligand-1 (PD-L1) and Programmed cell death 1 (PD-1) has been approved for treating bladder cancer and shows promising clinical benefits. However, the relatively low response rate highlights the need to seek an alternative strategy to traditional PD-1/PD-L1 targeting immunotherapy. In this study, we found that PLA2G7 is significantly elevated in bladder cancer and correlates with worse prognosis. In vitro experiments demonstrated that knockdown of PLA2G7 does not significantly affect the proliferation, migration, and invasion of bladder cancer cells. Flow cytometry detection, as well as protein and RNA detection, showed that knockdown of PLA2G7 significantly inhibits PD-L1 expression and suppresses the growth of transplanted tumors by promoting CD8 + T-cell infiltration. Further experiments showed that PLA2G7 regulates the JAK-STAT pathway to promote PD-L1 expression by upregulating the phosphorylation of STAT1 and STAT3. Meanwhile, results from syngeneic mouse models indicated that PLA2G7 suppression and anti-CTLA4 therapy have synergistic effects on tumor burden and mouse survival. In addition, we found that ETS1 promotes PLA2G7 overexpression in bladder cancer cells. In summary, our findings provide a novel immunotherapeutic strategy against bladder cancer through targeting the ETS1-PLA2G7-STAT1/STAT3-PD-L1 axis.
Collapse
Affiliation(s)
- Ding Peng
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, PR China
| | - Wuping Yang
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, PR China
| | - Tianyu Tang
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, PR China
| | - Anbang He
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, PR China
| | - Xin Xu
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, PR China.
| | - Taile Jing
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, PR China.
| | - Dan Xia
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang Province, 310003, PR China.
| |
Collapse
|
2
|
Shin S, Baek DS, Mellors JW, Dimitrov DS, Li W. Development of Fully Human Antibodies Targeting SIRPα and PLA2G7 for Cancer Therapy. Antibodies (Basel) 2025; 14:21. [PMID: 40136470 PMCID: PMC11939323 DOI: 10.3390/antib14010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/12/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Macrophages play an important role in eliminating diseased and damaged cells through programmed cell death. Signal regulatory protein alpha (SIRPα) is a crucial immune checkpoint primarily expressed on myeloid cells and macrophages. It initiates a 'do not eat me' signal when engaged with CD47, which is typically expressed at elevated levels on multiple solid tumors. The phospholipase A2 Group 7 (PLA2G7), which is mainly secreted by macrophages, interacts with oxidized low-density lipoprotein (oxLDL) and associates with several vascular diseases and cancers. Methods: To identify potent fully human monoclonal antibodies (mAbs) against human SIRPα and PLA2G7, we conducted bio-panning of phage antibody libraries. Results: We isolated one human Fab (1B3) and VH (1A3) for SIRPα, as well as one human Fab (1H8) and one VH (1A9) for PLA2G7; the 1B3 Fab and 1A3 VH are competitively bound to SIRPα, interfering with CD47 binding. The 1B3 IgG and 1A3 VH-Fc augmented macrophage-mediated phagocytic activity when combined with the anti-EGFR antibody, cetuximab. The anti-PLA2G7 antibodies exhibited high specificity for the PLA2G7 antigen and effectively blocked the PLA2G7 enzymatic activity with half-maximal inhibitory concentrations (IC50) in the single-digit nanomolar range. Additionally, 1H8 IgG and its derivative bispecific antibody exhibited the ability to block PLA2G7-mediated tumor cell migration. Conclusions: Our anti-SIRPα mAbs are expected to serve as potent and fully human immune checkpoint inhibitors of SIRPα, enhancing the antitumor responses of SIRPα-positive immune cells. Moreover, our anti-PLA2G7 mAbs represent promising fully human PLA2G7 enzymatic blockade antibodies with the potential to enhance both anti-tumor and anti-aging responses. Anti-SIRPα and PLA2G7 mAbs can modulate macrophage phagocytic activity and inflammatory responses against tumors.
Collapse
Affiliation(s)
- Seungmin Shin
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
| | - Du-San Baek
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
- GLPG US, 1401 Forbes Avenue, Pittsburgh, PA 15219, USA
| | - John W. Mellors
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
- GLPG US, 1401 Forbes Avenue, Pittsburgh, PA 15219, USA
| | - Dimiter S. Dimitrov
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
- GLPG US, 1401 Forbes Avenue, Pittsburgh, PA 15219, USA
| | - Wei Li
- Center for Antibody Therapeutics, Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; (D.-S.B.); (J.W.M.); (D.S.D.)
| |
Collapse
|
3
|
Bonnefont-Rousselot D. [Lipoprotein-associated phospholipase A 2 (Lp-PLA 2): Relevant biomarker and therapeutic target?]. ANNALES PHARMACEUTIQUES FRANÇAISES 2025; 83:45-57. [PMID: 39241907 DOI: 10.1016/j.pharma.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
Over the last fifteen years, numerous studies have sought to decipher the role of lipoprotein-associated phospholipase A2 (Lp-PLA2) in vascular inflammation-related diseases, notably atherosclerosis. Despite the disappointing results of clinical trials using the Lp-PLA2 inhibitor darapladib, new pathophysiological, epidemiological and genetic data have enabled the development of new inhibitors. Recent studies also show that Lp-PLA2 is involved in vascular inflammation-related diseases other than atherosclerosis (ischemic stroke, Alzheimer's disease and vascular dementia, diabetes, cancers…), and inhibition of Lp-PLA2 could have beneficial therapeutic in these diseases. This review aims to present new data on Lp-PLA2 and to evaluate its current interest as a biomarker but also as a therapeutic target.
Collapse
Affiliation(s)
- Dominique Bonnefont-Rousselot
- Service de biochimie métabolique, hôpitaux universitaires Pitié-Salpêtrière-Charles-Foix, AP-HP, 47-83, boulevard de l'Hôpital, 75651 Paris, France; Inserm, CNRS, UFR de pharmacie, UTCBS, université Paris Cité, Paris, France.
| |
Collapse
|
4
|
Zhang F, Liu W, Meng F, Jiang Q, Tang W, Liu Z, Lin X, Xue R, Zhang S, Dong L. Inhibiting PLA2G7 reverses the immunosuppressive function of intratumoral macrophages and augments immunotherapy response in hepatocellular carcinoma. J Immunother Cancer 2024; 12:e008094. [PMID: 38272562 PMCID: PMC10824072 DOI: 10.1136/jitc-2023-008094] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is an exceptionally immunosuppressive malignancy characterized by limited treatment options and a dismal prognosis. Macrophages constitute the primary and heterogeneous immune cell population within the HCC microenvironment. Our objective is to identify distinct subsets of macrophages implicated in the progression of HCC and their resistance to immunotherapy. METHODS Intratumoral macrophage-specific marker genes were identified via single-cell RNA sequencing analyses. The clinical relevance of phospholipase A2 Group VII (PLA2G7), a pivotal enzyme in phospholipid metabolism, was assessed in patients with HCC through immunohistochemistry and immunofluorescence. Flow cytometry and an in vitro co-culture system were used to elucidate the specific role of PLA2G7 in macrophages. Orthotopic and subcutaneous HCC mouse models were employed to evaluate the potential of the PLA2G7 inhibitor in complementing immune checkpoint blockade (ICB) therapy. RESULTS Single-cell RNA sequencing analyses disclosed predominant PLA2G7 expression in intratumoral macrophages within the HCC microenvironment. The macrophage-specific PLA2G7 was significantly correlated with poorer prognosis and immunotherapy resistance in patients with HCC. PLA2G7high macrophages represent a highly immunosuppressive subset and impede CD8 T-cell activation. Pharmacological inhibition of PLA2G7 by darapladib improved the therapeutic efficacy of anti-programmed cell death protein 1 antibodies in the HCC mouse models. CONCLUSIONS Macrophage-specific PLA2G7 serves as a novel biomarker capable of prognosticating immunotherapy responsiveness and inhibiting PLA2G7 has the potential to enhance the efficacy of ICB therapy for HCC.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Wenfeng Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Fansheng Meng
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Qiuyu Jiang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Wenqing Tang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhiyong Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiahui Lin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Si Zhang
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
5
|
Verdoia M, Rolla R, Gioscia R, Rognoni A, De Luca G. Lipoprotein associated- phospholipase A2 in STEMI vs. NSTE-ACS patients: a marker of cardiovascular atherosclerotic risk rather than thrombosis. J Thromb Thrombolysis 2023:10.1007/s11239-023-02801-1. [PMID: 37022507 DOI: 10.1007/s11239-023-02801-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2023] [Indexed: 04/07/2023]
Abstract
The precise role of Lipoprotein associated phospholipase A2 (Lp-PlA2) in the pathogenesis of acute coronary syndromes (ACS) and in the prediction of future cardiovascular events is still debated. So far, few data exist on the variations of Lp-PlA2 activity in ACS and especially in NSTE-ACS vs. STEMI patients, where thrombotic and atherosclerotic mechanisms could play a differential role. The aim of the present study was, then, to compare Lp-PlA2 activity according to the type of ACS presentation. METHODS A consecutive cohort of patients undergoing coronary angiography for acute coronary syndrome (ACS) were included and divided according to presentation for non ST-segment elevation-ACS or ST-segment elevation Myocardial Infarction (STEMI). Lp-PLA2 activity was assessed in blood samples drawn at admission using the Diazyme Lp-PlA2 Activity Assay. RESULTS We included in our study 117 patients, of whom 31 (26.5%) presented with STEMI. STEMI patients were significantly younger (p = 0.05), displayed a lower rate of hypertension (p = 0.002), previous MI (p = 0.001) and PCI (p = 0.01) and used less frequently statins (p = 0.01) and clopidogrel (p = 0.02). White blood cells and admission glycemia were increased in STEMI (p = 0.001, respectively). The prevalence and severity of CAD was not different according to ACS types, but for a higher prevalence of thrombus (p < 0.001) and lower TIMI flow (p = 0.002) in STEMI. The levels of Lp-PlA2 were significantly lower in STEMI as compared to NSTE-ACS patients, (132 ± 41.1 vs. 154.6 ± 40.9 nmol/min/mL, p = 0.01). In fact, the rate of patients with Lp-PlA2 above the median (148 nmol/min/mL) was significantly lower in STEMI patients as compared to NSTE-ACS (32.3% vs. 57%, p = 0.02, adjusted OR[95% CI] = 0.20[0.06-0.68], p = 0.010). Moreover, a direct linear relationship was observed between Lp-PlA2 and LDL-C (r = 0.47, p < 0.001), but not with inflammatory biomarkers. CONCLUSION The present study shows that among ACS patients, the levels of Lp-PlA2 are inversely associated with STEMI presentation and thrombotic coronary occlusion, being instead increased in NSTE-ACS patients, therefore potentially representing a marker of more aggressive chronic cardiovascular disease with an increased risk of recurrent cardiovascular events.
Collapse
Affiliation(s)
- Monica Verdoia
- Division of Cardiology, Nuovo Ospedale degli Infermi, ASL Biella, Biella, Italy
| | - Roberta Rolla
- Clinical Chemistry, Azienda Ospedaliera-Universitaria "Maggiore della Carità", Universit? del Piemonte Orientale, Novara, Italy
| | - Rocco Gioscia
- Division of Cardiology, Nuovo Ospedale degli Infermi, ASL Biella, Biella, Italy
| | - Andrea Rognoni
- Division of Cardiology, Nuovo Ospedale degli Infermi, ASL Biella, Biella, Italy
| | - Giuseppe De Luca
- Division of Cardiology, AOU "Policlinico G. Martino", Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.
- Division of Cardiology, Galeazzi-Sant'Ambrogio Hospital, Milan, Italy.
| |
Collapse
|
6
|
Wang J, Jiang M, Xiong A, Zhang L, Luo L, Liu Y, Liu S, Ran Q, Wu D, Xiong Y, He X, Leung ELH, Li G. Integrated analysis of single-cell and bulk RNA sequencing reveals pro-fibrotic PLA2G7 high macrophages in pulmonary fibrosis. Pharmacol Res 2022; 182:106286. [PMID: 35662628 DOI: 10.1016/j.phrs.2022.106286] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/09/2022] [Accepted: 05/29/2022] [Indexed: 10/18/2022]
Abstract
Pulmonary fibrosis (PF) is the pathological change of end-stage interstitial lung diseases with high mortality and limited therapeutic options. Lung macrophages have distinct subsets with divergent functions, and play critical roles in the pathogenesis of PF. In this study, integrative analysis of lung single-cell and bulk RNA-seq data from patients with fibrotic hypersensitivity pneumonitis and idiopathic pulmonary fibrosis was utilized to identify particular macrophage subsets during the development of PF. We find a specific macrophage subpopulation highly expressing PLA2G7 in fibrotic lungs. We performed additional single-cell RNA-seq analysis to identify analogous macrophage population in bleomycin (BLM)-induced mouse pulmonary fibrosis models. By in vitro and in vivo experiments, we further reveal the pro-fibrotic role for this PLA2G7high macrophage subset in fibroblast-to-myofibroblast transition (FMT) during pulmonary fibrosis. PLA2G7 promotes FMT via LPC/ATX/LPA/LPA2 axis in macrophages. Moreover, PLA2G7 is regulated by STAT1, and pharmacological inhibition of PLA2G7 by Darapladib ameliorates pulmonary fibrosis in BLM-induced mice. The results of this study support the view that PLA2G7high macrophage subpopulation contributes importantly to the pathogenesis of PF, which provides a potential way for targeted therapy.
Collapse
Affiliation(s)
- Junyi Wang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology, Taipa, Macao Special Administrative Region of China
| | - Manling Jiang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China
| | - Anying Xiong
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China
| | - Lei Zhang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology, Taipa, Macao Special Administrative Region of China
| | - Li Luo
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yao Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Shengbin Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Qin Ran
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China
| | - Dehong Wu
- Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China
| | - Ying Xiong
- Department of Pulmonary and Critical Care Medicine, Sichuan Friendship Hospital, Chengdu, China
| | - Xiang He
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China.
| | - Elaine Lai-Han Leung
- Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region of China.
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China.
| |
Collapse
|
7
|
González LM, Robles NR, Mota-Zamorano S, Arévalo-Lorido JC, Valdivielso JM, López-Gómez J, Gervasini G. Tag-SNPs in Phospholipase-Related Genes Modify the Susceptibility to Nephrosclerosis and its Associated Cardiovascular Risk. Front Pharmacol 2022; 13:817020. [PMID: 35586043 PMCID: PMC9108153 DOI: 10.3389/fphar.2022.817020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Nephrosclerosis patients have a high cardiovascular (CV) risk that is very often of more concern than the renal disease itself. We aimed to determine whether variants in phospholipase-related genes, associated with atherosclerosis and CV outcomes in the general population, could constitute biomarkers of nephrosclerosis and/or its associated CV risk. We screened 1,209 nephrosclerosis patients and controls for 86 tag-SNPs that were identified in the SCARB1, PLA2G4A, and PLA2G7 gene loci. Regression models were utilized to evaluate their effect on several clinical parameters. Most notably, rs10846744 and rs838880 in SCARB1 showed significant odds ratios (OR) of 0.66 (0.51–0.87), p = 0.003 and 1.48 (1.11–1.96), p = 0.007 for nephrosclerosis risk. PLA2G4A and PLA2G7 harboured several SNPs associated with atherosclerosis measurements in the patients, namely common carotid intima media thickness (ccIMT), presence of plaques, number of plaques detected and 2-years ccIMT progression (significant p-values ranging from 0.0004 to 0.047). Eight SNPs in PLA2G4A were independent risk factors for CV events in nephrosclerosis patients. Their addition to a ROC model containing classic risk factors significantly improved its predictive power from AUC = 69.1% (61.4–76.9) to AUC = 79.1% (73.1–85.1%), p = 0.047. Finally, PLA2G4A rs932476AA and rs6683619AA genotypes were associated with lower CV event-free survival after controlling for confounding variables [49.59 (47.97–51.21) vs. 51.81 (49.93–51.78) months, p = 0.041 and 46.46 (41.00–51.92) vs. 51.17 (50.25–52.08) months, p = 0.022, respectively]. Variability in phospholipase-related genes play a relevant role in nephrosclerosis and associated atherosclerosis measurements and CV events.
Collapse
Affiliation(s)
- Luz M. González
- Department of Medical and Surgical Therapeutics, Medical School, Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, Spain
| | - Nicolás R. Robles
- Service of Nephrology, Badajoz University Hospital, Badajoz, Spain
- RICORS2040 Renal Research Network, Madrid, Spain
| | - Sonia Mota-Zamorano
- Department of Medical and Surgical Therapeutics, Medical School, Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, Spain
- RICORS2040 Renal Research Network, Madrid, Spain
| | | | - José Manuel Valdivielso
- RICORS2040 Renal Research Network, Madrid, Spain
- Vascular and Renal Translational Research Group, UDETMA, IRBLleida, Lleida, Spain
| | - Juan López-Gómez
- Service of Clinical Analyses, Badajoz University Hospital, Badajoz, Spain
| | - Guillermo Gervasini
- Department of Medical and Surgical Therapeutics, Medical School, Institute of Molecular Pathology Biomarkers, University of Extremadura, Badajoz, Spain
- RICORS2040 Renal Research Network, Madrid, Spain
- *Correspondence: Guillermo Gervasini,
| |
Collapse
|
8
|
Abstract
Insights into the genetic basis of human disease are helping to address some of the key challenges in new drug development including the very high rates of failure. Here we review the recent history of an emerging, genomics-assisted approach to pharmaceutical research and development, and its relationship to Mendelian randomization (MR), a well-established analytical approach to causal inference. We demonstrate how human genomic data linked to pharmaceutically relevant phenotypes can be used for (1) drug target identification (mapping relevant drug targets to diseases), (2) drug target validation (inferring the likely effects of drug target perturbation), (3) evaluation of the effectiveness and specificity of compound-target engagement (inferring the extent to which the effects of a compound are exclusive to the target and distinguishing between on-target and off-target compound effects), and (4) the selection of end points in clinical trials (the diseases or conditions to be evaluated as trial outcomes). We show how genomics can help identify indication expansion opportunities for licensed drugs and repurposing of compounds developed to clinical phase that proved safe but ineffective for the original intended indication. We outline statistical and biological considerations in using MR for drug target validation (drug target MR) and discuss the obstacles and challenges for scaled applications of these genomics-based approaches.
Collapse
Affiliation(s)
- Amand F Schmidt
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, United Kingdom
- UCL British Heart Foundation Research Accelerator, London WC1E 6BT, United Kingdom
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, United Kingdom
- UCL British Heart Foundation Research Accelerator, London WC1E 6BT, United Kingdom
- Health Data Research UK, London NW1 2BE, United Kingdom
| | - Chris Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, United Kingdom
- UCL British Heart Foundation Research Accelerator, London WC1E 6BT, United Kingdom
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
- Health Data Research UK, London NW1 2BE, United Kingdom
| |
Collapse
|
9
|
Patoulias D, Stavropoulos K, Imprialos K, Athyros V, Grassos H, Doumas M, Faselis C. Inflammatory Markers in Cardiovascular Disease; Lessons Learned and Future Perspectives. Curr Vasc Pharmacol 2021; 19:323-342. [PMID: 32188386 DOI: 10.2174/1570161118666200318104434] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiovascular disease (CVD) still remains the leading cause of morbidity and mortality worldwide. It is now established that inflammation plays a crucial role in atherosclerosis and atherothrombosis, and thus, it is closely linked to cardiovascular disease. OBJECTIVE The aim of the present review is to summarize and critically appraise the most relevant evidence regarding the potential use of inflammatory markers in the field of CVD. METHODS We conducted a comprehensive research of the relevant literature, searching MEDLINE from its inception until November 2018, primarily for meta-analyses, randomized controlled trials and observational studies. RESULTS Established markers of inflammation, mainly C-reactive protein, have yielded significant results both for primary and secondary prevention of CVD. Newer markers, such as lipoprotein-associated phospholipase A2, lectin-like oxidized low-density lipoprotein receptor-1, cytokines, myeloperoxidase, cell adhesion molecules, matrix metalloproteinases, and the CD40/CD40 ligand system, have been largely evaluated in human studies, enrolling both individuals from the general population and patients with established CVD. Some markers have yielded conflicting results; however, others are now recognized not only as promising biomarkers of CVD, but also as potential therapeutic targets, establishing the role of anti-inflammatory and pleiotropic drugs in CVD. CONCLUSION There is significant evidence regarding the role of consolidated and novel inflammatory markers in the field of diagnosis and prognosis of CVD. However, multimarker model assessment, validation of cut-off values and cost-effectiveness analyses are required in order for those markers to be integrated into daily clinical practice.
Collapse
Affiliation(s)
- Dimitrios Patoulias
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | | | - Konstantinos Imprialos
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | - Vasilios Athyros
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | | | - Michael Doumas
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | - Charles Faselis
- VA Medical Center, and George Washington University, Washington, DC 20422, United States
| |
Collapse
|
10
|
Fani L, Georgakis MK, Ikram MA, Ikram MK, Malik R, Dichgans M. Circulating biomarkers of immunity and inflammation, risk of Alzheimer's disease, and hippocampal volume: a Mendelian randomization study. Transl Psychiatry 2021; 11:291. [PMID: 34001857 PMCID: PMC8129147 DOI: 10.1038/s41398-021-01400-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 02/08/2023] Open
Abstract
The aim of this study was to explore the association between genetically predicted circulating levels of immunity and inflammation, and the risk of Alzheimer's disease (AD) and hippocampal volume, by conducting a two-sample Mendelian Randomization Study. We identified 12 markers of immune cells and derived ratios (platelet count, eosinophil count, neutrophil count, basophil count, monocyte count, lymphocyte count, platelet-to-lymphocyte ratio, monocyte-to-lymphocyte ratio, CD4 count, CD8 count, CD4-to-CD8 ratio, and CD56) and 5 signaling molecules (IL-6, fibrinogen, CRP, and Lp-PLA2 activity and mass) as primary exposures of interest. Other genetically available immune biomarkers with a weaker a priori link to AD were considered secondary exposures. Associations with AD were evaluated in The International Genomics of Alzheimer's Project (IGAP) GWAS dataset (21,982 cases; 41,944 controls of European ancestry). For hippocampal volume, we extracted data from a GWAS meta-analysis on 33,536 participants of European ancestry. None of the primary or secondary exposures showed statistically significant associations with AD or with hippocampal volume following P-value correction for multiple comparisons using false discovery rate < 5% (Q-value < 0.05). CD4 count showed the strongest suggestive association with AD (odds ratio 1.32, P < 0.01, Q > 0.05). There was evidence for heterogeneity in the MR inverse variance-weighted meta-analyses as measured by Cochran Q, and weighted median and weighted mode for multiple exposures. Further cluster analyses did not reveal clusters of variants that could influence the risk factor in distinct ways. This study suggests that genetically predicted circulating biomarkers of immunity and inflammation are not associated with AD risk or hippocampal volume. Future studies should assess competing risk, explore in more depth the role of adaptive immunity in AD, in particular T cells and the CD4 subtype, and confirm these findings in other ethnicities.
Collapse
Affiliation(s)
- Lana Fani
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Marios K. Georgakis
- grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research, University Hospital LMU Munich, Munich, Germany
| | - M. Arfan Ikram
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - M. Kamran Ikram
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands ,grid.5645.2000000040459992XDepartment of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Rainer Malik
- grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research, University Hospital LMU Munich, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital LMU Munich, Munich, Germany.
| |
Collapse
|
11
|
Fras Z, Tršan J, Banach M. On the present and future role of Lp-PLA 2 in atherosclerosis-related cardiovascular risk prediction and management. Arch Med Sci 2021; 17:954-964. [PMID: 34336025 PMCID: PMC8314407 DOI: 10.5114/aoms.2020.98195] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/02/2020] [Indexed: 12/23/2022] Open
Abstract
Circulating concentration and activity of secretory phospholipase A2 (sPLA2) and lipoprotein-associated phospholipase A2 (Lp-PLA2) have been proven as biomarkers of increased risk of atherosclerosis-related cardiovascular disease (ASCVD). Lp-PLA2 might be part of the atherosclerotic process and may contribute to plaque destabilisation through inflammatory activity within atherosclerotic lesions. However, all attempts to translate the inhibition of phospholipase into clinically beneficial ASCVD risk reduction, including in randomised studies, by either non-specific inhibition of sPLA2 (by varespladib) or specific Lp-PLA2 inhibition by darapladib, unexpectedly failed. This gives us a strong imperative to continue research aimed at a better understanding of how Lp-PLA2 and sPLA2 regulate vascular inflammation and atherosclerotic plaque development. From the clinical viewpoint there is a need to establish and validate the existing and emerging novel anti-inflammatory therapeutic strategies to fight against ASCVD development, by using potentially better animal models and differently designed clinical trials in humans.
Collapse
Affiliation(s)
- Zlatko Fras
- Centre for Preventive Cardiology, Department of Vascular Medicine, Division of Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Chair of Internal Medicine, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Jure Tršan
- Centre for Preventive Cardiology, Department of Vascular Medicine, Division of Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Poland
- Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| |
Collapse
|
12
|
Ajala ON, Everett BM. Targeting Inflammation to Reduce Residual Cardiovascular Risk. Curr Atheroscler Rep 2020; 22:66. [DOI: 10.1007/s11883-020-00883-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
13
|
Perrot N, Thériault S, Rigade S, Chen HY, Dina C, Martinsson A, Boekholdt SM, Capoulade R, Le Tourneau T, Messika-Zeitoun D, Engert JC, Wareham NJ, Clavel MA, Pibarot P, Smith JG, Schott JJ, Mathieu P, Bossé Y, Thanassoulis G, Arsenault BJ. Lipoprotein-associated phospholipase A2 activity, genetics and calcific aortic valve stenosis in humans. Heart 2020; 106:1407-1412. [PMID: 32636298 DOI: 10.1136/heartjnl-2020-316722] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Lipoprotein-associated phospholipase A2 (Lp-PLA2) activity has been shown to predict calcific aortic valve stenosis (CAVS) outcomes. Our objective was to test the association between plasma Lp-PLA2 activity and genetically elevated Lp-PLA2 mass/activity with CAVS in humans. METHODS AND RESULTS Lp-PLA2 activity was measured in 890 patients undergoing cardiac surgery, including 476 patients undergoing aortic valve replacement for CAVS and 414 control patients undergoing coronary artery bypass grafting. After multivariable adjustment, Lp-PLA2 activity was positively associated with the presence of CAVS (OR=1.21 (95% CI 1.04 to 1.41) per SD increment). We selected four single nucleotide polymorphisms (SNPs) at the PLA2G7 locus associated with either Lp-PLA2 mass or activity (rs7756935, rs1421368, rs1805017 and rs4498351). Genetic association studies were performed in eight cohorts: Quebec-CAVS (1009 cases/1017 controls), UK Biobank (1350 cases/349 043 controls), European Prospective Investigation into Cancer and Nutrition-Norfolk (504 cases/20 307 controls), Genetic Epidemiology Research on Aging (3469 cases/51 723 controls), Malmö Diet and Cancer Study (682 cases/5963 controls) and three French cohorts (3123 cases/6532 controls), totalling 10 137 CAVS cases and 434 585 controls. A fixed-effect meta-analysis using the inverse-variance weighted method revealed that none of the four SNPs was associated with CAVS (OR=0.99 (95% CI 0.96 to 1.02, p=0.55) for rs7756935, 0.97 (95% CI 0.93 to 1.01, p=0.11) for rs1421368, 1.00 (95% CI 1.00 to 1.01, p=0.29) for rs1805017, and 1.00 (95% CI 0.97 to 1.04, p=0.87) for rs4498351). CONCLUSIONS Higher Lp-PLA2 activity is significantly associated with the presence of CAVS and might represent a biomarker of CAVS in patients with heart disease. Results of our genetic association study suggest that Lp-PLA2 is however unlikely to represent a causal risk factor or therapeutic target for CAVS.
Collapse
Affiliation(s)
- Nicolas Perrot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec city, Québec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| | - Sébastien Thériault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec city, Québec, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| | - Sidwell Rigade
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Hao Yu Chen
- McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Christian Dina
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Andreas Martinsson
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden.,Department of Cardiology, Sahlgrenska universitetssjukhuset, Goteborg, Sweden
| | - S Matthijs Boekholdt
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Romain Capoulade
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Thierry Le Tourneau
- Ultrasound and Cardiology Departments, University Hospital, Institut du Thorax, Nantes, France
| | - David Messika-Zeitoun
- University of Ottawa Heart Institute, APHP, Bichat Hospital, Ottawa, Ontario, Canada
| | - James C Engert
- McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Nicholas J Wareham
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Marie-Annick Clavel
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec city, Québec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec city, Québec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| | - J Gustav Smith
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | | | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec city, Québec, Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Québec City, Quebec, Canada
| | - Yohan Bossé
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec city, Québec, Canada.,Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Quebec, Canada
| | - George Thanassoulis
- McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec city, Québec, Canada .,Department of Medicine, Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
14
|
Liu Y, Wu X, Nie S, Zhou S, Xiao S, Gao X, Lin Z, Sun J, Huang Y. Methylation of Phospholipase A2 Group VII Gene Is Associated with Brain Arteriovenous Malformations in Han Chinese Populations. J Mol Neurosci 2020; 70:1056-1063. [DOI: 10.1007/s12031-020-01508-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 02/19/2020] [Indexed: 11/29/2022]
|
15
|
Zhou M, Chen M, Bai H, He GL, Liu QQ, Guan LB, Liu XH, Fan P. Association of the G994T and R92H genotypes of platelet-activating factor acetylhydrolase with risk of preeclampsia in Chinese women. Pregnancy Hypertens 2020; 20:19-26. [DOI: 10.1016/j.preghy.2020.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/02/2019] [Accepted: 02/23/2020] [Indexed: 10/24/2022]
|
16
|
The Beneficial Effects of Alpha Lipoic Acid Supplementation on Lp-PLA2 Mass and Its Distribution between HDL and apoB-Containing Lipoproteins in Type 2 Diabetic Patients: A Randomized, Double-Blind, Placebo-Controlled Trial. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5850865. [PMID: 32256955 PMCID: PMC7085885 DOI: 10.1155/2020/5850865] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/11/2020] [Indexed: 02/06/2023]
Abstract
Lipoprotein-associated phospholipase A2 (Lp-PLA2) is a new specific vascular inflammation biomarker that is carried by the lipoproteins in the blood and plays a prominent role in the pathogenesis of atherosclerosis. Increased Lp-PLA2 levels and impaired Lp-PLA2 distribution across high-density lipoprotein (HDL) and non-HDL lipoproteins have been reported in diabetic patients, which is associated with the increase in cardiovascular disease (CVD) risk. This study is aimed at investigating the effect of alpha lipoic acid (ALA), as an antioxidant with potential cardioprotective properties, on the Lp-PLA2 mass and its distribution in diabetic patients. In a double-blind, randomized, placebo-controlled clinical trial, seventy diabetic patients were randomly allocated to ALA (1200 mg ALA as two 600 mg capsules/day) and placebo (two maltodextrin capsules/day) groups. The serum levels of total Lp-PLA2 mass, HDL-Lp-PLA2, oxidized low-density lipoproteins (ox-LDL), apolipoprotein A1 (apo A1), lipid profiles, fasting blood sugar (FBS), and insulin were measured, and apolipoprotein B- (apoB-) associated Lp-PLA2 and homeostasis model of assessment index (HOMA-IR) were calculated at the baseline and after 8 weeks of intervention. ALA significantly decreased the ox-LDL, total Lp-PLA2 mass, apoB-associated Lp-PLA2, and percent of apoB-associated Lp-PLA2 and triglyceride and increased the percent of HDL-Lp-PLA2 compared with the placebo group but had no significant effect on HDL-Lp-PLA2 mass, apo A1, lipid profiles, and glycemic indices. There was a positive correlation between the reduction in the ox-LDL level and total Lp-PLA2 mass in the ALA group. In conclusion, ALA may decrease the CVD risk by reducing the ox-LDL and Lp-PLA2 mass and improving the Lp-PLA2 distribution among lipoproteins in type 2 diabetic patients.
Collapse
|
17
|
Wu C, Zhou T, Zhou Y, Han W. Association of Serum Lipoprotein-Associated Phospholipase A2 and A379V Gene Polymorphisms with Carotid Plaques. Genet Test Mol Biomarkers 2020; 24:131-137. [PMID: 32109154 DOI: 10.1089/gtmb.2019.0162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Objective: Lipoprotein-associated phospholipase A2 (LP-PLA2) is closely related to the development of atherosclerosis. The A379V gene polymorphism, located in exon 11 of the PLA2G7 gene, can affect LP-PLA2 levels and the inflammatory response. However, the association between the A379V polymorphism and formation of carotid plaques is unclear. Materials and Methods: A total of 516 ischemic stroke patients were classified according to carotid intima-media thickness as measured by ultrasound into the plaque group (n = 375, including 258 and 117 cases having vulnerable and stable plaques, respectively) and the nonplaque group (n = 141). The LP-PLA2 gene A379V polymorphism was determined by DNA sequencing, and Lp-PLA2 serum protein levels were determined simultaneously. Results: The serum Lp-PLA2 levels (p < 0.0005), CT+TT genotype frequency (odds ratio [OR]: 1.730, 95% confidence interval [CI]: 1.114-2.686, p = 0.014), and T allele frequency (OR: 1.592, 95% CI: 1.082-2.342, p = 0.018) in the plaque group were significantly higher than those in the nonplaque group. Lp-PLA2 serum levels in the vulnerable plaque subgroup were significantly higher than those in the stable plaque subgroup (p = 0.003). However, there were no significant differences in the frequency of the A379V polymorphism between the vulnerable and stable plaque subgroups. For all subjects, Lp-PLA2 serum levels for patients having a CC genotype were significantly lower than those for patients having a CT (p = 0.003), TT (p = 0.014), or CC+TT genotype (p = 0.001). Logistic regression showed that the Lp-PLA2 level was a risk factor for carotid plaque formation (OR: 1.024, 95% CI: 1.011-1.030, p = 0.001), but the A379V gene polymorphism was not (OR: 1.037, 95% CI: 0.357-3.012, p = 0.947). Conclusion: The A379V gene polymorphism might be associated with serum Lp-PLA2 levels and carotid plaque formation, but not with plaque vulnerability in a Chinese Han population. Serum Lp-PLA2 level was shown to be a risk factor for carotid plaque formation.
Collapse
Affiliation(s)
- Changzhu Wu
- Department of Neurology, Affiliated Hospital of Wenzhou Medical University, Taizhou Hospital, Taizhou, Zhejiang, People's Republic of China
| | - Ting Zhou
- Department of Neurology, Affiliated Hospital of Wenzhou Medical University, Taizhou Hospital, Taizhou, Zhejiang, People's Republic of China
| | - Yuanlin Zhou
- Department of Neurology, Affiliated Hospital of Wenzhou Medical University, Taizhou Hospital, Taizhou, Zhejiang, People's Republic of China
| | - Wensheng Han
- Department of Neurology, Affiliated Hospital of Wenzhou Medical University, Taizhou Hospital, Taizhou, Zhejiang, People's Republic of China
| |
Collapse
|
18
|
Gu X, Lin W, Xu Y, Che D, Tan Y, Lu Z, Pi L, Fu L, Zhou H, Jiang Z, Gu X. The rs1051931 G>A Polymorphism in the PLA2G7 Gene Confers Resistance to Immunoglobulin Therapy in Kawasaki Disease in a Southern Chinese Population. Front Pediatr 2020; 8:338. [PMID: 32656171 PMCID: PMC7324548 DOI: 10.3389/fped.2020.00338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 05/21/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Kawasaki disease (KD) is a common cardiovascular disease in infants and young children, with fever, rash, and conjunctivitis as the main clinical manifestations, which can lead to the occurrence of coronary aneurysms. Intravenous immunoglobulin (IVIG) is the preferred treatment for KD patients, but 10-20% of patients are resistant to IVIG. Lipoprotein-associated phospholipase A 2 (Lp-PLA2) is a potential therapeutic target for coronary atherosclerotic heart disease, and the polymorphism of Phospholipase A2 Group VII (PLA2G7) is closely related to the activity of Lp-PLA2, of which rs1051931 is the strongest. Therefore, the rs1051931 polymorphism may be a predictor of IVIG resistance in KD patients. Methods: A total of 760 KD cases, including 148 IVIG-resistant patients and 612 IVIG-responsive patients, were genotyped for rs1051931 in PLA2G7, we compared the effects of rs1051931 on IVIG treatment in KD patients by odds ratios (OR) and 95% confidence interval (CI). Results: The homozygous mutation AA may be a protective factor for IVIG resistance in KD patients (adjusted OR = 3.47, 95% CI = 1.14-10.57, P = 0.0284) and is more evident in patients with KD aged <60 months (adjusted OR = 3.68, 95% CI = 1.10-12.28, P = 0.0399). Conclusions: The PLA2G7 rs1051931 G>A polymorphism may be suitable as a biomarker for the diagnosis or prognosis of IVIG resistance in KD in a southern Chinese population.
Collapse
Affiliation(s)
- Xueping Gu
- Department of Blood Transfusion and Clinical Lab, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Wenchun Lin
- Department of Pneumology, Guangzhou Women and Children's Medical Center, Guangzhou Medical College, Guangzhou, China
| | - Yufen Xu
- Department of Clinical Biological Resource Bank, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Di Che
- Department of Clinical Biological Resource Bank, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Yaqian Tan
- Department of Clinical Biological Resource Bank, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Zhaoliang Lu
- Department of Clinical Biological Resource Bank, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Lei Pi
- Department of Clinical Biological Resource Bank, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Lanyan Fu
- Department of Clinical Biological Resource Bank, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Huazhong Zhou
- Department of Clinical Biological Resource Bank, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Zhiyong Jiang
- Department of Blood Transfusion and Clinical Lab, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Xiaoqiong Gu
- Department of Blood Transfusion and Clinical Lab, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China.,Department of Clinical Biological Resource Bank, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
19
|
Hingorani AD, Kuan V, Finan C, Kruger FA, Gaulton A, Chopade S, Sofat R, MacAllister RJ, Overington JP, Hemingway H, Denaxas S, Prieto D, Casas JP. Improving the odds of drug development success through human genomics: modelling study. Sci Rep 2019; 9:18911. [PMID: 31827124 PMCID: PMC6906499 DOI: 10.1038/s41598-019-54849-w] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 11/06/2019] [Indexed: 01/19/2023] Open
Abstract
Lack of efficacy in the intended disease indication is the major cause of clinical phase drug development failure. Explanations could include the poor external validity of pre-clinical (cell, tissue, and animal) models of human disease and the high false discovery rate (FDR) in preclinical science. FDR is related to the proportion of true relationships available for discovery (γ), and the type 1 (false-positive) and type 2 (false negative) error rates of the experiments designed to uncover them. We estimated the FDR in preclinical science, its effect on drug development success rates, and improvements expected from use of human genomics rather than preclinical studies as the primary source of evidence for drug target identification. Calculations were based on a sample space defined by all human diseases - the 'disease-ome' - represented as columns; and all protein coding genes - 'the protein-coding genome'- represented as rows, producing a matrix of unique gene- (or protein-) disease pairings. We parameterised the space based on 10,000 diseases, 20,000 protein-coding genes, 100 causal genes per disease and 4000 genes encoding druggable targets, examining the effect of varying the parameters and a range of underlying assumptions, on the inferences drawn. We estimated γ, defined mathematical relationships between preclinical FDR and drug development success rates, and estimated improvements in success rates based on human genomics (rather than orthodox preclinical studies). Around one in every 200 protein-disease pairings was estimated to be causal (γ = 0.005) giving an FDR in preclinical research of 92.6%, which likely makes a major contribution to the reported drug development failure rate of 96%. Observed success rate was only slightly greater than expected for a random pick from the sample space. Values for γ back-calculated from reported preclinical and clinical drug development success rates were also close to the a priori estimates. Substituting genome wide (or druggable genome wide) association studies for preclinical studies as the major information source for drug target identification was estimated to reverse the probability of late stage failure because of the more stringent type 1 error rate employed and the ability to interrogate every potential druggable target in the same experiment. Genetic studies conducted at much larger scale, with greater resolution of disease end-points, e.g. by connecting genomics and electronic health record data within healthcare systems has the potential to produce radical improvement in drug development success rate.
Collapse
Affiliation(s)
- Aroon D Hingorani
- Institute of Cardiovascular Science, University College London, London, UK.
- Health Data Research UK and UCL BHF Research Accelerator, London, UK.
| | - Valerie Kuan
- Institute of Cardiovascular Science, University College London, London, UK
- Health Data Research UK and UCL BHF Research Accelerator, London, UK
| | - Chris Finan
- Institute of Cardiovascular Science, University College London, London, UK
- Health Data Research UK and UCL BHF Research Accelerator, London, UK
| | | | - Anna Gaulton
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
| | - Sandesh Chopade
- Institute of Cardiovascular Science, University College London, London, UK
- Health Data Research UK and UCL BHF Research Accelerator, London, UK
| | - Reecha Sofat
- Health Data Research UK and UCL BHF Research Accelerator, London, UK
- Institute of Health Informatics, University College London, London, UK
| | | | - John P Overington
- Institute of Cardiovascular Science, University College London, London, UK
- Medicines Discovery Catapult, Mereside, Alderley Park, Alderley Edge, Cheshire, UK
| | - Harry Hemingway
- Health Data Research UK and UCL BHF Research Accelerator, London, UK
- Institute of Health Informatics, University College London, London, UK
| | - Spiros Denaxas
- Health Data Research UK and UCL BHF Research Accelerator, London, UK
- Institute of Health Informatics, University College London, London, UK
| | - David Prieto
- Institute of Health Informatics, University College London, London, UK
- Applied Statistics in Medical Research Group, Catholic University of Murcia (UCAM), Murcia, Spain
| | - Juan Pablo Casas
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), Veterans Administration, Boston, MA, USA
| |
Collapse
|
20
|
Schmidt AF, Holmes MV, Preiss D, Swerdlow DI, Denaxas S, Fatemifar G, Faraway R, Finan C, Valentine D, Fairhurst-Hunter Z, Hartwig FP, Horta BL, Hypponen E, Power C, Moldovan M, van Iperen E, Hovingh K, Demuth I, Norman K, Steinhagen-Thiessen E, Demuth J, Bertram L, Lill CM, Coassin S, Willeit J, Kiechl S, Willeit K, Mason D, Wright J, Morris R, Wanamethee G, Whincup P, Ben-Shlomo Y, McLachlan S, Price JF, Kivimaki M, Welch C, Sanchez-Galvez A, Marques-Vidal P, Nicolaides A, Panayiotou AG, Onland-Moret NC, van der Schouw YT, Matullo G, Fiorito G, Guarrera S, Sacerdote C, Wareham NJ, Langenberg C, Scott RA, Luan J, Bobak M, Malyutina S, Pająk A, Kubinova R, Tamosiunas A, Pikhart H, Grarup N, Pedersen O, Hansen T, Linneberg A, Jess T, Cooper J, Humphries SE, Brilliant M, Kitchner T, Hakonarson H, Carrell DS, McCarty CA, Lester KH, Larson EB, Crosslin DR, de Andrade M, Roden DM, Denny JC, Carty C, Hancock S, Attia J, Holliday E, Scott R, Schofield P, O'Donnell M, Yusuf S, Chong M, Pare G, van der Harst P, Said MA, Eppinga RN, Verweij N, Snieder H, Christen T, Mook-Kanamori DO, Gustafsson S, Lind L, Ingelsson E, Pazoki R, Franco O, Hofman A, Uitterlinden A, Dehghan A, Teumer A, Baumeister S, Dörr M, Lerch MM, Völker U, Völzke H, Ward J, Pell JP, Meade T, Christophersen IE, Maitland-van der Zee AH, Baranova EV, Young R, Ford I, Campbell A, Padmanabhan S, Bots ML, Grobbee DE, Froguel P, Thuillier D, Roussel R, Bonnefond A, Cariou B, Smart M, Bao Y, Kumari M, Mahajan A, Hopewell JC, Seshadri S, Dale C, Costa RPE, Ridker PM, Chasman DI, Reiner AP, Ritchie MD, Lange LA, Cornish AJ, Dobbins SE, Hemminki K, Kinnersley B, Sanson M, Labreche K, Simon M, Bondy M, Law P, Speedy H, Allan J, Li N, Went M, Weinhold N, Morgan G, Sonneveld P, Nilsson B, Goldschmidt H, Sud A, Engert A, Hansson M, Hemingway H, Asselbergs FW, Patel RS, Keating BJ, Sattar N, Houlston R, Casas JP, Hingorani AD. Phenome-wide association analysis of LDL-cholesterol lowering genetic variants in PCSK9. BMC Cardiovasc Disord 2019; 19:240. [PMID: 31664920 PMCID: PMC6820948 DOI: 10.1186/s12872-019-1187-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/19/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND We characterised the phenotypic consequence of genetic variation at the PCSK9 locus and compared findings with recent trials of pharmacological inhibitors of PCSK9. METHODS Published and individual participant level data (300,000+ participants) were combined to construct a weighted PCSK9 gene-centric score (GS). Seventeen randomized placebo controlled PCSK9 inhibitor trials were included, providing data on 79,578 participants. Results were scaled to a one mmol/L lower LDL-C concentration. RESULTS The PCSK9 GS (comprising 4 SNPs) associations with plasma lipid and apolipoprotein levels were consistent in direction with treatment effects. The GS odds ratio (OR) for myocardial infarction (MI) was 0.53 (95% CI 0.42; 0.68), compared to a PCSK9 inhibitor effect of 0.90 (95% CI 0.86; 0.93). For ischemic stroke ORs were 0.84 (95% CI 0.57; 1.22) for the GS, compared to 0.85 (95% CI 0.78; 0.93) in the drug trials. ORs with type 2 diabetes mellitus (T2DM) were 1.29 (95% CI 1.11; 1.50) for the GS, as compared to 1.00 (95% CI 0.96; 1.04) for incident T2DM in PCSK9 inhibitor trials. No genetic associations were observed for cancer, heart failure, atrial fibrillation, chronic obstructive pulmonary disease, or Alzheimer's disease - outcomes for which large-scale trial data were unavailable. CONCLUSIONS Genetic variation at the PCSK9 locus recapitulates the effects of therapeutic inhibition of PCSK9 on major blood lipid fractions and MI. While indicating an increased risk of T2DM, no other possible safety concerns were shown; although precision was moderate.
Collapse
Affiliation(s)
- Amand F Schmidt
- Institute of Cardiovascular Science, University College London, 222 Euston Road, London, NW1 2DA, UK.
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
- UCL's BHF Research Accelerator Centre, London, UK.
| | - Michael V Holmes
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - David Preiss
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel I Swerdlow
- Institute of Cardiovascular Science, University College London, 222 Euston Road, London, NW1 2DA, UK
- Department of Medicine, Imperial College London, London, UK
| | - Spiros Denaxas
- UCL's BHF Research Accelerator Centre, London, UK
- Health Data Research UK, University College London, 222 Euston Road, London, NW1 2DA, UK
- Institute of Health Informatics, University College London, 222 Euston Road, London, NW1 2DA, UK
- The Alan Turing Institute, British Library, 96 Euston Rd, London, NW1 2DB, UK
| | - Ghazaleh Fatemifar
- UCL's BHF Research Accelerator Centre, London, UK
- Health Data Research UK, University College London, 222 Euston Road, London, NW1 2DA, UK
- Institute of Health Informatics, University College London, 222 Euston Road, London, NW1 2DA, UK
| | - Rupert Faraway
- Institute of Cardiovascular Science, University College London, 222 Euston Road, London, NW1 2DA, UK
| | - Chris Finan
- Institute of Cardiovascular Science, University College London, 222 Euston Road, London, NW1 2DA, UK
- UCL's BHF Research Accelerator Centre, London, UK
| | - Dennis Valentine
- UCL's BHF Research Accelerator Centre, London, UK
- University College London, Farr Institute of Health Informatics, London, UK
| | - Zammy Fairhurst-Hunter
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | | | - Bernardo Lessa Horta
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | - Elina Hypponen
- Centre for Population Health Research, Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
- Population, Policy and Practice, UCL GOS Institute of Child Health, London, UK
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Christine Power
- Population, Policy and Practice, UCL GOS Institute of Child Health, London, UK
| | - Max Moldovan
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Erik van Iperen
- Durrer Center for Cardiovascular Research, Netherlands Heart Institute, Utrecht, The Netherlands
- Department of Clinical Epidemiology, Biostatistics And Bioinformatics, Academic Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Kees Hovingh
- Department of vascular medicine, Academic Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Ilja Demuth
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Lipid Clinic at the Interdisciplinary Metabolism Center, Berlin, Germany
- Charité - Universitätsmedizin Berlin, BCRT - Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Kristina Norman
- Institute of Nutritional Science, University of Potsdam, 14558, Nuthetal, Germany
- Geriatrics Research Group, Charité - Universitätsmedizin Berlin, 13347, Berlin, Germany
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558, Nuthetal, Germany
| | - Elisabeth Steinhagen-Thiessen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Lipid Clinic at the Interdisciplinary Metabolism Center, Berlin, Germany
| | | | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany
- Center for Lifespan Changes in Brain and Cognition (LCBC), Dept. Psychology, University of Oslo, Oslo, Norway
| | - Christina M Lill
- Genetic and Molecular Epidemiology Group, Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, Germany
- Institute of Human Genetics, Lübeck, Germany
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College, London, UK
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Johann Willeit
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Stefan Kiechl
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Karin Willeit
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
- Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Dan Mason
- Bradford Institute for Health Research, Bradford Royal Infirmary, Bradford, UK
| | - John Wright
- Bradford Institute for Health Research, Bradford Royal Infirmary, Bradford, UK
| | - Richard Morris
- Department Primary Care & Population Health, University College London, London, UK
| | - Goya Wanamethee
- Department Primary Care & Population Health, University College London, London, UK
| | - Peter Whincup
- Population Health Research Institute, St George's, University of London, London, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Stela McLachlan
- Centre for Population Health Sciences, The Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Jackie F Price
- Centre for Population Health Sciences, The Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, UCL Institute of Epidemiology and Health Care, University College London, London, UK
| | - Catherine Welch
- Department of Epidemiology and Public Health, UCL Institute of Epidemiology and Health Care, University College London, London, UK
| | - Adelaida Sanchez-Galvez
- Department of Epidemiology and Public Health, UCL Institute of Epidemiology and Health Care, University College London, London, UK
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Andrew Nicolaides
- Department of Vascular Surgery, Imperial College, London, United Kingdom
- Department of Surgery, Nicosia Medical School, University of Nicosia, Nicosia, Cyprus
| | - Andrie G Panayiotou
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus
| | - N Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Giuseppe Matullo
- Italian Institute for Genomic Medicine (IIGM), Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Fiorito
- Italian Institute for Genomic Medicine (IIGM), Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Simonetta Guarrera
- Italian Institute for Genomic Medicine (IIGM), Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, UK
| | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, UK
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, UK
| | - Martin Bobak
- Department of Epidemiology and Public Health, UCL Institute of Epidemiology and Health Care, University College London, London, UK
| | - Sofia Malyutina
- Novosibirsk State Medical University, Novosibirsk, Russian Federation
- Institute of Internal and Preventive Medicine, Siberian Branch of the Russian Academy of Medical Sciences, Novosibirsk, Russian Federation
| | - Andrzej Pająk
- Department of Epidemiology and Population Studies, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | | | | | - Hynek Pikhart
- Department of Epidemiology and Public Health, UCL Institute of Epidemiology and Health Care, University College London, London, UK
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan Linneberg
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, The Capital Region of Denmark, Denmark
| | - Tine Jess
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, The Capital Region of Denmark, Denmark
| | - Jackie Cooper
- Centre for Cardiovascular Genetics, Department of Medicine, University College London, London, UK
| | - Steve E Humphries
- Centre for Cardiovascular Genetics, Department of Medicine, University College London, London, UK
| | - Murray Brilliant
- Center for Human Genetics, Marshfield Clinic Research Institute, Marshfield, USA
| | - Terrie Kitchner
- Center for Human Genetics, Marshfield Clinic Research Institute, Marshfield, USA
| | | | - David S Carrell
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | | | | | - Eric B Larson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - David R Crosslin
- Department of Biomedical Informatics and Medical Education University of Washington Seattle, Seattle, WA, USA
| | | | - Dan M Roden
- Department of Medicine, Department of Pharmacology, Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | | - John Attia
- University of Newcastle, Newcastle, NSW, Australia
- Public Health Program, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Elizabeth Holliday
- University of Newcastle, Newcastle, NSW, Australia
- Public Health Program, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Rodney Scott
- University of Newcastle, Newcastle, NSW, Australia
| | - Peter Schofield
- Hunter New England Local Health District, Newcastle, NSW, Australia
| | | | - Salim Yusuf
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Michael Chong
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Guillaume Pare
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Pim van der Harst
- Durrer Center for Cardiovascular Research, Netherlands Heart Institute, Utrecht, The Netherlands
- Department of Clinical Epidemiology, Biostatistics And Bioinformatics, Academic Medical Center Amsterdam, Amsterdam, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M Abdullah Said
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ruben N Eppinga
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Tim Christen
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - D O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stefan Gustafsson
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Raha Pazoki
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Oscar Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Andre Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Sebastian Baumeister
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Chair of Epidemiology, Ludwig-Maximilians-Universität München, UNIKA-T Augsburg, Augsburg, Germany
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Markus M Lerch
- Department of Internal Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, G12 8RZ, Scotland, UK
| | - Jill P Pell
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, G12 8RZ, Scotland, UK
| | - Tom Meade
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Ingrid E Christophersen
- The Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
| | - Anke H Maitland-van der Zee
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Department of Respiratory Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Robin Young
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Archie Campbell
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Michiel L Bots
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus
| | - Diederick E Grobbee
- Cyprus International Institute for Environmental and Public Health, Cyprus University of Technology, Limassol, Cyprus
| | - Philippe Froguel
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000, Lille, France
- Department of Genomics of Common Disease, Imperial College London, W12 0NN, London, United Kingdom
| | - Dorothée Thuillier
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000, Lille, France
| | - Ronan Roussel
- INSERM, U-1138, Centre de Recherche des Cordeliers, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Départment de Diabétologie, Endocrinologie et Nutrition, Assistance Publique Hôpitaux de Paris, Hôpital Bicha, Paris, France
| | - Amélie Bonnefond
- CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, 59000, Lille, France
| | - Bertrand Cariou
- l'institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Melissa Smart
- Institute for Social and Economic Research, University of Essex, Essex, UK
| | - Yanchun Bao
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, England
| | - Meena Kumari
- Boston University School of Medicine, Boston, MA, USA
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, England
| | - Jemma C Hopewell
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | | | - Caroline Dale
- University College London, Farr Institute of Health Informatics, London, UK
| | | | - Paul M Ridker
- Harvard Medical School Center for Cardiovascular Disease Prevention Brigham and Women's Hospital, Boston, USA
| | - Daniel I Chasman
- Harvard Medical School Center for Cardiovascular Disease Prevention Brigham and Women's Hospital, Boston, USA
| | | | | | | | - Alex J Cornish
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Sara E Dobbins
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Kari Hemminki
- Div. Molecular Genetic Epidemiology German Cancer Research Center, Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
- Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Ben Kinnersley
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Marc Sanson
- The Institut du Cerveau et de la Moelle épinière - ICM, Paris, France
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, F-75013, Paris, France
| | - Karim Labreche
- The Institut du Cerveau et de la Moelle épinière - ICM, Paris, France
- Sorbonne Universités, UPMC Université Paris 06, UMR S 1127, F-75013, Paris, France
| | - Matthias Simon
- Department of Neurosurgery, Bethel Clinic, Kantensiek 11, 33617, Bielefeld, Germany
| | - Melissa Bondy
- Division of Hematology-Oncology, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Philip Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Helen Speedy
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - James Allan
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Ni Li
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Molly Went
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Niels Weinhold
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Gareth Morgan
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, 3075 EA, Rotterdam, the Netherlands
| | - Björn Nilsson
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, BMC B13, SE-221 84, Lund, Sweden
| | - Hartmut Goldschmidt
- University Clinic Heidelberg, Internal Medicine V and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Andreas Engert
- Department of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Markus Hansson
- Hematology Clinic, Skåne University Hospital, Skåne, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Harry Hemingway
- UCL's BHF Research Accelerator Centre, London, UK
- Health Data Research UK, University College London, 222 Euston Road, London, NW1 2DA, UK
- Institute of Health Informatics, University College London, 222 Euston Road, London, NW1 2DA, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, 222 Euston Road, London, NW1 2DA, UK
| | - Folkert W Asselbergs
- Institute of Cardiovascular Science, University College London, 222 Euston Road, London, NW1 2DA, UK
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- UCL's BHF Research Accelerator Centre, London, UK
- Health Data Research UK and Institute of Health Informatics, University College London, London, United Kingdom
| | - Riyaz S Patel
- Institute of Cardiovascular Science, University College London, 222 Euston Road, London, NW1 2DA, UK
- UCL's BHF Research Accelerator Centre, London, UK
- The Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | | | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Juan P Casas
- Massachusetts Veterans Epidemiology and Research Information Center (MAVERIC) Veterans Affairs Boston Healthcare System, Boston, USA
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, University College London, 222 Euston Road, London, NW1 2DA, UK
- UCL's BHF Research Accelerator Centre, London, UK
| |
Collapse
|
21
|
McPherson R. 2018 George Lyman Duff Memorial Lecture: Genetics and Genomics of Coronary Artery Disease: A Decade of Progress. Arterioscler Thromb Vasc Biol 2019; 39:1925-1937. [PMID: 31462092 PMCID: PMC6766359 DOI: 10.1161/atvbaha.119.311392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/06/2019] [Indexed: 11/16/2022]
Abstract
Recent studies have led to a broader understanding of the genetic architecture of coronary artery disease and demonstrate that it largely derives from the cumulative effect of multiple common risk alleles individually of small effect size rather than rare variants with large effects on coronary artery disease risk. The tools applied include genome-wide association studies encompassing over 200 000 individuals complemented by bioinformatic approaches including imputation from whole-genome data sets, expression quantitative trait loci analyses, and interrogation of ENCODE (Encyclopedia of DNA Elements), Roadmap Epigenetic Project, and other data sets. Over 160 genome-wide significant loci associated with coronary artery disease risk have been identified using the genome-wide association studies approach, 90% of which are situated in intergenic regions. Here, I will describe, in part, our research over the last decade performed in collaboration with a series of bright trainees and an extensive number of groups and individuals around the world as it applies to our understanding of the genetic basis of this complex disease. These studies include computational approaches to better understand missing heritability and identify causal pathways, experimental approaches, and progress in understanding at the molecular level the function of the multiple risk loci identified and potential applications of these genomic data in clinical medicine and drug discovery.
Collapse
Affiliation(s)
- Ruth McPherson
- From the Division of Cardiology, Atherogenomics Laboratory, Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, ON, Canada
| |
Collapse
|
22
|
Huang F, Wang K, Shen J. Lipoprotein-associated phospholipase A2: The story continues. Med Res Rev 2019; 40:79-134. [PMID: 31140638 PMCID: PMC6973114 DOI: 10.1002/med.21597] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/20/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022]
Abstract
Inflammation is thought to play an important role in the pathogenesis of vascular diseases. Lipoprotein-associated phospholipase A2 (Lp-PLA2) mediates vascular inflammation through the regulation of lipid metabolism in blood, thus, it has been extensively investigated to identify its role in vascular inflammation-related diseases, mainly atherosclerosis. Although darapladib, the most advanced Lp-PLA2 inhibitor, failed to meet the primary endpoints of two large phase III trials in atherosclerosis patients cotreated with standard medical care, the research on Lp-PLA2 has not been terminated. Novel pathogenic, epidemiologic, genetic, and crystallographic studies regarding Lp-PLA2 have been reported recently, while novel inhibitors were identified through a fragment-based lead discovery strategy. More strikingly, recent clinical and preclinical studies revealed that Lp-PLA2 inhibition showed promising therapeutic effects in diabetic macular edema and Alzheimer's disease. In this review, we not only summarized the knowledge of Lp-PLA2 established in the past decades but also emphasized new findings in recent years. We hope this review could be valuable for helping researchers acquire a much deeper insight into the nature of Lp-PLA2, identify more potent and selective Lp-PLA2 inhibitors, and discover the potential indications of Lp-PLA2 inhibitors.
Collapse
Affiliation(s)
- Fubao Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Kai Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Jianhua Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
23
|
Hu Q, Hao P, Liu Q, Dong M, Gong Y, Zhang C, Zhang Y. Mendelian randomization studies on atherosclerotic cardiovascular disease: evidence and limitations. SCIENCE CHINA-LIFE SCIENCES 2019; 62:758-770. [DOI: 10.1007/s11427-019-9537-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/26/2019] [Indexed: 12/26/2022]
|
24
|
Hallén J, Sreeharan N. Development of triglyceride-lowering drugs to address residual cardiovascular risk: strategic and clinical considerations. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2019; 4:237-242. [PMID: 30060063 DOI: 10.1093/ehjcvp/pvy029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 07/26/2018] [Indexed: 12/29/2022]
Abstract
The prevalence of hypertriglyceridaemia is high and growing in several parts of the world. Hypertriglyceridaemia has a well-defined association with the risk of atherosclerotic cardiovascular (CV) disease and triglycerides represent a potential target for drugs aimed at mitigating CV risk. So far, no triglyceride-lowering pharmacological strategy has succeeded in conclusively showing the ability to modify clinical outcomes. This article discusses strategic and clinical aspects of development of triglyceride-lowering drugs to address CV disease.
Collapse
Affiliation(s)
- Jonas Hallén
- Novartis Norge AS, Nydalen Alle 37, Oslo, Norway
| | - Nadarajah Sreeharan
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, James Clerk Maxwell Building, 57 Waterloo Road, London, UK
| |
Collapse
|
25
|
Roberts R. Mendelian Randomization Studies Promise to Shorten the Journey to FDA Approval. JACC Basic Transl Sci 2018; 3:690-703. [PMID: 30456340 PMCID: PMC6234613 DOI: 10.1016/j.jacbts.2018.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022]
Abstract
There has been a dearth of new drugs approved for cardiovascular disorders. The cost is prohibitive, averaging to $2.5 billion, and requiring 12.5 years. This is in large part due to the high failure rate, with only 5% approval by the Food and Drug Administration. Despite preclinical studies showing potential safety and efficacy, most fail when they go to clinical trials phase I to III. One cause for failure is the drug target, often discovered to be a biomarker rather than causative for the disease. Mendelian randomization (MR) studies would determine whether the drug target is causative and could save millions of dollars and time, and prevent unnecessary exposure to adverse drug effects. This was demonstrated in 3 clinical trials that were negative with 2 drugs, veraspladib and darapladib. MR studies during the trials showed the targets of secretory and lipoprotein-associated phospholipids A2 are not causative for coronary artery disease and predicted negative results. The requirement for MR studies is a genetic risk variant with altered function, randomized at conception that remains fixed throughout one’s lifetime. It is not confounded by dietary, lifestyle, or socioeconomic factors. It is more sensitive than randomized controlled trials because exposure to the risk factor is fixed for a lifetime. MR studies showed plasma high-density lipoprotein cholesterol is not a causative target of coronary artery disease, and neither is uric acid, C-reactive protein, and others. MR studies are highly sensitive in determining whether drug targets are causative, and are relatively easy, inexpensive, and not time consuming. It is recommended that drug targets undergo MR studies before proceeding to randomized controlled trials.
Collapse
Affiliation(s)
- Robert Roberts
- Department of Medicine, University of Arizona College of Medicine, Phoenix, Arizona
| |
Collapse
|
26
|
Young EP, Stitziel NO. Capitalizing on Insights from Human Genetics to Identify Novel Therapeutic Targets for Coronary Artery Disease. Annu Rev Med 2018; 70:19-32. [PMID: 30355262 DOI: 10.1146/annurev-med-041717-085853] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Coronary artery disease (CAD) is a major cause of morbidity and mortality. Unfortunately, despite decades of research focused on disease pathogenesis, we still lack a sufficient pharmacopeia for preventing CAD. The failure of many novel cardiovascular drugs to improve clinical outcomes reflects the major substantial challenge of drug development: identifying causal mechanisms that can be therapeutically manipulated to lower disease risk. Identifying genetic variants that are associated with risk of CAD has emerged as a clear path toward improving our understanding of the underlying mechanisms that lead to disease and to the development of new therapies. Here, we review the potential utility and limitations of using human genetics to guide the identification of therapeutic targets for CAD.
Collapse
Affiliation(s)
- Erica P Young
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA;
| | - Nathan O Stitziel
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA; .,Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri 63110, USA.,McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, Missouri 63108, USA;
| |
Collapse
|
27
|
Roberts R. Genetics-Current and Future Role in the Prevention and Management of Coronary Artery Disease. Curr Atheroscler Rep 2017; 18:78. [PMID: 27815829 DOI: 10.1007/s11883-016-0628-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The purpose of this study is to review genetic risk variants for coronary artery disease (CAD) and how they will change the management and prevention of CAD currently and in the future. RECENT FINDINGS Through the efforts of international consortia, 58 genetic risk variants for CAD of genome-wide significance have been replicated in appropriate independent populations. Only one third of these variants mediate their risk through known conventional risk factors for CAD. Thus, unknown mechanisms contribute to CAD. Secondly, the genetic risk is proportional to the total number of risk variants rather than the intensity of any risk factor. Thirdly, the availability of the genetic risk variants enables one to perform Mendelian randomization (MR) studies since they are randomized at conception, not confounded, fixed for life, and can be used to determine if a risk factor is causative or just a marker. MR can also be used to determine the safety and efficacy of a gene product targeted for drug therapy. Genetic risk variants have been shown to successfully risk stratify for CAD in both primary and secondary preventions. Contrary to dogma, MR documents that plasma HDL-C is not protective of CAD. The use of genetic risk score (GRS) for CAD is shown to be more effective in risk stratifying for CAD than the Framingham risk score and independent of the conventional risk factors including family history. Furthermore, the GRS predicts the response to statin therapy in primary and secondary preventions. The use of GRS could represent a paradigm shift in the prevention of CAD.
Collapse
Affiliation(s)
- Robert Roberts
- University of Arizona College of Medicine-Phoenix, 550 East Van Buren, Phoenix, AZ, 85004, USA.
| |
Collapse
|
28
|
Welsh P, Grassia G, Botha S, Sattar N, Maffia P. Targeting inflammation to reduce cardiovascular disease risk: a realistic clinical prospect? Br J Pharmacol 2017; 174:3898-3913. [PMID: 28409825 PMCID: PMC5660005 DOI: 10.1111/bph.13818] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 12/16/2022] Open
Abstract
Data from basic science experiments is overwhelmingly supportive of the causal role of immune-inflammatory response(s) at the core of atherosclerosis, and therefore, the theoretical potential to manipulate the inflammatory response to prevent cardiovascular events. However, extrapolation to humans requires care and we still lack definitive evidence to show that interfering in immune-inflammatory processes may safely lessen clinical atherosclerosis. In this review, we discuss key therapeutic targets in the treatment of vascular inflammation, placing basic research in a wider clinical perspective, as well as identifying outstanding questions. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Paul Welsh
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Gianluca Grassia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Shani Botha
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom campus, South Africa
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Pasquale Maffia
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
29
|
Lacey B, Herrington WG, Preiss D, Lewington S, Armitage J. The Role of Emerging Risk Factors in Cardiovascular Outcomes. Curr Atheroscler Rep 2017; 19:28. [PMID: 28477314 PMCID: PMC5419996 DOI: 10.1007/s11883-017-0661-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW This review discusses the recent evidence for a selection of blood-based emerging risk factors, with particular reference to their relation with coronary heart disease and stroke. RECENT FINDINGS For lipid-related emerging risk factors, recent findings indicate that increasing high-density lipoprotein cholesterol is unlikely to reduce cardiovascular risk, whereas reducing triglyceride-rich lipoproteins and lipoprotein(a) may be beneficial. For inflammatory and hemostatic biomarkers, genetic studies suggest that IL-6 (a pro-inflammatory cytokine) and several coagulation factors are causal for cardiovascular disease, but such studies do not support a causal role for C-reactive protein and fibrinogen. Patients with chronic kidney disease are at high cardiovascular risk with some of this risk not mediated by blood pressure. Randomized evidence (trials or Mendelian) suggests homocysteine and uric acid are unlikely to be key causal mediators of chronic kidney disease-associated risk and sufficiently large trials of interventions which modify mineral bone disease biomarkers are unavailable. Despite not being causally related to cardiovascular disease, there is some evidence that cardiac biomarkers (e.g. troponin) may usefully improve cardiovascular risk scores. Many blood-based factors are strongly associated with cardiovascular risk. Evidence is accumulating, mainly from genetic studies and clinical trials, on which of these associations are causal. Non-causal risk factors may still have value, however, when added to cardiovascular risk scores. Although much of the burden of vascular disease can be explained by 'classic' risk factors (e.g. smoking and blood pressure), studies of blood-based emerging factors have contributed importantly to our understanding of pathophysiological mechanisms of vascular disease, and new targets for potential therapies have been identified.
Collapse
Affiliation(s)
- Ben Lacey
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - William G Herrington
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - David Preiss
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- MRC Population Health Research Unit (MRC PHRU), Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Sarah Lewington
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
- MRC Population Health Research Unit (MRC PHRU), Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK
| | - Jane Armitage
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK.
- MRC Population Health Research Unit (MRC PHRU), Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford, OX3 7LF, UK.
| |
Collapse
|
30
|
Holmes MV, Ala-Korpela M, Smith GD. Mendelian randomization in cardiometabolic disease: challenges in evaluating causality. Nat Rev Cardiol 2017; 14:577-590. [PMID: 28569269 DOI: 10.1038/nrcardio.2017.78] [Citation(s) in RCA: 454] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mendelian randomization (MR) is a burgeoning field that involves the use of genetic variants to assess causal relationships between exposures and outcomes. MR studies can be straightforward; for example, genetic variants within or near the encoding locus that is associated with protein concentrations can help to assess their causal role in disease. However, a more complex relationship between the genetic variants and an exposure can make findings from MR more difficult to interpret. In this Review, we describe some of these challenges in interpreting MR analyses, including those from studies using genetic variants to assess causality of multiple traits (such as branched-chain amino acids and risk of diabetes mellitus); studies describing pleiotropic variants (for example, C-reactive protein and its contribution to coronary heart disease); and those investigating variants that disrupt normal function of an exposure (for example, HDL cholesterol or IL-6 and coronary heart disease). Furthermore, MR studies on variants that encode enzymes responsible for the metabolism of an exposure (such as alcohol) are discussed, in addition to those assessing the effects of variants on time-dependent exposures (extracellular superoxide dismutase), cumulative exposures (LDL cholesterol), and overlapping exposures (triglycerides and non-HDL cholesterol). We elaborate on the molecular features of each relationship, and provide explanations for the likely causal associations. In doing so, we hope to contribute towards more reliable evaluations of MR findings.
Collapse
Affiliation(s)
- Michael V Holmes
- Medical Research Council Population Health Research Unit, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK.,Clinical Trial Service Unit &Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Big Data Institute Building, Old Road Campus, Roosevelt Drive, Oxford OX3 7BN, UK.,National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospital, Old Road, Oxford OX3 7LE, UK.,Medical Research Council Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Mika Ala-Korpela
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, University of Oulu, Aapistie 5A, 90014, Oulu, Finland.,School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| |
Collapse
|
31
|
Khera AV, Kathiresan S. Genetics of coronary artery disease: discovery, biology and clinical translation. Nat Rev Genet 2017; 18:331-344. [PMID: 28286336 PMCID: PMC5935119 DOI: 10.1038/nrg.2016.160] [Citation(s) in RCA: 421] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Coronary artery disease is the leading global cause of mortality. Long recognized to be heritable, recent advances have started to unravel the genetic architecture of the disease. Common variant association studies have linked approximately 60 genetic loci to coronary risk. Large-scale gene sequencing efforts and functional studies have facilitated a better understanding of causal risk factors, elucidated underlying biology and informed the development of new therapeutics. Moving forwards, genetic testing could enable precision medicine approaches by identifying subgroups of patients at increased risk of coronary artery disease or those with a specific driving pathophysiology in whom a therapeutic or preventive approach would be most useful.
Collapse
Affiliation(s)
- Amit V Khera
- Division of Cardiology, Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital; Cardiovascular Disease Initiative, Broad Institute of Harvard and Massachusetts Institute of Technology, 185 Cambridge Street, CPZN 5.252, Boston, Massachusetts 02114, USA
| | - Sekar Kathiresan
- Division of Cardiology, Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital; Cardiovascular Disease Initiative, Broad Institute of Harvard and Massachusetts Institute of Technology, 185 Cambridge Street, CPZN 5.252, Boston, Massachusetts 02114, USA
| |
Collapse
|
32
|
Ma M, Zhai CX, Sun CX. Correlations Between LP-PLA2 Gene Polymorphisms and Susceptibility and Severity of Acute Pancreatitis in a Chinese Population. Genet Test Mol Biomarkers 2017; 21:206-212. [PMID: 28332853 DOI: 10.1089/gtmb.2016.0243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Min Ma
- Department of Emergency, Laiwu City People's Hospital, Laiwu, P.R. China
| | - Chun-Xia Zhai
- Department of Emergency, Laiwu City People's Hospital, Laiwu, P.R. China
| | - Cai-Xia Sun
- Department of Gastrointestinal, Colorectal and Anal Surgery, China–Japan Union Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
33
|
Finan C, Gaulton A, Kruger FA, Lumbers RT, Shah T, Engmann J, Galver L, Kelley R, Karlsson A, Santos R, Overington JP, Hingorani AD, Casas JP. The druggable genome and support for target identification and validation in drug development. Sci Transl Med 2017; 9:eaag1166. [PMID: 28356508 PMCID: PMC6321762 DOI: 10.1126/scitranslmed.aag1166] [Citation(s) in RCA: 483] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 01/27/2017] [Indexed: 12/11/2022]
Abstract
Target identification (determining the correct drug targets for a disease) and target validation (demonstrating an effect of target perturbation on disease biomarkers and disease end points) are important steps in drug development. Clinically relevant associations of variants in genes encoding drug targets model the effect of modifying the same targets pharmacologically. To delineate drug development (including repurposing) opportunities arising from this paradigm, we connected complex disease- and biomarker-associated loci from genome-wide association studies to an updated set of genes encoding druggable human proteins, to agents with bioactivity against these targets, and, where there were licensed drugs, to clinical indications. We used this set of genes to inform the design of a new genotyping array, which will enable association studies of druggable genes for drug target selection and validation in human disease.
Collapse
Affiliation(s)
- Chris Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Anna Gaulton
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K
| | - Felix A Kruger
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- BenevolentAI, 40 Churchway, London, U.K
| | - R Thomas Lumbers
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Tina Shah
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Jorgen Engmann
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Luana Galver
- Illumina Inc., 5200 Illumina Way, San Diego, CA 92122, USA
| | - Ryan Kelley
- Illumina Inc., 5200 Illumina Way, San Diego, CA 92122, USA
| | - Anneli Karlsson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K
| | - Rita Santos
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K
| | - John P Overington
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K.
- BenevolentAI, 40 Churchway, London, U.K
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K.
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Juan P Casas
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K.
| |
Collapse
|
34
|
Zhang R, Song Q, Liu H, Bai H, Zhang Y, Liu Q, Guan L, Fan P. Effect of the R92H and A379V genotypes of platelet-activating factor acetylhydrolase on its enzyme activity, oxidative stress and metabolic profile in Chinese women with polycystic ovary syndrome. Lipids Health Dis 2017; 16:57. [PMID: 28320416 PMCID: PMC5359970 DOI: 10.1186/s12944-017-0448-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/09/2017] [Indexed: 02/15/2023] Open
Abstract
Background The G994T polymorphism in platelet-activating factor acetylhydrolase (PAF-AH) gene is associated with the risk of polycystic ovary syndrome (PCOS). The aim of this study was to investigate the relationship between R92H and A379V variants of the PAF-AH gene and the risk of PCOS and to evaluate the effects of the genotypes on PAF-AH activities and clinical, metabolic and oxidative stress indexes in Chinese women. Methods A total of 862 patients with PCOS based on the Rotterdam consensus criteria and 750 control women from a population of Chinese Han nationality in the Chengdu area were studied from 2006–2015. PAF-AH genotypes were determined by PCR and restriction fragment length polymorphism analysis. Plasma PAF-AH, high-density lipoprotein (HDL)-associated PAF-AH (H-PAF-AH) and apolipoprotein (apo) B-containing lipoprotein-associated PAF-AH (apoB-PAF-AH) activities were measured using the trichloroacetic acid precipitation procedure with PAF C-16 as a substrate. Circulating markers of oxidative stress, including serum total oxidant status, total antioxidant capacity, oxidative stress index and malondialdehyde levels, and clinical and metabolic parameters were also analyzed. Results No significant differences were observed in the frequencies of R92H and A379V genotypes and alleles of the PAF-AH gene between PCOS and control groups (P > 0.05). Compared with patients with the 92RR genotype, patients with H allele of R92H (RH + HH genotype) had significantly higher plasma PAF-AH and apoB-PAF-AH activities (P < 0.05) and tended to exhibit increased H-PAF-AH activity (P = 0.063) after adjusted for age and BMI. However, when serum LDL-C, HDL-C, TG and HOMA index were added as covariates, the comparisons no longer remained statistical significance (P > 0.05). There were no significant differences in clinical, hormonal, metabolic and circulating oxidative stress parameters and the frequencies of PAF-AH G449T genotype according to PAF-AH R92H or A379V genotyping in patients with PCOS and control women. Conclusions There were no significant associations between R92H and A379V variants of PAF-AH gene and risk of PCOS in Chinese women. The increased plasma PAF-AH and apoB-PAF-AH activities in patients with H allele of R92H are related to the R92 → H variation, changes in plasma lipoprotein levels, insulin resistance, aging, and gaining weight and thus may be involved in the pathogenesis of PCOS and the increased risks of future cardiovascular diseases.
Collapse
Affiliation(s)
- Renjiao Zhang
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qi Song
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Hongwei Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yujin Zhang
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingqing Liu
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Linbo Guan
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
35
|
Gregson JM, Freitag DF, Surendran P, Stitziel NO, Chowdhury R, Burgess S, Kaptoge S, Gao P, Staley JR, Willeit P, Nielsen SF, Caslake M, Trompet S, Polfus LM, Kuulasmaa K, Kontto J, Perola M, Blankenberg S, Veronesi G, Gianfagna F, Männistö S, Kimura A, Lin H, Reilly DF, Gorski M, Mijatovic V, Munroe PB, Ehret GB, Thompson A, Uria-Nickelsen M, Malarstig A, Dehghan A, Vogt TF, Sasaoka T, Takeuchi F, Kato N, Yamada Y, Kee F, Müller-Nurasyid M, Ferrières J, Arveiler D, Amouyel P, Salomaa V, Boerwinkle E, Thompson SG, Ford I, Wouter Jukema J, Sattar N, Packard CJ, Shafi Majumder AA, Alam DS, Deloukas P, Schunkert H, Samani NJ, Kathiresan S, Nordestgaard BG, Saleheen D, Howson JMM, Di Angelantonio E, Butterworth AS, Danesh J. Genetic invalidation of Lp-PLA 2 as a therapeutic target: Large-scale study of five functional Lp-PLA 2-lowering alleles. Eur J Prev Cardiol 2017; 24:492-504. [PMID: 27940953 PMCID: PMC5460752 DOI: 10.1177/2047487316682186] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/24/2016] [Indexed: 01/12/2023]
Abstract
Aims Darapladib, a potent inhibitor of lipoprotein-associated phospholipase A2 (Lp-PLA2), has not reduced risk of cardiovascular disease outcomes in recent randomized trials. We aimed to test whether Lp-PLA2 enzyme activity is causally relevant to coronary heart disease. Methods In 72,657 patients with coronary heart disease and 110,218 controls in 23 epidemiological studies, we genotyped five functional variants: four rare loss-of-function mutations (c.109+2T > C (rs142974898), Arg82His (rs144983904), Val279Phe (rs76863441), Gln287Ter (rs140020965)) and one common modest-impact variant (Val379Ala (rs1051931)) in PLA2G7, the gene encoding Lp-PLA2. We supplemented de-novo genotyping with information on a further 45,823 coronary heart disease patients and 88,680 controls in publicly available databases and other previous studies. We conducted a systematic review of randomized trials to compare effects of darapladib treatment on soluble Lp-PLA2 activity, conventional cardiovascular risk factors, and coronary heart disease risk with corresponding effects of Lp-PLA2-lowering alleles. Results Lp-PLA2 activity was decreased by 64% ( p = 2.4 × 10-25) with carriage of any of the four loss-of-function variants, by 45% ( p < 10-300) for every allele inherited at Val279Phe, and by 2.7% ( p = 1.9 × 10-12) for every allele inherited at Val379Ala. Darapladib 160 mg once-daily reduced Lp-PLA2 activity by 65% ( p < 10-300). Causal risk ratios for coronary heart disease per 65% lower Lp-PLA2 activity were: 0.95 (0.88-1.03) with Val279Phe; 0.92 (0.74-1.16) with carriage of any loss-of-function variant; 1.01 (0.68-1.51) with Val379Ala; and 0.95 (0.89-1.02) with darapladib treatment. Conclusions In a large-scale human genetic study, none of a series of Lp-PLA2-lowering alleles was related to coronary heart disease risk, suggesting that Lp-PLA2 is unlikely to be a causal risk factor.
Collapse
Affiliation(s)
- John M Gregson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Daniel F Freitag
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Praveen Surendran
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Nathan O Stitziel
- Departments of Medicine and Genetics, Washington University School of Medicine, St Louis, USA
| | - Rajiv Chowdhury
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Stephen Burgess
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Stephen Kaptoge
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Pei Gao
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - James R Staley
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Peter Willeit
- Department of Public Health and Primary Care, University of Cambridge, UK
- Department of Neurology, Innsbruck Medical University, Austria
| | - Sune F Nielsen
- Copenhagen University Hospital, University of Copenhagen, Denmark
| | | | | | | | - Kari Kuulasmaa
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Jukka Kontto
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Markus Perola
- Institute of Molecular Medicine FIMM, University of Helsinki, Finland
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart Centre Hamburg, Germany
- University Medical Centre Hamburg Eppendorf, Hamburg, Germany
| | - Giovanni Veronesi
- Research Centre, Department of Clinical and Experimental Medicine, University of Insubria, Varese, Italy
| | - Francesco Gianfagna
- Research Centre, Department of Clinical and Experimental Medicine, University of Insubria, Varese, Italy
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Satu Männistö
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Honghuang Lin
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, USA
- The NHLBI’s Framingham Heart Study, Framingham, USA
| | - Dermot F Reilly
- Merck Research Laboratories, Genetics and Pharmacogenomics, Boston, USA
| | - Mathias Gorski
- Department of Genetic Epidemiology, University of Regensburg, Germany
- Department of Nephrology, University Hospital Regensburg, Germany
| | - Vladan Mijatovic
- Department of Life and Reproduction Sciences, University of Verona, Italy
| | | | - Patricia B Munroe
- Clinical Pharmacology and The Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
- NIHR Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, UK
| | - Georg B Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- Cardiology, Department of Medicine, Geneva University Hospital, Switzerland
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Thomas F Vogt
- Merck Research Laboratories, Cardiometabolic Disease, Kenilworth, USA
- CHDI Management/CHDI Foundation, Princeton, USA
| | - Taishi Sasaoka
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Yoshiji Yamada
- Department of Human Functional Genomics, Life Science Research Centre, Mie University, Japan
| | - Frank Kee
- UKCRC Centre of Excellence for Public Health, Queens University, Belfast, Ireland
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Department of Medicine I, Ludwig-Maximilians-University Munich, Germany
| | - Jean Ferrières
- Department of Epidemiology, UMR 1027-INSERM, Toulouse University-CHU Toulouse, France
| | - Dominique Arveiler
- Department of Epidemiology and Public Health, EA 3430, University of Strasbourg and Strasbourg University Hospital, France
| | - Philippe Amouyel
- Department of Epidemiology and Public Health, Institut Pasteur de Lille, France
| | - Veikko Salomaa
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, USA
| | - Simon G Thompson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | | | | | | | | | | | - Dewan S Alam
- Centre for Global Health Research, St Michael Hospital, Toronto, Canada
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Heribert Schunkert
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Deutsches Herzzentrum München, Technische Universität München, Germany
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, UK
| | - Sekar Kathiresan
- Broad Institute, Cambridge and Massachusetts General Hospital, Boston, USA
| | | | | | | | - Joanna MM Howson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Emanuele Di Angelantonio
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Adam S Butterworth
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - John Danesh
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- British Heart Foundation Cambridge Centre of Excellence, University of Cambridge, Cambridge, UK
- National Institute of Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| |
Collapse
|
36
|
A previously unreported impact of a PLA2G7 gene polymorphism on the plasma levels of lipoprotein-associated phospholipase A2 activity and mass. Sci Rep 2016; 6:37465. [PMID: 27905470 PMCID: PMC5131362 DOI: 10.1038/srep37465] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 10/27/2016] [Indexed: 12/30/2022] Open
Abstract
Lipoprotein-associated phospholipase A2 (Lp-PLA2) levels are associated with the development of atherosclerosis. We aimed to assess the genetic determinants of Lp-PLA2 activity and mass by genotyping multiple polymorphisms in PLA2G7, the gene encoding Lp-PLA2, among 1258 participants from the Chinese Multi-provincial Cohort Study-Beijing Project. The Sequenom MassARRAY system, Taqman assay and direct sequencing were adopted. For the first time, the rs13218408 polymorphism was found to be significantly associated with reduced Lp-PLA2 levels. We also confirmed the significant association of previously validated polymorphisms (rs1421378, rs1805018, rs16874954 and rs2216465), even after adjusting for traditional cardiovascular risk factors and for Bonferroni correction. Percentages of variance attributable to rs13218408 were 7.2% for activity and 13.3% for mass, and were secondary to those of rs16874954 (8.1% for activity and 16.9% for mass). A significant joint effect of rs13218408 and rs16874954 was observed on Lp-PLA2 activity (P = 0.058) and mass (P = 0.003), with their minor alleles together linking to the largest reduction in Lp-PLA2 levels (37.8% reduction in activity and 41.6% reduction in mass). Taken together, our findings show a significant association of a PLA2G7 polymorphism with Lp-PLA2 levels, which was previously unreported in any population. The functionality of this genetic variation deserves further investigations.
Collapse
|
37
|
Garg PK, Jorgensen NW, McClelland RL, Jenny NS, Criqui MH, Allison MA, Greenland P, Rosenson RS, Siscovick DS, Cushman M. Lipoprotein-associated phospholipase A 2 and risk of incident peripheral arterial disease in a multi-ethnic cohort: The Multi-Ethnic Study of Atherosclerosis. Vasc Med 2016; 22:5-12. [PMID: 28215109 DOI: 10.1177/1358863x16671424] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Prospective studies supporting a relationship between elevated lipoprotein-associated phospholipase A2 (Lp-PLA2) and incident peripheral arterial disease (PAD) are limited. We evaluated the association of Lp-PLA2 with incident PAD in a multi-ethnic cohort without clinical cardiovascular disease. A total of 4622 participants with measurement of Lp-PLA2 mass and Lp-PLA2 activity and an ankle-brachial index (ABI) between 0.9 and 1.4 were followed for the development of PAD (median follow-up = 9.3 years), defined as an ABI ⩽0.9 and decline from baseline ⩾0.15. There were 158 incident PAD events during follow-up. In adjusted logistic regression models, each higher standard deviation of both Lp-PLA2 activity and mass did not confer an increased risk of developing PAD [odds ratios, (95% confidence intervals)]: 0.92 (0.66-1.27) for Lp-PLA2 activity and 1.06 (0.85-1.34) for mass. Additionally, no significant interaction was found according to ethnicity: p=0.43 for Lp-PLA2 activity and p=0.55 for Lp-PLA2 mass. We found no evidence of an association between Lp-PLA2 and incident PAD.
Collapse
Affiliation(s)
- Parveen K Garg
- 1 Division of Cardiovascular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Neal W Jorgensen
- 2 Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Robyn L McClelland
- 2 Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Nancy S Jenny
- 3 Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Michael H Criqui
- 4 Department of Family Medicine & Public Health, University of California in San Diego, La Jolla, CA, USA
| | - Matthew A Allison
- 4 Department of Family Medicine & Public Health, University of California in San Diego, La Jolla, CA, USA
| | - Philip Greenland
- 5 Department of Medicine, Northwestern University, Chicago, IL, USA.,6 Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Robert S Rosenson
- 7 Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Mary Cushman
- 3 Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA.,9 Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| |
Collapse
|
38
|
Stitziel NO, Kathiresan S. Leveraging human genetics to guide drug target discovery. Trends Cardiovasc Med 2016; 27:352-359. [PMID: 27686272 DOI: 10.1016/j.tcm.2016.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 01/11/2023]
Abstract
Identifying appropriate molecular targets is a critical step in drug development. Despite many advantages, the traditional tools of observational epidemiology and cellular or animal models of disease can be misleading in identifying causal pathways likely to lead to successful therapeutics. Here, we review some favorable aspects of human genetics studies that have the potential to accelerate drug target discovery. These include using genetic studies to identify pathways relevant to human disease, leveraging human genetics to discern causal relationships between biomarkers and disease, and studying genetic variation in humans to predict the potential efficacy and safety of inhibitory compounds aimed at molecular targets. We present some examples taken from studies of plasma lipids and coronary artery disease to highlight how human genetics can accelerate therapeutics development.
Collapse
Affiliation(s)
- Nathan O Stitziel
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, Saint Louis, MO; Department of Genetics, Washington University School of Medicine, Saint Louis, MO; McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO.
| | - Sekar Kathiresan
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA; Department of Medicine, Harvard Medical School, Boston, MA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA.
| |
Collapse
|
39
|
Estrogen and promoter methylation in the regulation of PLA2G7 transcription. Gene 2016; 591:262-267. [PMID: 27450918 DOI: 10.1016/j.gene.2016.07.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 06/30/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022]
Abstract
In the current study, cell lines including HEK293, SW480, HPASMC, HPCASMC and HAEC were cultured with 5-aza-2-deoxycytidine (DAC) and 17-β-estradiol to investigate whether PLA2G7 transcription was under the control of promoter methylation and 17-β-estradiol. Luciferase reporter gene assays were used to evaluate whether reporter gene activity was enhanced by PLA2G7 promoter fragment. Gene expression and methylation were detected using RT-PCR and pyrosequencing methods, respectively. Endogenous PLA2G7 transcription levels were found to be significantly lower in vascular related cell lines than in the other cell lines. Luciferase reporter gene assays indicated that gene activity was significantly enhanced by PLA2G7 promoter fragment. PLA2G7 transcription was found to be up-regulated with the treatment of DAC. The 17-β-estradiol was found to down-regulate PLA2G7 transcription in all the cell lines. However, 17-β-estradiol did not have significant effect on PLA2G7 methylation. Further chromatin immunoprecipitation assay showed that 17-β-estradiol might regulate gene transcription by affecting the acetylated histone H3 and H4 marks on PLA2G7 promoter. Our results showed that PLA2G7 gene expression was co-regulated by 17-β-estradiol and promoter methylation. Our findings might provide molecular clues for gender disparity in the contribution of PLA2G7 to vascular related diseases such as coronary heart disease.
Collapse
|
40
|
Abstract
Genetic factors contribute importantly to the risk of coronary artery disease (CAD), and in the past decade, there has been major progress in this area. The tools applied include genome-wide association studies encompassing >200,000 individuals complemented by bioinformatic approaches, including 1000 Genomes imputation, expression quantitative trait locus analyses, and interrogation of Encyclopedia of DNA Elements, Roadmap, and other data sets. close to 60 common SNPs (minor allele frequency>0.05) associated with CAD risk and reaching genome-wide significance (P<5 × 10(-8)) have been identified. Furthermore, a total of 202 independent signals in 109 loci have achieved a false discovery rate (q<0.05) and together explain 28% of the estimated heritability of CAD. These data have been used successfully to create genetic risk scores that can improve risk prediction beyond conventional risk factors and identify those individuals who will benefit most from statin therapy. Such information also has important applications in clinical medicine and drug discovery by using a Mendelian randomization approach to interrogate the causal nature of many factors found to associate with CAD risk in epidemiological studies. In contrast to genome-wide association studies, whole-exome sequencing has provided valuable information directly relevant to genes with known roles in plasma lipoprotein metabolism but has, thus far, failed to identify other rare coding variants linked to CAD. Overall, recent studies have led to a broader understanding of the genetic architecture of CAD and demonstrate that it largely derives from the cumulative effect of multiple common risk alleles individually of small effect size rather than rare variants with large effects on CAD risk. Despite this success, there has been limited progress in understanding the function of the novel loci; the majority of which are in noncoding regions of the genome.
Collapse
Affiliation(s)
- Ruth McPherson
- From the Department of Medicine, Atherogenomics Laboratory, Division of Cardiology, Ruddy Canadian Cardiovascular Genetics Center, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.M.); and Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.T.-H.).
| | - Anne Tybjaerg-Hansen
- From the Department of Medicine, Atherogenomics Laboratory, Division of Cardiology, Ruddy Canadian Cardiovascular Genetics Center, University of Ottawa Heart Institute, Ottawa, Ontario, Canada (R.M.); and Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.T.-H.)
| |
Collapse
|
41
|
Millwood IY, Bennett DA, Walters RG, Clarke R, Waterworth D, Johnson T, Chen Y, Yang L, Guo Y, Bian Z, Hacker A, Yeo A, Parish S, Hill MR, Chissoe S, Peto R, Cardon L, Collins R, Li L, Chen Z. A phenome-wide association study of a lipoprotein-associated phospholipase A2 loss-of-function variant in 90 000 Chinese adults. Int J Epidemiol 2016; 45:1588-1599. [PMID: 27301456 PMCID: PMC5100610 DOI: 10.1093/ije/dyw087] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2016] [Indexed: 12/20/2022] Open
Abstract
Background: Lipoprotein-associated phospholipase A2 (Lp-PLA2) has been implicated in development of atherosclerosis; however, recent randomized trials of Lp-PLA2 inhibition reported no beneficial effects on vascular diseases. In East Asians, a loss-of-function variant in the PLA2G7 gene can be used to assess the effects of genetically determined lower Lp-PLA2. Methods:PLA2G7 V279F (rs76863441) was genotyped in 91 428 individuals randomly selected from the China Kadoorie Biobank of 0.5 M participants recruited in 2004–08 from 10 regions of China, with 7 years’ follow-up. Linear regression was used to assess effects of V279F on baseline traits. Logistic regression was conducted for a range of vascular and non-vascular diseases, including 41 ICD-10 coded disease categories. Results:PLA2G7 V279F frequency was 5% overall (range 3–7% by region), and 9691 (11%) participants had at least one loss-of-function variant. V279F was not associated with baseline blood pressure, adiposity, blood glucose or lung function. V279F was not associated with major vascular events [7141 events; odds ratio (OR) = 0.98 per F variant, 95% confidence interval (CI) 0.90-1.06] or other vascular outcomes, including major coronary events (922 events; 0.96, 0.79-1.18) and stroke (5967 events; 1.00, 0.92-1.09). Individuals with V279F had lower risks of diabetes (7031 events; 0.91, 0.84-0.98) and asthma (182 events; 0.53, 0.28-0.98), but there was no association after adjustment for multiple testing. Conclusions: Lifelong lower Lp-PLA2 activity was not associated with major risks of vascular or non-vascular diseases in Chinese adults. Using functional genetic variants in large-scale prospective studies with linkage to a range of health outcomes is a valuable approach to inform drug development and repositioning.
Collapse
Affiliation(s)
- Iona Y Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Derrick A Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Robert Clarke
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Dawn Waterworth
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Toby Johnson
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Yu Guo
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China and
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China and
| | - Alex Hacker
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Astrid Yeo
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Sarah Parish
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Michael R Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Stephanie Chissoe
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Richard Peto
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Lon Cardon
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Rory Collins
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Liming Li
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China and.,Department of Epidemiology & Biostatistics, Peking University Health Science Centre, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | | |
Collapse
|
42
|
Haghdoost F, Gharzi M, Faez F, Hosseinzadeh E, Tajaddini M, Rafiei L, Asgari F, Banihashemi M, Masjedi SS, Zandifar A, Haghjooy-Javanmard S. Association between Ala379Val polymorphism of lipoprotein-associated phospholipase A2 and migraine without aura in Iranian population. IRANIAN JOURNAL OF NEUROLOGY 2016; 15:80-4. [PMID: 27326362 PMCID: PMC4912673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND Migraine is a common neurovascular disorder with multifactorial and polygenic inheritance. The aim of this study was to investigate the association of a migraine without aura and Ala379Val polymorphism of lipoprotein-associated phospholipase A2 (Lp-PLA2) gene in the Iranian population. METHODS In this study, 103 migraine patients and 100 healthy controls were enrolled. DNA samples were extracted and the Ala379Val polymorphism of Lp-PLA2 gene was investigated. To assess severity of a headache, patients filled out the headache impact test (HIT-6) and migraine severity (MIGSEV) questionnaires. RESULTS Allele V had significantly lower frequency in the case group than control subjects [P = 0.001, odds ratio (OR) = 0.25, confidence interval (CI): 0.15-0.40]. The frequency of migraine patients that were a carrier of V allele (V/V and A/V) was statistically significant lower than the control group (P = 0.003, OR = 2.39, CI: 1.35-4.23). There was no significant difference of alleles frequency between three grades of MIGSEV (P = 0.316). Furthermore, total HIT-6 score was not significantly different between different genotypes (P = 0.466). CONCLUSION Our results showed that Ala379Val gene polymorphism of LP-PLA2 is associated with lower risk of migraine but not with severity of headaches in an Iranian population.
Collapse
Affiliation(s)
- Faraidoon Haghdoost
- Medical Student Research Center AND Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Gharzi
- Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farough Faez
- Pharmacy Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elinaz Hosseinzadeh
- Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Laleh Rafiei
- Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Asgari
- Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahboobeh Banihashemi
- Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Samaneh Sadat Masjedi
- Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Zandifar
- Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
43
|
Chen X, Wang K, Xu W, Ma Q, Chen M, Du L, Mo M, Wang Y, Shen J. Discovery of Potent and Orally Active Lipoprotein-Associated Phospholipase A2 (Lp-PLA2) Inhibitors as a Potential Therapy for Diabetic Macular Edema. J Med Chem 2016; 59:2674-87. [PMID: 26927682 DOI: 10.1021/acs.jmedchem.5b01930] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lipoprotein-associated phospholipase A2 (Lp-PLA2) is considered to be a promising therapeutic target for several inflammation-associated diseases. Herein, we describe the discovery of a series of pyrimidone derivatives as Lp-PLA2 inhibitors. Systematic structural modifications led to the identification of several pyrimidone compounds with promising in vitro inhibitory potency and pharmacokinetic properties. Compound 14c, selected for in vivo evaluation, demonstrated decent pharmacokinetic profiles and robust inhibitory potency against Lp-PLA2 in Sprague-Dawley (SD) rats. Furthermore, 14c significantly inhibited retinal thickening in STZ-induced diabetic SD rats as a model of diabetic macular edema (DME) after oral dosing for 4 weeks. Taken together, these results suggested that 14c can serve as a valuable lead in the search for new Lp-PLA2 inhibitors for prevention and/or treatment of DME.
Collapse
Affiliation(s)
- Xinde Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Kai Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Wenwei Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Quanxin Ma
- Laboratory Animal Research Center, Zhejiang Chinese Medical University , Hangzhou 310053, China
| | - Minli Chen
- Laboratory Animal Research Center, Zhejiang Chinese Medical University , Hangzhou 310053, China
| | - Lili Du
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Mingguang Mo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yiping Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Jianhua Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| |
Collapse
|
44
|
Centurión OA. Serum biomarkers and source of inflammation in acute coronary syndromes and percutaneous coronary interventions. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2016; 17:119-28. [DOI: 10.1016/j.carrev.2016.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 01/05/2016] [Accepted: 01/13/2016] [Indexed: 11/17/2022]
|
45
|
Mendelian Randomization for the Identification of Causal Pathways in Atherosclerotic Vascular Disease. Cardiovasc Drugs Ther 2016; 30:41-9. [DOI: 10.1007/s10557-016-6640-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
46
|
Waters DD, Chau KHY. Tribulations of Recent Cardiology Trials, the Audacity of Hope, and HOPE-3. Can J Cardiol 2015; 32:275-7. [PMID: 26577896 DOI: 10.1016/j.cjca.2015.08.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 08/19/2015] [Accepted: 08/19/2015] [Indexed: 01/23/2023] Open
Affiliation(s)
- David D Waters
- Division of Cardiology, San Francisco General Hospital, and the Department of Medicine, University of California, San Francisco, San Francisco, California, USA.
| | - Katherine Hsin-Yu Chau
- Division of Cardiology, San Francisco General Hospital, and the Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
47
|
Orho-Melander M. Genetics of coronary heart disease: towards causal mechanisms, novel drug targets and more personalized prevention. J Intern Med 2015; 278:433-46. [PMID: 26477595 DOI: 10.1111/joim.12407] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Coronary heart disease (CHD) is an archetypical multifactorial disorder that is influenced by genetic susceptibility as well as both modifiable and nonmodifiable risk factors, and their interactions. Advances during recent years in the field of multifactorial genetics, in particular genomewide association studies (GWASs) and their meta-analyses, have provided the statistical power to identify and replicate genetic variants in more than 50 risk loci for CHD and in several hundreds of loci for cardiometabolic risk factors for CHD such as blood lipids and lipoproteins. Although for a great majority of these loci both the causal variants and mechanisms remain unknown, progress in identifying the causal variants and underlying mechanisms has already been made for several genetic loci. Furthermore, identification of rare loss-of-function variants in genes such as PCSK9, NPC1L1, APOC3 and APOA5, which cause a markedly decreased risk of CHD and no adverse side effects, illustrates the power of translating genetic findings into novel mechanistic information and provides some optimism for the future of developing novel drugs, given the many genes associated with CHD in GWASs. Finally, Mendelian randomization can be used to reveal or exclude causal relationships between heritable biomarkers and CHD, and such approaches have already provided evidence of causal relationships between CHD and LDL cholesterol, triglycerides/remnant particles and lipoprotein(a), and indicated a lack of causality for HDL cholesterol, C-reactive protein and lipoprotein-associated phospholipase A2. Together, these genetic findings are beginning to lead to promising new drug targets and novel interventional strategies and thus have great potential to improve prevention, prediction and therapy of CHD.
Collapse
Affiliation(s)
- M Orho-Melander
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden
| |
Collapse
|
48
|
Maiolino G, Bisogni V, Rossitto G, Rossi GP. Lipoprotein-associated phospholipase A2 prognostic role in atherosclerotic complications. World J Cardiol 2015; 7:609-620. [PMID: 26516415 PMCID: PMC4620072 DOI: 10.4330/wjc.v7.i10.609] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/11/2015] [Accepted: 09/28/2015] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis manifests itself clinically at advanced stages when plaques undergo hemorrhage and/or rupture with superimposed thrombosis, thus abruptly stopping blood supply. Identification of markers of plaque destabilization at a pre-clinical stage is, therefore, a major goal of cardiovascular research. Promising results along this line were provided by studies investigating the lipoprotein-associated phospholipase A2 (Lp-PLA2), a member of phospholipase A2 proteins family that plays a key role in the metabolism of pro-inflammatory phospholipids, as oxidized low-density lipoproteins, and in the generation of pro-atherogenic metabolites, including lysophosphatidylcholine and oxidized free fatty acids. We herein review the experimental and clinical studies supporting use of Lp-PLA2 activity for predicting cardiovascular events. To his end we considered not only Lp-PLA2 activity and mass, but also Lp-PLA2 gene variations and their association with incident coronary artery disease, stroke, and cardiovascular mortality. Based on these evidences the major scientific societies have included in their guidelines the measurement of Lp-PLA2 activity among the biomarkers that are useful in risk stratification of adult asymptomatic patients at intermediate cardiovascular risk. The results of two recently published major clinical trials with the Lp-PLA2 inhibitor darapladib, which seem to challenge the pathogenic role of Lp-PLA2, will also be discussed.
Collapse
|
49
|
[Lp-PLA2, a biomarker of vascular inflammation and vulnerability of atherosclerosis plaques]. ANNALES PHARMACEUTIQUES FRANÇAISES 2015; 74:190-7. [PMID: 26499399 DOI: 10.1016/j.pharma.2015.09.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 09/09/2015] [Indexed: 01/21/2023]
Abstract
A chronic inflammation is involved in various stages of development of the atherosclerotic plaques. Among the emerging biomarkers of atherogenesis, the lipoprotein-associated phospholipase A2 (Lp-PLA2), formerly known as PAF-acetylhydrolase (McIntyre et al., 2009), hydrolyses the oxidized short chain phospholipids of low-density lipoproteins (LDL), thereby releasing pro-inflammatory mediators (lysophospholipids and oxidized fatty acids). Lp-PLA2, produced by monocytes/macrophages and T-lymphocytes, and mainly associated with LDL (Gazi et al., 2005), is predominantly expressed in the necrotic center of the atherosclerotic plaques and in the macrophage-rich areas (Kolodgie et al., 2006). It would have a predictive role of cardiovascular (CV) events in relation to the vulnerability of atherosclerotic plaques. Determination of Lp-PLA2 has been proposed in the assessment of the CV risk, to ensure a better stratification of populations at intermediate risk for targeted therapy (Davidson et al., 2008). Its proatherogenic role suggested that inhibition of its activity could ensure a better vascular protection in combination with cholesterol-lowering agents. Nevertheless, Lp-PLA2 is not yet a fully validated marker for use in daily clinical practice, especially since the studies using an inhibitor of Lp-PLA2 (darapladib) (STABILITY Investigators et al., 2014; O'Donoghue et al., 2014) did not show any reduction in coronary events. Lp-PLA2 could have a site-specific role in plaque inflammation and development (Fenning et al., 2015). High Lp-PLA2 activity could reflect a response to pro-inflammatory stress characteristic of atherosclerosis (Marathe et al., 2014). This presentation aims at clarifying the involvement of Lp-PLA2 in the pathophysiology of atherosclerosis, and at assessing its interest both as a biomarker for the onset of CV events and as a therapeutic target.
Collapse
|
50
|
Talmud PJ, Holmes MV. Deciphering the Causal Role of sPLA2s and Lp-PLA2 in Coronary Heart Disease. Arterioscler Thromb Vasc Biol 2015; 35:2281-9. [PMID: 26338298 DOI: 10.1161/atvbaha.115.305234] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/19/2015] [Indexed: 11/16/2022]
Abstract
Over the last 10 to 15 years, animal and human observational studies have identified elevated levels of both proinflammatory secretory phospholipase A2-IIA and lipoprotein-associated phospholipase A2 as potential risk factors for coronary heart disease. However, Mendelian randomization, a genetic tool to test causality of a biomarker, and phase III randomized controlled trials of inhibitors of theses enzymes (varespladib and darapladib) converged to indicate that elevated levels are unlikely to be themselves causal of coronary heart disease and that inhibition had little or no clinical utility. The concordance of findings from Mendelian randomization and clinical trials suggests that for these 2 drugs, and for other novel biomarkers in future, validation of potential therapeutic targets by genetic studies (such as Mendelian randomization) before embarking on costly phase III randomized controlled trials could increase efficiency and offset the high risk of drug development, thereby facilitating discovery of new therapeutics and mitigating against the exuberant costs of drug development.
Collapse
Affiliation(s)
- Philippa J Talmud
- From the Center for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK (P.J.T.); and Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK (M.V.H.).
| | - Michael V Holmes
- From the Center for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London, London, UK (P.J.T.); and Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK (M.V.H.)
| |
Collapse
|