1
|
Surendran A, Zhang H, Stamenkovic A, Ravandi A. Lipidomics and cardiovascular disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167806. [PMID: 40122185 DOI: 10.1016/j.bbadis.2025.167806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of mortality worldwide, necessitating innovative approaches for early detection and personalized interventions. Lipidomics, leveraging advanced mass spectrometry techniques, has become instrumental in deciphering lipid-mediated mechanisms in CVDs. This review explores the application of lipidomics in identifying biomarkers for myocardial infarction, heart failure, stroke, and calcific aortic valve stenosis (CAVS). This review examines the technological advancements in shotgun lipidomics and LC/MS, which provide unparalleled insights into lipid composition and function. Key lipid biomarkers, including ceramides and lysophospholipids, have been linked to disease progression and therapeutic outcomes. Integrating lipidomics with genomic and proteomic data reveals the molecular underpinnings of CVDs, enhancing risk prediction and intervention strategies. This review positions lipidomics as a transformative tool in reshaping cardiovascular research and clinical practice.
Collapse
Affiliation(s)
- Arun Surendran
- Mass Spectrometry Core Facility, BRIC-Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Hannah Zhang
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Manitoba, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Manitoba, Canada; Precision Cardiovascular Medicine Group, St. Boniface Hospital Research, Manitoba, Canada
| | - Aleksandra Stamenkovic
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Manitoba, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Manitoba, Canada; Precision Cardiovascular Medicine Group, St. Boniface Hospital Research, Manitoba, Canada
| | - Amir Ravandi
- Cardiovascular Lipidomics Laboratory, St. Boniface Hospital, Albrechtsen Research Centre, Manitoba, Canada; Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Manitoba, Canada; Precision Cardiovascular Medicine Group, St. Boniface Hospital Research, Manitoba, Canada.
| |
Collapse
|
2
|
Vo NX, Pham HL, Bui TT, Bui TT. Systematic Review on Efficacy, Effectiveness, and Safety of Pitavastatin in Dyslipidemia in Asia. Healthcare (Basel) 2024; 13:59. [PMID: 39791666 PMCID: PMC11720254 DOI: 10.3390/healthcare13010059] [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: 11/29/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Objectives: Dyslipidemia, a significant risk factor for cardiovascular disease (CVD), is marked by abnormal lipid levels, such as the elevated lowering of low-density lipoprotein cholesterol (LDL-C). Statins are the first-line treatment for LDL-C reduction. Pitavastatin (PIT) has shown potential in lowering LDL-C and improving high-density lipoprotein cholesterol (HDL-C). This review assesses pitavastatin's efficacy, effectiveness, and safety in dyslipidemia management in Asia. Methods: A systematic review was conducted using PubMed, Cochrane, and Embase databases up to November 2024, adhering to Preferred Reporting Items of Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Seventeen studies (12 RCTs and 5 non-RCTs) were analyzed, focusing on LDL-C reduction, safety profiles, and adverse events. The quality of the studies was assessed using checklists to ensure the selection of the best studies and to limit bias. Results: Pitavastatin doses (1-4 mg) reduced LDL-C by 28-47%, comparable to atorvastatin, rosuvastatin, and simvastatin. The 2 mg dose matched atorvastatin's 10 mg dose in efficacy for both short-term (35-42%) and long-term (28-36%) use. LDL-C target achievement rates were 75-95%. Adverse events, including mild myalgia and elevated liver enzymes, were rare, and discontinuation rates were low. Conclusions: Pitavastatin is an effective and safe alternative to traditional statins for dyslipidemia management in Asia. Further research on long-term outcomes and high-risk groups is warranted.
Collapse
Affiliation(s)
- Nam Xuan Vo
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam;
| | - Huong Lai Pham
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam;
| | - Tan Trong Bui
- Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam;
| | - Tien Thuy Bui
- Faculty of Pharmacy, Le Van Thinh Hospital, Ho Chi Minh City 700000, Vietnam;
| |
Collapse
|
3
|
Abrahams T, Nicholls SJ. Perspectives on the success of plasma lipidomics in cardiovascular drug discovery and future challenges. Expert Opin Drug Discov 2024; 19:281-290. [PMID: 38402906 DOI: 10.1080/17460441.2023.2292039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 12/04/2023] [Indexed: 02/27/2024]
Abstract
INTRODUCTION Plasma lipidomics has emerged as a powerful tool in cardiovascular drug discovery by providing insights into disease mechanisms, identifying potential biomarkers for diagnosis and prognosis, and discovering novel targets for drug development. Widespread application of plasma lipidomics is hampered by technological limitations and standardization and requires a collaborative approach to maximize its use in cardiovascular drug discovery. AREAS COVERED This review provides an overview of the utility of plasma lipidomics in cardiovascular drug discovery and discusses the challenges and future perspectives of this rapidly evolving field. The authors discuss the role of lipidomics in understanding the molecular mechanisms of CVD, identifying novel biomarkers for diagnosis and prognosis, and discovering new therapeutic targets for drug development. Furthermore, they highlight the challenges faced in data analysis, standardization, and integration with other omics approaches and propose future directions for the field. EXPERT OPINION Plasma lipidomics holds great promise for improving the diagnosis, treatment, and prevention of CVD. While challenges remain in standardization and technology, ongoing research and collaboration among scientists and clinicians will undoubtedly help overcome these obstacles. As lipidomics evolves, its impact on cardiovascular drug discovery and clinical practice is expected to grow, ultimately benefiting patients and healthcare systems worldwide.
Collapse
Affiliation(s)
- Timothy Abrahams
- From the Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Stephen J Nicholls
- From the Victorian Heart Institute, Monash University, Melbourne, Australia
| |
Collapse
|
4
|
Garrett TJ, Puchowicz MA, Park EA, Dong Q, Farage G, Childress R, Guingab J, Simpson CL, Sen S, Brogdon EC, Buchanan LM, Raghow R, Elam MB. Effect of statin treatment on metabolites, lipids and prostanoids in patients with Statin Associated Muscle Symptoms (SAMS). PLoS One 2023; 18:e0294498. [PMID: 38100464 PMCID: PMC10723679 DOI: 10.1371/journal.pone.0294498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/02/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Between 5-10% of patients discontinue statin therapy due to statin-associated adverse reactions, primarily statin associated muscle symptoms (SAMS). The absence of a clear clinical phenotype or of biomarkers poses a challenge for diagnosis and management of SAMS. Similarly, our incomplete understanding of the pathogenesis of SAMS hinders the identification of treatments for SAMS. Metabolomics, the profiling of metabolites in biofluids, cells and tissues is an important tool for biomarker discovery and provides important insight into the origins of symptomatology. In order to better understand the pathophysiology of this common disorder and to identify biomarkers, we undertook comprehensive metabolomic and lipidomic profiling of plasma samples from patients with SAMS who were undergoing statin rechallenge as part of their clinical care. METHODS AND FINDINGS We report our findings in 67 patients, 28 with SAMS (cases) and 39 statin-tolerant controls. SAMS patients were studied during statin rechallenge and statin tolerant controls were studied while on statin. Plasma samples were analyzed using untargeted LC-MS metabolomics and lipidomics to detect differences between cases and controls. Differences in lipid species in plasma were observed between cases and controls. These included higher levels of linoleic acid containing phospholipids and lower ether lipids and sphingolipids. Reduced levels of acylcarnitines and altered amino acid profile (tryptophan, tyrosine, proline, arginine, and taurine) were observed in cases relative to controls. Pathway analysis identified significant increase of urea cycle metabolites and arginine and proline metabolites among cases along with downregulation of pathways mediating oxidation of branched chain fatty acids, carnitine synthesis, and transfer of acetyl groups into mitochondria. CONCLUSIONS The plasma metabolome of patients with SAMS exhibited reduced content of long chain fatty acids and increased levels of linoleic acid (18:2) in phospholipids, altered energy production pathways (β-oxidation, citric acid cycle and urea cycles) as well as reduced levels of carnitine, an essential mediator of mitochondrial energy production. Our findings support the hypothesis that alterations in pro-inflammatory lipids (arachidonic acid pathway) and impaired mitochondrial energy metabolism underlie the muscle symptoms of patients with statin associated muscle symptoms (SAMS).
Collapse
Affiliation(s)
- Timothy J. Garrett
- Southeast Center for Integrated Metabolomics (SECIM), Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Michelle A. Puchowicz
- Pediatrics-Obesity, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Edwards A. Park
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Qingming Dong
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Gregory Farage
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Richard Childress
- Endocrine Section, Memphis Veteran’s Affairs Medical Center, Memphis, Tennessee, United States of America
| | - Joy Guingab
- Southeast Center for Integrated Metabolomics (SECIM), Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Claire L. Simpson
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Saunak Sen
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Elizabeth C. Brogdon
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Logan M. Buchanan
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Rajendra Raghow
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Marshall B. Elam
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
- Cardiology Section, Memphis Veteran’s Affairs Medical Center, Memphis, Tennessee, United States of America
| |
Collapse
|
5
|
Cerda A, Bortolin RH, Yoshinaga MY, Freitas RCCD, Dagli-Hernandez C, Borges JB, Oliveira VFD, Gonçalves RM, Faludi AA, Bastos GM, Hirata RDC, Hirata MH. Lipidomic analysis identified potential predictive biomarkers of statin response in subjects with Familial hypercholesterolemia. Chem Phys Lipids 2023; 257:105348. [PMID: 37827478 DOI: 10.1016/j.chemphyslip.2023.105348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023]
Abstract
Familial hypercholesterolemia (FH) is a disorder of lipid metabolism that causes elevated low-density lipoprotein cholesterol (LDL-c) and increased premature atherosclerosis risk. Statins inhibit endogenous cholesterol biosynthesis, which reduces LDL-c plasma levels and prevent from cardiovascular events. This study aimed to explore the effects of statin treatment on serum lipidomic profile and to identify biomarkers of response in subjects with FH. Seventeen adult FH patients underwent a 6-week washout followed by 4-week treatment with atorvastatin (80 mg/day) or rosuvastatin (40 mg/day). LDL-c response was considered good (40-70 % reduction, n = 9) or poor (3-33 % reduction, n = 8). Serum lipidomic profile was analyzed by ultra-high-performance liquid chromatography combined with electrospray ionization tandem time-of-flight mass spectrometry, and data were analyzed using MetaboAnalyst v5.0. Lipidomic analysis identified 353 lipids grouped into 16 classes. Statin treatment reduced drastically 8 of 13 lipid classes, generating a characteristic lipidomic profile with a significant contribution of phosphatidylinositols (PI) 16:0/18:2, 18:0/18:1 and 18:0/18:2; and triacylglycerols (TAG) 18:2x2/18:3, 18:1/18:2/18:3, 16:1/18:2x2, 16:1/18:2/18:3 and 16:1/18:2/Arachidonic acid (p-adjusted <0.05). Biomarker analysis implemented in MetaboAnalyst subsequently identified PI 16:1/18:0, 16:0/18:2 and 18:0/18:2 as predictors of statin response with and receiver operating characteristic (ROC) areas under the curve of 0.98, 0.94 and 0.91, respectively. In conclusion, statins extensively modulate the overall serum lipid composition of FH individuals and these findings suggest that phosphatidyl-inositol molecules are potential predictive biomarkers of statin response.
Collapse
Affiliation(s)
- Alvaro Cerda
- Department of Basic Sciences, Center of Excellence in Translational Medicine, CEMT-BIOREN, Universidad de La Frontera, Temuco 4810296, Chile
| | - Raul Hernandes Bortolin
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; Department of Cardiology, Boston Children's Hospital/Harvard Medical School, Boston, MA 02115, United States
| | - Marcos Yukio Yoshinaga
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, São Paulo 05508-000, Brazil
| | - Renata Caroline Costa de Freitas
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil; Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, United States
| | - Carolina Dagli-Hernandez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Jessica Bassani Borges
- Department of Research, Hospital Beneficiencia Portuguesa de Sao Paulo, Sao Paulo 01323-001, Brazil
| | - Victor Fernandes de Oliveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | | | - Andre Arpad Faludi
- Medical Division, Institute of Cardiology Dante Pazzanese, Sao Paulo 04012-909, Brazil
| | - Gisele Medeiros Bastos
- Department of Research, Hospital Beneficiencia Portuguesa de Sao Paulo, Sao Paulo 01323-001, Brazil
| | - Rosario Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil.
| |
Collapse
|
6
|
Beaufrère H, Barboza T, Burnett A, Stark KD, Wood RD. Effects of Atorvastatin and Rosuvastatin on Blood Lipids in Quaker Parrots ( Myiopsitta monachus). J Avian Med Surg 2023; 37:199-208. [PMID: 37962313 DOI: 10.1647/22-00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Statin drugs are the most effective class of hypolipidemic and antiatherosclerotic drugs, with atorvastatin and rosuvastatin being the most effective. While the use of statins would be a tremendous asset in the treatment of dyslipidemia and lipid-accumulation disorders in birds, there are only limited data available regarding their use and effectiveness in psittacine species. Two consecutive randomized crossover trials on Quaker parrots (Myiopsitta monachus) were performed to study the effect of atorvastatin and rosuvastatin. Ten birds were used in an initial balanced crossover experiment with 5 oral treatments (control; atorvastatin 10 mg/kg q12h and q24h; rosuvastatin 10 mg/kg q12h and q24h) for 2 weeks each. Plasma lipidomics and lipoprotein profiling were performed after each treatment. Twelve birds were used in a second experiment consisting of 2 parallel crossover studies, each with 6 birds either fed their regular diet or a 0.3% cholesterol diet. In the 2 parallel crossover studies, the treatment group was administered atorvastatin 20 mg/kg orally q12h and the control group a placebo suspension orally q12h. Plasma lipidomics, lipoprotein profiles, and 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase activity were subsequently measured. Results were analyzed with serial linear mixed models and trends were assessed graphically. No statistically significant effect of any statin treatment was detected on plasma lipids, lipoproteins, creatinine kinase, or HMG-CoA reductase activity. In the first trial, all the rosuvastatin treatments led to some nonsignificant decreases in several triacylglycerol species, while in the second trial this was only observed in the birds on atorvastatin 20 mg/kg q12h being fed their regular diet. Quaker parrots may require much higher doses of statin drugs to show significant and clinically useful lipid-lowering effects.
Collapse
Affiliation(s)
- Hugues Beaufrère
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616, USA,
| | - Trinita Barboza
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| | - Alysha Burnett
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | - Ken D Stark
- Department of Kinesiology and Health Studies, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1
| | - R Darren Wood
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| |
Collapse
|
7
|
Pharmacometabolomics for the Study of Lipid-Lowering Therapies: Opportunities and Challenges. Int J Mol Sci 2023; 24:ijms24043291. [PMID: 36834701 PMCID: PMC9960554 DOI: 10.3390/ijms24043291] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Lipid-lowering therapies are widely used to prevent the development of atherosclerotic cardiovascular disease (ASCVD) and related mortality worldwide. "Omics" technologies have been successfully applied in recent decades to investigate the mechanisms of action of these drugs, their pleiotropic effects, and their side effects, aiming to identify novel targets for future personalized medicine with an improvement of the efficacy and safety associated with the treatment. Pharmacometabolomics is a branch of metabolomics that is focused on the study of drug effects on metabolic pathways that are implicated in the variation of response to the treatment considering also the influences from a specific disease, environment, and concomitant pharmacological therapies. In this review, we summarized the most significant metabolomic studies on the effects of lipid-lowering therapies, including the most commonly used statins and fibrates to novel drugs or nutraceutical approaches. The integration of pharmacometabolomics data with the information obtained from the other "omics" approaches could help in the comprehension of the biological mechanisms underlying the use of lipid-lowering drugs in view of defining a precision medicine to improve the efficacy and reduce the side effects associated with the treatment.
Collapse
|
8
|
Platelet Redox Imbalance in Hypercholesterolemia: A Big Problem for a Small Cell. Int J Mol Sci 2022; 23:ijms231911446. [PMID: 36232746 PMCID: PMC9570056 DOI: 10.3390/ijms231911446] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022] Open
Abstract
The imbalance between reactive oxygen species (ROS) synthesis and their scavenging by anti-oxidant defences is the common soil of many disorders, including hypercholesterolemia. Platelets, the smallest blood cells, are deeply involved in the pathophysiology of occlusive arterial thrombi associated with myocardial infarction and stroke. A great deal of evidence shows that both increased intraplatelet ROS synthesis and impaired ROS neutralization are implicated in the thrombotic process. Hypercholesterolemia is recognized as cause of atherosclerosis, cerebro- and cardiovascular disease, and, closely related to this, is the widespread acceptance that it strongly contributes to platelet hyperreactivity via direct oxidized LDL (oxLDL)-platelet membrane interaction via scavenger receptors such as CD36 and signaling pathways including Src family kinases (SFK), mitogen-activated protein kinases (MAPK), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. In turn, activated platelets contribute to oxLDL generation, which ends up propagating platelet activation and thrombus formation through a mechanism mediated by oxidative stress. When evaluating the effect of lipid-lowering therapies on thrombogenesis, a large body of evidence shows that the effects of statins and proprotein convertase subtilisin/kexin type 9 inhibitors are not limited to the reduction of LDL-C but also to the down-regulation of platelet reactivity mainly by mechanisms sensitive to intracellular redox balance. In this review, we will focus on the role of oxidative stress-related mechanisms as a cause of platelet hyperreactivity and the pathophysiological link of the pleiotropism of lipid-lowering agents to the beneficial effects on platelet function.
Collapse
|
9
|
Garshick MS, Block R, Drenkova K, Tawil M, James G, Brenna JT. Statin therapy upregulates arachidonic acid status via enhanced endogenous synthesis in patients with plaque psoriasis. Prostaglandins Leukot Essent Fatty Acids 2022; 180:102428. [PMID: 35490599 PMCID: PMC9870621 DOI: 10.1016/j.plefa.2022.102428] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 01/26/2023]
Abstract
Circulating fatty acids (FA) may be important in the psoriatic pro-inflammatory phenotype. FADS1 converts linoleic acid (LA) to arachidonic acid (AA), a precursor to potent signaling molecules. HMG-CoA reductase inhibitors (statins) increase FADS1/2 expression in vitro. Psoriasis patients (42 ± 14 years/age, 47% male) were randomized to 40 mg of atorvastatin (n = 20) or nothing (n = 10) for two weeks and plasma FA measured pre and post treatment. After treatment, LDL-C was 44% lower in the statin compared to the no-treatment group. Statins increased FADS1/2 expression, and lowered LA 12% (33% - > 29%, p<0.001) and raised AA 14% (7.7% - > 9.0%, p<0.01) with no change in the no-treatment group. In psoriasis, statins enhance AA and decrease LA, consistent with the action of enhanced FADS expression in vivo. Therapies intended to blunt the effects of AA on platelet aggregation, such as aspirin or omega-3 fatty acids, may require dose adjustment when co-administered with atorvastatin. NCT: NCT03228017.
Collapse
Affiliation(s)
- Michael S Garshick
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, NYU Langone Health, United States; Leon H. Charney Division of Cardiology, Department of Medicine, NYU Langone Health, United States; Cardiovascular Research Center, NYU Langone Health, United States.
| | - Robert Block
- Department of Public Health Sciences and Cardiology Division, Department of Medicine, University of Rochester, United States
| | - Kamelia Drenkova
- Cardiovascular Research Center, NYU Langone Health, United States
| | - Michael Tawil
- Cardiovascular Research Center, NYU Langone Health, United States
| | - Genevieve James
- Dell Pediatric Research Institute and Dept of Nutrition, University of Texas at Austin, United States
| | - J Thomas Brenna
- Dell Pediatric Research Institute and Dept of Nutrition, University of Texas at Austin, United States
| |
Collapse
|
10
|
Lipidomics in Understanding Pathophysiology and Pharmacologic Effects in Inflammatory Diseases: Considerations for Drug Development. Metabolites 2022; 12:metabo12040333. [PMID: 35448520 PMCID: PMC9030008 DOI: 10.3390/metabo12040333] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 01/26/2023] Open
Abstract
The lipidome has a broad range of biological and signaling functions, including serving as a structural scaffold for membranes and initiating and resolving inflammation. To investigate the biological activity of phospholipids and their bioactive metabolites, precise analytical techniques are necessary to identify specific lipids and quantify their levels. Simultaneous quantification of a set of lipids can be achieved using high sensitivity mass spectrometry (MS) techniques, whose technological advancements have significantly improved over the last decade. This has unlocked the power of metabolomics/lipidomics allowing the dynamic characterization of metabolic systems. Lipidomics is a subset of metabolomics for multianalyte identification and quantification of endogenous lipids and their metabolites. Lipidomics-based technology has the potential to drive novel biomarker discovery and therapeutic development programs; however, appropriate standards have not been established for the field. Standardization would improve lipidomic analyses and accelerate the development of innovative therapies. This review aims to summarize considerations for lipidomic study designs including instrumentation, sample stabilization, data validation, and data analysis. In addition, this review highlights how lipidomics can be applied to biomarker discovery and drug mechanism dissection in various inflammatory diseases including cardiovascular disease, neurodegeneration, lung disease, and autoimmune disease.
Collapse
|
11
|
Silveira AMR, Duarte GHB, Fernandes AMADP, Garcia PHD, Vieira NR, Antonio MA, Carvalho PDO. Serum Predose Metabolic Profiling for Prediction of Rosuvastatin Pharmacokinetic Parameters in Healthy Volunteers. Front Pharmacol 2021; 12:752960. [PMID: 34867363 PMCID: PMC8633954 DOI: 10.3389/fphar.2021.752960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/13/2021] [Indexed: 11/23/2022] Open
Abstract
Rosuvastatin is a well-known lipid-lowering agent generally used for hypercholesterolemia treatment and coronary artery disease prevention. There is a substantial inter-individual variability in the absorption of statins usually caused by genetic polymorphisms leading to a variation in the corresponding pharmacokinetic parameters, which may affect drug therapy safety and efficacy. Therefore, the investigation of metabolic markers associated with rosuvastatin inter-individual variability is exceedingly relevant for drug therapy optimization and minimizing side effects. This work describes the application of pharmacometabolomic strategies using liquid chromatography coupled to mass spectrometry to investigate endogenous plasma metabolites capable of predicting pharmacokinetic parameters in predose samples. First, a targeted method for the determination of plasma concentration levels of rosuvastatin was validated and applied to obtain the pharmacokinetic parameters from 40 enrolled individuals; then, predose samples were analyzed using a metabolomic approach to search for associations between endogenous metabolites and the corresponding pharmacokinetic parameters. Data processing using machine learning revealed some candidates including sterols and bile acids, carboxylated metabolites, and lipids, suggesting the approach herein described as promising for personalized drug therapy.
Collapse
Affiliation(s)
| | | | | | | | - Nelson Rogerio Vieira
- Integrated Unit of Pharmacology and Gastroenterology (UNIFAG), São Francisco University-USF, Bragança Paulista, Brazil
| | - Marcia Aparecida Antonio
- Integrated Unit of Pharmacology and Gastroenterology (UNIFAG), São Francisco University-USF, Bragança Paulista, Brazil
| | | |
Collapse
|
12
|
Zhang S, Yuan L, Li H, Han L, Jing W, Wu X, Ullah S, Liu R, Wu Y, Xu J. The Novel Interplay between Commensal Gut Bacteria and Metabolites in Diet-Induced Hyperlipidemic Rats Treated with Simvastatin. J Proteome Res 2021; 21:808-821. [PMID: 34365791 DOI: 10.1021/acs.jproteome.1c00252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hyperlipidemia is one kind of metabolic syndrome for which the treatment commonly includes simvastatin (SV). Individuals vary widely in statin responses, and growing evidence implicates gut microbiome involvement in this variability. However, the associated molecular mechanisms between metabolic improvement and microbiota composition following SV treatment are still not fully understood. In this study, combinatory approaches using ultrahigh-performance liquid chromatography coupled with hybrid triple quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF MS/MS)-based metabolomic profiling, PCR-denaturing gradient gel electrophoresis (PCR-DGGE), quantitative PCR (qPCR), and 16S rRNA gene sequencing-based gut microbiota profiling were performed to investigate the interplay of endogenous metabolites and the gut microbiota related to SV treatment. A total of 6 key differential endogenous metabolites were identified that affect the metabolism of amino acids (phenylalanine and tyrosine), unsaturated fatty acids (linoleic acid and 9-hydroxyoctadecadienoic acid (9-HODE)), and the functions of gut microbial metabolism. Moreover, a total of 22 differentially abundant taxa were obtained following SV treatment. Three bacterial taxa were identified to be involved in SV treatment, namely, Bacteroidaceae, Prevotellaceae, and Porphyromonadaceae. These findings suggested that the phenylalanine and tyrosine-associated amino acid metabolism pathways, as well as the linoleic acid and 9-HODE-associated unsaturated fatty acid metabolism pathways, which are involved in gut flora interactions, might be potential therapeutic targets for improvement in SV hypolipidemic efficacy. The mass spectrometric data have been deposited to MassIVE (https://massive.ucsd.edu/ProteoSAFe/static/massive.jsp). Username: MSV000087842_reviewer. Password: hardworkingzsr.
Collapse
Affiliation(s)
- Siruo Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.,Department of Clinical Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, PR China
| | - Lu Yuan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Huan Li
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Lei Han
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Wanghui Jing
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Xiaokang Wu
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Shakir Ullah
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Ruina Liu
- College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Yonghong Wu
- Department of Medical Technology, Xi'an Medical University, Xi'an, Shaanxi 710021, PR China
| | - Jiru Xu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| |
Collapse
|
13
|
Fu J, Zhang Y, Liu J, Lian X, Tang J, Zhu F. Pharmacometabonomics: data processing and statistical analysis. Brief Bioinform 2021; 22:6236068. [PMID: 33866355 DOI: 10.1093/bib/bbab138] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/09/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
Individual variations in drug efficacy, side effects and adverse drug reactions are still challenging that cannot be ignored in drug research and development. The aim of pharmacometabonomics is to better understand the pharmacokinetic properties of drugs and monitor the drug effects on specific metabolic pathways. Here, we systematically reviewed the recent technological advances in pharmacometabonomics for better understanding the pathophysiological mechanisms of diseases as well as the metabolic effects of drugs on bodies. First, the advantages and disadvantages of all mainstream analytical techniques were compared. Second, many data processing strategies including filtering, missing value imputation, quality control-based correction, transformation, normalization together with the methods implemented in each step were discussed. Third, various feature selection and feature extraction algorithms commonly applied in pharmacometabonomics were described. Finally, the databases that facilitate current pharmacometabonomics were collected and discussed. All in all, this review provided guidance for researchers engaged in pharmacometabonomics and metabolomics, and it would promote the wide application of metabolomics in drug research and personalized medicine.
Collapse
Affiliation(s)
- Jianbo Fu
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Ying Zhang
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Jin Liu
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Xichen Lian
- College of Pharmaceutical Sciences in Zhejiang University, China
| | - Jing Tang
- Department of Bioinformatics in Chongqing Medical University, China
| | - Feng Zhu
- College of Pharmaceutical Sciences in Zhejiang University, China
| |
Collapse
|
14
|
A pilot randomized trial of atorvastatin as adjunct therapy in patients with acute venous thromboembolism. Blood Coagul Fibrinolysis 2021; 32:16-22. [PMID: 33196511 DOI: 10.1097/mbc.0000000000000968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Venous thromboembolism (VTE) is the third most common cardiovascular disease and optimizing treatment is essential. In this single-center pilot study, we sought to investigate the effects of statins in addition to anticoagulation in patients with acute VTE. We enrolled patients over 18 with an acute proximal lower extremity deep vein thrombosis with or without pulmonary embolism. Patients were randomized to anticoagulation alone (with either warfarin or rivaroxaban) or anticoagulation and atorvastatin 40 mg daily and followed for 9 months. The primary objective was to determine if adjunct atorvastatin reduced thrombin generation, measured by endogenous thrombin potential and/or peak thrombin concentration. Secondary endpoints included recurrent VTE, arterial thrombosis, bleeding events, lipidomic profiles, and symptoms of post thrombotic syndrome. A total of 21 patients were enrolled (11 anticoagulation only and 10 anticoagulation and atorvastatin) over 3.5 years. Endogenous thrombin potential or peak thrombin was not significantly recued with the addition of atorvastatin. Atorvastatin did significantly reduce the mean LDLs at 3 months, without reduction of either d-dimer or high-sensitivity-C reactive protein. Given the low recruitment rate, continuation of the study was deemed futile and the study was terminated early. Barriers to enrollment and completion of study included the many ineligible patients by exclusion criteria (e.g., preexisting statin use, active malignancy, etc.) and high rate of lost follow-up. The pilot study was terminated early but could inform obstacles for future studies investigating the effects of statins in the management of patients with VTE.
Collapse
|
15
|
|
16
|
Shon JC, Kim WC, Ryu R, Wu Z, Seo JS, Choi MS, Liu KH. Plasma Lipidomics Reveals Insights into Anti-Obesity Effect of Chrysanthemum morifolium Ramat Leaves and Its Constituent Luteolin in High-Fat Diet-Induced Dyslipidemic Mice. Nutrients 2020; 12:nu12102973. [PMID: 33003339 PMCID: PMC7650530 DOI: 10.3390/nu12102973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023] Open
Abstract
The Chrysanthemum morifolium Ramat (CM) is widely used as a traditional medicine and herbal tea by the Asian population for its health benefits related to obesity. However, compared to the flowers of CM, detailed mechanisms underlying the beneficial effects of its leaves on obesity and dyslipidemia have not yet been elucidated. Therefore, to investigate the lipidomic biomarkers responsible for the pharmacological effects of CM leaf extract (CLE) in plasma of mice fed a high-fat diet (HFD), the plasma of mice fed a normal diet (ND), HFD, HFD plus CLE 1.5% diet, and HFD plus luteolin 0.003% diet (LU) for 16 weeks were analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) combined with multivariate analysis. In our analysis, the ND, HFD, CLE, and LU groups were clearly differentiated by partial least-squares discriminant analysis (PLS-DA) score plots. The major metabolites contributing to this differentiation were cholesteryl esters (CEs), lysophosphatidylcholines (LPCs), phosphatidylcholines (PCs), ceramides (CERs), and sphingomyelins (SMs). The levels of plasma CEs, LPCs, PCs, SMs, and CERs were significantly increased in the HFD group compared to those in the ND group, and levels of these lipids recovered to normal after administration of CLE or LU. Furthermore, changes in hepatic mRNA expression levels involved in the Kennedy pathway and sphingolipid biosynthesis were also suppressed by treatment with CLE or LU. In conclusion, this study examined the beneficial effects of CLE and LU on obesity and dyslipidemia, which were demonstrated as reduced synthesis of lipotoxic intermediates. These results may provide valuable insights towards evaluating the therapeutic effects of CLE and LU and understanding obesity-related diseases.
Collapse
Affiliation(s)
- Jong Cheol Shon
- Environmental Chemistry Research Group, Korea Institute of Toxicology, Jinju 52834, Korea; (J.C.S.); (J.-S.S.)
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (W.C.K.); (Z.W.)
| | - Won Cheol Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (W.C.K.); (Z.W.)
| | - Ri Ryu
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea;
| | - Zhexue Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (W.C.K.); (Z.W.)
| | - Jong-Su Seo
- Environmental Chemistry Research Group, Korea Institute of Toxicology, Jinju 52834, Korea; (J.C.S.); (J.-S.S.)
| | - Myung-Sook Choi
- Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (M.-S.C.); (K.-H.L.); Tel.: +82-53-950-6232 (M.-S.C.); +82-53-950-8567 (K.-H.L.); Fax: +82-53-950-8557 (M.-S.C. & K.-H.L.)
| | - Kwang-Hyeon Liu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (W.C.K.); (Z.W.)
- Correspondence: (M.-S.C.); (K.-H.L.); Tel.: +82-53-950-6232 (M.-S.C.); +82-53-950-8567 (K.-H.L.); Fax: +82-53-950-8557 (M.-S.C. & K.-H.L.)
| |
Collapse
|
17
|
Current Concepts in Pharmacometabolomics, Biomarker Discovery, and Precision Medicine. Metabolites 2020; 10:metabo10040129. [PMID: 32230776 PMCID: PMC7241083 DOI: 10.3390/metabo10040129] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
Pharmacometabolomics (PMx) studies use information contained in metabolic profiles (or metabolome) to inform about how a subject will respond to drug treatment. Genome, gut microbiome, sex, nutrition, age, stress, health status, and other factors can impact the metabolic profile of an individual. Some of these factors are known to influence the individual response to pharmaceutical compounds. An individual’s metabolic profile has been referred to as his or her “metabotype.” As such, metabolomic profiles obtained prior to, during, or after drug treatment could provide insights about drug mechanism of action and variation of response to treatment. Furthermore, there are several types of PMx studies that are used to discover and inform patterns associated with varied drug responses (i.e., responders vs. non-responders; slow or fast metabolizers). The PMx efforts could simultaneously provide information related to an individual’s pharmacokinetic response during clinical trials and be used to predict patient response to drugs making pharmacometabolomic clinical research valuable for precision medicine. PMx biomarkers can also be discovered and validated during FDA clinical trials. Using biomarkers during medical development is described in US Law under the 21st Century Cures Act. Information on how to submit biomarkers to the FDA and their context of use is defined herein.
Collapse
|
18
|
Takeda H, Izumi Y, Tamura S, Koike T, Koike Y, Shiomi M, Bamba T. Lipid Profiling of Serum and Lipoprotein Fractions in Response to Pitavastatin Using an Animal Model of Familial Hypercholesterolemia. J Proteome Res 2020; 19:1100-1108. [PMID: 31965805 DOI: 10.1021/acs.jproteome.9b00602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Statins are widely used for the treatment of atherosclerotic cardiovascular diseases. They inhibit cholesterol biosynthesis in the liver and cause pleiotropic effects, including anti-inflammatory and antioxidant effects. To develop novel therapeutic drugs, the effect of blood-borne lipid molecules on the pleiotropic effects of statins must be elucidated. Myocardial infarction-prone Watanabe heritable hyperlipidemic (WHHLMI) rabbits, an animal model for hypercholesterolemia, are suitable for the determination of lipid molecules in the blood in response to statins because their lipoprotein metabolism is similar to that of humans. Herein, lipid molecules were investigated by lipidome analysis in response to pitavastatin using WHHLMI rabbits. Various lipid molecules in the blood were measured using a supercritical fluid chromatography triple quadrupole mass spectrometry. Cholesterol and cholesterol ester blood concentrations decreased by reducing the secretion of very low density lipoproteins from the liver. Independent of the inhibition effects of cholesterol biosynthesis, the concentrations of some lipids with anti-inflammation and antioxidant effects (phospholipid molecules with n-6 fatty acid side chains, lysophosphatidylcholines, phosphatidylethanolamine plasmalogens, and ceramide molecules) were significantly altered. These findings may lead to further investigation of the mechanism of statin action.
Collapse
Affiliation(s)
- Hiroaki Takeda
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shohei Tamura
- Institute for Experimental Animals, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tomonari Koike
- Institute for Experimental Animals, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yui Koike
- Institute for Experimental Animals, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masashi Shiomi
- Institute for Experimental Animals, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.,Division of Comparative Pathophysiology, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
19
|
Liu J, Lahousse L, Nivard MG, Bot M, Chen L, van Klinken JB, Thesing CS, Beekman M, van den Akker EB, Slieker RC, Waterham E, van der Kallen CJH, de Boer I, Li-Gao R, Vojinovic D, Amin N, Radjabzadeh D, Kraaij R, Alferink LJM, Murad SD, Uitterlinden AG, Willemsen G, Pool R, Milaneschi Y, van Heemst D, Suchiman HED, Rutters F, Elders PJM, Beulens JWJ, van der Heijden AAWA, van Greevenbroek MMJ, Arts ICW, Onderwater GLJ, van den Maagdenberg AMJM, Mook-Kanamori DO, Hankemeier T, Terwindt GM, Stehouwer CDA, Geleijnse JM, 't Hart LM, Slagboom PE, van Dijk KW, Zhernakova A, Fu J, Penninx BWJH, Boomsma DI, Demirkan A, Stricker BHC, van Duijn CM. Integration of epidemiologic, pharmacologic, genetic and gut microbiome data in a drug-metabolite atlas. Nat Med 2020; 26:110-117. [PMID: 31932804 DOI: 10.1038/s41591-019-0722-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 11/27/2019] [Indexed: 12/17/2022]
Abstract
Progress in high-throughput metabolic profiling provides unprecedented opportunities to obtain insights into the effects of drugs on human metabolism. The Biobanking BioMolecular Research Infrastructure of the Netherlands has constructed an atlas of drug-metabolite associations for 87 commonly prescribed drugs and 150 clinically relevant plasma-based metabolites assessed by proton nuclear magnetic resonance. The atlas includes a meta-analysis of ten cohorts (18,873 persons) and uncovers 1,071 drug-metabolite associations after evaluation of confounders including co-treatment. We show that the effect estimates of statins on metabolites from the cross-sectional study are comparable to those from intervention and genetic observational studies. Further data integration links proton pump inhibitors to circulating metabolites, liver function, hepatic steatosis and the gut microbiome. Our atlas provides a tool for targeted experimental pharmaceutical research and clinical trials to improve drug efficacy, safety and repurposing. We provide a web-based resource for visualization of the atlas (http://bbmri.researchlumc.nl/atlas/).
Collapse
Affiliation(s)
- Jun Liu
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands. .,Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Lies Lahousse
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.,Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Michel G Nivard
- Department of Biological Psychology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Mariska Bot
- Department of Biological Psychology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Lianmin Chen
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands.,Department of Pediatrics, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan Bert van Klinken
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.,Department of Clinical Chemistry, Laboratory Genetic Metabolic Disease, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Carisha S Thesing
- Department of Biological Psychology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Marian Beekman
- Department of Biomedical Data Sciences, section of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Erik Ben van den Akker
- Department of Biomedical Data Sciences, section of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Pattern Recognition and Bioinformatics, Delft University of Technology, Delft, the Netherlands.,Leiden Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Roderick C Slieker
- Amsterdam Public Health Research Institute, Amsterdam, the Netherlands.,Department of Epidemiology and Biostatistics, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Eveline Waterham
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Carla J H van der Kallen
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands.,School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Irene de Boer
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Djawad Radjabzadeh
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Robert Kraaij
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Louise J M Alferink
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Sarwa Darwish Murad
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.,Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Rene Pool
- Department of Biological Psychology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Yuri Milaneschi
- Department of Biological Psychology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - H Eka D Suchiman
- Department of Biomedical Data Sciences, section of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Femke Rutters
- Amsterdam Public Health Research Institute, Amsterdam, the Netherlands.,Department of Epidemiology and Biostatistics, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands
| | - Petra J M Elders
- Amsterdam Public Health Research Institute, Amsterdam, the Netherlands.,Department of General Practice and Elderly Care Medicine, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands
| | - Joline W J Beulens
- Amsterdam Public Health Research Institute, Amsterdam, the Netherlands.,Department of Epidemiology and Biostatistics, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands
| | - Amber A W A van der Heijden
- Amsterdam Public Health Research Institute, Amsterdam, the Netherlands.,Department of General Practice and Elderly Care Medicine, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands.,School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Ilja C W Arts
- School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands.,Department of Epidemiology, Maastricht University, Maastricht, the Netherlands.,Maastricht Center for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | | | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas Hankemeier
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands.,Netherlands Metabolomics Center, Leiden, the Netherlands
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands.,School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Johanna M Geleijnse
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Leen M 't Hart
- Amsterdam Public Health Research Institute, Amsterdam, the Netherlands.,Department of Biomedical Data Sciences, section of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Epidemiology and Biostatistics, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, section of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.,Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands.,Department of Pediatrics, University Medical Center Groningen, Groningen, the Netherlands
| | - Brenda W J H Penninx
- Department of Biological Psychology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, the Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Ayşe Demirkan
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.,Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands.,Section of Statistical Multi-omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford, UK
| | - Bruno H C Stricker
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.,Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.,Inspectorate of Healthcare, The Hague, the Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands. .,Nuffield Department of Population Health, University of Oxford, Oxford, UK. .,Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
20
|
Goracci L, Valeri A, Sciabola S, Aleo MD, Moritz W, Lichtenberg J, Cruciani G. A Novel Lipidomics-Based Approach to Evaluating the Risk of Clinical Hepatotoxicity Potential of Drugs in 3D Human Microtissues. Chem Res Toxicol 2019; 33:258-270. [PMID: 31820940 DOI: 10.1021/acs.chemrestox.9b00364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The importance of adsorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis is expected to grow substantially due to recent failures in detecting severe toxicity issues of new chemical entities during preclinical/clinical development. Traditionally, safety risk assessment studies for humans have been conducted in animals during advanced preclinical or clinical phase of drug development. However, potential drug toxicity in humans now needs to be detected in the drug discovery process as soon as possible without reliance on animal studies. The "omics", such as genomics, proteomics, and metabolomics, have recently entered pharmaceutical research in both drug discovery and drug development, but to the best of our knowledge, no applications in high-throughput safety risk assessment have been attempted so far. This paper reports an innovative method to anticipate adverse drug effects in an early discovery phase based on lipid fingerprints using human three-dimensional microtissues. The risk of clinical hepatotoxicity potential was evaluated for a data set of 22 drugs belonging to five different therapeutic chemical classes and with various drug-induced liver injury effect. The treatment of microtissues with repeated doses of each drug allowed collecting lipid fingerprints for five time points (2, 4, 7, 9, and 11 days), and multivariate statistical analysis was applied to search for correlations with the hepatotoxic effect. The method allowed clustering of the drugs based on their hepatotoxic effect, and the observed lipid impairments for a number of drugs was confirmed by literature sources. Compared to traditional screening methods, here multiple interconnected variables (lipids) are measured simultaneously, providing a snapshot of the cellular status from the lipid perspective at a molecular level. Applied here to hepatotoxicity, the proposed workflow can be applied to several tissues, being tridimensional microtissues from various origins.
Collapse
Affiliation(s)
- Laura Goracci
- Department of Chemistry, Biology, and Biotechnology , University of Perugia , Perugia 06123 , Italy
| | | | - Simone Sciabola
- Medicinal Chemistry , Biogen , 115 Broadway Street , Cambridge , Massachusetts 02139 , United States
| | - Michael D Aleo
- Drug Safety R&D , Pfizer Worldwide Research and Development , Groton , Connecticut 06340 , United States
| | | | | | - Gabriele Cruciani
- Department of Chemistry, Biology, and Biotechnology , University of Perugia , Perugia 06123 , Italy
| |
Collapse
|
21
|
Kaddurah-Daouk R, Hankemeier T, Scholl EH, Baillie R, Harms A, Stage C, Dalhoff KP, Jűrgens G, Taboureau O, Nzabonimpa GS, Motsinger-Reif AA, Thomsen R, Linnet K, Rasmussen HB. Pharmacometabolomics Informs About Pharmacokinetic Profile of Methylphenidate. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 7:525-533. [PMID: 30169917 PMCID: PMC6118295 DOI: 10.1002/psp4.12309] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/17/2018] [Indexed: 12/29/2022]
Abstract
Carboxylesterase 1 (CES1) metabolizes methylphenidate and other drugs. CES1 gene variation only partially explains pharmacokinetic (PK) variability. Biomarkers predicting the PKs of drugs metabolized by CES1 are needed. We identified lipids in plasma from 44 healthy subjects that correlated with CES1 activity as determined by PK parameters of methylphenidate including a ceramide (q value = 0.001) and a phosphatidylcholine (q value = 0.005). Carriers of the CES1 143E allele had decreased methylphenidate metabolism and altered concentration of this phosphatidylcholine (q value = 0.040) and several high polyunsaturated fatty acid lipids (PUFAs). The half‐maximal inhibitory concentration (IC50) values of chenodeoxycholate and taurocholate were 13.55 and 19.51 μM, respectively, consistent with a physiological significance. In silico analysis suggested that bile acid inhibition of CES1 involved both binding to the active and superficial sites of the enzyme. We initiated identification of metabolites predicting PKs of drugs metabolized by CES1 and suggest lipids to regulate or be regulated by this enzyme.
Collapse
Affiliation(s)
- Rima Kaddurah-Daouk
- Duke Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina, USA.,Duke Institute for Brain Sciences, Duke University, Durham, North Carolina, USA
| | - Thomas Hankemeier
- Division of Analytical Biosciences, Leiden Academic Centre for Drug Research, Leiden, The Netherlands.,Netherlands Metabolomics Centre, Leiden, The Netherlands
| | - Elizabeth H Scholl
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| | | | - Amy Harms
- Division of Analytical Biosciences, Leiden Academic Centre for Drug Research, Leiden, The Netherlands.,Netherlands Metabolomics Centre, Leiden, The Netherlands
| | - Claus Stage
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg University Hospital, Frederiksberg, Denmark
| | - Kim P Dalhoff
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg University Hospital, Frederiksberg, Denmark
| | - Gesche Jűrgens
- Clinical Pharmacological Unit, Zealand University Hospital, Roskilde, Denmark
| | - Olivier Taboureau
- INSERM, UMRS 973, MTi, Université Paris Diderot, Paris Cedex, France
| | - Grace S Nzabonimpa
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Alison A Motsinger-Reif
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA.,Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| | - Ragnar Thomsen
- Section of Forensic Chemistry, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Kristian Linnet
- Section of Forensic Chemistry, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Henrik B Rasmussen
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, Roskilde, Denmark.,Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | | | | |
Collapse
|
22
|
Jiménez-Rojo N, Riezman H. On the road to unraveling the molecular functions of ether lipids. FEBS Lett 2019; 593:2378-2389. [PMID: 31166014 DOI: 10.1002/1873-3468.13465] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022]
Abstract
Ether lipids are glycerolipids further classified into alkyl-ether and alkenyl-ether (also termed plasmalogens) lipids. The two ether lipid subclasses share the first steps of their synthesis. However, alkyl-ether and alkenyl-ether lipids differ in their structure and physico-chemical properties (featuring different head groups) and, thus, probably in their functions. Ether lipids have intermittent distribution across the evolutionary tree and defects in their synthesis have been shown to perturb cellular homeostasis and lead to disease in humans. Here, we review their structure, their interactions with other lipids, and their potential roles in cellular functions, such as membrane homeostasis and membrane trafficking. Moreover, we discuss still unclear aspects of these lipids such as their subcellular distribution, and the need to unravel their molecular functions as well as how novel tools to study lipid biology will help clarify these aspects.
Collapse
Affiliation(s)
- Noemi Jiménez-Rojo
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, Switzerland
| | - Howard Riezman
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, Switzerland
| |
Collapse
|
23
|
Jia Z, Tie C, Wang C, Wu C, Zhang J. Perturbed Lipidomic Profiles in Rats With Chronic Cerebral Ischemia Are Regulated by Xiao-Xu-Ming Decoction. Front Pharmacol 2019; 10:264. [PMID: 30941043 PMCID: PMC6433774 DOI: 10.3389/fphar.2019.00264] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 03/04/2019] [Indexed: 01/03/2023] Open
Abstract
Chronic cerebral ischemia (CCI) is a serious human health condition with lacking therapeutic agents. Moreover, its mechanism of action remains elusive, and thus novel treatment options are required. Lipid metabolism disorder are closely related to CCI. In this study, a CCI-rats model was established by the permanent occlusion of rat bilateral common carotid arteries, and then the rats were treated with a Xiao-Xu-Ming decoction (XXMD). Lipidomic profiling was conducted in both plasma and brain o determine the effects of the injury and therapy on lipid metabolism. Sphingolipid (particularly long acyl chain and total ceramides), glyceryl phosphatide, and glyceride profiles significantly changed in the brain after model induction and again after dosing. A total of 35 potential biomarkers were found in the brain and four were found in the plasma, representing both CCI injury and XXMD action. Correlations between endogenous lipids and exogenous XXMD compounds were analyzed using linear regression. Two exogenous compounds (cimifugin and 5-O-methylvisamminol) in the brain and 17 exogenous compounds in the plasma, which may represent the active constituents in XXMD, were significantly associated with lipid metabolism. This study provides a new perspective on the potential mechanism of CCI and its treatment with XXMD, as well as on discovering effective components in traditional Chinese medicines.
Collapse
Affiliation(s)
| | | | | | | | - Jinlan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
Everett JR. Pharmacometabonomics: The Prediction of Drug Effects Using Metabolic Profiling. Handb Exp Pharmacol 2019; 260:263-299. [PMID: 31823071 DOI: 10.1007/164_2019_316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metabonomics, also known as metabolomics, is concerned with the study of metabolite profiles in humans, animals, plants and other systems in order to assess their health or other status and their responses to experimental interventions. Metabonomics is thus widely used in disease diagnosis and in understanding responses to therapies such as drug administration. Pharmacometabonomics, also known as pharmacometabolomics, is a related methodology but with a prognostic as opposed to diagnostic thrust. Pharmacometabonomics aims to predict drug effects including efficacy, safety, metabolism and pharmacokinetics, prior to drug administration, via an analysis of pre-dose metabolite profiles. This article will review the development of pharmacometabonomics as a new field of science that has much promise in helping to deliver more effective personalised medicine, a major goal of twenty-first century healthcare.
Collapse
Affiliation(s)
- Jeremy R Everett
- Medway Metabonomics Research Group, University of Greenwich, Kent, UK.
| |
Collapse
|
25
|
Balashova EE, Maslov DL, Lokhov PG. A Metabolomics Approach to Pharmacotherapy Personalization. J Pers Med 2018; 8:jpm8030028. [PMID: 30189667 PMCID: PMC6164342 DOI: 10.3390/jpm8030028] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/17/2018] [Accepted: 09/03/2018] [Indexed: 12/27/2022] Open
Abstract
The optimization of drug therapy according to the personal characteristics of patients is a perspective direction in modern medicine. One of the possible ways to achieve such personalization is through the application of "omics" technologies, including current, promising metabolomics methods. This review demonstrates that the analysis of pre-dose metabolite biofluid profiles allows clinicians to predict the effectiveness of a selected drug treatment for a given individual. In the review, it is also shown that the monitoring of post-dose metabolite profiles could allow clinicians to evaluate drug efficiency, the reaction of the host to the treatment, and the outcome of the therapy. A comparative description of pharmacotherapy personalization (pharmacogenomics, pharmacoproteomics, and therapeutic drug monitoring) and personalization based on the analysis of metabolite profiles for biofluids (pharmacometabolomics) is also provided.
Collapse
Affiliation(s)
- Elena E Balashova
- Institute of Biomedical Chemistry, Pogodinskaya St. 10, Moscow 119121, Russia.
| | - Dmitry L Maslov
- Institute of Biomedical Chemistry, Pogodinskaya St. 10, Moscow 119121, Russia.
| | - Petr G Lokhov
- Institute of Biomedical Chemistry, Pogodinskaya St. 10, Moscow 119121, Russia.
| |
Collapse
|
26
|
Regulation of endogenic metabolites by rosuvastatin in hyperlipidemia patients: An integration of metabolomics and lipidomics. Chem Phys Lipids 2018; 214:69-83. [DOI: 10.1016/j.chemphyslip.2018.05.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/25/2018] [Accepted: 05/27/2018] [Indexed: 01/13/2023]
|
27
|
Shahin MH, Gong Y, Frye RF, Rotroff DM, Beitelshees AL, Baillie RA, Chapman AB, Gums JG, Turner ST, Boerwinkle E, Motsinger-Reif A, Fiehn O, Cooper-DeHoff RM, Han X, Kaddurah-Daouk R, Johnson JA. Sphingolipid Metabolic Pathway Impacts Thiazide Diuretics Blood Pressure Response: Insights From Genomics, Metabolomics, and Lipidomics. J Am Heart Assoc 2017; 7:e006656. [PMID: 29288159 PMCID: PMC5778957 DOI: 10.1161/jaha.117.006656] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 11/01/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND Although hydrochlorothiazide (HCTZ) is a well-established first-line antihypertensive in the United States, <50% of HCTZ treated patients achieve blood pressure (BP) control. Thus, identifying biomarkers that could predict the BP response to HCTZ is critically important. In this study, we utilized metabolomics, genomics, and lipidomics to identify novel pathways and biomarkers associated with HCTZ BP response. METHODS AND RESULTS First, we conducted a pathway analysis for 13 metabolites we recently identified to be significantly associated with HCTZ BP response. From this analysis, we found the sphingolipid metabolic pathway as the most significant pathway (P=5.8E-05). Testing 78 variants, within 14 genes involved in the sphingolipid metabolic canonical pathway, with the BP response to HCTZ identified variant rs6078905, within the SPTLC3 gene, as a novel biomarker significantly associated with the BP response to HCTZ in whites (n=228). We found that rs6078905 C-allele carriers had a better BP response to HCTZ versus noncarriers (∆SBP/∆DBP: -11.4/-6.9 versus -6.8/-3.5 mm Hg; ∆SBP P=6.7E-04; ∆DBP P=4.8E-04). Additionally, in blacks (n=148), we found genetic signals in the SPTLC3 genomic region significantly associated with the BP response to HCTZ (P<0.05). Last, we observed that rs6078905 significantly affects the baseline level of 4 sphingomyelins (N24:2, N24:3, N16:1, and N22:1; false discovery rate <0.05), from which N24:2 sphingomyelin has a significant correlation with both HCTZ DBP-response (r=-0.42; P=7E-03) and SBP-response (r=-0.36; P=2E-02). CONCLUSIONS This study provides insight into potential pharmacometabolomic and genetic mechanisms underlying HCTZ BP response and suggests that SPTLC3 is a potential determinant of the BP response to HCTZ. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00246519.
Collapse
Affiliation(s)
- Mohamed H Shahin
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Reginald F Frye
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Daniel M Rotroff
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC
| | | | | | | | - John G Gums
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | | | - Eric Boerwinkle
- Human Genetics Center and Institute for Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | | | - Oliver Fiehn
- Genome Center, University of California at Davis, CA
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi-Arabia
| | - Rhonda M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| | - Xianlin Han
- Sanford-Burnham Medical Research Institute, Orlando, FL
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioural Sciences and Department of Medicine, Duke University, Durham, NC
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, FL
| |
Collapse
|
28
|
Everett JR. NMR-based pharmacometabonomics: A new paradigm for personalised or precision medicine. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2017; 102-103:1-14. [PMID: 29157489 DOI: 10.1016/j.pnmrs.2017.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 04/23/2017] [Accepted: 04/24/2017] [Indexed: 06/07/2023]
Abstract
Metabolic profiling by NMR spectroscopy or hyphenated mass spectrometry, known as metabonomics or metabolomics, is an important tool for systems-based approaches in biology and medicine. The experiments are typically done in a diagnostic fashion where changes in metabolite profiles are interpreted as a consequence of an intervention or event; be that a change in diet, the administration of a drug, physical exertion or the onset of a disease. By contrast, pharmacometabonomics takes a prognostic approach to metabolic profiling, in order to predict the effects of drug dosing before it occurs. Differences in pre-dose metabolite profiles between groups of subjects are used to predict post-dose differences in response to drug administration. Thus the paradigm is inverted and pharmacometabonomics is the metabolic equivalent of pharmacogenomics. Although the field is still in its infancy, it is expected that pharmacometabonomics, alongside pharmacogenomics, will assist with the delivery of personalised or precision medicine to patients, which is a critical goal of 21st century healthcare.
Collapse
Affiliation(s)
- Jeremy R Everett
- Medway Metabonomics Group, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK.
| |
Collapse
|
29
|
Novel Applications of Metabolomics in Personalized Medicine: A Mini-Review. Molecules 2017; 22:molecules22071173. [PMID: 28703775 PMCID: PMC6152045 DOI: 10.3390/molecules22071173] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 12/20/2022] Open
Abstract
Interindividual variability in drug responses and disease susceptibility is common in the clinic. Currently, personalized medicine is highly valued, the idea being to prescribe the right medicine to the right patient. Metabolomics has been increasingly applied in evaluating the therapeutic outcomes of clinical drugs by correlating the baseline metabolic profiles of patients with their responses, i.e., pharmacometabonomics, as well as prediction of disease susceptibility among population in advance, i.e., patient stratification. The accelerated advance in metabolomics technology pinpoints the huge potential of its application in personalized medicine. In current review, we discussed the novel applications of metabolomics with typical examples in evaluating drug therapy and patient stratification, and underlined the potential of metabolomics in personalized medicine in the future.
Collapse
|
30
|
He X, Zheng N, He J, Liu C, Feng J, Jia W, Li H. Gut Microbiota Modulation Attenuated the Hypolipidemic Effect of Simvastatin in High-Fat/Cholesterol-Diet Fed Mice. J Proteome Res 2017; 16:1900-1910. [PMID: 28378586 PMCID: PMC5687503 DOI: 10.1021/acs.jproteome.6b00984] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The hypolipidemic effect of simvastatin varies greatly among patients. In the current study, we investigated the gut microbial-involved mechanisms underlying the different responses to simvastatin. Male C57BL/6J mice were divided into control (Con), high-fat/cholesterol diet (HFD), antibiotic (AB), simvastatin (SV) and antibiotic_simvastatin (AB_SV) groups, respectively. At the end of the experiment, serum samples were collected for lipids and metabolomic analysis, and liver tissues for histology, gene and protein expression analysis. The results showed that antibiotic treatment not only altered the composition of gut microbiota, but attenuated the hypolipidemic effect of SV. A total of 16 differential metabolites between SV and HFD groups were identified with metabolomics, while most of them showed no statistical differences between AB_SV and HFD groups, and similar changes were also observed in bile acids profile. The expressions of several genes and proteins involved in regulating bile acids synthesis were significantly reversed by SV, but not AB_SV in HFD fed mice. In summary, our current study indicated that the hypolipidemic effect of SV was correlated with the composition of the gut microbiota, and the attenuated hypolipidemic effect of SV by gut microbiota modulation was associated with a suppression of bile acids synthesis from cholesterol.
Collapse
Affiliation(s)
- Xuyun He
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ningning Zheng
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaojiao He
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Can Liu
- Laboratory medicine of Southern Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Jing Feng
- Laboratory medicine of Southern Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Wei Jia
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, 96813, USA
| | - Houkai Li
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
31
|
Kulkarni H, Mamtani M, Blangero J, Curran JE. Lipidomics in the Study of Hypertension in Metabolic Syndrome. Curr Hypertens Rep 2017; 19:7. [PMID: 28168678 DOI: 10.1007/s11906-017-0705-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
32
|
Elbadawi-Sidhu M, Baillie RA, Zhu H, Chen YDI, Goodarzi MO, Rotter JI, Krauss RM, Fiehn O, Kaddurah-Daouk R. Pharmacometabolomic signature links simvastatin therapy and insulin resistance. Metabolomics 2017; 13:11. [PMID: 29732238 PMCID: PMC5931366 DOI: 10.1007/s11306-016-1141-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 11/26/2016] [Indexed: 02/05/2023]
Abstract
Introduction Statins, widely prescribed drugs for treatment of cardiovascular disease, inhibit the biosynthesis of low density lipoprotein cholesterol (LDL-C). Despite providing major benefits, sub populations of patients experience adverse effects, including muscle myopathy and development of type II diabetes mellitus (T2DM) that may result in premature discontinuation of treatment. There are no reliable biomarkers for predicting clinical side effects in vulnerable individuals. Pharmacometabolomics provides powerful tools for identifying global biochemical changes induced by statin treatment, providing insights about drug mechanism of action, development of side effects and basis of variation of response. Objective To determine whether statin-induced changes in intermediary metabolism correlated with statin-induced hyperglycemia and insulin resistance; to identify pre-drug treatment metabolites predictive of post-drug treatment increased diabetic risk. Methods Drug-naïve patients were treated with 40 mg/day simvastatin for 6 weeks in the Cholesterol and Pharmacogenetics (CAP) study; metabolomics by gas chromatography-time-of-flight mass-spectrometry (GC-TOF-MS) was performed on plasma pre and post treatment on 148 of the 944 participants. Results Six weeks of simvastatin treatment resulted in 6.9% of patients developing hyperglycemia and 25% developing changes consistent with development of pre-diabetes. Altered beta cell function was observed in 53% of patients following simvastatin therapy and insulin resistance was observed in 54% of patients. We identified initial signature of simvastatin-induced insulin resistance, including ethanolamine, hydroxylamine, hydroxycarbamate and isoleucine which, upon further replication and expansion, could be predictive biomarkers of individual susceptibility to simvastatin-induced new onset pre-type II diabetes mellitus. No patients were clinically diagnosed with T2DM. Conclusion Within this short 6 weeks study, some patients became hyperglycemic and/or insulin resistant. Diabetic markers were associated with decarboxylated small aminated metabolites as well as a branched chain amino acid directly linked to glucose metabolism and fatty acid biosynthesis. Pharmacometabolomics provides powerful tools for precision medicine by predicting development of drug adverse effects in sub populations of patients. Metabolic profiling prior to start of drug therapy may empower physicians with critical information when prescribing medication and determining prognosis.
Collapse
Affiliation(s)
- Mona Elbadawi-Sidhu
- West Coast Metabolomics Center, Genome Center, University of California - Davis, Davis, CA, USA
| | | | - Hongjie Zhu
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research, Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research, Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ronald M Krauss
- Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, Genome Center, University of California - Davis, Davis, CA, USA
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rima Kaddurah-Daouk
- Department of Internal Medicine; Department of Psychiatry and Behavioral Sciences, Duke Institute for Brain Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
33
|
Wu Q, Lai XL, Zhao HX, Zhu ZY, Hong ZY, Guo ZY, Chai YF. A metabolomics approach for predicting the response to intravenous iron therapy in peritoneal dialysis patients with anemia. RSC Adv 2017. [DOI: 10.1039/c6ra24152b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Anemia is an almost universal complication of chronic kidney disease (CKD), and nearly all patients with end-stage renal disease (ESRD) and approximately 70% of those with earlier stages of CKD receive treatment for anemia.
Collapse
Affiliation(s)
- Qiong Wu
- School of Pharmacy
- Second Military Medical University
- Shanghai
- China 200433
| | - Xue-li Lai
- Department of Nephrology
- Changhai Hospital
- Second Military Medical University
- Shanghai
- China 200433
| | - Hong-xia Zhao
- Analysis and Measurement Center
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Zhen-yu Zhu
- Analysis and Measurement Center
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Zhan-ying Hong
- School of Pharmacy
- Second Military Medical University
- Shanghai
- China 200433
| | - Zhi-yong Guo
- Department of Nephrology
- Changhai Hospital
- Second Military Medical University
- Shanghai
- China 200433
| | - Yi-feng Chai
- School of Pharmacy
- Second Military Medical University
- Shanghai
- China 200433
| |
Collapse
|
34
|
Maslov D, Balashova E, Lokhov P, Archakov A. Pharmacometabonomics – the novel way to personalized drug therapy. ACTA ACUST UNITED AC 2017; 63:115-123. [DOI: 10.18097/pbmc20176302115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review is devoted to pharmacometabonomics - a new branch of science focused on personalization of drug therapy through the comprehensive analysis of metabolites of patient's biological fluids. It considers the history of pharmacometabonomic, positioning to other “-omic” sciences, and system approach, realized by this science, in determination of individual therapeutic dose of the drugs and also a technical implementation of pharmacometabonomic based on direct mass spectrometry of blood plasma metabolites. Special attention is paid to a comparative analysis of pharmacometabonomics and other main approaches to personalized therapy in the clinic, such as pharmacogenetics and therapeutic drug monitoring. Finally, prospects of pharmacometabonomics applications in clinical practice were also discussed.
Collapse
Affiliation(s)
- D.L. Maslov
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | - P.G. Lokhov
- Institute of Biomedical Chemistry, Moscow, Russia
| | | |
Collapse
|
35
|
Sethi S, Brietzke E. Recent advances in lipidomics: Analytical and clinical perspectives. Prostaglandins Other Lipid Mediat 2017; 128-129:8-16. [DOI: 10.1016/j.prostaglandins.2016.12.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 10/20/2022]
|
36
|
Kantae V, Krekels EHJ, Esdonk MJV, Lindenburg P, Harms AC, Knibbe CAJ, Van der Graaf PH, Hankemeier T. Integration of pharmacometabolomics with pharmacokinetics and pharmacodynamics: towards personalized drug therapy. Metabolomics 2016; 13:9. [PMID: 28058041 PMCID: PMC5165030 DOI: 10.1007/s11306-016-1143-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/26/2016] [Indexed: 02/05/2023]
Abstract
Personalized medicine, in modern drug therapy, aims at a tailored drug treatment accounting for inter-individual variations in drug pharmacology to treat individuals effectively and safely. The inter-individual variability in drug response upon drug administration is caused by the interplay between drug pharmacology and the patients' (patho)physiological status. Individual variations in (patho)physiological status may result from genetic polymorphisms, environmental factors (including current/past treatments), demographic characteristics, and disease related factors. Identification and quantification of predictors of inter-individual variability in drug pharmacology is necessary to achieve personalized medicine. Here, we highlight the potential of pharmacometabolomics in prospectively informing on the inter-individual differences in drug pharmacology, including both pharmacokinetic (PK) and pharmacodynamic (PD) processes, and thereby guiding drug selection and drug dosing. This review focusses on the pharmacometabolomics studies that have additional value on top of the conventional covariates in predicting drug PK. Additionally, employing pharmacometabolomics to predict drug PD is highlighted, and we suggest not only considering the endogenous metabolites as static variables but to include also drug dose and temporal changes in drug concentration in these studies. Although there are many endogenous metabolite biomarkers identified to predict PK and more often to predict PD, validation of these biomarkers in terms of specificity, sensitivity, reproducibility and clinical relevance is highly important. Furthermore, the application of these identified biomarkers in routine clinical practice deserves notable attention to truly personalize drug treatment in the near future.
Collapse
Affiliation(s)
- Vasudev Kantae
- Division of Analytical Biosciences, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Elke H. J. Krekels
- Division of Pharmacology, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Michiel J. Van Esdonk
- Division of Pharmacology, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Peter Lindenburg
- Division of Analytical Biosciences, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Amy C. Harms
- Division of Analytical Biosciences, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Catherijne A. J. Knibbe
- Division of Pharmacology, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Piet H. Van der Graaf
- Division of Pharmacology, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Certara QSP, Canterbury Innovation Centre, Canterbury, UK
| | - Thomas Hankemeier
- Division of Analytical Biosciences, Systems Pharmacology Cluster, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
37
|
Rankin NJ, Preiss D, Welsh P, Sattar N. Applying metabolomics to cardiometabolic intervention studies and trials: past experiences and a roadmap for the future. Int J Epidemiol 2016; 45:1351-1371. [PMID: 27789671 PMCID: PMC5100629 DOI: 10.1093/ije/dyw271] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2016] [Indexed: 12/22/2022] Open
Abstract
Metabolomics and lipidomics are emerging methods for detailed phenotyping of small molecules in samples. It is hoped that such data will: (i) enhance baseline prediction of patient response to pharmacotherapies (beneficial or adverse); (ii) reveal changes in metabolites shortly after initiation of therapy that may predict patient response, including adverse effects, before routine biomarkers are altered; and( iii) give new insights into mechanisms of drug action, particularly where the results of a trial of a new agent were unexpected, and thus help future drug development. In these ways, metabolomics could enhance research findings from intervention studies. This narrative review provides an overview of metabolomics and lipidomics in early clinical intervention studies for investigation of mechanisms of drug action and prediction of drug response (both desired and undesired). We highlight early examples from drug intervention studies associated with cardiometabolic disease. Despite the strengths of such studies, particularly the use of state-of-the-art technologies and advanced statistical methods, currently published studies in the metabolomics arena are largely underpowered and should be considered as hypothesis-generating. In order for metabolomics to meaningfully improve stratified medicine approaches to patient treatment, there is a need for higher quality studies, with better exploitation of biobanks from randomized clinical trials i.e. with large sample size, adjudicated outcomes, standardized procedures, validation cohorts, comparison witth routine biochemistry and both active and control/placebo arms. On the basis of this review, and based on our research experience using clinically established biomarkers, we propose steps to more speedily advance this area of research towards potential clinical impact.
Collapse
Affiliation(s)
- Naomi J Rankin
- BHF Glasgow Cardiovascular Research Centre
- Glasgow Polyomics, University of Glasgow, Glasgow, UK
| | - David Preiss
- Clinical Trials Service Unit and Epidemiological Studies Unit, University of Oxford, Oxford, UK
| | - Paul Welsh
- BHF Glasgow Cardiovascular Research Centre
| | | |
Collapse
|
38
|
Everett JR. From Metabonomics to Pharmacometabonomics: The Role of Metabolic Profiling in Personalized Medicine. Front Pharmacol 2016; 7:297. [PMID: 27660611 PMCID: PMC5014868 DOI: 10.3389/fphar.2016.00297] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/23/2016] [Indexed: 01/08/2023] Open
Abstract
Variable patient responses to drugs are a key issue for medicine and for drug discovery and development. Personalized medicine, that is the selection of medicines for subgroups of patients so as to maximize drug efficacy and minimize toxicity, is a key goal of twenty-first century healthcare. Currently, most personalized medicine paradigms rely on clinical judgment based on the patient's history, and on the analysis of the patients' genome to predict drug effects i.e., pharmacogenomics. However, variability in patient responses to drugs is dependent upon many environmental factors to which human genomics is essentially blind. A new paradigm for predicting drug responses based on individual pre-dose metabolite profiles has emerged in the past decade: pharmacometabonomics, which is defined as “the prediction of the outcome (for example, efficacy or toxicity) of a drug or xenobiotic intervention in an individual based on a mathematical model of pre-intervention metabolite signatures.” The new pharmacometabonomics paradigm is complementary to pharmacogenomics but has the advantage of being sensitive to environmental as well as genomic factors. This review will chart the discovery and development of pharmacometabonomics, and provide examples of its current utility and possible future developments.
Collapse
Affiliation(s)
- Jeremy R Everett
- Medway Metabonomics Research Group, University of Greenwich Kent, UK
| |
Collapse
|
39
|
Beger RD, Dunn W, Schmidt MA, Gross SS, Kirwan JA, Cascante M, Brennan L, Wishart DS, Oresic M, Hankemeier T, Broadhurst DI, Lane AN, Suhre K, Kastenmüller G, Sumner SJ, Thiele I, Fiehn O, Kaddurah-Daouk R, for “Precision Medicine and Pharmacometabolomics Task Group”-Metabolomics Society Initiative. Metabolomics enables precision medicine: "A White Paper, Community Perspective". Metabolomics 2016; 12:149. [PMID: 27642271 PMCID: PMC5009152 DOI: 10.1007/s11306-016-1094-6] [Citation(s) in RCA: 405] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/08/2016] [Indexed: 01/12/2023]
Abstract
INTRODUCTION BACKGROUND TO METABOLOMICS Metabolomics is the comprehensive study of the metabolome, the repertoire of biochemicals (or small molecules) present in cells, tissues, and body fluids. The study of metabolism at the global or "-omics" level is a rapidly growing field that has the potential to have a profound impact upon medical practice. At the center of metabolomics, is the concept that a person's metabolic state provides a close representation of that individual's overall health status. This metabolic state reflects what has been encoded by the genome, and modified by diet, environmental factors, and the gut microbiome. The metabolic profile provides a quantifiable readout of biochemical state from normal physiology to diverse pathophysiologies in a manner that is often not obvious from gene expression analyses. Today, clinicians capture only a very small part of the information contained in the metabolome, as they routinely measure only a narrow set of blood chemistry analytes to assess health and disease states. Examples include measuring glucose to monitor diabetes, measuring cholesterol and high density lipoprotein/low density lipoprotein ratio to assess cardiovascular health, BUN and creatinine for renal disorders, and measuring a panel of metabolites to diagnose potential inborn errors of metabolism in neonates. OBJECTIVES OF WHITE PAPER—EXPECTED TREATMENT OUTCOMES AND METABOLOMICS ENABLING TOOL FOR PRECISION MEDICINE We anticipate that the narrow range of chemical analyses in current use by the medical community today will be replaced in the future by analyses that reveal a far more comprehensive metabolic signature. This signature is expected to describe global biochemical aberrations that reflect patterns of variance in states of wellness, more accurately describe specific diseases and their progression, and greatly aid in differential diagnosis. Such future metabolic signatures will: (1) provide predictive, prognostic, diagnostic, and surrogate markers of diverse disease states; (2) inform on underlying molecular mechanisms of diseases; (3) allow for sub-classification of diseases, and stratification of patients based on metabolic pathways impacted; (4) reveal biomarkers for drug response phenotypes, providing an effective means to predict variation in a subject's response to treatment (pharmacometabolomics); (5) define a metabotype for each specific genotype, offering a functional read-out for genetic variants: (6) provide a means to monitor response and recurrence of diseases, such as cancers: (7) describe the molecular landscape in human performance applications and extreme environments. Importantly, sophisticated metabolomic analytical platforms and informatics tools have recently been developed that make it possible to measure thousands of metabolites in blood, other body fluids, and tissues. Such tools also enable more robust analysis of response to treatment. New insights have been gained about mechanisms of diseases, including neuropsychiatric disorders, cardiovascular disease, cancers, diabetes and a range of pathologies. A series of ground breaking studies supported by National Institute of Health (NIH) through the Pharmacometabolomics Research Network and its partnership with the Pharmacogenomics Research Network illustrate how a patient's metabotype at baseline, prior to treatment, during treatment, and post-treatment, can inform about treatment outcomes and variations in responsiveness to drugs (e.g., statins, antidepressants, antihypertensives and antiplatelet therapies). These studies along with several others also exemplify how metabolomics data can complement and inform genetic data in defining ethnic, sex, and gender basis for variation in responses to treatment, which illustrates how pharmacometabolomics and pharmacogenomics are complementary and powerful tools for precision medicine. CONCLUSIONS KEY SCIENTIFIC CONCEPTS AND RECOMMENDATIONS FOR PRECISION MEDICINE Our metabolomics community believes that inclusion of metabolomics data in precision medicine initiatives is timely and will provide an extremely valuable layer of data that compliments and informs other data obtained by these important initiatives. Our Metabolomics Society, through its "Precision Medicine and Pharmacometabolomics Task Group", with input from our metabolomics community at large, has developed this White Paper where we discuss the value and approaches for including metabolomics data in large precision medicine initiatives. This White Paper offers recommendations for the selection of state of-the-art metabolomics platforms and approaches that offer the widest biochemical coverage, considers critical sample collection and preservation, as well as standardization of measurements, among other important topics. We anticipate that our metabolomics community will have representation in large precision medicine initiatives to provide input with regard to sample acquisition/preservation, selection of optimal omics technologies, and key issues regarding data collection, interpretation, and dissemination. We strongly recommend the collection and biobanking of samples for precision medicine initiatives that will take into consideration needs for large-scale metabolic phenotyping studies.
Collapse
Affiliation(s)
- Richard D. Beger
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079 USA
| | - Warwick Dunn
- School of Biosciences, Phenome Centre Birmingham and Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Michael A. Schmidt
- Advanced Pattern Analysis and Countermeasures Group, Research Innovation Center, Colorado State University, Fort Collins, CO 80521 USA
| | - Steven S. Gross
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10021 USA
| | - Jennifer A. Kirwan
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain
- Institute of Biomedicine of Universitat de Barcelona (IBUB) and CSIC-Associated Unit, Barcelona, Spain
| | | | - David S. Wishart
- Departments of Computing Science and Biological Sciences, University of Alberta, Edmonton, AB Canada
| | - Matej Oresic
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Thomas Hankemeier
- Division of Analytical Biosciences and Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University & Netherlands Metabolomics Centre, Leiden, The Netherlands
| | | | - Andrew N. Lane
- Center for Environmental Systems Biochemistry, Department Toxicology and Cancer Biology, Markey Cancer Center, Lexington, KY USA
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Doha, Qatar
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Center Munich, Oberschleißheim, Germany
| | - Susan J. Sumner
- Discovery Sciences, RTI International, Research Triangle Park, Durham, NC USA
| | - Ines Thiele
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, Campus Belval, Esch-Sur-Alzette, Luxembourg
| | - Oliver Fiehn
- West Coast Metabolomics Center, UC Davis, Davis, CA USA
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rima Kaddurah-Daouk
- Psychiatry and Behavioral Sciences, Duke Internal Medicine and Duke Institute for Brain Sciences and Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Box 3903, Durham, NC 27710 USA
| | - for “Precision Medicine and Pharmacometabolomics Task Group”-Metabolomics Society Initiative
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079 USA
- School of Biosciences, Phenome Centre Birmingham and Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
- Advanced Pattern Analysis and Countermeasures Group, Research Innovation Center, Colorado State University, Fort Collins, CO 80521 USA
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10021 USA
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Av Diagonal 643, 08028 Barcelona, Spain
- Institute of Biomedicine of Universitat de Barcelona (IBUB) and CSIC-Associated Unit, Barcelona, Spain
- UCD Institute of Food and Health, UCD, Belfield, Dublin Ireland
- Departments of Computing Science and Biological Sciences, University of Alberta, Edmonton, AB Canada
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
- Division of Analytical Biosciences and Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University & Netherlands Metabolomics Centre, Leiden, The Netherlands
- School of Science, Edith Cowan University, Perth, Australia
- Center for Environmental Systems Biochemistry, Department Toxicology and Cancer Biology, Markey Cancer Center, Lexington, KY USA
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Doha, Qatar
- Institute of Bioinformatics and Systems Biology, Helmholtz Center Munich, Oberschleißheim, Germany
- Discovery Sciences, RTI International, Research Triangle Park, Durham, NC USA
- University of Luxembourg, Luxembourg Centre for Systems Biomedicine, Campus Belval, Esch-Sur-Alzette, Luxembourg
- West Coast Metabolomics Center, UC Davis, Davis, CA USA
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
- Psychiatry and Behavioral Sciences, Duke Internal Medicine and Duke Institute for Brain Sciences and Center for Applied Genomics and Precision Medicine, Duke University Medical Center, Box 3903, Durham, NC 27710 USA
| |
Collapse
|
40
|
Burt T, Nandal S. Pharmacometabolomics in Early-Phase Clinical Development. Clin Transl Sci 2016; 9:128-38. [PMID: 27127917 PMCID: PMC5351331 DOI: 10.1111/cts.12396] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/01/2016] [Indexed: 12/28/2022] Open
Affiliation(s)
- T Burt
- Burt Consultancy, Durham, North Carolina, USA
| | - S Nandal
- Department of Medical Oncology Novartis (Singapore) Pte Ltd, Singapore
| |
Collapse
|
41
|
Jutley GS, Young SP. Metabolomics to identify biomarkers and as a predictive tool in inflammatory diseases. Best Pract Res Clin Rheumatol 2016; 29:770-82. [PMID: 27107512 DOI: 10.1016/j.berh.2016.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
There is an overwhelming need for a simple, reliable tool that aids clinicians in diagnosing, assessing disease activity and treating rheumatic conditions. Identification of biomarkers in partially understood inflammatory disorders has long been sought after as the Holy Grail of Rheumatology. Given the complex nature of inflammatory conditions, it has been difficult to earmark the potential biomarkers. Metabolomics, however, is promising in providing new insights into inflammatory conditions and also identifying such biomarkers. Metabolomic studies have generally revealed increased energy requirements for by-products of a hypoxic environment, leading to a characteristic metabolic fingerprint. Here, we discuss the significance of such studies and their potential as a biomarker.
Collapse
Affiliation(s)
- Gurpreet Singh Jutley
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Stephen P Young
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
42
|
Deidda M, Piras C, Bassareo PP, Cadeddu Dessalvi C, Mercuro G. Metabolomics, a promising approach to translational research in cardiology. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.ijcme.2015.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
43
|
Ellero-Simatos S, Beitelshees AL, Lewis JP, Yerges-Armstrong LM, Georgiades A, Dane A, Harms AC, Strassburg K, Guled F, Hendriks MMWB, Horenstein RB, Shuldiner AR, Hankemeier T, Kaddurah-Daouk R. Oxylipid Profile of Low-Dose Aspirin Exposure: A Pharmacometabolomics Study. J Am Heart Assoc 2015; 4:e002203. [PMID: 26504148 PMCID: PMC4845113 DOI: 10.1161/jaha.115.002203] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background While aspirin is a well‐established and generally effective anti‐platelet agent, considerable inter‐individual variation in drug response exists, for which mechanisms are not completely understood. Metabolomics allows for extensive measurement of small molecules in biological samples, enabling detailed mapping of pathways involved in drug response. Methods and Results We used a mass‐spectrometry‐based metabolomics platform to investigate the changes in the serum oxylipid metabolome induced by an aspirin intervention (14 days, 81 mg/day) in healthy subjects (n=156). We observed a global decrease in serum oxylipids in response to aspirin (25 metabolites decreased out of 30 measured) regardless of sex. This decrease was concomitant with a significant decrease in serum linoleic acid levels (−19%, P=1.3×10−5), one of the main precursors for oxylipid synthesis. Interestingly, several linoleic acid‐derived oxylipids were not significantly associated with arachidonic‐induced ex vivo platelet aggregation, a widely accepted marker of aspirin response, but were significantly correlated with platelet reactivity in response to collagen. Conclusions Together, these results suggest that linoleic acid‐derived oxylipids may contribute to the non‐COX1 mediated variability in response to aspirin. Pharmacometabolomics allowed for more comprehensive interrogation of mechanisms of action of low dose aspirin and of variation in aspirin response.
Collapse
Affiliation(s)
- Sandrine Ellero-Simatos
- Analytical Biosciences Division, Leiden Academic Centre for Drug Research, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.) Netherlands Metabolomics Centre, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.)
| | - Amber L Beitelshees
- Program Personalized and Genomic Medicine, Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD (A.L.B., J.P.L., L.M.Y.A., R.B.H., A.R.S.)
| | - Joshua P Lewis
- Program Personalized and Genomic Medicine, Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD (A.L.B., J.P.L., L.M.Y.A., R.B.H., A.R.S.)
| | - Laura M Yerges-Armstrong
- Program Personalized and Genomic Medicine, Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD (A.L.B., J.P.L., L.M.Y.A., R.B.H., A.R.S.)
| | | | - Adrie Dane
- Analytical Biosciences Division, Leiden Academic Centre for Drug Research, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.) Netherlands Metabolomics Centre, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.)
| | - Amy C Harms
- Analytical Biosciences Division, Leiden Academic Centre for Drug Research, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.) Netherlands Metabolomics Centre, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.)
| | - Katrin Strassburg
- Analytical Biosciences Division, Leiden Academic Centre for Drug Research, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.) Netherlands Metabolomics Centre, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.)
| | - Faisa Guled
- Analytical Biosciences Division, Leiden Academic Centre for Drug Research, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.) Netherlands Metabolomics Centre, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.)
| | - Margriet M W B Hendriks
- Analytical Biosciences Division, Leiden Academic Centre for Drug Research, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.) Netherlands Metabolomics Centre, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.)
| | - Richard B Horenstein
- Program Personalized and Genomic Medicine, Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD (A.L.B., J.P.L., L.M.Y.A., R.B.H., A.R.S.)
| | - Alan R Shuldiner
- Program Personalized and Genomic Medicine, Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD (A.L.B., J.P.L., L.M.Y.A., R.B.H., A.R.S.)
| | - Thomas Hankemeier
- Analytical Biosciences Division, Leiden Academic Centre for Drug Research, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.) Netherlands Metabolomics Centre, Leiden, The Netherlands (S.E.S., A.D., A.C.H., K.S., F.G., M.B.H., T.H.)
| | - Rima Kaddurah-Daouk
- Duke University Medical Center, Durham, NC (A.G., R.K.D.) Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC (R.K.D.) Duke Institute for Brain Sciences, Duke University, Durham, NC (R.K.D.) Institute of Genome Science and Policy, Durham, NC (R.K.D.)
| | | |
Collapse
|
44
|
Visioli F. Lipidomics to Assess Omega 3 Bioactivity. J Clin Med 2015; 4:1753-60. [PMID: 26371049 PMCID: PMC4600157 DOI: 10.3390/jcm4091753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/19/2015] [Accepted: 08/31/2015] [Indexed: 11/16/2022] Open
Abstract
How can we resolve the conflict between the strong epidemiological evidence pointing to the usefulness of fish—and, thus, omega 3—consumption with the debacle of supplementation trials? One potential explanation is that the null results obtained thus far are the consequences of ill-contrived investigations that do not allow us to conclude on the effects (or lack thereof) of omega 3 fatty acid supplementation. One potential solution is through the use of lipidomics, which should prove very useful to screen suitable patients and to correlate plasma (or red blood cells, or whole blood, or phospholipid) fatty acid profile with outcomes. This has never been done in omega 3 trials. The wise use of lipidomics should be essential part of future omega 3 trials and would help in untangling this current riddle.
Collapse
Affiliation(s)
- Francesco Visioli
- Department of Molecular Medicine, University of Padova, Via 8 Febbraio, 2-35122 Padova, Italy.
| |
Collapse
|
45
|
Huang Q, Aa J, Jia H, Xin X, Tao C, Liu L, Zou B, Song Q, Shi J, Cao B, Yong Y, Wang G, Zhou G. A Pharmacometabonomic Approach To Predicting Metabolic Phenotypes and Pharmacokinetic Parameters of Atorvastatin in Healthy Volunteers. J Proteome Res 2015. [PMID: 26216528 DOI: 10.1021/acs.jproteome.5b00440] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Qing Huang
- China Pharmaceutical
University, Nanjing 210009, China
- Jiangsu Institute
for Food and Drug Control, Nanjing 210008, China
| | - Jiye Aa
- China Pharmaceutical
University, Nanjing 210009, China
| | - Huning Jia
- China Pharmaceutical
University, Nanjing 210009, China
- Department
of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Xiaoqing Xin
- China Pharmaceutical
University, Nanjing 210009, China
- Department
of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Chunlei Tao
- Anhui University
of Chinese Medicine, Hefei 230038, China
| | - Linsheng Liu
- Clinical
Pharmacology Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Bingjie Zou
- Department
of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Qinxin Song
- China Pharmaceutical
University, Nanjing 210009, China
| | - Jian Shi
- China Pharmaceutical
University, Nanjing 210009, China
| | - Bei Cao
- China Pharmaceutical
University, Nanjing 210009, China
| | - Yonghong Yong
- The First Affiliated
Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Guangji Wang
- China Pharmaceutical
University, Nanjing 210009, China
| | - Guohua Zhou
- Department
of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| |
Collapse
|
46
|
Hinterwirth H, Stegemann C, Mayr M. Lipidomics: quest for molecular lipid biomarkers in cardiovascular disease. ACTA ACUST UNITED AC 2015; 7:941-54. [PMID: 25516624 DOI: 10.1161/circgenetics.114.000550] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Lipidomics is the comprehensive analysis of molecular lipid species, including their quantitation and metabolic pathways. The huge diversity of native lipids and their modifications make lipidomic analyses challenging. The method of choice for sensitive detection and quantitation of molecular lipid species is mass spectrometry, either by direct infusion (shotgun lipidomics) or coupled with liquid chromatography. Although shotgun lipidomics allows for high-throughput analysis, low-abundant lipid species are not detected. Previous separation of lipid species by liquid chromatography increases ionization efficiency and is better suited for quantifying low abundant and isomeric lipid species. In this review, we will discuss the potential of lipidomics for cardiovascular research. To date, cardiovascular research predominantly focuses on the role of lipid classes rather than molecular entities. An in-depth knowledge about the molecular lipid species that contribute to the pathophysiology of cardiovascular diseases may provide better biomarkers and novel therapeutic targets for cardiovascular disease.
Collapse
Affiliation(s)
- Helmut Hinterwirth
- From the King's British Heart Foundation Centre, King's College, London, United Kingdom
| | - Christin Stegemann
- From the King's British Heart Foundation Centre, King's College, London, United Kingdom
| | - Manuel Mayr
- From the King's British Heart Foundation Centre, King's College, London, United Kingdom.
| |
Collapse
|
47
|
Abstract
Identifying the mechanisms that convert a healthy vascular wall to an atherosclerotic wall is of major importance since the consequences may lead to a shortened lifespan. Classical risk factors (age, smoking, obesity, diabetes mellitus, hypertension, and dyslipidemia) may result in the progression of atherosclerotic lesions by processes including inflammation and lipid accumulation. Thus, the evaluation of blood lipids and the full lipid complement produced by cells, organisms, or tissues (lipidomics) is an issue of importance. In this review, we shall describe the recent progress in vascular health research using lipidomic advances. We will begin with an overview of vascular wall biology and lipids, followed by a short analysis of lipidomics. Finally, we shall focus on the clinical implications of lipidomics and studies that have examined lipidomic approaches and vascular health.
Collapse
Affiliation(s)
- Genovefa Kolovou
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece
| | - Vana Kolovou
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece ; Molecular Immunology Laboratory, Onassis Cardiac Surgery Center, Athens, Greece
| | - Sophie Mavrogeni
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece
| |
Collapse
|
48
|
Kaddurah-Daouk R, Weinshilboum R. Metabolomic Signatures for Drug Response Phenotypes: Pharmacometabolomics Enables Precision Medicine. Clin Pharmacol Ther 2015; 98:71-5. [PMID: 25871646 DOI: 10.1002/cpt.134] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 03/31/2015] [Accepted: 03/31/2015] [Indexed: 12/15/2022]
Abstract
The scaling up of data in clinical pharmacology and the merger of systems biology and pharmacology has led to the emergence of a new discipline of Quantitative and Systems Pharmacology (QSP). This new research direction might significantly advance the discovery, development, and clinical use of therapeutic drugs. Research communities from computational biology, systems biology, and biological engineering--working collaboratively with pharmacologists, geneticists, biochemists, and analytical chemists--are creating and modeling large data on drug effects that is transforming our understanding of how these drugs work at a network level. In this review, we highlight developments in a new and rapidly growing field--pharmacometabolomics--in which large biochemical data-capturing effects of genome, gut microbiome, and environment exposures is revealing information about metabotypes and treatment outcomes, and creating metabolic signatures as new potential biomarkers. Pharmacometabolomics informs and complements pharmacogenomics and together they provide building blocks for QSP.
Collapse
Affiliation(s)
- R Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA.,Duke Institute for Brain Sciences, Duke University, Durham, North Carolina, USA
| | - R Weinshilboum
- Mayo Clinic, Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
49
|
Dehairs J, Derua R, Rueda-Rincon N, Swinnen JV. Lipidomics in drug development. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 13:33-38. [PMID: 26190681 DOI: 10.1016/j.ddtec.2015.03.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 03/25/2015] [Accepted: 03/26/2015] [Indexed: 06/04/2023]
Abstract
Numerous human pathologies, including common conditions such as obesity, diabetes, cardiovascular disease, cancer, inflammatory disease and neurodegeneration, involve changes in lipid metabolism. Likewise, a growing number of drugs are being developed that directly or indirectly affect lipid metabolic pathways. Instead of classical and cumbrous radiochemical analyses, lipid profiling by mass spectrometry (MS)-based lipidomics holds great potential as companion diagnostic in several steps along the drug development process. In this review we describe some typical lipidomics set-ups and illustrate how these technologies can be implemented in target discovery, compound screening, in vitro and in vivo preclinical testing, toxicity testing of drugs, and prediction and monitoring of response.
Collapse
Affiliation(s)
- Jonas Dehairs
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, B-3000 Leuven, Belgium
| | - Rita Derua
- KU Leuven - University of Leuven, Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, B-3000 Leuven, Belgium
| | - Natalia Rueda-Rincon
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, B-3000 Leuven, Belgium
| | - Johannes V Swinnen
- KU Leuven - University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, B-3000 Leuven, Belgium.
| |
Collapse
|
50
|
Lipidome and transcriptome profiling of pneumolysin intoxication identifies networks involved in statin-conferred protection of airway epithelial cells. Sci Rep 2015; 5:10624. [PMID: 26023727 PMCID: PMC4448502 DOI: 10.1038/srep10624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/22/2015] [Indexed: 12/12/2022] Open
Abstract
Pneumonia remains one of the leading causes of death in both adults and children worldwide. Despite the adoption of a wide variety of therapeutics, the mortality from community-acquired pneumonia has remained relatively constant. Although viral and fungal acute airway infections can result in pneumonia, bacteria are the most common cause of community-acquired pneumonia, with Streptococcus pneumoniae isolated in nearly 50% of cases. Pneumolysin is a cholesterol-dependent cytolysin or pore-forming toxin produced by Streptococcus pneumonia and has been shown to play a critical role in bacterial pathogenesis. Airway epithelium is the initial site of many bacterial contacts and its barrier and mucosal immunity functions are central to infectious lung diseases. In our studies, we have shown that the prior exposure to statins confers significant resistance of airway epithelial cells to the cytotoxicity of pneumolysin. We decided to take this study one step further, assessing changes in both the transcriptome and lipidome of human airway epithelial cells exposed to toxin, statin or both. Our current work provides the first global view in human airway epithelial cells of both the transcriptome and the lipid interactions that result in cellular protection from pneumolysin.
Collapse
|