1
|
Torta F, Hoffmann N, Burla B, Alecu I, Arita M, Bamba T, Bennett SAL, Bertrand-Michel J, Brügger B, Cala MP, Camacho-Muñoz D, Checa A, Chen M, Chocholoušková M, Cinel M, Chu-Van E, Colsch B, Coman C, Connell L, Sousa BC, Dickens AM, Fedorova M, Eiríksson FF, Gallart-Ayala H, Ghorasaini M, Giera M, Guan XL, Haid M, Hankemeier T, Harms A, Höring M, Holčapek M, Hornemann T, Hu C, Hülsmeier AJ, Huynh K, Jones CM, Ivanisevic J, Izumi Y, Köfeler HC, Lam SM, Lange M, Lee JC, Liebisch G, Lippa K, Lopez-Clavijo AF, Manzi M, Martinefski MR, Math RGH, Mayor S, Meikle PJ, Monge ME, Moon MH, Muralidharan S, Nicolaou A, Nguyen-Tran T, O'Donnell VB, Orešič M, Ramanathan A, Riols F, Saigusa D, Schock TB, Schwartz-Zimmermann H, Shui G, Singh M, Takahashi M, Thorsteinsdóttir M, Tomiyasu N, Tournadre A, Tsugawa H, Tyrrell VJ, van der Gugten G, Wakelam MO, Wheelock CE, Wolrab D, Xu G, Xu T, Bowden JA, Ekroos K, Ahrends R, Wenk MR. Concordant inter-laboratory derived concentrations of ceramides in human plasma reference materials via authentic standards. Nat Commun 2024; 15:8562. [PMID: 39362843 PMCID: PMC11449902 DOI: 10.1038/s41467-024-52087-x] [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: 11/26/2023] [Accepted: 08/27/2024] [Indexed: 10/05/2024] Open
Abstract
In this community effort, we compare measurements between 34 laboratories from 19 countries, utilizing mixtures of labelled authentic synthetic standards, to quantify by mass spectrometry four clinically used ceramide species in the NIST (National Institute of Standards and Technology) human blood plasma Standard Reference Material (SRM) 1950, as well as a set of candidate plasma reference materials (RM 8231). Participants either utilized a provided validated method and/or their method of choice. Mean concentration values, and intra- and inter-laboratory coefficients of variation (CV) were calculated using single-point and multi-point calibrations, respectively. These results are the most precise (intra-laboratory CVs ≤ 4.2%) and concordant (inter-laboratory CVs < 14%) community-derived absolute concentration values reported to date for four clinically used ceramides in the commonly analyzed SRM 1950. We demonstrate that calibration using authentic labelled standards dramatically reduces data variability. Furthermore, we show how the use of shared RM can correct systematic quantitative biases and help in harmonizing lipidomics. Collectively, the results from the present study provide a significant knowledge base for translation of lipidomic technologies to future clinical applications that might require the determination of reference intervals (RIs) in various human populations or might need to estimate reference change values (RCV), when analytical variability is a key factor for recall during multiple testing of individuals.
Collapse
Affiliation(s)
- Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore, 169857, Singapore
| | - Nils Hoffmann
- Institute for Bio- and Geosciences (IBG-5), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Bo Burla
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Irina Alecu
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Ottawa Brain and Mind Research Institute, Department of Biochemistry, Microbiology, and Immunology, and Department of Chemistry, Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, K1H 8M5, Canada
| | - Makoto Arita
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Takeshi Bamba
- Division of Metabolomics Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 3‑1‑1, Maidashi, Higashi‑ku, Fukuoka, 812‑8582, Japan
| | - Steffany A L Bennett
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Ottawa Brain and Mind Research Institute, Department of Biochemistry, Microbiology, and Immunology, and Department of Chemistry, Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, K1H 8M5, Canada
| | | | - Britta Brügger
- Heidelberg University Biochemistry Center (BZH), Im Neuenheimer Feld 328, 69120, Heidelberg, Germany
| | - Mónica P Cala
- Metabolomics Core Facility-MetCore, Universidad de los Andes, Bogotá, 111711, Colombia
| | - Dolores Camacho-Muñoz
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9NT, United Kingdom
| | - Antonio Checa
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Michael Chen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Michaela Chocholoušková
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10, Pardubice, Czech Republic
| | - Michelle Cinel
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Emeline Chu-Van
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif sur Yvette, France
| | - Benoit Colsch
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif sur Yvette, France
| | - Cristina Coman
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | | | - Bebiana C Sousa
- Babraham Institute, Babraham Research Campus, Cambridge, MA, CB22 3AT, USA
| | - Alex M Dickens
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
- Department of Chemistry, University of Turku, Turku, Finland
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, 04013, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, 04013, Leipzig, Germany
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine Carl Gustav Carus of TU Dresden, 01307, Dresden, Germany
| | - Finnur Freyr Eiríksson
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
- ArcticMass, Reykjavik, Iceland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Mohan Ghorasaini
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333ZA, Leiden, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333ZA, Leiden, The Netherlands
| | - Xue Li Guan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| | - Mark Haid
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Amy Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Marcus Höring
- University Hospital of Regensburg, Institute of Clinical Chemistry and Laboratory Medicine, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Michal Holčapek
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10, Pardubice, Czech Republic
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Zurich, 8952, Schlieren, Switzerland
| | - Chunxiu Hu
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Andreas J Hülsmeier
- Institute of Clinical Chemistry, University Zurich, 8952, Schlieren, Switzerland
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Christina M Jones
- Chemical Science Division, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Yoshihiro Izumi
- Division of Metabolomics Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 3‑1‑1, Maidashi, Higashi‑ku, Fukuoka, 812‑8582, Japan
| | - Harald C Köfeler
- Core Facility Mass Spectrometry, Medical University of Graz, 8010, Graz, Austria
| | - Sin Man Lam
- LipidALL Technologies, Changzhou, 213000, Jiangshu, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mike Lange
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, 04013, Leipzig, Germany
- Center for Biotechnology and Biomedicine, University of Leipzig, 04013, Leipzig, Germany
| | - Jong Cheol Lee
- Department of Chemistry, Yonsei University, Seoul, 03722, South Korea
| | - Gerhard Liebisch
- University Hospital of Regensburg, Institute of Clinical Chemistry and Laboratory Medicine, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Katrice Lippa
- Chemical Science Division, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | | | - Malena Manzi
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD, Ciudad de Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes, 2160 C1428EGA, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Departamento de Desarrollo Analítico y Control de Procesos, Instituto Nacional de Tecnología Industrial, Av. General Paz 5445, B1650WAB, Buenos Aires, Argentina
| | - Manuela R Martinefski
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD, Ciudad de Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, Junin 954, Junin, C1113AAD, CABA, Argentina
| | - Raviswamy G H Math
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka, 560065, India
| | - Satyajit Mayor
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka, 560065, India
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, 3086, Australia
| | - María Eugenia Monge
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD, Ciudad de Buenos Aires, Argentina
| | - Myeong Hee Moon
- Department of Chemistry, Yonsei University, Seoul, 03722, South Korea
| | - Sneha Muralidharan
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka, 560065, India
| | - Anna Nicolaou
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9NT, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9NT, United Kingdom
| | - Thao Nguyen-Tran
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Ottawa Brain and Mind Research Institute, Department of Biochemistry, Microbiology, and Immunology, and Department of Chemistry, Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, K1H 8M5, Canada
| | - Valerie B O'Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 702 81, Örebro, Sweden
| | - Arvind Ramanathan
- Institute for Stem Cell Science and Regenerative Medicine, 560065, Bangalore, India
| | - Fabien Riols
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8573, Japan
| | - Tracey B Schock
- Chemical Science Division, National Institute of Standards and Technology, Charleston, SC, 29412, USA
| | - Heidi Schwartz-Zimmermann
- Christian Doppler Laboratory for Innovative Gut Health Concepts of Livestock, Institute of Bioanalytics and Agro-Metabolomics, Department of Agrobiotechnology (IFATulln), University of Natural Resources and Life Sciences, Vienna (BOKU), Konrad-Lorenz-Str. 20, 3430, Tulln, Austria
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Madhulika Singh
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Masatomo Takahashi
- Division of Metabolomics Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 3‑1‑1, Maidashi, Higashi‑ku, Fukuoka, 812‑8582, Japan
| | - Margrét Thorsteinsdóttir
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
- ArcticMass, Reykjavik, Iceland
| | - Noriyuki Tomiyasu
- Division of Metabolomics Center, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, 3‑1‑1, Maidashi, Higashi‑ku, Fukuoka, 812‑8582, Japan
| | | | - Hiroshi Tsugawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- RIKEN Center for Sustainable Resource Science, Yokohama, Japan
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Victoria J Tyrrell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Grace van der Gugten
- St. Paul's Hospital, Department of Pathology and Laboratory Medicine, Vancouver, BC, Canada
| | - Michael O Wakelam
- Babraham Institute, Babraham Research Campus, Cambridge, MA, CB22 3AT, USA
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Denise Wolrab
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10, Pardubice, Czech Republic
| | - Guowang Xu
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Tianrun Xu
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - John A Bowden
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Kim Ekroos
- Lipidomics Consulting Ltd., Espoo, Finland.
| | - Robert Ahrends
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria.
| | - Markus R Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore.
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
2
|
Ganjali S, Cardenia V, Bonciolini A, Santos RD, Al-Rasadi K, Sahebkar A. Lipidomic profiling in patients with familial hypercholesterolemia: Abnormalities in glycerolipids and oxysterols. Clin Biochem 2024; 131-132:110812. [PMID: 39197573 DOI: 10.1016/j.clinbiochem.2024.110812] [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: 06/19/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/01/2024]
Abstract
OBJECTIVES AND AIM This study aimed to identify precise biomarkers and develop targeted therapeutic strategies for preventing premature atherosclerotic cardiovascular disease in patients with familial hypercholesterolemia (FH) by investigating the quantitative and qualitative abnormalities in the metabolic network of lipids in these patients using an advanced lipidomics platform. DESIGN & METHODS The study population comprised 18 homozygous (HoFH), 18 heterozygous (HeFH) FH patients, and 20 healthy controls. Cholesterol oxidation products (oxysterol, COPs) and main lipid classes were determined using gas chromatography-mass spectrometry. Results were expressed as percentages of total fat matter for lipid classes and percentages of total COPs for oxysterols. The principal component analysis (PCA) was also carried out, to highlight the correlation between studied parameters and groups investigated. RESULTS Patients (both HoFH and HeFH) showed lower content of free fatty acids (FFAs) and greater values of triacylglycerols (TAGs) in comparison to controls. HoFH showed lower monoacylglycerols (P<0.01) and higher free cholesterol (FC) (P<0.05) when compared to HeFH and controls. The total content of COPs ranged from 1.96 to 4.25 mg/dL, from 2.27 to 4.05 mg/dL, and from 0.79 to 4.12 mg/dL in healthy controls, HoFH and HeFH groups, respectively, with no significant differences between patients and controls. In general, the 7α-hydroxycholesterol (7α-HC) was greater than other COPs. However, no significant differences were found between the three studied groups. Moreover, an opposite trend was observed between 7α-HC and 7-ketocholesterol (7-KC). Additionally, when PCA was carried out, the first two PCs explained 92.13 % of the total variance, of which the PC1 describes 53.94 % of variance mainly correlated to TAGs, diacylglycerols (DAGs), and 7-KC. On the other hand, the PC2 was correlated primarily for FFAs, FC and esterified sterols (E-STE). CONCLUSIONS In conclusion, abnormal levels of TAGs, DAGs and 7-KC were associated with HeFH while HoFH was associated with the abnormal amount of E-STE.
Collapse
Affiliation(s)
- Shiva Ganjali
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Australia
| | - Vladimiro Cardenia
- Department of Agricultural, Forest and Food Sciences, University of Turin, 10095 Grugliasco, Italy
| | - Ambra Bonciolini
- Department of Agricultural, Forest and Food Sciences, University of Turin, 10095 Grugliasco, Italy
| | - Raul D Santos
- Lipid Clinic Heart Institute (InCor) University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Khalid Al-Rasadi
- Medical Research Centre, Sultan Qaboos University, Muscat, Oman; Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Amirhossein Sahebkar
- Biotechnology Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Applied Biomedical Research Centre, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Wu J, Giles C, Dakic A, Beyene HB, Huynh K, Wang T, Meikle T, Olshansky G, Salim A, Duong T, Watts GF, Hung J, Hui J, Cadby G, Beilby J, Blangero J, Moses EK, Shaw JE, Magliano DJ, Zhu D, Yang JY, Grieve SM, Wilson A, Chow CK, Vernon ST, Gray MP, Figtree GA, Carrington MJ, Inouye M, Marwick TH, Meikle PJ. Lipidomic Risk Score to Enhance Cardiovascular Risk Stratification for Primary Prevention. J Am Coll Cardiol 2024; 84:434-446. [PMID: 39048275 DOI: 10.1016/j.jacc.2024.04.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 03/22/2024] [Accepted: 04/15/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Accurate risk stratification is vital for primary prevention of cardiovascular disease (CVD). However, traditional tools such as the Framingham Risk Score (FRS) may underperform within the diverse intermediate-risk group, which includes individuals requiring distinct management strategies. OBJECTIVES This study aimed to develop a lipidomic-enhanced risk score (LRS), specifically targeting risk prediction and reclassification within the intermediate group, benchmarked against the FRS. METHODS The LRS was developed via a machine learning workflow using ridge regression on the Australian Diabetes, Obesity, and Lifestyle Study (AusDiab; n = 10,339). It was externally validated with the Busselton Health Study (n = 4,492), and its predictive utility for coronary artery calcium scoring (CACS)-based outcomes was independently validated in the BioHEART cohort (n = 994). RESULTS LRS significantly improved discrimination metrics for the intermediate-risk group in both AusDiab and Busselton Health Study cohorts (all P < 0.001), increasing the area under the curve for CVD events by 0.114 (95% CI: 0.1123-0.1157) and 0.077 (95% CI: 0.0755-0.0785), with a net reclassification improvement of 0.36 (95% CI: 0.21-0.51) and 0.33 (95% CI: 0.15-0.49), respectively. For CACS-based outcomes in BioHEART, LRS achieved a significant area under the curve improvement of 0.02 over the FRS (0.76 vs 0.74; P < 1.0 × 10-5). A simplified, clinically applicable version of LRS was also created that had comparable performance to the original LRS. CONCLUSIONS LRS, augmenting the FRS, presents potential to improve intermediate-risk stratification and to predict atherosclerotic markers using a simple blood test, suitable for clinical application. This could facilitate the triage of individuals for noninvasive imaging such as CACS, fostering precision medicine in CVD prevention and management.
Collapse
Affiliation(s)
- Jingqin Wu
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Victoria, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Aleksandar Dakic
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Habtamu B Beyene
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Tingting Wang
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Thomas Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Victoria, Australia
| | - Gavriel Olshansky
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Victoria, Australia
| | - Agus Salim
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Thy Duong
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Gerald F Watts
- Medical School, University of Western Australia, Perth, Western Australia, Australia; Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Joseph Hung
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Jennie Hui
- PathWest Laboratory Medicine of Western Australia, Nedlands, Western Australia, Australia; School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia; School of Population and Global Health, University of Western Australia, Crawley, Western Australia, Australia
| | - Gemma Cadby
- School of Population and Global Health, University of Western Australia, Crawley, Western Australia, Australia
| | - John Beilby
- School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - John Blangero
- South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Eric K Moses
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jonathan E Shaw
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Dantong Zhu
- School of Mathematics and Statistics, The University of Sydney, Camperdown, New South Wales, Australia; Kolling Institute of Medical Research, The University of Sydney, St Leonards, New South Wales, Australia
| | - Jean Y Yang
- School of Mathematics and Statistics, The University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Stuart M Grieve
- Department of Radiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia; Imaging and Phenotyping Laboratory, Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Andrew Wilson
- Menzies Centre for Health Policy and Economics, Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Clara K Chow
- Westmead Applied Research Centre, Faculty of Medicine and Health, University of Sydney, Westmead, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Stephen T Vernon
- Kolling Institute of Medical Research, The University of Sydney, St Leonards, New South Wales, Australia
| | - Michael P Gray
- Kolling Institute of Medical Research, The University of Sydney, St Leonards, New South Wales, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, The University of Sydney, St Leonards, New South Wales, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia; Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | | | - Mike Inouye
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, United Kingdom
| | - Thomas H Marwick
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, Victoria, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Beyene HB, Huynh K, Wang T, Paul S, Cinel M, Mellett NA, Olshansky G, Meikle TG, Watts GF, Hung J, Hui J, Beilby J, Blangero J, Moses EK, Shaw JE, Magliano DJ, Giles C, Meikle PJ. Development and validation of a plasmalogen score as an independent modifiable marker of metabolic health: population based observational studies and a placebo-controlled cross-over study. EBioMedicine 2024; 105:105187. [PMID: 38861870 PMCID: PMC11215217 DOI: 10.1016/j.ebiom.2024.105187] [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: 02/12/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Decreased levels of circulating ethanolamine plasmalogens [PE(P)], and a concurrent increase in phosphatidylethanolamine (PE) are consistently reported in various cardiometabolic conditions. Here we devised, a plasmalogen score (Pls Score) that mirrors a metabolic signal that encompasses the levels of PE(P) and PE and captures the natural variation in circulating plasmalogens and perturbations in their metabolism associated with disease, diet, and lifestyle. METHODS We utilised, plasma lipidomes from the Australian Obesity, Diabetes and Lifestyle study (AusDiab; n = 10,339, 55% women) a nationwide cohort, to devise the Pls Score and validated this in the Busselton Health Study (BHS; n = 4,492, 56% women, serum lipidome) and in a placebo-controlled crossover trial involving Shark Liver Oil (SLO) supplementation (n = 10, 100% men). We examined the association of the Pls Score with cardiometabolic risk factors, type 2 diabetes mellitus (T2DM), cardiovascular disease and all-cause mortality (over 17 years). FINDINGS In a model, adjusted for age, sex and BMI, individuals in the top quintile of the Pls Score (Q5) relative to Q1 had an OR of 0.31 (95% CI 0.21-0.43), 0.39 (95% CI 0.25-0.61) and 0.42 (95% CI 0.30-0.57) for prevalent T2DM, incident T2DM and prevalent cardiovascular disease respectively, and a 34% lower mortality risk (HR = 0.66; 95% CI 0.56-0.78). Significant associations between diet and lifestyle habits and Pls Score exist and these were validated through dietary supplementation of SLO that resulted in a marked change in the Pls Score. INTERPRETATION The Pls Score as a measure that captures the natural variation in circulating plasmalogens, was not only inversely related to cardiometabolic risk and all-cause mortality but also associate with diet and lifestyle. Our results support the potential utility of the Pls Score as a biomarker for metabolic health and its responsiveness to dietary interventions. Further research is warranted to explore the underlying mechanisms and optimise the practical implementation of the Pls Score in clinical and population settings. FUNDING National Health and Medical Research Council (NHMRC grant 233200), National Health and Medical Research Council of Australia (Project grant APP1101320), Health Promotion Foundation of Western Australia, and National Health and Medical Research Council of Australia Senior Research Fellowship (#1042095).
Collapse
Affiliation(s)
- Habtamu B Beyene
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Tingting Wang
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Sudip Paul
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Michelle Cinel
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | | | - Thomas G Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
| | - Gerald F Watts
- Medical School, University of Western Australia, Perth, WA, Australia; Cardiometabolic Service, Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA, Australia
| | - Joseph Hung
- Medical School, University of Western Australia, Perth, WA, Australia
| | - Jennie Hui
- PathWest Laboratory Medicine of Western Australia, Queen Elizabeth II Medical Centre, Nedlands, WA, Australia; School of Population and Global Health, University of Western Australia, Crawley, WA, Australia
| | - John Beilby
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - John Blangero
- South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Eric K Moses
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia; Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Jonathan E Shaw
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia.
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia; Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia; Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Gerhardtova I, Cizmarova I, Jankech T, Olesova D, Jampilek J, Parrak V, Nemergutova K, Sopko L, Piestansky J, Kovac A. Implementation of Modern Therapeutic Drug Monitoring and Lipidomics Approaches in Clinical Practice: A Case Study with Colistin Treatment. Pharmaceuticals (Basel) 2024; 17:753. [PMID: 38931420 PMCID: PMC11206893 DOI: 10.3390/ph17060753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Nowadays, lipidomics plays a crucial role in the investigation of novel biomarkers of various diseases. Its implementation into the field of clinical analysis led to the identification of specific lipids and/or significant changes in their plasma levels in patients suffering from cancer, Alzheimer's disease, sepsis, and many other diseases and pathological conditions. Profiling of lipids and determination of their plasma concentrations could also be helpful in the case of drug therapy management, especially in combination with therapeutic drug monitoring (TDM). Here, for the first time, a combined approach based on the TDM of colistin, a last-resort antibiotic, and lipidomic profiling is presented in a case study of a critically ill male patient suffering from Pseudomonas aeruginosa-induced pneumonia. Implementation of innovative analytical approaches for TDM (online combination of capillary electrophoresis with tandem mass spectrometry, CZE-MS/MS) and lipidomics (liquid chromatography-tandem mass spectrometry, LC-MS/MS) was demonstrated. The CZE-MS/MS strategy confirmed the chosen colistin drug dosing regimen, leading to stable colistin concentrations in plasma samples. The determined colistin concentrations in plasma samples reached the required minimal inhibitory concentration of 1 μg/mL. The complex lipidomics approach led to monitoring 545 lipids in collected patient plasma samples during and after the therapy. Some changes in specific individual lipids were in good agreement with previous lipidomics studies dealing with sepsis. The presented case study represents a good starting point for identifying particular individual lipids that could correlate with antimicrobial and inflammation therapeutic management.
Collapse
Affiliation(s)
- Ivana Gerhardtova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (I.G.); (T.J.); (D.O.); (J.J.); (V.P.)
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University Bratislava, Ilkovicova 6, 842 15 Bratislava, Slovakia
| | - Ivana Cizmarova
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojarov 10, 832 32 Bratislava, Slovakia;
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University Bratislava, Odbojarov 10, 832 32 Bratislava, Slovakia
| | - Timotej Jankech
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (I.G.); (T.J.); (D.O.); (J.J.); (V.P.)
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University Bratislava, Ilkovicova 6, 842 15 Bratislava, Slovakia
| | - Dominika Olesova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (I.G.); (T.J.); (D.O.); (J.J.); (V.P.)
- Institute of Experimental Endocrinology, Biomedical Research Center SAS, Dubravska cesta 9, 845 10 Bratislava, Slovakia
| | - Josef Jampilek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (I.G.); (T.J.); (D.O.); (J.J.); (V.P.)
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University Bratislava, Ilkovicova 6, 842 15 Bratislava, Slovakia
| | - Vojtech Parrak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (I.G.); (T.J.); (D.O.); (J.J.); (V.P.)
- Clinic of Hematology and Transfusiology, St. Cyril and Methodius Hospital, Antolska 11, 851 07 Bratislava, Slovakia; (K.N.); (L.S.)
| | - Kristina Nemergutova
- Clinic of Hematology and Transfusiology, St. Cyril and Methodius Hospital, Antolska 11, 851 07 Bratislava, Slovakia; (K.N.); (L.S.)
| | - Ladislav Sopko
- Clinic of Hematology and Transfusiology, St. Cyril and Methodius Hospital, Antolska 11, 851 07 Bratislava, Slovakia; (K.N.); (L.S.)
| | - Juraj Piestansky
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (I.G.); (T.J.); (D.O.); (J.J.); (V.P.)
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University Bratislava, Odbojarov 10, 832 32 Bratislava, Slovakia
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojarov 10, 832 32 Bratislava, Slovakia
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia; (I.G.); (T.J.); (D.O.); (J.J.); (V.P.)
- Department of Pharmacology and Toxicology, University of Veterinary Medicine and Pharmacy in Kosice, Komenského 68/73, 041 81 Kosice, Slovakia
| |
Collapse
|
6
|
Su H, Masters CL, Bush AI, Barnham KJ, Reid GE, Vella LJ. Exploring the significance of lipids in Alzheimer's disease and the potential of extracellular vesicles. Proteomics 2024; 24:e2300063. [PMID: 37654087 DOI: 10.1002/pmic.202300063] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Lipids play a significant role in maintaining central nervous system (CNS) structure and function, and the dysregulation of lipid metabolism is known to occur in many neurological disorders, including Alzheimer's disease. Here we review what is currently known about lipid dyshomeostasis in Alzheimer's disease. We propose that small extracellular vesicle (sEV) lipids may provide insight into the pathophysiology and progression of Alzheimer's disease. This stems from the recognition that sEV likely contributes to disease pathogenesis, but also an understanding that sEV can serve as a source of potential biomarkers. While the protein and RNA content of sEV in the CNS diseases have been studied extensively, our understanding of the lipidome of sEV in the CNS is still in its infancy.
Collapse
Affiliation(s)
- Huaqi Su
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Kevin J Barnham
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Gavin E Reid
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura J Vella
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
7
|
Hristova-Panusheva K, Xenodochidis C, Georgieva M, Krasteva N. Nanoparticle-Mediated Drug Delivery Systems for Precision Targeting in Oncology. Pharmaceuticals (Basel) 2024; 17:677. [PMID: 38931344 PMCID: PMC11206252 DOI: 10.3390/ph17060677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Nanotechnology has emerged as a transformative force in oncology, facilitating advancements in site-specific cancer therapy and personalized oncomedicine. The development of nanomedicines explicitly targeted to cancer cells represents a pivotal breakthrough, allowing the development of precise interventions. These cancer-cell-targeted nanomedicines operate within the intricate milieu of the tumour microenvironment, further enhancing their therapeutic efficacy. This comprehensive review provides a contemporary perspective on precision cancer medicine and underscores the critical role of nanotechnology in advancing site-specific cancer therapy and personalized oncomedicine. It explores the categorization of nanoparticle types, distinguishing between organic and inorganic variants, and examines their significance in the targeted delivery of anticancer drugs. Current insights into the strategies for developing actively targeted nanomedicines across various cancer types are also provided, thus addressing relevant challenges associated with drug delivery barriers. Promising future directions in personalized cancer nanomedicine approaches are delivered, emphasising the imperative for continued optimization of nanocarriers in precision cancer medicine. The discussion underscores translational research's need to enhance cancer patients' outcomes by refining nanocarrier technologies in nanotechnology-driven, site-specific cancer therapy.
Collapse
Affiliation(s)
- Kamelia Hristova-Panusheva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (K.H.-P.); (C.X.)
| | - Charilaos Xenodochidis
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (K.H.-P.); (C.X.)
| | - Milena Georgieva
- Institute of Molecular Biology “Acad. R. Tsanev”, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria;
| | - Natalia Krasteva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (K.H.-P.); (C.X.)
| |
Collapse
|
8
|
Wan Z, Su J, Zhu X, Liu X, Guo Y, Xiang D, Zhou X, Peng X, Tao R, Cao Q, Lang G, Huang Y, Zhu B. Distinct Lipidomic Profiles between People Living with HIV Treated with E/C/F/TAF or B/F/TAF: An Open-Label Prospective Cohort Study. Infect Dis Ther 2024; 13:727-744. [PMID: 38489119 PMCID: PMC11058159 DOI: 10.1007/s40121-024-00943-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/15/2024] [Indexed: 03/17/2024] Open
Abstract
INTRODUCTION Elvitegravir/cobicistat/emtricitabine/tenofovir alafenamide (E/C/F/TAF) has been increasingly replaced by bictegravir/emtricitabine/tenofovir alafenamide (B/F/TAF) in the treatment of human immunodeficiency virus (HIV) owing to its more favorable pharmacokinetics and fewer drug-drug interactions. However, the effect of this switch on plasma lipids and lipidomic profiles remains poorly characterized. METHODS HIV infected patients on an E/C/F/TAF regimen were recruited into the study and followed up every 12 weeks. Participants were divided into E/C/F/TAF and B/F/TAF groups depending on whether they were switched to B/F/TAF during follow-up. Clinical information and blood samples were collected at 0, 12, and 24 weeks, and lipidomic analysis was performed using liquid chromatography mass spectrometry. RESULTS No significant differences were observed between the groups at baseline. At week 24, patients switched to B/F/TAF had lower triglyceride [mmol/L; 1.23 (0.62) versus 2.03 (0.75), P = 0.001] and very low-density lipoprotein cholesterol [mmol/L; 0.64 (0.26) versus 0.84 (0.32), P = 0.037) compared with patients who continued E/C/F/TAF therapy. Small decrease from baseline in Framingham general cardiovascular risk score (FRS) was observed in the B/F/TAF arm [week (W) 0: 2.59 (1.57) versus W24: 2.18 (1.01), P = 0.043]. Lipidomic analysis indicated that E/C/F/TAF treatment increased the levels of several diglycerides (DGs), triacylglycerols (TAGs), and lyso-phosphatidylcholines (LPCs), whereas switching to B/F/TAF led to increased sphingolipids and glycerophospholipids. After adjusting for demographic and clinical parameters, only DG (16:0/18:2), DG (18:2/22:6), DG (18:3/18:2), DG (20:5/18:2), TAG (18:3/18:2/21:5), TAG (20:5/18:2/22:6), and LPC (22:6) were found to be significantly associated with FRS (regression coefficient of 0.17-6.02, P < 0.05). Most of these FRS associate lipid species were significantly elevated in individuals treated with E/C/F/TAF instead of individuals treated with B/F/TAF. CONCLUSION E/C/F/TAF promotes the accumulation of lipid species closely associated with cardiovascular disease (CVD) risk among people living with HIV, whereas B/F/TAF has a decreased impact on CVD-related lipid profile and is associated with lower CVD risk. A graphical abstract is available with this article. TRIAL REGISTRATION ClinicalTrials.gov; identifier, NCT06019273.
Collapse
Affiliation(s)
- Zhikai Wan
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Junwei Su
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Xueling Zhu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Xiang Liu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Yongzheng Guo
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Dairong Xiang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Xiaotang Zhou
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Xiaorong Peng
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Ran Tao
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Qing Cao
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Guanjing Lang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Ying Huang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China
| | - Biao Zhu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Ewald S, Nasuhidehnavi A, Feng TY, Lesani M, McCall LI. The intersection of host in vivo metabolism and immune responses to infection with kinetoplastid and apicomplexan parasites. Microbiol Mol Biol Rev 2024; 88:e0016422. [PMID: 38299836 PMCID: PMC10966954 DOI: 10.1128/mmbr.00164-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024] Open
Abstract
SUMMARYProtozoan parasite infection dramatically alters host metabolism, driven by immunological demand and parasite manipulation strategies. Immunometabolic checkpoints are often exploited by kinetoplastid and protozoan parasites to establish chronic infection, which can significantly impair host metabolic homeostasis. The recent growth of tools to analyze metabolism is expanding our understanding of these questions. Here, we review and contrast host metabolic alterations that occur in vivo during infection with Leishmania, trypanosomes, Toxoplasma, Plasmodium, and Cryptosporidium. Although genetically divergent, there are commonalities among these pathogens in terms of metabolic needs, induction of the type I immune responses required for clearance, and the potential for sustained host metabolic dysbiosis. Comparing these pathogens provides an opportunity to explore how transmission strategy, nutritional demand, and host cell and tissue tropism drive similarities and unique aspects in host response and infection outcome and to design new strategies to treat disease.
Collapse
Affiliation(s)
- Sarah Ewald
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Azadeh Nasuhidehnavi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| |
Collapse
|
10
|
Sarkar S, Roy D, Chatterjee B, Ghosh R. Clinical advances in analytical profiling of signature lipids: implications for severe non-communicable and neurodegenerative diseases. Metabolomics 2024; 20:37. [PMID: 38459207 DOI: 10.1007/s11306-024-02100-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 02/06/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Lipids play key roles in numerous biological processes, including energy storage, cell membrane structure, signaling, immune responses, and homeostasis, making lipidomics a vital branch of metabolomics that analyzes and characterizes a wide range of lipid classes. Addressing the complex etiology, age-related risk, progression, inflammation, and research overlap in conditions like Alzheimer's Disease, Parkinson's Disease, Cardiovascular Diseases, and Cancer poses significant challenges in the quest for effective therapeutic targets, improved diagnostic markers, and advanced treatments. Mass spectrometry is an indispensable tool in clinical lipidomics, delivering quantitative and structural lipid data, and its integration with technologies like Liquid Chromatography (LC), Magnetic Resonance Imaging (MRI), and few emerging Matrix-Assisted Laser Desorption Ionization- Imaging Mass Spectrometry (MALDI-IMS) along with its incorporation into Tissue Microarray (TMA) represents current advances. These innovations enhance lipidomics assessment, bolster accuracy, and offer insights into lipid subcellular localization, dynamics, and functional roles in disease contexts. AIM OF THE REVIEW The review article summarizes recent advancements in lipidomic methodologies from 2019 to 2023 for diagnosing major neurodegenerative diseases, Alzheimer's and Parkinson's, serious non-communicable cardiovascular diseases and cancer, emphasizing the role of lipid level variations, and highlighting the potential of lipidomics data integration with genomics and proteomics to improve disease understanding and innovative prognostic, diagnostic and therapeutic strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW Clinical lipidomic studies are a promising approach to track and analyze lipid profiles, revealing their crucial roles in various diseases. This lipid-focused research provides insights into disease mechanisms, biomarker identification, and potential therapeutic targets, advancing our understanding and management of conditions such as Alzheimer's Disease, Parkinson's Disease, Cardiovascular Diseases, and specific cancers.
Collapse
Affiliation(s)
- Sutanu Sarkar
- Amity Institute of Biotechnology (AIBNK), Amity University, Rajarhat, Newtown Action Area 2, Kolkata, 700135, West Bengal, India
| | - Deotima Roy
- Amity Institute of Biotechnology (AIBNK), Amity University, Rajarhat, Newtown Action Area 2, Kolkata, 700135, West Bengal, India
| | - Bhaskar Chatterjee
- Amity Institute of Biotechnology (AIBNK), Amity University, Rajarhat, Newtown Action Area 2, Kolkata, 700135, West Bengal, India
| | - Rajgourab Ghosh
- Amity Institute of Biotechnology (AIBNK), Amity University, Rajarhat, Newtown Action Area 2, Kolkata, 700135, West Bengal, India.
| |
Collapse
|
11
|
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
|
12
|
McKenzie CM, Marinkovich M, Armién AG, Leger JS, Armando AM, Dennis EA, Quehenberger O, Righton A. Lipid storage disease in 4 sibling superb birds-of-paradise ( Lophorina superba). Vet Pathol 2024; 61:288-297. [PMID: 37842940 PMCID: PMC11032166 DOI: 10.1177/03009858231203314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Pedigree analysis, clinical, gross, microscopic, ultrastructural, and lipidomic findings in 4 female superb bird-of-paradise (SBOP, Lophorina superba) siblings led to the diagnosis of a primary inherited glycerolipid storage disease. These birds were the offspring of a related breeding pair (inbreeding coefficient = 0.1797) and are the only known SBOPs to display this constellation of lesions. The birds ranged from 0.75 to 4.3 years of age at the time of death. Two birds were euthanized and 1 died naturally due to the disease, and 1 died of head trauma with no prior clinical signs. Macroscopic findings included hepatomegaly and pallor (4/4), cardiac and renal pallor (2/4), and coelomic effusion (1/4). Microscopic examination found marked tissue distortion due to cytoplasmic lipid vacuoles in hepatocytes (4/4), cardiomyocytes (4/4), renal tubular epithelial cells (4/4), parathyroid gland principal cells (2/2), exocrine pancreatic cells (3/3), and the glandular cells of the ventriculus and proventriculus (3/3). Ultrastructurally, the lipids were deposited in single to coalescing or fused droplets lined by an inconspicuous or discontinuous monolayer membrane. Lipidomic profiling found that the cytoplasmic lipid deposits were primarily composed of triacylglycerols. Future work, including sequencing of the SBOP genome and genotyping, will be required to definitively determine the underlying genetic mechanism of this disease.
Collapse
|
13
|
Scheinberg T, Lin HM, Fitzpatrick M, Azad AA, Bonnitcha P, Davies A, Heller G, Huynh K, Mak B, Mahon K, Sullivan D, Meikle PJ, Horvath LG. PCPro: a clinically accessible, circulating lipid biomarker signature for poor-prognosis metastatic prostate cancer. Prostate Cancer Prostatic Dis 2024; 27:136-143. [PMID: 37147359 PMCID: PMC10876475 DOI: 10.1038/s41391-023-00666-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/23/2023] [Accepted: 03/30/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND Using comprehensive plasma lipidomic profiling from men with metastatic castration-resistant prostate cancer (mCRPC), we have previously identified a poor-prognostic lipid profile associated with shorter overall survival (OS). In order to translate this biomarker into the clinic, these men must be identifiable via a clinically accessible, regulatory-compliant assay. METHODS A single regulatory-compliant liquid chromatography-mass spectrometry assay of candidate lipids was developed and tested on a mCRPC Discovery cohort of 105 men. Various risk-score Cox regression prognostic models of OS were built using the Discovery cohort. The model with the highest concordance index (PCPro) was chosen for validation and tested on an independent Validation cohort of 183 men. RESULTS PCPro, the lipid biomarker, contains Cer(d18:1/18:0), Cer(d18:1/24:0), Cer(d18:1/24:1), triglycerides and total cholesterol. Within the Discovery and Validation cohorts, men who were PCPro positive had significantly shorter OS compared to those who were PCPro negative (Discovery: median OS 12.0 months vs 24.2 months, hazard ratio (HR) 3.75 [95% confidence interval (CI) 2.29-6.15], p < 0.001, Validation: median OS 13.0 months vs 25.7 months, HR = 2.13 [95% CI 1.46-3.12], p < 0.001). CONCLUSIONS We have developed PCPro, a lipid biomarker assay capable of prospectively identifying men with mCRPC with a poor prognosis. Prospective clinical trials are required to determine if men who are PCPro positive will benefit from therapeutic agents targeting lipid metabolism.
Collapse
Affiliation(s)
- Tahlia Scheinberg
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia
- Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- University of Sydney, Camperdown, NSW, Australia
| | - Hui-Ming Lin
- Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Michael Fitzpatrick
- NSW Health Pathology, Department of Chemical Pathology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Arun A Azad
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Paul Bonnitcha
- University of Sydney, Camperdown, NSW, Australia
- NSW Health Pathology, Department of Chemical Pathology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Amy Davies
- Department of Medical Oncology, Monash Health, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| | | | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiovascular Research Translation and implementation, La Trobe University, Melbourne, VIC, Australia
| | - Blossom Mak
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia
- Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- University of Sydney, Camperdown, NSW, Australia
| | - Kate Mahon
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia
- Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- University of Sydney, Camperdown, NSW, Australia
| | - David Sullivan
- NSW Health Pathology, Department of Chemical Pathology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiovascular Research Translation and implementation, La Trobe University, Melbourne, VIC, Australia
| | - Lisa G Horvath
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia.
- Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.
- University of Sydney, Camperdown, NSW, Australia.
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
| |
Collapse
|
14
|
Yu J, Ren J, Ren Y, Wu Y, Zeng Y, Zhang Q, Xiao X. Using metabolomics and proteomics to identify the potential urine biomarkers for prediction and diagnosis of gestational diabetes. EBioMedicine 2024; 101:105008. [PMID: 38368766 PMCID: PMC10882130 DOI: 10.1016/j.ebiom.2024.105008] [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: 11/28/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/20/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common metabolic complications during pregnancy, threatening both maternal and fetal health. Prediction and diagnosis of GDM is not unified. Finding effective biomarkers for GDM is particularly important for achieving early prediction, accurate diagnosis and timely intervention. Urine, due to its accessibility in large quantities, noninvasive collection and easy preparation, has become a good sample for biomarker identification. In recent years, a number of studies using metabolomics and proteomics approaches have identified differential expressed urine metabolites and proteins in GDM patients. In this review, we summarized these potential urine biomarkers for GDM prediction and diagnosis and elucidated their role in development of GDM.
Collapse
Affiliation(s)
- Jie Yu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jing Ren
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yaolin Ren
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yifan Wu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuan Zeng
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
15
|
Conde-Torres D, Blanco-González A, Seco-González A, Suárez-Lestón F, Cabezón A, Antelo-Riveiro P, Piñeiro Á, García-Fandiño R. Unraveling lipid and inflammation interplay in cancer, aging and infection for novel theranostic approaches. Front Immunol 2024; 15:1320779. [PMID: 38361953 PMCID: PMC10867256 DOI: 10.3389/fimmu.2024.1320779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
The synergistic relationships between Cancer, Aging, and Infection, here referred to as the CAIn Triangle, are significant determinants in numerous health maladies and mortality rates. The CAIn-related pathologies exhibit close correlations with each other and share two common underlying factors: persistent inflammation and anomalous lipid concentration profiles in the membranes of affected cells. This study provides a comprehensive evaluation of the most pertinent interconnections within the CAIn Triangle, in addition to examining the relationship between chronic inflammation and specific lipidic compositions in cellular membranes. To tackle the CAIn-associated diseases, a suite of complementary strategies aimed at diagnosis, prevention, and treatment is proffered. Our holistic approach is expected to augment the understanding of the fundamental mechanisms underlying these diseases and highlight the potential of shared features to facilitate the development of novel theranostic strategies.
Collapse
Affiliation(s)
- Daniel Conde-Torres
- Departamento de Física Aplicada, Facultade de Física, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Organic Chemistry Department, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Alexandre Blanco-González
- Departamento de Física Aplicada, Facultade de Física, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Organic Chemistry Department, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- MD.USE Innovations S.L., Edificio Emprendia, Santiago de Compostela, Spain
| | - Alejandro Seco-González
- Organic Chemistry Department, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Fabián Suárez-Lestón
- Departamento de Física Aplicada, Facultade de Física, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Organic Chemistry Department, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- MD.USE Innovations S.L., Edificio Emprendia, Santiago de Compostela, Spain
| | - Alfonso Cabezón
- Organic Chemistry Department, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Paula Antelo-Riveiro
- Departamento de Física Aplicada, Facultade de Física, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Organic Chemistry Department, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ángel Piñeiro
- Departamento de Física Aplicada, Facultade de Física, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Rebeca García-Fandiño
- Organic Chemistry Department, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
16
|
Demicheva E, Dordiuk V, Polanco Espino F, Ushenin K, Aboushanab S, Shevyrin V, Buhler A, Mukhlynina E, Solovyova O, Danilova I, Kovaleva E. Advances in Mass Spectrometry-Based Blood Metabolomics Profiling for Non-Cancer Diseases: A Comprehensive Review. Metabolites 2024; 14:54. [PMID: 38248857 PMCID: PMC10820779 DOI: 10.3390/metabo14010054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Blood metabolomics profiling using mass spectrometry has emerged as a powerful approach for investigating non-cancer diseases and understanding their underlying metabolic alterations. Blood, as a readily accessible physiological fluid, contains a diverse repertoire of metabolites derived from various physiological systems. Mass spectrometry offers a universal and precise analytical platform for the comprehensive analysis of blood metabolites, encompassing proteins, lipids, peptides, glycans, and immunoglobulins. In this comprehensive review, we present an overview of the research landscape in mass spectrometry-based blood metabolomics profiling. While the field of metabolomics research is primarily focused on cancer, this review specifically highlights studies related to non-cancer diseases, aiming to bring attention to valuable research that often remains overshadowed. Employing natural language processing methods, we processed 507 articles to provide insights into the application of metabolomic studies for specific diseases and physiological systems. The review encompasses a wide range of non-cancer diseases, with emphasis on cardiovascular disease, reproductive disease, diabetes, inflammation, and immunodeficiency states. By analyzing blood samples, researchers gain valuable insights into the metabolic perturbations associated with these diseases, potentially leading to the identification of novel biomarkers and the development of personalized therapeutic approaches. Furthermore, we provide a comprehensive overview of various mass spectrometry approaches utilized in blood metabolomics research, including GC-MS, LC-MS, and others discussing their advantages and limitations. To enhance the scope, we propose including recent review articles supporting the applicability of GC×GC-MS for metabolomics-based studies. This addition will contribute to a more exhaustive understanding of the available analytical techniques. The Integration of mass spectrometry-based blood profiling into clinical practice holds promise for improving disease diagnosis, treatment monitoring, and patient outcomes. By unraveling the complex metabolic alterations associated with non-cancer diseases, researchers and healthcare professionals can pave the way for precision medicine and personalized therapeutic interventions. Continuous advancements in mass spectrometry technology and data analysis methods will further enhance the potential of blood metabolomics profiling in non-cancer diseases, facilitating its translation from the laboratory to routine clinical application.
Collapse
Affiliation(s)
- Ekaterina Demicheva
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Ekaterinburg 620049, Russia
| | - Vladislav Dordiuk
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
| | - Fernando Polanco Espino
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
| | - Konstantin Ushenin
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Autonomous Non-Profit Organization Artificial Intelligence Research Institute (AIRI), Moscow 105064, Russia
| | - Saied Aboushanab
- Institute of Chemical Engineering, Ural Federal University, Ekaterinburg 620002, Russia; (S.A.); (V.S.); (E.K.)
| | - Vadim Shevyrin
- Institute of Chemical Engineering, Ural Federal University, Ekaterinburg 620002, Russia; (S.A.); (V.S.); (E.K.)
| | - Aleksey Buhler
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
| | - Elena Mukhlynina
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Ekaterinburg 620049, Russia
| | - Olga Solovyova
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Ekaterinburg 620049, Russia
| | - Irina Danilova
- Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg 620075, Russia; (V.D.); (F.P.E.); (K.U.); (A.B.); (E.M.); (O.S.); (I.D.)
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Ekaterinburg 620049, Russia
| | - Elena Kovaleva
- Institute of Chemical Engineering, Ural Federal University, Ekaterinburg 620002, Russia; (S.A.); (V.S.); (E.K.)
| |
Collapse
|
17
|
Farage G, Zhao C, Choi HY, Garrett TJ, Kechris K, Elam MB, Sen Ś. Matrix Linear Models for connecting metabolite composition to individual characteristics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.19.572450. [PMID: 38187579 PMCID: PMC10769268 DOI: 10.1101/2023.12.19.572450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
High-throughput metabolomics data provide a detailed molecular window into biological processes. We consider the problem of assessing how the association of metabolite levels with individual (sample) characteristics such as sex or treatment may depend on metabolite characteristics such as pathway. Typically this is one in a two-step process: In the first step we assess the association of each metabolite with individual characteristics. In the second step an enrichment analysis is performed by metabolite characteristics among significant associations. We combine the two steps using a bilinear model based on the matrix linear model (MLM) framework we have previously developed for high-throughput genetic screens. Our framework can estimate relationships in metabolites sharing known characteristics, whether categorical (such as type of lipid or pathway) or numerical (such as number of double bonds in triglycerides). We demonstrate how MLM offers flexibility and interpretability by applying our method to three metabolomic studies. We show that our approach can separate the contribution of the overlapping triglycerides characteristics, such as the number of double bonds and the number of carbon atoms. The proposed method have been implemented in the open-source Julia package, MatrixLM. Data analysis scripts with example data analyses are also available.
Collapse
Affiliation(s)
- Gregory Farage
- Department of Preventive Medicine, Division of Biostatistics, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Chenhao Zhao
- Department of Preventive Medicine, Division of Biostatistics, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Hyo Young Choi
- Department of Preventive Medicine, Division of Biostatistics, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Katerina Kechris
- Department of Biostatistics & Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Marshall B Elam
- Department of Pharmacology and of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Śaunak Sen
- Department of Preventive Medicine, Division of Biostatistics, University of Tennessee Health Science Center Memphis, TN 38163
| |
Collapse
|
18
|
Ly R, Torres LC, Ly N, Britz-McKibbin P. Expanding Lipidomic Coverage in Multisegment Injection-Nonaqueous Capillary Electrophoresis-Mass Spectrometry via a Convenient and Quantitative Methylation Strategy. Anal Chem 2023; 95:17513-17524. [PMID: 37991882 DOI: 10.1021/acs.analchem.3c02605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Orthogonal separation techniques coupled to high-resolution mass spectrometry are required for characterizing the human lipidome, given its inherent chemical and structural complexity. However, electrophoretic separations remain largely unrecognized in contemporary lipidomics research compared to established chromatographic and ion mobility methods. Herein, we introduce a novel derivatization protocol based on 3-methyl-1-p-tolyltriazene (MTT) as a safer alternative to diazomethane for quantitative phospholipid (PL) methylation (∼90%), which enables their rapid analysis by multisegment injection-nonaqueous capillary electrophoresis-mass spectrometry (MSI-NACE-MS). Isobaric interferences and ion suppression effects were minimized by performing an initial reaction using 9-fluorenylmethyoxycarbonyl chloride prior to MTT and a subsequent back extraction in hexane. This charge-switch derivatization strategy expands lipidome coverage when using MSI-NACE-MS under positive ion mode with improved resolution, greater sensitivity, and higher throughput (∼3.5 min/sample), notably for zwitterionic PLs that are analyzed as their cationic phosphate methyl esters. Our method was validated by analyzing methyl-tert-butyl ether extracts of reference human plasma, which enabled a direct comparison of 48 phosphatidylcholine and 27 sphingomyelin species previously reported in an interlaboratory lipidomics harmonization study. The potential for plasma PL quantification by MSI-NACE-MS via a serial dilution of NIST SRM-1950 was also demonstrated based on estimation of relative response factors using their reported consensus concentrations. Moreover, lipid identification was supported by modeling predictable changes in the electrophoretic mobility for cationic PLs in conjunction with MS/MS. Overall, this work offers a practical derivatization protocol to expand lipidome coverage in CE-MS beyond the analysis of hydrophilic/polar metabolites under aqueous buffer conditions.
Collapse
Affiliation(s)
- Ritchie Ly
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, Canada L8S 4M1
| | - Lucas Christian Torres
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, Canada L8S 4M1
| | - Nicholas Ly
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, Canada L8S 4M1
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario, Canada L8S 4M1
| |
Collapse
|
19
|
Chicanne G, Darcourt J, Bertrand-Michel J, Garcia C, Ribes A, Payrastre B. What can we learn from the platelet lipidome? Platelets 2023; 34:2182180. [PMID: 36880158 DOI: 10.1080/09537104.2023.2182180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Besides their proteome, platelets use, in all responses to the environmental cues, a huge and diverse family of hydrophobic and amphipathic small molecules involved in structural, metabolic and signaling functions; the lipids. Studying how platelet lipidome changes modulate platelet function is an old story constantly renewed through the impressive technical advances allowing the discovery of new lipids, functions and metabolic pathways. Technical progress in analytical lipidomic profiling by top-of-the-line approaches such as nuclear magnetic resonance and gas chromatography or liquid chromatography coupled to mass spectrometry enables either large-scale analysis of lipids or targeted lipidomics. With the support of bioinformatics tools and databases, it is now possible to investigate thousands of lipids over a concentration range of several orders of magnitude. The lipidomic landscape of platelets is considered a treasure trove, not only able to expand our knowledge of platelet biology and pathologies but also to bring diagnostic and therapeutic opportunities. The aim of this commentary article is to summarize the advances in the field and to highlight what lipidomics can tell us about platelet biology and pathophysiology.
Collapse
Affiliation(s)
- Gaëtan Chicanne
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France
| | - Jean Darcourt
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France
| | - Justine Bertrand-Michel
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Cédric Garcia
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,Laboratory of Haematology, University Hospital of Toulouse, Toulouse, France
| | - Agnès Ribes
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,Laboratory of Haematology, University Hospital of Toulouse, Toulouse, France
| | - Bernard Payrastre
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France.,Laboratory of Haematology, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
20
|
Beyene HB, Giles C, Huynh K, Wang T, Cinel M, Mellett NA, Olshansky G, Meikle TG, Watts GF, Hung J, Hui J, Cadby G, Beilby J, Blangero J, Moses EK, Shaw JE, Magliano DJ, Meikle PJ. Metabolic phenotyping of BMI to characterize cardiometabolic risk: evidence from large population-based cohorts. Nat Commun 2023; 14:6280. [PMID: 37805498 PMCID: PMC10560260 DOI: 10.1038/s41467-023-41963-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/26/2023] [Indexed: 10/09/2023] Open
Abstract
Obesity is a risk factor for type 2 diabetes and cardiovascular disease. However, a substantial proportion of patients with these conditions have a seemingly normal body mass index (BMI). Conversely, not all obese individuals present with metabolic disorders giving rise to the concept of "metabolically healthy obese". We use lipidomic-based models for BMI to calculate a metabolic BMI score (mBMI) as a measure of metabolic dysregulation associated with obesity. Using the difference between mBMI and BMI (mBMIΔ), we identify individuals with a similar BMI but differing in their metabolic health and disease risk profiles. Exercise and diet associate with mBMIΔ suggesting the ability to modify mBMI with lifestyle intervention. Our findings show that, the mBMI score captures information on metabolic dysregulation that is independent of the measured BMI and so provides an opportunity to assess metabolic health to identify "at risk" individuals for targeted intervention and monitoring.
Collapse
Affiliation(s)
- Habtamu B Beyene
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Tingting Wang
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia
| | - Michelle Cinel
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | | | - Thomas G Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, WA, Australia
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, WA, Australia
| | - Joseph Hung
- School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Jennie Hui
- PathWest Laboratory Medicine of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- School of Population and Global Health, University of Western Australia, Crawley, WA, Australia
| | - Gemma Cadby
- School of Population and Global Health, University of Western Australia, Crawley, WA, Australia
| | - John Beilby
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - John Blangero
- South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Eric K Moses
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Jonathan E Shaw
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC, Australia.
- Baker Department of Cardiometabolic Health, Melbourne University, Melbourne, VIC, Australia.
| |
Collapse
|
21
|
Shields PG. Role of untargeted omics biomarkers of exposure and effect for tobacco research. ADDICTION NEUROSCIENCE 2023; 7:100098. [PMID: 37396411 PMCID: PMC10310069 DOI: 10.1016/j.addicn.2023.100098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Tobacco research remains a clear priority to improve individual and population health, and has recently become more complex with emerging combustible and noncombustible tobacco products. The use of omics methods in prevention and cessation studies are intended to identify new biomarkers for risk, compared risks related to other products and never use, and compliance for cessation and reinitation. to assess the relative effects of tobacco products to each other. They are important for the prediction of reinitiation of tobacco use and relapse prevention. In the research setting, both technical and clinical validation is required, which presents a number of complexities in the omics methodologies from biospecimen collection and sample preparation to data collection and analysis. When the results identify differences in omics features, networks or pathways, it is unclear if the results are toxic effects, a healthy response to a toxic exposure or neither. The use of surrogate biospecimens (e.g., urine, blood, sputum or nasal) may or may not reflect target organs such as the lung or bladder. This review describes the approaches for the use of omics in tobacco research and provides examples of prior studies, along with the strengths and limitations of the various methods. To date, there is little consistency in results, likely due to small number of studies, limitations in study size, the variability in the analytic platforms and bioinformatic pipelines, differences in biospecimen collection and/or human subject study design. Given the demonstrated value for the use of omics in clinical medicine, it is anticipated that the use in tobacco research will be similarly productive.
Collapse
Affiliation(s)
- Peter G. Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH
| |
Collapse
|
22
|
Wu Q, Huang Y, Kong X, Jia B, Lu X, Chen Y, Huang Z, Li YY, Dai W. DBLiPro: A Database for Lipids and Proteins in Human Lipid Metabolism. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:350-359. [PMID: 37589022 PMCID: PMC10425311 DOI: 10.1007/s43657-023-00099-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 08/18/2023]
Abstract
To help researchers in the field of biology, medicine, chemistry, and materials science to use lipidomic data conveniently, there is an urgent need to develop a platform that provides a systematic knowledgebase of human lipid metabolism and lipidome-centric omics analysis tools. DBLiPro is a user-friendly webserver allowing for access to human metabolism-related lipids and proteins knowledge database and an interactive bioinformatics integrative analysis workflow for lipidomics, transcriptomics, and proteomics data. In DBLiPro, there are 3109 lipid-associated proteins (LAPs) and 2098 lipid metabolites in the knowledge base section, which were obtained from Uniprot, Kyoto Encyclopedia of Genes and Genomes (KEGG) and were further annotated by information from other public resources in the knowledge base section, such as RaftProt and PubChem. DBLiPro offers a step-by-step interactive analysis workflow for lipidomics, transcriptomics, proteomics, and their integrating multi-omics analysis focusing on the human lipid metabolism. In summary, DBLiPro is capable of helping users discover key molecules (lipids and proteins) in human lipid metabolism and investigate lipid-protein functions underlying mechanisms based on their own omics data. The DBLiPro is freely available at http://lipid.cloudna.cn/home.
Collapse
Affiliation(s)
- Qian Wu
- NHC Key Laboratory of Reproduction Regulation & Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Fudan University, No. 779, Old Hu Min Road, Shanghai, 200237 China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203 China
| | - Yuanyuan Huang
- Beijing Cloudna Technology Co., Ltd., No. 53, Yanqi Street, Beijing, 100101 China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203 China
| | - Xiangya Kong
- Beijing Cloudna Technology Co., Ltd., No. 53, Yanqi Street, Beijing, 100101 China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203 China
| | - Ben Jia
- NHC Key Laboratory of Reproduction Regulation & Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Fudan University, No. 779, Old Hu Min Road, Shanghai, 200237 China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203 China
| | - Xiaoting Lu
- NHC Key Laboratory of Reproduction Regulation & Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Fudan University, No. 779, Old Hu Min Road, Shanghai, 200237 China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203 China
| | - Yunqin Chen
- NHC Key Laboratory of Reproduction Regulation & Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Fudan University, No. 779, Old Hu Min Road, Shanghai, 200237 China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203 China
| | - Zechi Huang
- Beijing Cloudna Technology Co., Ltd., No. 53, Yanqi Street, Beijing, 100101 China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203 China
| | - Yuan-Yuan Li
- NHC Key Laboratory of Reproduction Regulation & Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Fudan University, No. 779, Old Hu Min Road, Shanghai, 200237 China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203 China
| | - Wentao Dai
- NHC Key Laboratory of Reproduction Regulation & Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Fudan University, No. 779, Old Hu Min Road, Shanghai, 200237 China
- Shanghai Key Laboratory of Gastric Neoplasms, Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, 201203 China
| |
Collapse
|
23
|
Mingo-Casas P, Sanchez-Céspedes J, Blázquez AB, Casas J, Balsera-Manzanero M, Herrero L, Vázquez A, Pachón J, Aguilar-Guisado M, Cisneros JM, Saiz JC, Martín-Acebes MA. Lipid signatures of West Nile virus infection unveil alterations of sphingolipid metabolism providing novel biomarkers. Emerg Microbes Infect 2023:2231556. [PMID: 37377355 DOI: 10.1080/22221751.2023.2231556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
West Nile virus (WNV) is a neurotropic flavivirus transmitted by the bites of infected mosquitoes. Severe forms of West Nile disease (WND) can curse with meningitis, encephalitis or acute flaccid paralysis. A better understanding of the physiopathology associated with disease progression is mandatory to find biomarkers and effective therapies. In this scenario, blood derivatives (plasma and serum) constitute the more commonly used biofluids due to its ease of collection and high value for diagnostic purposes. Therefore, the potential impact of this virus in the circulating lipidome was addressed combining the analysis of samples from experimentally infected mice and naturally WND patients. Our results unveil dynamic alterations in the lipidome that define specific metabolic fingerprints of different infection stages. Concomitant with neuroinvasion in mice, the lipid landscape was dominated by a metabolic reprograming that resulted in significant elevations of circulating sphingolipids (ceramides, dihydroceramides and dihydrosphingomyelins), phosphatidylethanolamines and triacylglycerols. Remarkably, patients suffering from WND also displayed an elevation of ceramides, dihydroceramides, lactosylceramides and monoacylglycerols in their sera. The dysregulation of sphingolipid metabolism by WNV may provide new therapeutic opportunities and supports the potential of certain lipids as novel peripheral biomarkers of WND progression.
Collapse
Affiliation(s)
- Patricia Mingo-Casas
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Javier Sanchez-Céspedes
- Department of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana-Belén Blázquez
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Josefina Casas
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Balsera-Manzanero
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
| | - Lura Herrero
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Vázquez
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Jerónimo Pachón
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
- Department of Medicine, School of Medicine, University of Seville, Seville, Spain
| | - Manuela Aguilar-Guisado
- Department of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - José Miguel Cisneros
- Department of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan-Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Miguel A Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| |
Collapse
|
24
|
Babu M, Snyder M. Multi-Omics Profiling for Health. Mol Cell Proteomics 2023; 22:100561. [PMID: 37119971 PMCID: PMC10220275 DOI: 10.1016/j.mcpro.2023.100561] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023] Open
Abstract
The world has witnessed a steady rise in both non-infectious and infectious chronic diseases, prompting a cross-disciplinary approach to understand and treating disease. Current medical care focuses on treating people after they become patients rather than preventing illness, leading to high costs in treating chronic and late-stage diseases. Additionally, a "one-size-fits all" approach to health care does not take into account individual differences in genetics, environment, or lifestyle factors, decreasing the number of people benefiting from interventions. Rapid advances in omics technologies and progress in computational capabilities have led to the development of multi-omics deep phenotyping, which profiles the interaction of multiple levels of biology over time and empowers precision health approaches. This review highlights current and emerging multi-omics modalities for precision health and discusses applications in the following areas: genetic variation, cardio-metabolic diseases, cancer, infectious diseases, organ transplantation, pregnancy, and longevity/aging. We will briefly discuss the potential of multi-omics approaches in disentangling host-microbe and host-environmental interactions. We will touch on emerging areas of electronic health record and clinical imaging integration with muti-omics for precision health. Finally, we will briefly discuss the challenges in the clinical implementation of multi-omics and its future prospects.
Collapse
Affiliation(s)
- Mohan Babu
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA.
| |
Collapse
|
25
|
Zhang J, Yang S, Wang J, Xu Y, Zhao H, Lei J, Zhou Y, Chen Y, Wu L, Li Y. Equivalent carbon number based targeted odd chain fatty acyl lipidomics reveals triacylglycerol profiling in clinical colon cancer. J Lipid Res 2023:100393. [PMID: 37257561 PMCID: PMC10331287 DOI: 10.1016/j.jlr.2023.100393] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
Odd chain fatty acids (OCFAs) present in very low level at nearly 1% of total fatty acids in human plasma and thus their functions were usually ignored. Recent epidemiological studies have shown that OCFAs are inversely associated with a variety of disease risks. However, the contribution of OCFAs incorporated into complex lipids remains elusive. Here, we developed a targeted odd chain fatty acyl containing lipidomics method based on equivalent carbon number and retention time prediction. The method displayed good reproducibility and robustness as shown by peak width at half height within 0.7 min and coefficient of variation (CV) under 20%. A total number of 776 lipid species with odd chain fatty acyl residues could be detected in the electrospray ionization (ESI) mode of reverse phase liquid chromatography-mass spectrometry, of which 309 lipids were further validated using multiple MRM transitiions. Using this method, we quantified odd chain fatty acyl containing lipidome in tissues from 12 colon cancer patients, revealing the remodeling of triacylglycerol (TAG). The dynamics of odd chain fatty acyl lipids were further consolidated by the association with genomic and proteomic feature of altered catabolism of branched chain amino acids and TAG endogenous synthesis in colon cancer. This lipidomics approach will be applicable for screening of dysregulated odd chain fatty acyl lipids, which enriches and improves the methods for diagnosis and prognosis evaluation of cancer using lipidomics.
Collapse
Affiliation(s)
- Jiangang Zhang
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Shuai Yang
- Department of Pathology, the 958th Hospital, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Jingchun Wang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Yanquan Xu
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Huakan Zhao
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Juan Lei
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yu Zhou
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yu Chen
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China.
| |
Collapse
|
26
|
Chen P, Wang R, Liu F, Li S, Gu Y, Wang L, Yuan Y. Schizandrin C regulates lipid metabolism and inflammation in liver fibrosis by NF-κB and p38/ERK MAPK signaling pathways. Front Pharmacol 2023; 14:1092151. [PMID: 37288106 PMCID: PMC10242051 DOI: 10.3389/fphar.2023.1092151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/25/2023] [Indexed: 06/09/2023] Open
Abstract
Liver fibrosis is considered a sustained wound healing response and metabolic syndrome, and its therapy is of great significance for chronic liver disease. Schizandrin C, as one lignan from hepatic protectant Schisandra chinensis, can depress the oxidative effect and lipid peroxidation, and protect against liver injury. In this study, C57BL/6J mice were used to estimate a liver fibrosis model by CCl4, and Schizandrin C exerted an anti-hepatic fibrosis effect, as evidenced by decreased alanine aminotransferase, aspartate aminotransferase and total bilirubin activities in serum, lower hydroxyproline content, recuperative structure and less collagen accumulation in the liver. In addition, Schizandrin C reduced the expressions of alpha-smooth muscle actin and type Ι collagen in the liver. In vitro experiments also revealed that Schizandrin C attenuated hepatic stellate cell activation in both LX-2 and HSC-T6 cells. Furthermore, lipidomics and quantitative real-time PCR analysis revealed that Schizandrin C regulated the lipid profile and related metabolic enzymes in the liver. In addition, the mRNA levels of inflammation factors were downregulated by Schizandrin C treatment, accompanied by lower protein levels of IκB-Kinase-β, nuclear factor kappa-B p65, and phospho-nuclear factor kappa-B p65. Finally, Schizandrin C inhibited the phosphorylation of p38 MAP kinase and extracellular signal-regulated protein kinase, which were activated in the CCl4 fibrotic liver. Taken together, Schizandrin C can regulate lipid metabolism and inflammation to ameliorate liver fibrosis by nuclear factor kappa-B and p38/ERK MAPK signaling pathways. These findings supported Schizandrin C as a potential drug for liver fibrosis.
Collapse
Affiliation(s)
- Panpan Chen
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Rong Wang
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Fangbin Liu
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Shengnan Li
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yanqiu Gu
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Ross DH, Guo J, Bilbao A, Huan T, Smith RD, Zheng X. Evaluating Software Tools for Lipid Identification from Ion Mobility Spectrometry-Mass Spectrometry Lipidomics Data. Molecules 2023; 28:molecules28083483. [PMID: 37110719 PMCID: PMC10142755 DOI: 10.3390/molecules28083483] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The unambiguous identification of lipids is a critical component of lipidomics studies and greatly impacts the interpretation and significance of analyses as well as the ultimate biological understandings derived from measurements. The level of structural detail that is available for lipid identifications is largely determined by the analytical platform being used. Mass spectrometry (MS) coupled with liquid chromatography (LC) is the predominant combination of analytical techniques used for lipidomics studies, and these methods can provide fairly detailed lipid identification. More recently, ion mobility spectrometry (IMS) has begun to see greater adoption in lipidomics studies thanks to the additional dimension of separation that it provides and the added structural information that can support lipid identification. At present, relatively few software tools are available for IMS-MS lipidomics data analysis, which reflects the still limited adoption of IMS as well as the limited software support. This fact is even more pronounced for isomer identifications, such as the determination of double bond positions or integration with MS-based imaging. In this review, we survey the landscape of software tools that are available for the analysis of IMS-MS-based lipidomics data and we evaluate lipid identifications produced by these tools using open-access data sourced from the peer-reviewed lipidomics literature.
Collapse
Affiliation(s)
- Dylan H Ross
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Jian Guo
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99354, USA
- Department of Chemistry, Faculty of Science, University of British Columbia, Vancouver Campus, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| | - Aivett Bilbao
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Tao Huan
- Department of Chemistry, Faculty of Science, University of British Columbia, Vancouver Campus, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| | - Richard D Smith
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Xueyun Zheng
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| |
Collapse
|
28
|
Kadoguchi T, Shimada K, Fukui N, Tanaka N, Tsuno H, Shiozawa T, Fukao K, Nishitani‐Yokoyama M, Isoda K, Matsushita S, Yokoyama N, Daida H. Accumulation of polyunsaturated fatty acid-derived metabolites in the sarcopenic muscle of aging mice. Geriatr Gerontol Int 2023; 23:297-303. [PMID: 36811314 PMCID: PMC11503557 DOI: 10.1111/ggi.14561] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 12/26/2022] [Accepted: 01/27/2023] [Indexed: 02/24/2023]
Abstract
AIM Although it is known that advanced age alters skeletal muscle lipid metabolism, the role(s) of polyunsaturated fatty acid-derived metabolites (mostly eicosanoids and docosanoids) in sarcopenia are not clear. We therefore examined the changes in the metabolites of arachidonic acid, eicosapentaenoic acid and docosahexaenoic acid in the sarcopenic muscle of aged mice. METHODS We used 6- and 24-month-old male C57BL/6J mice as healthy and sarcopenic muscle models, respectively. Skeletal muscles were removed from the lower limb and subjected to a liquid chromatography-tandem mass spectrometry analysis. RESULTS The liquid chromatography-tandem mass spectrometry analysis detected distinct changes of metabolites in the muscles of the aged mice. Of the 63 metabolites identified, nine were significantly higher in the sarcopenic muscle of aged mice compared with the healthy muscle of young mice. In particular, prostaglandin E2 , prostaglandin F2a , thromboxane B2 , 5-hydroxyeicosatetraenoic acid, and 15-oxo-eicosatetraenoic acid (arachidonic acid-derived metabolites), 12-hydroxy-eicosapentaenoic acid and 14,15-epoxy-eicosatetraenoic acid (eicosapentaenoic acid-derived metabolites) and 10-hydroxydocosa-hexaenoic acid and 14-hydroxyoctadeca-pentaenoic acid (docosahexaenoic acid-derived metabolites) were significantly higher in aged tissue compared with young tissue (all P < 0.05). CONCLUSIONS We observed the accumulation of metabolites in the sarcopenic muscle of aged mice. Our results may provide new insights into the pathogenesis and progression of aging- or disease-related sarcopenia. Geriatr Gerontol Int 2023; 23: 297-303.
Collapse
Affiliation(s)
- Tomoyasu Kadoguchi
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
- Sportology CenterJuntendo University Graduate School of MedicineTokyoJapan
| | - Kazunori Shimada
- Department of Cardiovascular Biology and MedicineJuntendo University Graduate School of MedicineTokyoJapan
| | - Naoshi Fukui
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
- Clinical Research Center, National Hospital Organization Sagamihara HospitalSagamiharaJapan
| | - Nobuho Tanaka
- Clinical Research Center, National Hospital Organization Sagamihara HospitalSagamiharaJapan
| | - Hirotaka Tsuno
- Clinical Research Center, National Hospital Organization Sagamihara HospitalSagamiharaJapan
| | - Tomoyuki Shiozawa
- Department of Cardiovascular Biology and MedicineJuntendo University Graduate School of MedicineTokyoJapan
- Department of Cardiovascular MedicineJuntendo University Shizuoka HospitalIzunokuniJapan
| | - Kosuke Fukao
- Department of Cardiovascular Biology and MedicineJuntendo University Graduate School of MedicineTokyoJapan
- Graduate School of Health and Sports ScienceJuntendo UniversityChibaJapan
| | - Miho Nishitani‐Yokoyama
- Department of Cardiovascular Biology and MedicineJuntendo University Graduate School of MedicineTokyoJapan
| | - Kikuo Isoda
- Department of Cardiovascular Biology and MedicineJuntendo University Graduate School of MedicineTokyoJapan
| | - Satoshi Matsushita
- Department of Cardiovascular SurgeryJuntendo University Graduate School of MedicineTokyoJapan
| | - Norihiko Yokoyama
- Department of Cardiovascular SurgeryJuntendo University Graduate School of MedicineTokyoJapan
| | - Hiroyuki Daida
- Sportology CenterJuntendo University Graduate School of MedicineTokyoJapan
- Department of Cardiovascular Biology and MedicineJuntendo University Graduate School of MedicineTokyoJapan
- Faculty of Health ScienceJuntendo UniversityTokyoJapan
| |
Collapse
|
29
|
Dorochow E, Gurke R, Rischke S, Geisslinger G, Hahnefeld L. Effects of Different Storage Conditions on Lipid Stability in Mice Tissue Homogenates. Metabolites 2023; 13:metabo13040504. [PMID: 37110163 PMCID: PMC10144362 DOI: 10.3390/metabo13040504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Lipids are biomolecules involved in numerous (patho-)physiological processes and their elucidation in tissue samples is of particular interest. However, tissue analysis goes hand in hand with many challenges and the influence of pre-analytical factors can intensively change lipid concentrations ex vivo, compromising the results of the whole research project. Here, we study the influence of pre-analytical factors on lipid profiles during the processing of homogenized tissues. Homogenates from four different mice tissues (liver, kidney, heart, spleen) were stored at room temperature as well as in ice water for up to 120 min and analyzed via ultra-high-performance liquid chromatography-high-resolution mass spectrometry (UHPLC-HRMS). Lipid class ratios were calculated since their suitability as indicators for sample stability has been previously illustrated. Only approx. 40% of lipid class ratios were unchanged after 35 min, which was further reduced to 25% after 120 min during storage at room temperature. In contrast, lipids in tissue homogenates were generally stable when samples were kept in ice water, as more than 90% of investigated lipid class ratios remained unchanged after 35 min. Ultimately, swift processing of tissue homogenates under cooled conditions represents a viable option for lipid analysis and pre-analytical factors require more attention to achieve reliable results.
Collapse
Affiliation(s)
- Erika Dorochow
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Robert Gurke
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Correspondence: (R.G.); (L.H.)
| | - Samuel Rischke
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Lisa Hahnefeld
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe University, Theodor Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Correspondence: (R.G.); (L.H.)
| |
Collapse
|
30
|
Kvasnička A, Najdekr L, Dobešová D, Piskláková B, Ivanovová E, Friedecký D. Clinical lipidomics in the era of the big data. Clin Chem Lab Med 2023; 61:587-598. [PMID: 36592414 DOI: 10.1515/cclm-2022-1105] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/16/2022] [Indexed: 01/03/2023]
Abstract
Lipidomics as a branch of metabolomics provides unique information on the complex lipid profile in biological materials. In clinically focused studies, hundreds of lipids together with available clinical information proved to be an effective tool in the discovery of biomarkers and understanding of pathobiochemistry. However, despite the introduction of lipidomics nearly twenty years ago, only dozens of big data studies using clinical lipidomics have been published to date. In this review, we discuss the lipidomics workflow, statistical tools, and the challenges of standartisation. The consequent summary divided into major clinical areas of cardiovascular disease, cancer, diabetes mellitus, neurodegenerative and liver diseases is demonstrating the importance of clinical lipidomics. In these publications, the potential of lipidomics for prediction, diagnosis or finding new targets for the treatment of selected diseases can be seen. The first of these results have already been implemented in clinical practice in the field of cardiovascular diseases, while in other areas we can expect the application of the results summarized in this review in the near future.
Collapse
Affiliation(s)
- Aleš Kvasnička
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| | - Lukáš Najdekr
- Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czechia
| | - Dana Dobešová
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| | - Barbora Piskláková
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| | - Eliška Ivanovová
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| | - David Friedecký
- Laboratory for Inherited Metabolic Disorders, Department of Clinical Biochemistry, University Hospital, Olomouc, Czechia
- Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czechia
| |
Collapse
|
31
|
Huynh K, Duong T, Mellett NA, Cinel M, Giles C, Meikle PJ. Comprehensive Targeted Lipidomic Profiling for Research and Clinical Applications. Methods Mol Biol 2023; 2628:489-504. [PMID: 36781803 DOI: 10.1007/978-1-0716-2978-9_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Mass spectrometry remains one of the gold standard approaches in examining the lipidome in biological samples. Recently, advancements in chromatography and mass spectrometry approaches have enabled broad coverage of the lipidome. However, many limitations still exist, and lipidomic analysis often requires a fine balance between coverage of the lipidome, structural detail, and sample throughput. For biomedical and clinical research using human samples, the diversity and natural variation between different individuals necessitate larger sample numbers to identify significant associations with clinical outcomes and account for potential confounding factors. Here we describe a targeted lipidomics workflow that enables reproducible profiling of thousands of plasma samples in a systematic manner, while maintaining good structural detail and high coverage of the lipidome.
Collapse
Affiliation(s)
- Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia.,Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Thy Duong
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | - Michelle Cinel
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia.,Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. .,Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia. .,Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia.
| |
Collapse
|
32
|
Staab TA, McIntyre G, Wang L, Radeny J, Bettcher L, Guillen M, Peck MP, Kalil AP, Bromley SP, Raftery D, Chan JP. The lipidomes of C. elegans with mutations in asm-3/acid sphingomyelinase and hyl-2/ceramide synthase show distinct lipid profiles during aging. Aging (Albany NY) 2023; 15:650-674. [PMID: 36787434 PMCID: PMC9970312 DOI: 10.18632/aging.204515] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023]
Abstract
Lipid metabolism affects cell and physiological functions that mediate animal healthspan and lifespan. Lipidomics approaches in model organisms have allowed us to better understand changes in lipid composition related to age and lifespan. Here, using the model C. elegans, we examine the lipidomes of mutants lacking enzymes critical for sphingolipid metabolism; specifically, we examine acid sphingomyelinase (asm-3), which breaks down sphingomyelin to ceramide, and ceramide synthase (hyl-2), which synthesizes ceramide from sphingosine. Worm asm-3 and hyl-2 mutants have been previously found to be long- and short-lived, respectively. We analyzed longitudinal lipid changes in wild type animals compared to mutants at 1-, 5-, and 10-days of age. We detected over 700 different lipids in several lipid classes. Results indicate that wildtype animals exhibit increased triacylglycerols (TAG) at 10-days compared to 1-day, and decreased lysophoshatidylcholines (LPC). We find that 10-day hyl-2 mutants have elevated total polyunsaturated fatty acids (PUFA) and increased LPCs compared to 10-day wildtype animals. These changes mirror another short-lived model, the daf-16/FOXO transcription factor that is downstream of the insulin-like signaling pathway. In addition, we find that hyl-2 mutants have poor oxidative stress response, supporting a model where mutants with elevated PUFAs may accumulate more oxidative damage. On the other hand, 10-day asm-3 mutants have fewer TAGs. Intriguingly, asm-3 mutants have a similar lipid composition as the long-lived, caloric restriction model eat-2/mAChR mutant. Together, these analyses highlight the utility of lipidomic analyses to characterize metabolic changes during aging in C. elegans.
Collapse
Affiliation(s)
- Trisha A. Staab
- Department of Biology, Marian University, Indianapolis, IN 46222, USA
| | - Grace McIntyre
- Department of Biology, Marian University, Indianapolis, IN 46222, USA
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Joycelyn Radeny
- Department of Biology, Juniata College, Huntingdon, PA 16652, USA
| | - Lisa Bettcher
- Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98195, USA
| | - Melissa Guillen
- Department of Biology, Marian University, Indianapolis, IN 46222, USA
| | - Margaret P. Peck
- Department of Biology, Juniata College, Huntingdon, PA 16652, USA
| | - Azia P. Kalil
- Department of Biology, Juniata College, Huntingdon, PA 16652, USA
| | | | - Daniel Raftery
- Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98195, USA
| | - Jason P. Chan
- Department of Biology, Marian University, Indianapolis, IN 46222, USA
| |
Collapse
|
33
|
Scheinberg T, Mak B, Butler L, Selth L, Horvath LG. Targeting lipid metabolism in metastatic prostate cancer. Ther Adv Med Oncol 2023; 15:17588359231152839. [PMID: 36743527 PMCID: PMC9893394 DOI: 10.1177/17588359231152839] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023] Open
Abstract
Despite key advances in the treatment of prostate cancer (PCa), a proportion of men have de novo resistance, and all will develop resistance to current therapeutics over time. Aberrant lipid metabolism has long been associated with prostate carcinogenesis and progression, but more recently there has been an explosion of preclinical and clinical data which is informing new clinical trials. This review explores the epidemiological links between obesity and metabolic syndrome and PCa, the evidence for altered circulating lipids in PCa and their potential role as biomarkers, as well as novel therapeutic strategies for targeting lipids in men with PCa, including therapies widely used in cardiovascular disease such as statins, metformin and lifestyle modification, as well as novel targeted agents such as sphingosine kinase inhibitors, DES1 inhibitors and agents targeting FASN and beta oxidation.
Collapse
Affiliation(s)
- Tahlia Scheinberg
- Medical Oncology, Chris O’Brien Lifehouse, Camperdown NSW, Australia,Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia,University of Sydney, Camperdown, NSW, Australia
| | - Blossom Mak
- Medical Oncology, Chris O’Brien Lifehouse, Camperdown NSW, Australia,Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia,University of Sydney, Camperdown, NSW, Australia
| | - Lisa Butler
- Prostate Cancer Research Group, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia,South Australian Immunogenomics Cancer Institute and Freemason’s Centre for Male Health and Wellbeing, University of Adelaide, South Australia, Australia
| | - Luke Selth
- South Australian Immunogenomics Cancer Institute and Freemason’s Centre for Male Health and Wellbeing, University of Adelaide, South Australia, Australia,Dame Roma Mitchell Cancer Research Labs, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia,Flinders Health and Medical Research Institute, Flinders University, College of Medicine and Public Health, Bedford Park, Australia
| | | |
Collapse
|
34
|
Zandl-Lang M, Plecko B, Köfeler H. Lipidomics-Paving the Road towards Better Insight and Precision Medicine in Rare Metabolic Diseases. Int J Mol Sci 2023; 24:ijms24021709. [PMID: 36675224 PMCID: PMC9866746 DOI: 10.3390/ijms24021709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Even though the application of Next-Generation Sequencing (NGS) has significantly facilitated the identification of disease-associated mutations, the diagnostic rate of rare diseases is still below 50%. This causes a diagnostic odyssey and prevents specific treatment, as well as genetic counseling for further family planning. Increasing the diagnostic rate and reducing the time to diagnosis in children with unclear disease are crucial for a better patient outcome and improvement of quality of life. In many cases, NGS reveals variants of unknown significance (VUS) that need further investigations. The delineation of novel (lipid) biomarkers is not only crucial to prove the pathogenicity of VUS, but provides surrogate parameters for the monitoring of disease progression and therapeutic interventions. Lipids are essential organic compounds in living organisms, serving as building blocks for cellular membranes, energy storage and signaling molecules. Among other disorders, an imbalance in lipid homeostasis can lead to chronic inflammation, vascular dysfunction and neurodegenerative diseases. Therefore, analyzing lipids in biological samples provides great insight into the underlying functional role of lipids in healthy and disease statuses. The method of choice for lipid analysis and/or huge assemblies of lipids (=lipidome) is mass spectrometry due to its high sensitivity and specificity. Due to the inherent chemical complexity of the lipidome and the consequent challenges associated with analyzing it, progress in the field of lipidomics has lagged behind other omics disciplines. However, compared to the previous decade, the output of publications on lipidomics has increased more than 17-fold within the last decade and has, therefore, become one of the fastest-growing research fields. Combining multiple omics approaches will provide a unique and efficient tool for determining pathogenicity of VUS at the functional level, and thereby identifying rare, as well as novel, genetic disorders by molecular techniques and biochemical analyses.
Collapse
Affiliation(s)
- Martina Zandl-Lang
- Division of General Pediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Barbara Plecko
- Division of General Pediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Harald Köfeler
- Core Facility Mass Spectrometry, ZMF, Medical University of Graz, 8036 Graz, Austria
- Correspondence:
| |
Collapse
|
35
|
Zeković M, Bumbaširević U, Živković M, Pejčić T. Alteration of Lipid Metabolism in Prostate Cancer: Multifaceted Oncologic Implications. Int J Mol Sci 2023; 24:ijms24021391. [PMID: 36674910 PMCID: PMC9863986 DOI: 10.3390/ijms24021391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Cancer is increasingly recognized as an extraordinarily heterogeneous disease featuring an intricate mutational landscape and vast intra- and intertumor variability on both genetic and phenotypic levels. Prostate cancer (PCa) is the second most prevalent malignant disease among men worldwide. A single metabolic program cannot epitomize the perplexing reprogramming of tumor metabolism needed to sustain the stemness of neoplastic cells and their prominent energy-consuming functional properties, such as intensive proliferation, uncontrolled growth, migration, and invasion. In cancerous tissue, lipids provide the structural integrity of biological membranes, supply energy, influence the regulation of redox homeostasis, contribute to plasticity, angiogenesis and microenvironment reshaping, mediate the modulation of the inflammatory response, and operate as signaling messengers, i.e., lipid mediators affecting myriad processes relevant for the development of the neoplasia. Comprehensive elucidation of the lipid metabolism alterations in PCa, the underlying regulatory mechanisms, and their implications in tumorigenesis and the progression of the disease are gaining growing research interest in the contemporary urologic oncology. Delineation of the unique metabolic signature of the PCa featuring major aberrant pathways including de novo lipogenesis, lipid uptake, storage and compositional reprogramming may provide novel, exciting, and promising avenues for improving diagnosis, risk stratification, and clinical management of such a complex and heterogeneous pathology.
Collapse
Affiliation(s)
- Milica Zeković
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Uros Bumbaširević
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marko Živković
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Tomislav Pejčić
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence:
| |
Collapse
|
36
|
Belkin TG, Tham YK, McMullen JR. Lipids regulated by exercise and PI3K: potential role as biomarkers and therapeutic targets for cardiovascular disease. CURRENT OPINION IN PHYSIOLOGY 2023. [DOI: 10.1016/j.cophys.2023.100633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
37
|
Bishop LM, Fiehn O. Comprehensive lipidomic profiling by plasma separation cards. Anal Bioanal Chem 2023; 415:193-201. [PMID: 36316462 PMCID: PMC10448968 DOI: 10.1007/s00216-022-04399-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/04/2022] [Accepted: 10/21/2022] [Indexed: 01/07/2023]
Abstract
Large-scale lipidomic analyses have been limited by the cost and accessibility of traditional venipuncture sampling. Microsampling techniques offer a less-invasive and more accessible alternative. From a single drop of blood, plasma separation cards (PSC) deliver two volumetric dried plasma samples which are studied here for profiling endogenous blood lipids. Six lots of EDTA-treated human whole blood were used to compare PSC, dried blood spot analyses (DBS), and classic wet plasma extractions. Six replicate extractions were performed for each lot. Nontargeted lipidomics was performed by liquid chromatography-high resolution tandem mass spectrometry. Lipids were annotated by accurate mass/retention time matching and MS/MS spectral library matching using peak intensities for quantitation. Four hundred ninety-eight compounds covering 24 lipid subclasses were annotated. Inter-lot repeatability was evaluated by the percent relative standard deviation (%RSD) for each lot, giving median %RSD values across the lots at 14.6% for PSC, 9.3% for DBS, and 8.6% for wet plasma. Strong correlations of lipid peak intensities between wet plasma and PSCs were observed, but less for DBS. Lipid recovery and stability were comparable between the PSC and DBS samples, with roughly 60% of annotated lipids stable at room temperature after 28 days. Overall, PSCs provide a better alternative for quantitative blood lipidomic analyses compared to dried blood spots. However, problems with lipid stability for samples handled and shipped at room temperature are currently unavoidable outside of a clinical setting. Data transferability and comparability to standard plasma is lipid and lipid class dependent.
Collapse
Affiliation(s)
- Lauren M Bishop
- Department of Chemistry, University of California Davis, Davis, CA, USA
- West Coast Metabolomics Center, University of California Davis, Davis, CA, USA
| | - Oliver Fiehn
- Department of Chemistry, University of California Davis, Davis, CA, USA.
- West Coast Metabolomics Center, University of California Davis, Davis, CA, USA.
| |
Collapse
|
38
|
Wang W, Song L. Landscape of lipidomics in cardiovascular medicine from 2012 to 2021: A systematic bibliometric analysis and literature review. Medicine (Baltimore) 2022; 101:e32599. [PMID: 36596038 PMCID: PMC9803420 DOI: 10.1097/md.0000000000032599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Lipidomics has shaped our knowledge of how lipids play a central role in cardiovascular diseases (CVD), whereas there is a lack of a summary of existing research findings. This study performed a bibliometric analysis of lipidomics research in cardiovascular medicine to reveal the core countries, institutions, key researchers, important references, major journals, research hotspots and frontiers in this field. From 2012 to 2021, a total of 761 articles were obtained from the Web of Science Core Collection database. There is a steady increase of publications yearly. The United States and China are on the top of the list regarding article output. The institutions with the most publications were the Baker Heart and Diabetes Institute, the Chinese Academy of Sciences and Harvard Medical School. Peter J Meikle was both the most published and most co-cited author. The major journal in this field is Journal of lipid research. Keyword co-occurrence analysis indicated that coronary heart disease, mass spectrometry, risk, fatty acid, and insulin resistance have become hot topics in this field and keyword burst detection suggests that metabolomics, activation, liver, low density lipoprotein are the frontiers of research in recent years. Collectively, lipidomics in CVD is still in its infancy with a steady increase yearly. More in-depth studies in this area are warranted in the future.
Collapse
Affiliation(s)
- Wenting Wang
- Department of Cardiovascular Disease, Affiliated Hangzhou Chest Hospital, Zhejiang University School of Medicine, Hangzhou, China
- * Correspondence: Wenting Wang, Department of Cardiology, Affiliated Hangzhou Chest Hospital, Zhejiang University School of Medicine, 208 Huancheng East Road, Hangzhou 310003, China (e-mail: )
| | - Lei Song
- Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
39
|
Bautista JS, Falabella M, Flannery PJ, Hanna MG, Heales SJ, Pope SA, Pitceathly RD. Advances in methods to analyse cardiolipin and their clinical applications. Trends Analyt Chem 2022; 157:116808. [PMID: 36751553 PMCID: PMC7614147 DOI: 10.1016/j.trac.2022.116808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cardiolipin (CL) is a mitochondria-exclusive phospholipid, primarily localised within the inner mitochondrial membrane, that plays an essential role in mitochondrial architecture and function. Aberrant CL content, structure, and localisation have all been linked to impaired mitochondrial activity and are observed in the pathophysiology of cancer and neurological, cardiovascular, and metabolic disorders. The detection, quantification, and localisation of CL species is a valuable tool to investigate mitochondrial dysfunction and the pathophysiological mechanisms underpinning several human disorders. CL is measured using liquid chromatography, usually combined with mass spectrometry, mass spectrometry imaging, shotgun lipidomics, ion mobility spectrometry, fluorometry, and radiolabelling. This review summarises available methods to analyse CL, with a particular focus on modern mass spectrometry, and evaluates their advantages and limitations. We provide guidance aimed at selecting the most appropriate technique, or combination of techniques, when analysing CL in different model systems, and highlight the clinical contexts in which measuring CL is relevant.
Collapse
Affiliation(s)
- Javier S. Bautista
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Micol Falabella
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Padraig J. Flannery
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK,Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK
| | - Michael G. Hanna
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK,NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Simon J.R. Heales
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK,NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK,Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Simon A.S. Pope
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London, UK,Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Robert D.S. Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK,NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK, Corresponding author. Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK. (R.D.S. Pitceathly)
| |
Collapse
|
40
|
Zhou Y, Chen J, Li S, Chen A, Dai C, Liu M, Lu D, Chen Z, Wang X, Qian J, Ge J. Prognostic implication of lipidomics in patients with coronary total occlusion undergoing PCI. Eur J Clin Invest 2022; 52:e13826. [PMID: 35723949 PMCID: PMC9786902 DOI: 10.1111/eci.13826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/17/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Predictors of prognosis in patients with coronary chronic total occlusion (CTO) undergoing elective percutaneous coronary intervention (PCI) have remained lacking. Lipidomic profiling enables researchers to associate lipid species with disease progression and may improve the prediction of cardiovascular events. METHODS In the present study, 781 lipids were measured by targeted lipidomic profiling in 350 individuals (50 healthy controls, 50 patients with coronary artery disease and 250 patients with CTO). L1-regularized logistic regression was used to identify lipid species associated with adverse cardiovascular events and create predicting models, which were verified by 10-fold cross-validation (200 repeats). Comparisons were made between a traditional model constructed with clinical characteristics alone and a combined model built with both lipidomic data and traditional factors. RESULTS Twenty-four lipid species were dysregulated exclusively in patients with CTO, most of which belonged to sphingomyelin (SM) and triacylglycerol (TAG). Compared with traditional risk factors, new model combining lipids and traditional factors had significantly improved performance in predicting adverse cardiovascular events in CTO patients after PCI (area under the curve, 0.870 vs. 0.726, p < .05; Akaike information criterion, 129 versus 156; net reclassification improvement, 0.312, p < .001; integrated discrimination improvement, 0.244, p < .001). Nomogram was built based on the incorporated model and proved efficient by Kaplan-Meier method. CONCLUSIONS Lipidomic profiling revealed lipid species which may participate in the formation of CTO and could contribute to the risk stratification in CTO patients undergoing PCI.
Collapse
Affiliation(s)
- You Zhou
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| | - Jinxiang Chen
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| | - Su Li
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| | - Ao Chen
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| | - Chunfeng Dai
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| | - Muyin Liu
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| | - Danbo Lu
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| | - Zhangwei Chen
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| | - Xiangdong Wang
- Shanghai Institute of Clinical BioinformaticsFudan University Center of Clinical Bioinformatics; Shanghai Respiratory Research InstituteShanghaiPeople's Republic of China
| | - Juying Qian
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| | - Junbo Ge
- Department of CardiologyShanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University; National Clinical Research Center for Interventional Medicine; Shanghai Clinical Research Center for Interventional MedicineShanghaiPeople's Republic of China
| |
Collapse
|
41
|
Li HY, Zou Y, Elsheikha HM, Xu Y, Cai L, Xie SC, Zhu XQ, Zheng WB. Lipidomic changes in the liver of beagle dogs associated with Toxocara canis infection. Front Cell Infect Microbiol 2022; 12:890589. [PMID: 36176575 PMCID: PMC9514057 DOI: 10.3389/fcimb.2022.890589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
A global lipidomic analysis using liquid chromatography–tandem mass spectrometry was performed on the liver of beagle dogs infected with Toxocara canis to profile hepatic lipid species at 12 h post-infection (hpi), 24 hpi, and 36 days post-infection (dpi). This analysis identified six categories and 42 subclasses of lipids, including 173, 64, and 116 differentially abundant lipid species at 12 hpi, 24 hpi, and 36 dpi, respectively. Many of the identified lysophospholipids, such as lysophosphatidylglycerol, lysophosphatidylserine, and lysophosphatidylcholine, may contribute to the migration and development of T. canis during the early infection stage. Pathway analysis revealed significant alterations of several immune-inflammatory pathways, such as the B-cell receptor signaling pathway, the NF-kappa B signaling pathway, and the C-type lectin receptor signaling pathway at 12 and 24 hpi. These findings demonstrate the value of lipidomic profiling in revealing the extent of changes in the composition and abundance of hepatic lipidome caused by T. canis infection and their relevance to the pathophysiology of toxocariasis in beagle dogs.
Collapse
Affiliation(s)
- Hao-Yu Li
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Yang Zou
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Hany M. Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Yue Xu
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Lang Cai
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Shi-Chen Xie
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Xing-Quan Zhu
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
- Key Laboratory of Veterinary Public Health of Higher Education of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
- *Correspondence: Xing-Quan Zhu, ; Wen-Bin Zheng,
| | - Wen-Bin Zheng
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
- *Correspondence: Xing-Quan Zhu, ; Wen-Bin Zheng,
| |
Collapse
|
42
|
Gonzalez-Riano C, Santos M, Díaz M, García-Beltran C, Lerin C, Barbas C, Ibáñez L, Sánchez-Infantes D. Birth Weight and Early Postnatal Outcomes: Association with the Cord Blood Lipidome. Nutrients 2022; 14:3760. [PMID: 36145136 PMCID: PMC9505183 DOI: 10.3390/nu14183760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
Being born small or large for gestational age (SGA and LGA, respectively), combined with suboptimal early postnatal outcomes, can entail future metabolic alterations. The exact mechanisms underlying such risks are not fully understood. Lipids are a highly diverse class of molecules that perform multiple structural and metabolic functions. Dysregulation of lipid metabolism underlies the onset and progression of many disorders leading to pathological states. The aim of this pilot study was to investigate the relationships between birth weight, early postnatal outcomes, and cord blood serum lipidomes. We performed a non-targeted lipidomics-based approach to ascertain differences in cord blood lipid species among SGA, LGA, and appropriate-for-GA (AGA) newborns. Moreover, we longitudinally assessed (at birth and at ages of 4 and 12 months) weight and length, body composition (DXA), and clinical parameters. We disclosed distinct cord blood lipidome patterns in SGA, LGA, and AGA newborns; target lipid species distinctly modulated in each SGA, AGA, and LGA individual were associated with parameters related to growth and glucose homeostasis. The distinct lipidome patterns observed in SGA, AGA, and LGA newborns may play a role in adipose tissue remodeling and future metabolic risks. Maternal dietary interventions may potentially provide long-term benefits for the metabolic health of the offspring.
Collapse
Affiliation(s)
- Carolina Gonzalez-Riano
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Marcelo Santos
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Marta Díaz
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina García-Beltran
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carles Lerin
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Lourdes Ibáñez
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - David Sánchez-Infantes
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain
| |
Collapse
|
43
|
Salita T, Rustam YH, Mouradov D, Sieber OM, Reid GE. Reprogrammed Lipid Metabolism and the Lipid-Associated Hallmarks of Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14153714. [PMID: 35954376 PMCID: PMC9367418 DOI: 10.3390/cancers14153714] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Colorectal cancer (CRC) is the third-most diagnosed cancer and the second-leading cause of cancer-related deaths worldwide. Limitations in early and accurate diagnosis of CRC gives rise to poor patient survival. Advancements in analytical techniques have improved our understanding of the cellular and metabolic changes occurring in CRC and potentiate avenues for improved diagnostic and therapeutic strategies. Lipids are metabolites with important biological functions; however, their role in CRC is poorly understood. Here, we provide an in-depth review of the recent literature concerning lipid alterations in CRC and propose eight lipid metabolism-associated hallmarks of CRC. Abstract Lipids have diverse structures, with multifarious regulatory functions in membrane homeostasis and bioenergetic metabolism, in mediating functional protein–lipid and protein–protein interactions, as in cell signalling and proliferation. An increasing body of evidence supports the notion that aberrant lipid metabolism involving remodelling of cellular membrane structure and changes in energy homeostasis and signalling within cancer-associated pathways play a pivotal role in the onset, progression, and maintenance of colorectal cancer (CRC) and their tumorigenic properties. Recent advances in analytical lipidome analysis technologies have enabled the comprehensive identification and structural characterization of lipids and, consequently, our understanding of the role they play in tumour progression. However, despite progress in our understanding of cancer cell metabolism and lipidomics, the key lipid-associated changes in CRC have yet not been explicitly associated with the well-established ‘hallmarks of cancer’ defined by Hanahan and Weinberg. In this review, we summarize recent findings that highlight the role of reprogrammed lipid metabolism in CRC and use this growing body of evidence to propose eight lipid metabolism-associated hallmarks of colorectal cancer, and to emphasize their importance and linkages to the established cancer hallmarks.
Collapse
Affiliation(s)
- Timothy Salita
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia; (T.S.); (Y.H.R.)
- Personalized Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
| | - Yepy H. Rustam
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia; (T.S.); (Y.H.R.)
| | - Dmitri Mouradov
- Personalized Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
| | - Oliver M. Sieber
- Personalized Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
- Correspondence: (O.M.S.); (G.E.R.)
| | - Gavin E. Reid
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia; (T.S.); (Y.H.R.)
- School of Chemistry, University of Melbourne, Melbourne, VIC 3010, Australia
- Bio21 Molecular Science & Biotechnology Institute, University of Melbourne, Parkville, VIC 3010, Australia
- Correspondence: (O.M.S.); (G.E.R.)
| |
Collapse
|
44
|
De Angelis A, Park SH, Opella SJ. Magnetically Aligned Lipid Bilayers with High Cholesterol for Solid-State NMR of Membrane Proteins. Biochemistry 2022; 61:1561-1571. [PMID: 35849647 DOI: 10.1021/acs.biochem.2c00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Phospholipid bicelles are valuable membrane model systems to study membrane proteins by NMR and other physicochemical techniques. The range of bicelle compositions that are compatible with uniaxial alignment of the lipid bilayers in a magnetic field is still limited with regard to the addition of large amounts (>20%) of cholesterol and/or sphingolipids. Here, we demonstrate that n-dodecyl-β-D-melibioside (DDMB), which was recently introduced as a detergent to produce sphingolipid-cholesterol-rich isotropic bicelles for solution NMR studies, can also be used to produce magnetically alignable lipid bilayers with high cholesterol content that are well suited for solid-state NMR of membrane proteins. Remarkably, DDMB enables the preparation of high q bicelles that contain 50% mol cholesterol while retaining their ability to form a stable, well-aligned liquid crystalline bilayer phase in a magnetic field. We show that the intact 46-residue membrane-bound form of Pf1 bacteriophage coat protein and a truncated construct of the membrane protein Vpu from HIV-1 (residues 2-30) in DDMB bicelles are well aligned and undergo fast and uniaxial rotational diffusion about the bilayer normal, similarly to what is observed in other bicelle and macrodisc systems. We also demonstrate a spectroscopic method that measures the increase in the thickness of DMPC bilayers that results from the addition of cholesterol, using the PISA-wheel spectral patterns of trans-membrane helices as a molecular goniometer. For example, we find that the hydrophobic thickness of DMPC bilayers is increased by approximately 2.5 Å in the presence of 35% mol cholesterol.
Collapse
Affiliation(s)
- Anna De Angelis
- Department of Chemistry and Biochemistry, University of California, La Jolla, San Diego, California 92093-0307 United States
| | - Sang Ho Park
- Department of Chemistry and Biochemistry, University of California, La Jolla, San Diego, California 92093-0307 United States
| | - Stanley J Opella
- Department of Chemistry and Biochemistry, University of California, La Jolla, San Diego, California 92093-0307 United States
| |
Collapse
|
45
|
Duan L, Scheidemantle G, Lodge M, Cummings MJ, Pham E, Wang X, Kennedy A, Liu X. Prioritize biologically relevant ions for data-independent acquisition (BRI-DIA) in LC-MS/MS-based lipidomics analysis. Metabolomics 2022; 18:55. [PMID: 35842862 DOI: 10.1007/s11306-022-01913-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/27/2022] [Indexed: 10/17/2022]
Abstract
INTRODUCTION Data-dependent acquisition (DDA) is the most commonly used MS/MS scan method for lipidomics analysis on orbitrap-based instrument. However, MS instrument associated software decide the top N precursors for fragmentation, resulting in stochasticity of precursor selection and compromised consistency and reproducibility. We introduce a novel workflow using biologically relevant lipids to construct inclusion list for data-independent acquisition (DIA), named as BRI-DIA workflow. OBJECTIVES To ensure consistent coverage of biologically relevant lipids in LC-MS/MS-based lipidomics analysis. METHODS Biologically relevant ion list was constructed based on LIPID MAPS and lipidome atlas in MS-DIAL 4. Lipids were extracted from mouse tissues and used to assess different MS/MS scan workflow (DDA, BRI-DIA, and hybrid mode) on LC-Orbitrap Exploris 480 mass spectrometer. RESULTS DDA resulted in more MS/MS events, but the total number of unique lipids identified by three methods (DDA, BRI-DIA, and hybrid MS/MS scan mode) is comparable (580 unique lipids across 44 lipid subclasses in mouse liver). Major cardiolipin molecular species were identified by data generated using BRI-DIA and hybrid methods and allowed calculation of cardiolipin compositions, while identification of the most abundant cardiolipin CL72:8 was missing in data generated using DDA method, leading to wrong calculation of cardiolipin composition. CONCLUSION The method of using inclusion list comprised of biologically relevant lipids in DIA MS/MS scan is as efficient as traditional DDA method in profiling lipids, but offers better consistency of lipid identification, compared to DDA method. This study was performed using Orbitrap Exploris 480, and we will further evaluate this workflow on other platforms, and if verified by future work, this biologically relevant ion fragmentation workflow could be routinely used in many studies to improve MS/MS identification capacities.
Collapse
Affiliation(s)
- Likun Duan
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Grace Scheidemantle
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Mareca Lodge
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Magdalina J Cummings
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27695, USA
| | - Eva Pham
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Xiaoqiu Wang
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27695, USA
| | - Arion Kennedy
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Xiaojing Liu
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
46
|
Olshansky G, Giles C, Salim A, Meikle PJ. Challenges and opportunities for prevention and removal of unwanted variation in lipidomic studies. Prog Lipid Res 2022; 87:101177. [PMID: 35780914 DOI: 10.1016/j.plipres.2022.101177] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/19/2022] [Accepted: 06/26/2022] [Indexed: 10/17/2022]
Abstract
Large 'omics studies are of particular interest to population and clinical research as they allow elucidation of biological pathways that are often out of reach of other methodologies. Typically, these information rich datasets are produced from multiple coordinated profiling studies that may include lipidomics, metabolomics, proteomics or other strategies to generate high dimensional data. In lipidomics, the generation of such data presents a series of unique technological and logistical challenges; to maximize the power (number of samples) and coverage (number of analytes) of the dataset while minimizing the sources of unwanted variation. Technological advances in analytical platforms, as well as computational approaches, have led to improvement of data quality - especially with regard to instrumental variation. In the small scale, it is possible to control systematic bias from beginning to end. However, as the size and complexity of datasets grow, it is inevitable that unwanted variation arises from multiple sources, some potentially unknown and out of the investigators control. Increases in cohort sizes and complexity has led to new challenges in sample collection, handling, storage, and preparation stages. If not considered and dealt with appropriately, this unwanted variation may undermine the quality of the data and reliability of any subsequent analysis. Here we review the various experimental phases where unwanted variation may be introduced and review general strategies and approaches to handle this variation, specifically addressing issues relevant to lipidomics studies.
Collapse
Affiliation(s)
- Gavriel Olshansky
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Corey Giles
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Agus Salim
- Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC 3010, Australia; School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia; Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
47
|
Brial F, Hedjazi L, Sonomura K, Al Hageh C, Zalloua P, Matsuda F, Gauguier D. Genetic Architecture of Untargeted Lipidomics in Cardiometabolic-Disease Patients Combines Strong Polygenic Control and Pleiotropy. Metabolites 2022; 12:metabo12070596. [PMID: 35888720 PMCID: PMC9322850 DOI: 10.3390/metabo12070596] [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: 06/05/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Analysis of the genetic control of small metabolites provides powerful information on the regulation of the endpoints of genome expression. We carried out untargeted liquid chromatography−high-resolution mass spectrometry in 273 individuals characterized for pathophysiological elements of the cardiometabolic syndrome. We quantified 3013 serum lipidomic features, which we used in both genome-wide association studies (GWAS), using a panel of over 2.5 M imputed single-nucleotide polymorphisms (SNPs), and metabolome-wide association studies (MWAS) with phenotypes. Genetic analyses showed that 926 SNPs at 551 genetic loci significantly (q-value < 10−8) regulate the abundance of 74 lipidomic features in the group, with evidence of monogenic control for only 22 of these. In addition to this strong polygenic control of serum lipids, our results underscore instances of pleiotropy, when a single genetic locus controls the abundance of several distinct lipid features. Using the LIPID MAPS database, we assigned putative lipids, predominantly fatty acyls and sterol lipids, to 77% of the lipidome signals mapped to the genome. We identified significant correlations between lipids and clinical and biochemical phenotypes. These results demonstrate the power of untargeted lipidomic profiling for high-density quantitative molecular phenotyping in human-genetic studies and illustrate the complex genetic control of lipid metabolism.
Collapse
Affiliation(s)
- Francois Brial
- Center for Genomic Medicine, Graduate School of Medicine Kyoto University, Kyoto 606-8501, Japan; (F.B.); (F.M.)
- INSERM UMR 1124, Université Paris Cité, 45 rue des Saint-Pères, 75006 Paris, France
| | | | - Kazuhiro Sonomura
- Life Science Research Center, Technology Research Laboratory, Shimadzu Corporation, Kyoto 606-8501, Japan;
| | - Cynthia Al Hageh
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 17666, United Arab Emirates; (C.A.H.); (P.Z.)
| | - Pierre Zalloua
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 17666, United Arab Emirates; (C.A.H.); (P.Z.)
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine Kyoto University, Kyoto 606-8501, Japan; (F.B.); (F.M.)
- McGill University and Genome Quebec Innovation Centre, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada
| | - Dominique Gauguier
- Center for Genomic Medicine, Graduate School of Medicine Kyoto University, Kyoto 606-8501, Japan; (F.B.); (F.M.)
- INSERM UMR 1124, Université Paris Cité, 45 rue des Saint-Pères, 75006 Paris, France
- McGill University and Genome Quebec Innovation Centre, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada
- Correspondence:
| |
Collapse
|
48
|
Vvedenskaya O, Holčapek M, Vogeser M, Ekroos K, Meikle PJ, Bendt AK. Clinical lipidomics – A community-driven roadmap to translate research into clinical applications. J Mass Spectrom Adv Clin Lab 2022; 24:1-4. [PMID: 35199094 PMCID: PMC8844780 DOI: 10.1016/j.jmsacl.2022.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 12/29/2022] Open
Abstract
Overview of current state of mass spectrometry based lipidomics. Highlighting ongoing efforts towards harmonization. Invitation to join international community.
Lipid metabolites, beyond triglycerides and cholesterol, have been shown to have vast potential for applications in clinical applications, with substantial societal and economical value. To successfully evolve from the current research-grade methods to assays suitable for routine clinical applications, a harmonization – if not standardization – of these mass spectrometry-based workflows is necessary. Input on clinical needs and technological capabilities must be obtained from all relevant stakeholders, including wet lab scientists, informaticians and data scientists, manufacturers, and medical professionals. In order to build bridges between this diverse group of professionals, the International Lipidomics Society and its Clinical Lipidomics Interest Group were created. This opinion article is intended to provide an overview of international efforts to tackle the issues of workflow harmonization, and to serve as an open invitation for others to join this growing community.
Collapse
Affiliation(s)
- Olga Vvedenskaya
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Spectroswiss Sarl, Lausanne, Switzerland
| | - Michal Holčapek
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Pardubice, Czech Republic
| | - Michael Vogeser
- Institute for Laboratory Medicine in the Munich University Clinic, Munich, Germany
| | - Kim Ekroos
- Lipidomics Consulting Ltd., Esbo, Finland
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, Melbourne Victoria, Australia
| | - Anne K. Bendt
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore
- Corresponding author.
| |
Collapse
|
49
|
Watts GF, Chan DC. Microplastics, cardiometabolic risk, genetics and Alzheimer's disease. Curr Opin Endocrinol Diabetes Obes 2022; 29:85-86. [PMID: 35197423 DOI: 10.1097/med.0000000000000718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Gerald F Watts
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia
- Lipid Disorders Clinic, Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Dick C Chan
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia
| |
Collapse
|
50
|
Gao J, Gu X, Zhang M, Zu X, Shen F, Hou X, Hao E, Bai G. Ferulic acid targets ACSL1 to ameliorate lipid metabolic disorders in db/db mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|