1
|
Tolle G, Serreli G, Deiana M, Moi L, Zavattari P, Pantaleo A, Manis C, El Faqir MA, Caboni P. Lipidomics of Caco-2 Cells Under Simulated Microgravity Conditions. Int J Mol Sci 2024; 25:12638. [PMID: 39684348 DOI: 10.3390/ijms252312638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Microgravity may profoundly impact the cardiovascular system, skeletal muscle system, and immune system of astronauts. At the cellular level, microgravity may also affect cell proliferation, differentiation, and growth, as well as lipid metabolism. In this work, we investigated lipid changes in Caco-2 cells cultured in a clinostat for 24 h under simulated microgravity conditions (SMC). Complex lipids were measured using a UHPLC-QTOF/MS platform, and the data were subjected to multivariate analysis. Under SMC, levels of ceramides Cer 18:0;O2/16:0, Cer 18:1;O2/16:0, Cer 18:1; O2/22:0, Cer 18:1;O2/24:0, and Cer 18:2;O2/24:0 were found to be upregulated, while sphingomyelins SM 16:1;O2/16:0, SM 16:1;O2/18:1, SM 18:1;O2/24:0, and SM 18:2;O2/24:0 were found to be downregulated. On the other hand, considering that sphingolipids are involved in the process of inflammation, we also treated Caco-2 cells with dextran sodium sulfate (DSS) to induce cell inflammation and lipopolysaccharide (LPS) to induce cell immune responses. As a result, we observed similar lipid dysregulation, indicating that SMC may exert a condition similar to inflammation. Our lipidomics strategy provides new insights into the altered metabolic pathway of ceramides and sphingomyelins of Caco-2 cells under SMC.
Collapse
Affiliation(s)
- Giulia Tolle
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Gabriele Serreli
- Unit of Experimental Pathology, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Monica Deiana
- Unit of Experimental Pathology, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Loredana Moi
- Unit of Biology and Genetics, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Patrizia Zavattari
- Unit of Biology and Genetics, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Antonella Pantaleo
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy
| | - Cristina Manis
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Mohammed Amine El Faqir
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100 Sassari, Italy
| | - Pierluigi Caboni
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Italy
| |
Collapse
|
2
|
Gutierrez-Monreal MA, Wolff CA, Rijos EE, Viggars MR, Douglas CM, Pagala V, Peng J, Hunt LC, Ding H, Huo Z, Demontis F, Esser KA. Targeted Bmal1 restoration in muscle prolongs lifespan with systemic health effects in aging model. JCI Insight 2024; 9:e174007. [PMID: 39352748 PMCID: PMC11601919 DOI: 10.1172/jci.insight.174007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/26/2024] [Indexed: 10/04/2024] Open
Abstract
Disruption of the circadian clock in skeletal muscle worsens local and systemic health, leading to decreased muscle strength, metabolic dysfunction, and aging-like phenotypes. Whole-body knockout mice that lack Bmal1, a key component of the molecular clock, display premature aging. Here, by using adeno-associated viruses, we rescued Bmal1 expression specifically in the skeletal muscle fibers of Bmal1-KO mice and found that this engaged the circadian clock and clock output gene expression, contributing to extended lifespan. Time course phenotypic analyses found that muscle strength, mobility, and glucose tolerance were improved with no effects on muscle mass or fiber size or type. A multiomics approach at 2 ages further determined that restored muscle Bmal1 improved glucose handling pathways while concomitantly reducing lipid and protein metabolic pathways. The improved glucose tolerance and metabolic flexibility resulted in the systemic reduction of inflammatory signatures across peripheral tissues, including liver, lung, and white adipose fat. Together, these findings highlight the critical role of muscle Bmal1 and downstream target genes for skeletal muscle homeostasis with considerable implications for systemic health.
Collapse
Affiliation(s)
| | | | - Eduardo E. Rijos
- Department of Physiology and Aging
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, USA
| | | | | | - Vishwajeeth Pagala
- Department of Structural Biology, Center for Proteomics and Metabolomics, and
| | - Junmin Peng
- Department of Structural Biology, Center for Proteomics and Metabolomics, and
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Liam C. Hunt
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Biology, Rhodes College, Memphis, Tennessee, USA
| | - Haocheng Ding
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Karyn A. Esser
- Department of Physiology and Aging
- Myology Institute, and
| |
Collapse
|
3
|
Estrada-Pérez AR, García-Vázquez JB, Mendoza-Figueroa HL, Rosales-Hernández MC, Fernández-Pomares C, Correa-Basurto J. Untargeted LC-MS/MS Metabolomics Study of HO-AAVPA and VPA on Breast Cancer Cell Lines. Int J Mol Sci 2023; 24:14543. [PMID: 37833990 PMCID: PMC10572250 DOI: 10.3390/ijms241914543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer (BC) is one of the biggest health problems worldwide, characterized by intricate metabolic and biochemical complexities stemming from pronounced variations across dysregulated molecular pathways. If BC is not diagnosed early, complications may lead to death. Thus, the pursuit of novel therapeutic avenues persists, notably focusing on epigenetic pathways such as histone deacetylases (HDACs). The compound N-(2-hydroxyphenyl)-2-propylpentanamide (HO-AAVPA), a derivative of valproic acid (VPA), has emerged as a promising candidate warranting pre-clinical investigation. HO-AAVPA is an HDAC inhibitor with antiproliferative effects on BC, but its molecular mechanism has yet to be deciphered. Furthermore, in the present study, we determined the metabolomic effects of HO-AAVPA and VPA on cells of luminal breast cancer (MCF-7) and triple-negative breast cancer (MDA-MB-231) subtypes. The LC-MS untargeted metabolomic study allowed for the simultaneous measurement of multiple metabolites and pathways, identifying that both compounds affect glycerophospholipid and sphingolipid metabolism in the MCF-7 and MDA-MB-231 cell lines, suggesting that other biological targets were different from HDACs. In addition, there are different dysregulate metabolites, possibly due to the physicochemical differences between HO-AAVPA and VPA.
Collapse
Affiliation(s)
- Alan Rubén Estrada-Pérez
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México 11340, Mexico
| | - Juan Benjamín García-Vázquez
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México 11340, Mexico
| | - Humberto L. Mendoza-Figueroa
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México 11340, Mexico
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México 11340, Mexico
| | - Cynthia Fernández-Pomares
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México 11340, Mexico
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México 11340, Mexico
| |
Collapse
|
4
|
Islam KU, Anwar S, Patel AA, Mirdad MT, Mirdad MT, Azmi MI, Ahmad T, Fatima Z, Iqbal J. Global Lipidome Profiling Revealed Multifaceted Role of Lipid Species in Hepatitis C Virus Replication, Assembly, and Host Antiviral Response. Viruses 2023; 15:v15020464. [PMID: 36851679 PMCID: PMC9965260 DOI: 10.3390/v15020464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Hepatitis C virus (HCV) is a major human pathogen that requires a better understanding of its interaction with host cells. There is a close association of HCV life cycle with host lipid metabolism. Lipid droplets (LDs) have been found to be crucial organelles that support HCV replication and virion assembly. In addition to their role in replication, LDs also have protein-mediated antiviral properties that are activated during HCV infection. Studies have shown that HCV replicates well in cholesterol and sphingolipid-rich membranes, but the ways in which HCV alters host cell lipid dynamics are not yet known. In this study, we performed a kinetic study to check the enrichment of LDs at different time points of HCV infection. Based on the LD enrichment results, we selected early and later time points of HCV infection for global lipidomic study. Early infection represents the window period for HCV sensing and host immune response while later infection represents the establishment of viral RNA replication, virion assembly, and egress. We identified the dynamic profile of lipid species at early and later time points of HCV infection by global lipidomic study using mass spectrometry. At early HCV infection, phosphatidylinositol phospholipids (PIPs), lysophosphatidic acid (LPA), triacyl glycerols (TAG), phosphatidylcholine (PC), and trihexosylceramides (Hex3Cer) were observed to be enriched. Similarly, free fatty acids (FFA), phosphatidylethanolamine (PE), N-acylphosphatidylethanolamines (NAPE), and tri acylglycerols were enriched at later time points of HCV infection. Lipids enriched at early time of infection may have role in HCV sensing, viral attachment, and immune response as LPA and PIPs are important for immune response and viral attachment, respectively. Moreover, lipid species observed at later infection may contribute to HCV replication and virion assembly as PE, FFA, and triacylglycerols are known for the similar function. In conclusion, we identified lipid species that exhibited dynamic profile across early and later time points of HCV infection compared to mock cells, which could be therapeutically relevant in the design of more specific and effective anti-viral therapies.
Collapse
Affiliation(s)
- Khursheed Ul Islam
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Saleem Anwar
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ayyub A. Patel
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| | | | | | - Md Iqbal Azmi
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Tanveer Ahmad
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Zeeshan Fatima
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Gurugram 122413, India
- Correspondence: (Z.F.); (J.I.)
| | - Jawed Iqbal
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
- Correspondence: (Z.F.); (J.I.)
| |
Collapse
|
5
|
El Sobky SA, Aboud NK, El Assaly NM, Fawzy IO, El-Ekiaby N, Abdelaziz AI. Regulation of lipid droplet (LD) formation in hepatocytes via regulation of SREBP1c by non-coding RNAs. Front Med (Lausanne) 2022; 9:903856. [PMID: 36203751 PMCID: PMC9530594 DOI: 10.3389/fmed.2022.903856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/25/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Increased de novo lipogenesis (DNL) is one of the key factors contributing to fat accumulation and non-alcoholic fatty liver disease (NAFLD). Among the critical transcription factors (TFs) regulating DNL is mTOR and its downstream lipogenic TF, SREBP1c. In recent years, it has been established that non-coding RNAs (ncRNAs) play role in both biological processes and disease pathogenesis. Our group has previously characterized microRNAs that can target and regulate the expression of both mTOR and SREBP1c. Accordingly, this study aimed to broaden our understanding of the role of ncRNAs in regulating the mTOR/SREBP1c axis to elucidate the role of the non-coding transcriptome in DNL and lipid droplet (LD) formation. Hence, short ncRNA, miR-615-5p, and long non-coding RNA (lncRNA), H19, were chosen as they were previously proven to target mTOR by our group and in the published literature, respectively. Methodology Huh-7 cells were treated with 800 μM oleic acid (OA) to promote LD formation. Transfection of miR-615-5p mimics or H19 over-expression vectors was performed, followed by the measurement of their downstream targets, mTOR and SREBP, on the mRNA level by quantitative real-time PCR (qRT-PCR), and on the protein level by Western blot. To determine the functional impact of miR-615-5p and H19 on LD formation and triglyceride (TG) accumulation, post-transfection LDs were stained, imaged, and characterized, and TGs were extracted and quantified. Results miR-615-5p was able to reduce mTOR and SREBP1c significantly on both the mRNA and protein levels compared to control cells, while H19 caused a reduction of both targets on the protein level only. Both miR-615-5p and H19 were able to significantly reduce the LD count and total area, as well as TG levels compared to control cells. Conclusion To conclude, this study shows, for the first time, the impact of miR-615-5p and H19 on the mTOR/SREBP1c axis, and thus, their functional impact on LDs and TG accumulation. These findings might pave the way for using ncRNAs as potential therapeutic targets in the management of fatty liver.
Collapse
Affiliation(s)
| | | | - Nihal M. El Assaly
- Clinical Chemistry Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Injie O. Fawzy
- School of Medicine, Newgiza University (NGU), Giza, Egypt
| | - Nada El-Ekiaby
- School of Medicine, Newgiza University (NGU), Giza, Egypt
| | - Ahmed I. Abdelaziz
- School of Medicine, Newgiza University (NGU), Giza, Egypt
- *Correspondence: Ahmed I. Abdelaziz
| |
Collapse
|
6
|
Ma C, Hoffmann FW, Marciel MP, Page KE, Williams-Aduja MA, Akana ENL, Gojanovich GS, Gerschenson M, Urschitz J, Moisyadi S, Khadka VS, Rozovsky S, Deng Y, Horgen FD, Hoffmann PR. Upregulated ethanolamine phospholipid synthesis via selenoprotein I is required for effective metabolic reprogramming during T cell activation. Mol Metab 2021; 47:101170. [PMID: 33484950 PMCID: PMC7881273 DOI: 10.1016/j.molmet.2021.101170] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/02/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE T cell activation triggers metabolic reprogramming to meet increased demands for energy and metabolites required for cellular proliferation. Ethanolamine phospholipid synthesis has emerged as a regulator of metabolic shifts in stem cells and cancer cells, which led us to investigate its potential role during T cell activation. METHODS As selenoprotein I (SELENOI) is an enzyme participating in two metabolic pathways for the synthesis of phosphatidylethanolamine (PE) and plasmenyl PE, we generated SELENOI-deficient mouse models to determine loss-of-function effects on metabolic reprogramming during T cell activation. Ex vivo and in vivo assays were carried out along with metabolomic, transcriptomic, and protein analyses to determine the role of SELENOI and the ethanolamine phospholipids synthesized by this enzyme in cell signaling and metabolic pathways that promote T cell activation and proliferation. RESULTS SELENOI knockout (KO) in mouse T cells led to reduced de novo synthesis of PE and plasmenyl PE during activation and impaired proliferation. SELENOI KO did not affect T cell receptor signaling, but reduced activation of the metabolic sensor AMPK. AMPK was inhibited by high [ATP], consistent with results showing SELENOI KO causing ATP accumulation, along with disrupted metabolic pathways and reduced glycosylphosphatidylinositol (GPI) anchor synthesis/attachment CONCLUSIONS: T cell activation upregulates SELENOI-dependent PE and plasmenyl PE synthesis as a key component of metabolic reprogramming and proliferation.
Collapse
Affiliation(s)
- Chi Ma
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - FuKun W Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Michael P Marciel
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Kathleen E Page
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA
| | | | - Ellis N L Akana
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA
| | - Greg S Gojanovich
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Johann Urschitz
- Department of Anatomy, Physiology and Biochemistry, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Stefan Moisyadi
- Department of Anatomy, Physiology and Biochemistry, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Vedbar S Khadka
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Sharon Rozovsky
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI 96744, USA
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| |
Collapse
|
7
|
Domenick TM, Gill EL, Vedam-Mai V, Yost RA. Mass Spectrometry-Based Cellular Metabolomics: Current Approaches, Applications, and Future Directions. Anal Chem 2020; 93:546-566. [PMID: 33146525 DOI: 10.1021/acs.analchem.0c04363] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Taylor M Domenick
- Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Emily L Gill
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104-4283, United States.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-4283, United States
| | - Vinata Vedam-Mai
- Department of Neurology, University of Florida, Gainesville, Florida 32610, United States
| | - Richard A Yost
- Department of Chemistry, University of Florida, Gainesville, Florida 32611-7200, United States
| |
Collapse
|
8
|
Taylor NJ, Gaynanova I, Eschrich SA, Welsh EA, Garrett TJ, Beecher C, Sharma R, Koomen JM, Smalley KSM, Messina JL, Kanetsky PA. Metabolomics of primary cutaneous melanoma and matched adjacent extratumoral microenvironment. PLoS One 2020; 15:e0240849. [PMID: 33108391 PMCID: PMC7591037 DOI: 10.1371/journal.pone.0240849] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Melanoma causes the vast majority of deaths attributable to skin cancer, largely due to its propensity for metastasis. To date, few studies have examined molecular changes between primary cutaneous melanoma and adjacent putatively normal skin. To broaden temporal inferences related to initiation of disease, we performed a metabolomics investigation of primary melanoma and matched extratumoral microenvironment (EM) tissues; and, to make inferences about progressive disease, we also compared unmatched metastatic melanoma tissues to EM tissues. METHODS Ultra-high performance liquid chromatography-mass spectrometry-based metabolic profiling was performed on frozen human tissues. RESULTS We observed 824 metabolites as differentially abundant among 33 matched tissue samples, and 1,118 metabolites as differentially abundant between metastatic melanoma (n = 46) and EM (n = 34) after false discovery rate (FDR) adjustment (p<0.01). No significant differences in metabolite abundances were noted comparing primary and metastatic melanoma tissues. CONCLUSIONS Overall, pathway-based results significantly distinguished melanoma tissues from EM in the metabolism of: ascorbate and aldarate, propanoate, tryptophan, histidine, and pyrimidine. Within pathways, the majority of individual metabolite abundances observed in comparisons of primary melanoma vs. EM and metastatic melanoma vs. EM were directionally consistent. This observed concordance suggests most identified compounds are implicated in the initiation or maintenance of melanoma.
Collapse
Affiliation(s)
- Nicholas J. Taylor
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, Texas, United States of America
| | - Irina Gaynanova
- Department of Statistics, Texas A&M University, College Station, Texas, United States of America
| | - Steven A. Eschrich
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Eric A. Welsh
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Timothy J. Garrett
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Chris Beecher
- IROA Technologies, Chapel Hill, North Carolina, United States of America
| | - Ritin Sharma
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - John M. Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Keiran S. M. Smalley
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Jane L. Messina
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| | - Peter A. Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, United States of America
| |
Collapse
|
9
|
Rai SK, Bril F, Hatch HM, Xu Y, Shelton L, Kalavalapalli S, Click A, Lee D, Beecher C, Kirby A, Kong K, Trevino J, Jha A, Jatav S, Kriti K, Luthra S, Garrett TJ, Guingab-Cagmat J, Plant D, Bose P, Cusi K, Hromas RA, Tischler AS, Powers JF, Gupta P, Bibb J, Beuschlein F, Robledo M, Calsina B, Timmers H, Taieb D, Kroiss M, Richter S, Langton K, Eisenhofer G, Bergeron R, Pacak K, Tevosian SG, Ghayee HK. Targeting pheochromocytoma/paraganglioma with polyamine inhibitors. Metabolism 2020; 110:154297. [PMID: 32562798 PMCID: PMC7482423 DOI: 10.1016/j.metabol.2020.154297] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Pheochromocytomas (PCCs) and paragangliomas (PGLs) are neuroendocrine tumors that are mostly benign. Metastatic disease does occur in about 10% of cases of PCC and up to 25% of PGL, and for these patients no effective therapies are available. Patients with mutations in the succinate dehydrogenase subunit B (SDHB) gene tend to have metastatic disease. We hypothesized that a down-regulation in the active succinate dehydrogenase B subunit should result in notable changes in cellular metabolic profile and could present a vulnerability point for successful pharmacological targeting. METHODS Metabolomic analysis was performed on human hPheo1 cells and shRNA SDHB knockdown hPheo1 (hPheo1 SDHB KD) cells. Additional analysis of 115 human fresh frozen samples was conducted. In vitro studies using N1,N11-diethylnorspermine (DENSPM) and N1,N12- diethylspermine (DESPM) treatments were carried out. DENSPM efficacy was assessed in human cell line derived mouse xenografts. RESULTS Components of the polyamine pathway were elevated in hPheo1 SDHB KD cells compared to wild-type cells. A similar observation was noted in SDHx PCC/PGLs tissues compared to their non-mutated counterparts. Specifically, spermidine, and spermine were significantly elevated in SDHx-mutated PCC/PGLs, with a similar trend in hPheo1 SDHB KD cells. Polyamine pathway inhibitors DENSPM and DESPM effectively inhibited growth of hPheo1 cells in vitro as well in mouse xenografts. CONCLUSIONS This study demonstrates overactive polyamine pathway in PCC/PGL with SDHB mutations. Treatment with polyamine pathway inhibitors significantly inhibited hPheo1 cell growth and led to growth suppression in xenograft mice treated with DENSPM. These studies strongly implicate the polyamine pathway in PCC/PGL pathophysiology and provide new foundation for exploring the role for polyamine analogue inhibitors in treating metastatic PCC/PGL. PRéCIS: Cell line metabolomics on hPheo1 cells and PCC/PGL tumor tissue indicate that the polyamine pathway is activated. Polyamine inhibitors in vitro and in vivo demonstrate that polyamine inhibitors are promising for malignant PCC/PGL treatment. However, further research is warranted.
Collapse
Affiliation(s)
- Sudhir Kumar Rai
- Department of Medicine, Division of Endocrinology, University of Florida, Gainesville, FL, USA
| | - Fernando Bril
- Department of Medicine, Division of Endocrinology, University of Florida and Malcom Randall VA Medical Center, Gainesville, FL, USA
| | - Heather M Hatch
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Yiling Xu
- Department of Medicine, Division of Endocrinology, University of Florida, Gainesville, FL, USA
| | - Laura Shelton
- Scientific Project Development, Human Metabolome Technologies, Boston, MA, USA
| | - Srilaxmi Kalavalapalli
- Department of Medicine, Division of Endocrinology, University of Florida, Gainesville, FL, USA
| | - Arielle Click
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Austin Kirby
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Kimi Kong
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jose Trevino
- Department of Surgery, University of Florida, Gainesville, FL, USA
| | | | | | | | | | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Joy Guingab-Cagmat
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Daniel Plant
- Department of Physiological Sciences, University of Florida, Malcom Randall VA Medical Center, Gainesville, FL, USA
| | - Prodip Bose
- Department of Physiological Sciences, University of Florida, Malcom Randall VA Medical Center, Gainesville, FL, USA
| | - Kenneth Cusi
- Department of Medicine, Division of Endocrinology, University of Florida and Malcom Randall VA Medical Center, Gainesville, FL, USA
| | - Robert A Hromas
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Arthur S Tischler
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, MA, USA
| | - James F Powers
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, MA, USA
| | - Priyanka Gupta
- Department of Surgery, University of Alabama, Birmingham, AL, USA
| | - James Bibb
- Department of Surgery, University of Alabama, Birmingham, AL, USA
| | - Felix Beuschlein
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, UniversitätsSpital Zurich, Zurich, Switzerland
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Henri Timmers
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - David Taieb
- Department of Nuclear Medicine, La Timone University Hospital, European Center for Research in Medical Imaging, Aix Marseille Université, Marseille, France
| | - Matthias Kroiss
- Department of Internal Medicine, Division of Endocrinology and Diabetology, University Hospital Würzburg, University of Würzburg, Würzburg, Germany
| | - Susan Richter
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Katharina Langton
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Division of Clinical Neurochemistry, Institute of Clinical Chemistry and Laboratory Medicine, and Department of Medicine, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Raymond Bergeron
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, USA
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Sergei G Tevosian
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA.
| | - Hans K Ghayee
- Department of Medicine, Division of Endocrinology, University of Florida and Malcom Randall VA Medical Center, Gainesville, FL, USA.
| |
Collapse
|
10
|
Choi SC, Brown J, Gong M, Ge Y, Zadeh M, Li W, Croker BP, Michailidis G, Garrett TJ, Mohamadzadeh M, Morel L. Gut microbiota dysbiosis and altered tryptophan catabolism contribute to autoimmunity in lupus-susceptible mice. Sci Transl Med 2020; 12:eaax2220. [PMID: 32641487 PMCID: PMC7739186 DOI: 10.1126/scitranslmed.aax2220] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 10/04/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022]
Abstract
The autoimmune disease systemic lupus erythematosus (SLE) is characterized by the production of pathogenic autoantibodies. It has been postulated that gut microbial dysbiosis may be one of the mechanisms involved in SLE pathogenesis. Here, we demonstrate that the dysbiotic gut microbiota of triple congenic (TC) lupus-prone mice (B6.Sle1.Sle2.Sle3) stimulated the production of autoantibodies and activated immune cells when transferred into germfree congenic C57BL/6 (B6) mice. Fecal transfer to B6 mice induced autoimmune phenotypes only when the TC donor mice exhibited autoimmunity. Autoimmune pathogenesis was mitigated by horizontal transfer of the gut microbiota between co-housed lupus-prone TC mice and control congenic B6 mice. Metabolomic screening identified an altered distribution of tryptophan metabolites in the feces of TC mice including an increase in kynurenine, which was alleviated after antibiotic treatment. Low dietary tryptophan prevented autoimmune pathology in TC mice, whereas high dietary tryptophan exacerbated disease. Reducing dietary tryptophan altered gut microbial taxa in both lupus-prone TC mice and control B6 mice. Consequently, fecal transfer from TC mice fed a high tryptophan diet, but not a low tryptophan diet, induced autoimmune phenotypes in germfree B6 mice. The interplay of gut microbial dysbiosis, tryptophan metabolism and host genetic susceptibility in lupus-prone mice suggest that aberrant tryptophan metabolism may contribute to autoimmune activation in this disease.
Collapse
Affiliation(s)
- Seung-Chul Choi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Josephine Brown
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Minghao Gong
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32610, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Yong Ge
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32610, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mojgan Zadeh
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32610, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Wei Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Byron P Croker
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - George Michailidis
- Department of Statistics and the Informatics Institute, University of Florida, Gainesville, FL 32610, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mansour Mohamadzadeh
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32610, USA.
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
11
|
Teclemariam ET, Pergande MR, Cologna SM. Considerations for mass spectrometry-based multi-omic analysis of clinical samples. Expert Rev Proteomics 2020; 17:99-107. [PMID: 31996049 DOI: 10.1080/14789450.2020.1724540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: The role of mass spectrometry in biomolecule analysis has become paramount over the last several decades ranging in the analysis across model systems and human specimens. Accordingly, the presence of mass spectrometers in clinical laboratories has also expanded alongside the number of researchers investigating the protein, lipid, and metabolite composition of an array of biospecimens. With this increase in the number of omic investigations, it is important to consider the entire experimental strategy from sample collection and storage, data collection and analysis.Areas covered: In this short review, we outline considerations for working with clinical (e.g. human) specimens including blood, urine, and cerebrospinal fluid, with emphasis on sample handling, profiling composition, targeted measurements and relevance to disease. Discussions of integrated genomic or transcriptomic datasets are not included. A brief commentary is also provided regarding new technologies with clinical relevance.Expert opinion: The role of mass spectrometry to investigate clinically related specimens is on the rise and the ability to integrate multiple omics datasets from mass spectrometry measurements will be crucial to further understanding human health and disease.
Collapse
Affiliation(s)
- Esei T Teclemariam
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Melissa R Pergande
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, USA.,Laboratory of Integrated Neuroscience, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
12
|
Palaskas NJ, Garcia JD, Shirazi R, Shin DS, Puig-Saus C, Braas D, Ribas A, Graeber TG. Global alteration of T-lymphocyte metabolism by PD-L1 checkpoint involves a block of de novo nucleoside phosphate synthesis. Cell Discov 2019; 5:62. [PMID: 31798961 PMCID: PMC6877514 DOI: 10.1038/s41421-019-0130-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/18/2019] [Indexed: 12/29/2022] Open
Abstract
Metabolic obstacles of the tumor microenvironment remain a challenge to T-cell-mediated cancer immunotherapies. To better understand the interplay of immune checkpoint signaling and immune metabolism, this study developed and used an optimized metabolite extraction protocol for non-adherent primary human T-cells, to broadly profile in vitro metabolic changes effected by PD-1 signaling by mass spectrometry-based metabolomics and isotopomer analysis. Inhibitory signaling reduced aerobic glycolysis and glutaminolysis. A general scarcity across the panel of metabolites measured supported widespread metabolic regulation by PD-1. Glucose carbon fate analysis supported tricarboxylic acid cycle reliance on pyruvate carboxylation, catabolic-state fluxes into acetyl-CoA and succinyl-CoA, and a block in de novo nucleoside phosphate synthesis that was accompanied by reduced mTORC1 signaling. Nonetheless, exogenous administration of nucleosides was not sufficient to ameliorate proliferation of T-cells in the context of multiple metabolic insufficiencies due to PD-L1 treatment. Carbon fate analysis did not support the use of primarily glucose-derived carbons to fuel fatty acid beta oxidation, in contrast to reports on T-memory cells. These findings add to our understanding of metabolic dysregulation by PD-1 signaling and inform the effort to rationally develop metabolic interventions coupled with immune-checkpoint blockade for increased treatment efficacy.
Collapse
Affiliation(s)
- Nicolaos Jay Palaskas
- 1Division of Hematology and Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA.,2Department of Molecular, Cellular, and Integrative Physiology, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Jacob David Garcia
- 3Department of Molecular, Cellular, and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Roksana Shirazi
- 4Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Daniel Sanghoon Shin
- 1Division of Hematology and Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA.,2Department of Molecular, Cellular, and Integrative Physiology, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Cristina Puig-Saus
- 1Division of Hematology and Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Daniel Braas
- 2Department of Molecular, Cellular, and Integrative Physiology, University of California Los Angeles, Los Angeles, CA 90095 USA.,5Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, CA 90095 USA.,6Metabolomics Center, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Antoni Ribas
- 1Division of Hematology and Oncology, Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA.,2Department of Molecular, Cellular, and Integrative Physiology, University of California Los Angeles, Los Angeles, CA 90095 USA.,7Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095 USA.,8Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095 USA.,9Division of Surgical Oncology, Department of Surgery, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Thomas Glen Graeber
- 2Department of Molecular, Cellular, and Integrative Physiology, University of California Los Angeles, Los Angeles, CA 90095 USA.,5Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, CA 90095 USA.,6Metabolomics Center, University of California Los Angeles, Los Angeles, CA 90095 USA.,7Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095 USA.,8Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
13
|
Lu X, Lodi A, Konopleva M, Tiziani S. Three-Dimensional Leukemia Co-Culture System for In Vitro High-Content Metabolomics Screening. SLAS DISCOVERY 2019; 24:817-828. [PMID: 31345091 DOI: 10.1177/2472555219860446] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Metabolomics is increasingly applied to investigate different individuals and time-dependent responses to environmental stimuli. Rapid data acquisition and improved detection limits of direct infusion mass spectrometry (DIMS) are paving the way for applications of metabolomics in preclinical screening, opening new opportunities in drug discovery and personalized medicine. Three-dimensional (3D) cell culture systems, which mimic the in vivo cell microenvironment, are well recognized as tissue and organ substitutes. Here, we investigated cell viability and induction of reactive oxygen species (ROS) in stromal cells cultured in various 3D systems as well as the standard monolayer culture to evaluate which system provides the most favorable growing conditions. The selected 3D system was then tested for use in 3D co-culture of leukemia and stromal cells for DIMS-based high-throughput/high-content metabolic drug screens. The NanobioMatrix-poly(ε-caprolactone) (NBM-PCL) scaffold resulted in the lowest ROS production, supported rapid cell proliferation, and was suitable for the 96- and 384-well plate formats. Doxorubicin treatment in leukemia co-cultured with stromal cells induced some unique metabolic responses that drastically differed from those observed in leukemia cells alone. The DIMS results also showed that the drug-induced metabolic modulations in both normal and cancer cells were weakened by co-culturing even at high treatment doses, thereby demonstrating the value of the 3D co-culture high-content metabolic drug screen. In conclusion, we optimized a high sample throughput method for 3D co-culture with a DIMS-based high-content metabolic drug screen and drug development.
Collapse
Affiliation(s)
- Xiyuan Lu
- 1 Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, USA.,2 Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Alessia Lodi
- 1 Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, USA.,2 Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Marina Konopleva
- 3 Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stefano Tiziani
- 1 Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, USA.,2 Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
14
|
Abstract
Lipids exert key structural, metabolic, and signaling functions in cells. Lipid diversity found in cells and tissues is regulated principally by metabolic enzymes whose activity is modulated posttranslationally to shape head group and fatty acyl composition of membrane lipids. Methodologies capable of monitoring in vivo changes in the lipidome are needed to assign substrate specificity of metabolic enzymes, which represents a key step toward understanding structure-function of lipids in living systems. The resulting lipid annotations also serve as important biomarkers for understanding mode of action for pharmacological agents targeting metabolic enzymes in cells and animal models. In this chapter, we describe a general metabolomics workflow to complement (chemo)proteomic efforts to modulate lipid pathways for basic science and translational applications.
Collapse
Affiliation(s)
- Timothy B Ware
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Myungsun Shin
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Ku-Lung Hsu
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States; Department of Pharmacology, University of Virginia, Charlottesville, VA, United States; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, United States; University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, United States.
| |
Collapse
|
15
|
Mathon C, Bovard D, Dutertre Q, Sendyk S, Bentley M, Hoeng J, Knorr A. Impact of sample preparation upon intracellular metabolite measurements in 3D cell culture systems. Metabolomics 2019; 15:92. [PMID: 31190156 PMCID: PMC6561993 DOI: 10.1007/s11306-019-1551-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/29/2019] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Interest in cell culture metabolomics has increased greatly in recent years because of its many potential applications and advantages (e.g., in toxicology). The first critical step for exploring the cellular metabolome is sample preparation. For metabolomics studies, an ideal sample preparation would extract a maximum number of metabolites and would enable reproducible, accurate analysis of a large number of samples and replicates. In addition, it would provide consistent results across several studies over a relatively long time frame. OBJECTIVES This study was conducted to evaluate the impact of sample preparation strategies on monitoring intracellular metabolite responses, highlighting the potential critical step(s) in order to finally improve the quality of metabolomics studies. METHODS The sample preparation strategies were evaluated by calculating the sample preparation effect, matrix factor, and process efficiency (PE) for 16 tobacco exposition-related metabolites, including nicotine, nicotine-derived nitrosamine ketone, their major metabolites, and glutathione, using isotopically-labelled internal standards. Samples were analyzed by liquid chromatography (LC) coupled to high-resolution mass spectrometry (HRMS). RESULTS A sample drying step increased losses or variability for some selected metabolites. By avoiding evaporation, good sample preparation recovery was obtained for these compounds. For some metabolites, the cell or culture type impacted PE and matrix factor. CONCLUSION In our sample preparation protocol, the drying-reconstitution step was identified as the main cause of metabolite losses or increased data variability during metabolomics analysis by LC-HRMS. Furthermore, PE was affected by the type of matrix. Isotopologue internal standards fully compensate losses or enhancements.
Collapse
Affiliation(s)
- Caroline Mathon
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland.
- Unit of Toxicology, CURML, Lausanne-Geneva, Switzerland.
| | - David Bovard
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Quentin Dutertre
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Sandra Sendyk
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Mark Bentley
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Arno Knorr
- PMI R&D, Philip Morris Products S.A., Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| |
Collapse
|
16
|
Castellanos A, Ramirez CE, Michalkova V, Nouzova M, Noriega FG, Francisco FL. Three Dimensional Secondary Ion Mass Spectrometry Imaging (3D-SIMS) of Aedes aegypti ovarian follicles. JOURNAL OF ANALYTICAL ATOMIC SPECTROMETRY 2019; 34:874-883. [PMID: 31680712 PMCID: PMC6824543 DOI: 10.1039/c8ja00425k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The mobilization of nutrient reserves into the ovaries of Aedes aegypti mosquitoes after sugar-feeding plays a vital role in the female's reproductive maturation. In the present work, three-dimensional secondary ion mass spectrometry imaging (3D-SIMS) was used to generate ultrahigh spatial resolution (~1 μm) chemical maps and study the composition and spatial distribution of lipids at the single ovarian follicle level (~100 μm in size). 3D-Mass Spectrometry Imaging (3D-MSI) allowed the identification of cellular types in the follicle (oocyte, nurse and follicular cells) using endogenous markers, and revealed that most of the triacyglycerides (TGs) were compartmentalized in the oocyte region. By comparing follicles from water-fed and sugar-fed females (n=2), 3D-MSI-Time of Flight-SIMS showed that TGs were more abundant in ovarian follicles of sugar-fed females; despite relative sample reproducibility per feeding condition, more biological replicates will better support the trends observed. While the current 3D-MSI-TOF-SIMS does not permit MS/MS analysis of the lipid species, complementary LC-MS/MS analysis of the ovarian follicles aided tentative lipid assignments of the SIMS data. The combination of these MS approaches is giving us a first glimpse of the distribution of functionally relevant ovarian lipid molecules at the cellular level. These new tools can be used to investigate the roles of different lipids on follicle fitness and overall mosquito reproductive output.
Collapse
Affiliation(s)
- Anthony Castellanos
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, United States
| | - Cesar E. Ramirez
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, United States
| | - Veronika Michalkova
- Department of Biological Sciences, Florida International University, Miami, Florida, 33199, United States
| | - Marcela Nouzova
- Department of Biological Sciences, Florida International University, Miami, Florida, 33199, United States
- Institute of Parasitology, Biology Centre CAS, Ceske, Budejovice, Czech Republic; and
| | - Fernando G. Noriega
- Department of Biological Sciences, Florida International University, Miami, Florida, 33199, United States
- Biomolecular Sciences Institute, Florida International University, Miami, Florida, 33199, United States
| | - Fernández-Lima Francisco
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, United States
- Biomolecular Sciences Institute, Florida International University, Miami, Florida, 33199, United States
- Corresponding author: Francisco A. Fernández-Lima, Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th St AHC4-233, Miami, FL 33199, USA;
| |
Collapse
|
17
|
Stephenson DJ, MacKnight HP, Hoeferlin LA, Park M, Allegood J, Cardona CL, Chalfant CE. A rapid and adaptable lipidomics method for quantitative UPLC-mass spectrometric analysis of phosphatidylethanolamine and phosphatidylcholine in vitro, and in cells. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2019; 11:1765-1776. [PMID: 31788037 PMCID: PMC6884326 DOI: 10.1039/c9ay00052f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Phosphatidylethanolamine (PE) and phosphatidylcholine (PC) are highly prevalent phospholipids in mammalian membranes. There are currently no methods for detection of minute levels of these phospholipids or simultaneously with products of the utilization of these phospholipid substrates by phospholipase A2 (PLA2) enzymes. To examine the substrate utilization of PE and PC by PLA2, we developed a method to accurately detect and measure specific forms of PE and PC as low as 50 femtomoles. Validation of this method consisted of an enzymatic assay to monitor docosahexaenoic acid and arachidonic acid release from the hydrolysis of PE and PC by group IV phospholipase A2 (cPLA2α) coupled to the generation of lyso-PE (LPE) and lyso-PC (LPC). In addition, the PE and PC profiles of RAW 264.7 macrophages were monitored with zymosan/lipopolysaccharide-treatment. Finally, genetic validation for the specificity of the method consisted of the downregulation of two biosynthetic enzymes responsible for the production of PE and PC, choline kinase A (CHKA) and ethanolamine kinase 1 (ETNK1). This new UPLC ESI-MS/MS method provides accurate and highly sensitive detection of PE and PC species containing AA and DHA allowing for the specific examination of the substrate utilization of these phospholipids by PLA2 in vitro and in cells.
Collapse
Affiliation(s)
- Daniel J. Stephenson
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University (VCU), Richmond VA, 23298
| | - H. Patrick MacKnight
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University (VCU), Richmond VA, 23298
| | - L. Alexis Hoeferlin
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University (VCU), Richmond VA, 23298
- VCU Massey Cancer Center, Cancer Cell Signaling Program, Virginia Commonwealth University, Richmond VA, 23298
| | - Margaret Park
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620
- The Moffitt Cancer Center, Tampa, FL 33620
| | - Jeremy Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University (VCU), Richmond VA, 23298
| | - Christopher L. Cardona
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620
| | - Charles E. Chalfant
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612
| |
Collapse
|
18
|
Koomen DC, Guingab-Cagmat JD, Oliveira PS, Fang B, Liu M, Welsh EA, Meads MB, Nguyen T, Meke L, Eschrich SA, Shain KH, Garrett TJ, Koomen JM. Proteometabolomics of Melphalan Resistance in Multiple Myeloma. Methods Mol Biol 2019; 1996:273-296. [PMID: 31127562 DOI: 10.1007/978-1-4939-9488-5_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Drug resistance remains a critical problem for the treatment of multiple myeloma (MM), which can serve as a specific example for a highly prevalent unmet medical need across almost all cancer types. In MM, the therapeutic arsenal has expanded and diversified, yet we still lack in-depth molecular understanding of drug mechanisms of action and cellular pathways to therapeutic escape. For those reasons, preclinical models of drug resistance are developed and characterized using different approaches to gain insights into tumor biology and elucidate mechanisms of drug resistance. For MM, numerous drugs are used for treatment, including conventional chemotherapies (e.g., melphalan or L-phenylalanine nitrogen mustard), proteasome inhibitors (e.g., Bortezomib), and immunomodulators (e.g., Lenalidomide). These agents have diverse effects on the myeloma cells, and several mechanisms of drug resistance have been previously described. The disparity of these mechanisms and the complexity of these biological processes lead to the formation of complicated hypotheses that require omics approaches for efficient and effective analysis of model systems that can then be interpreted for patient benefit. Here, we describe the combination of metabolomics and proteomics to assess melphalan resistance in MM by examining three specific areas: drug metabolism, modulation of endogenous metabolites to assist in therapeutic escape, and changes in protein activity gauged by ATP probe uptake.
Collapse
Affiliation(s)
| | | | | | - Bin Fang
- Moffitt Cancer Center, Tampa, FL, USA
| | - Min Liu
- Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lu D, Xue L, Feng C, Jin Y, Wu C, Xie C, Gonzalez FJ, Wang G, Zhou Z. A systemic workflow for profiling metabolome and lipidome in tissue. J Chromatogr A 2018; 1589:105-115. [PMID: 30638710 DOI: 10.1016/j.chroma.2018.12.061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/13/2018] [Accepted: 12/27/2018] [Indexed: 12/16/2022]
Abstract
Simple metabolome and lipidome sample preparation procedures involving two successive extractions using small pieces of tissue, and a subsequent metabolite identification (MetID) strategy were developed. The sample preparation can significantly circumvent incomplete analysis due to insufficient amounts of tissue as a result of splitting into several aliquots for multiple measurements, with advantages over the similar previously reported methods in metabolite coverage, extraction efficiency, method robustness and friendly experimental operation. A MetID strategy, based on the integration of MS information mining (including adduct ions, in-source CID, MS information from both ESI (+) and ESI (-), characteristic fragmentation ions (CFIs), constant neutral losses (CNLs) and multimers) and in silico MS simulation, was demonstrated. A large number of adduct ions (83 features), in-source CID (123 features), ESI (+/-) ionization (20 features), CFIs& CNLs (more than 120 features) and multimers (17 features) were mined by manually or in silico recognition/filtering, which provide the most suspicious structures for subsequent in silico MS simulation. The unknown features presented the same score distribution as the known (83 features) features with scores ≥25% (geomean score: 52%) and with satisfactory match for the main ions of interest. The MS/MS noise and fragment ions of coeluted quasi-molecular ions of interest are the main reason for the low score in the simulation. Manual check/evaluation is always suggested for the simulation with a score less than 50%. This strategy presents satisfactory performance with 2.5 times more metabolites structurally characterized compared with that of the traditional method based on accurate-mass-based MS and MS/MS library matching. This strategy would be useful for potentially identifying metabolites without available MS/MS information in the library.
Collapse
Affiliation(s)
- Dasheng Lu
- Shanghai Municipal Center for Disease Control and Prevention, 1380 Zhongshan West Road, Shanghai, 200336, China; School of Public Health/MOE Key Lab for Public Health, Fudan University, Shanghai, 200032, China; Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Liming Xue
- Shanghai Municipal Center for Disease Control and Prevention, 1380 Zhongshan West Road, Shanghai, 200336, China
| | - Chao Feng
- Shanghai Municipal Center for Disease Control and Prevention, 1380 Zhongshan West Road, Shanghai, 200336, China
| | - Yu'e Jin
- Shanghai Municipal Center for Disease Control and Prevention, 1380 Zhongshan West Road, Shanghai, 200336, China
| | - Chunhua Wu
- School of Public Health/MOE Key Lab for Public Health, Fudan University, Shanghai, 200032, China
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Guoquan Wang
- Shanghai Municipal Center for Disease Control and Prevention, 1380 Zhongshan West Road, Shanghai, 200336, China.
| | - Zhijun Zhou
- School of Public Health/MOE Key Lab for Public Health, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
20
|
Ulmer CZ, Jones CM, Yost RA, Garrett TJ, Bowden JA. Optimization of Folch, Bligh-Dyer, and Matyash sample-to-extraction solvent ratios for human plasma-based lipidomics studies. Anal Chim Acta 2018; 1037:351-357. [PMID: 30292311 PMCID: PMC6261534 DOI: 10.1016/j.aca.2018.08.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 07/17/2018] [Accepted: 08/02/2018] [Indexed: 11/22/2022]
Abstract
In order to profile the lipidome for untargeted lipidomics applications, analysis by ultra-high performance liquid chromatography - high resolution mass spectrometry (UHPLC-HRMS) typically requires the extraction of lipid content from sample matrices using matrix-specific conditions. The Folch, Bligh-Dyer, and Matyash extraction methods, while promising approaches, were originally tailored to specific matrices (brain tissue, fish muscle, and E. coli, respectively). Each of these methods have specific solvent ratios that must be adhered to achieve optimal extraction. Thus, the sample-to-solvent ratios for these methods should be optimized for the sample matrix of interest prior to employment. This study evaluated the appropriate sample-to-extraction solvent ratios for human plasma-based lipidomics studies. An advantage of employing biphasic lipid extractions is the ability to investigate both the aqueous and organic layers for increased analyte coverage in untargeted studies. Therefore, this work also evaluated the multi-omic capability of each lipid extraction method for plasma in an effort to provide a workflow capable of increasing analyte coverage in a single extraction, thus providing a more complete understanding of complex biological systems. In plasma, a decrease in sample-to-solvent ratios from 1:4, 1:10, 1:20, to 1:100 (v/v) resulted in a gradual increase in the peak area of a diverse range of metabolite (aqueous layer) and lipid (organic layer) species for each extraction method up to the 1:20(v/v) sample-to-solvent ratio. The Bligh-Dyer and Folch methods yielded the highest peak areas at every plasma sample-to-solvent ratios for both metabolite and lipid species. Depending on the lipid class of interest, the Folch or Bligh-Dyer method is best suited for analysis of human plasma at a 1:20 (v/v) sample to total solvent ratio.
Collapse
Affiliation(s)
- Candice Z Ulmer
- National Institute of Standards and Technology, Chemical Sciences Division, Marine Biochemical Sciences Group, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC, 29412, USA.
| | - Christina M Jones
- National Institute of Standards and Technology, Chemical Sciences Division, Organic Chemical Measurement Science Group, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| | - Richard A Yost
- University of Florida, Department of Chemistry, 125 Buckman Drive, Gainesville, FL, 32611, USA; University of Florida, Department of Pathology, Immunology and Laboratory Medicine, Gainesville, FL, 32610, USA
| | - Timothy J Garrett
- University of Florida, Department of Pathology, Immunology and Laboratory Medicine, Gainesville, FL, 32610, USA
| | - John A Bowden
- National Institute of Standards and Technology, Chemical Sciences Division, Marine Biochemical Sciences Group, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC, 29412, USA; University of Florida, Center for Environmental and Human Toxicology, Department of Physiological Sciences, College of Veterinary Medicine, Gainesville, FL, 32611, USA
| |
Collapse
|
21
|
Tríbulo P, Balzano-Nogueira L, Conesa A, Siqueira LG, Hansen PJ. Changes in the uterine metabolome of the cow during the first 7 days after estrus. Mol Reprod Dev 2018; 86:75-87. [PMID: 30383328 PMCID: PMC6322963 DOI: 10.1002/mrd.23082] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 10/26/2018] [Indexed: 12/22/2022]
Abstract
The uterine microenvironment during the first 7 days after ovulation accommodates and facilitates sperm transit to the oviduct and constitutes the sole source of nutrients required for the development of preimplantation embryos. Knowledge of the composition of uterine fluid is largely incomplete. Using untargeted mass spectrometry, we characterized the uterine metabolome during the first 7 days of the estrous cycle. Bovine uteri were collected on Days 0 (N = 4), 3 (
N = 4), 5 (
N = 3), and 7 (
N = 4) relative to ovulation and flushed with Dulbecco’s phosphate‐buffered saline. A total of 1,993 molecular features were detected of which 184 peaks with putative identification represent 147 unique metabolites, including amino acids, benzoic acids, lipid molecules, carbohydrates, purines, pyrimidines, vitamins, and other intermediate and secondary metabolites. Results revealed changes in the uterine metabolome as the cow transitions from ovulation to Day 7 of the estrous cycle. The majority of metabolites that changed with day reached maximum intensity on either Day 5 or 7 relative to ovulation. Moreover, several metabolites found in the uterine fluid have signaling capabilities and some have been shown to affect preimplantation embryonic development. In conclusion, the metabolome of the bovine uterus changes during early stages of the estrous cycle and is likely to participate in the regulation of preimplantation embryonic development. Data reported here will serve as the basis for future studies aiming to evaluate maternal regulation of preimplantation embryonic development and optimal conditions for the culture of embryos.
Collapse
Affiliation(s)
- Paula Tríbulo
- Department of Animal Sciences, University of Florida, Gainesville, Florida.,D.H. Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, Florida
| | | | - Ana Conesa
- Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida.,Genetics Institute, University of Florida, Gainesville, Florida
| | - Luiz G Siqueira
- Department of Animal Sciences, University of Florida, Gainesville, Florida.,Embrapa Gado de Leite, Juiz de Fora, Brazil
| | - Peter J Hansen
- Department of Animal Sciences, University of Florida, Gainesville, Florida.,D.H. Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, Florida.,Genetics Institute, University of Florida, Gainesville, Florida
| |
Collapse
|
22
|
Zong L, Pi Z, Liu S, Liu Z, Song F. Metabolomics analysis of multidrug-resistant breast cancer cellsin vitrousing methyl-tert-butyl ether method. RSC Adv 2018; 8:15831-15841. [PMID: 35539507 PMCID: PMC9080077 DOI: 10.1039/c7ra12952a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/21/2018] [Indexed: 11/21/2022] Open
Abstract
MTBE-based cellular lipidomics to investigate the mechanisms of multidrug resistance of breast cancer.
Collapse
Affiliation(s)
- Li Zong
- National Center of Mass Spectrometry in Changchun
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
| | - Zifeng Pi
- National Center of Mass Spectrometry in Changchun
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
| | - Shu Liu
- National Center of Mass Spectrometry in Changchun
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
| | - Zhiqiang Liu
- National Center of Mass Spectrometry in Changchun
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
| | - Fengrui Song
- National Center of Mass Spectrometry in Changchun
- Jilin Province Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
| |
Collapse
|
23
|
Katragkou A, Williams M, Sternberg S, Pantazatos D, Roilides E, Walsh TJ. Micafungin alters the amino acid, nucleic acid and central carbon metabolism of Candida albicans at subinhibitory concentrations: novel insights into mechanisms of action. J Antimicrob Chemother 2017; 72:712-716. [PMID: 28039272 PMCID: PMC5890779 DOI: 10.1093/jac/dkw478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/16/2016] [Accepted: 10/03/2016] [Indexed: 12/24/2022] Open
Abstract
Background Echinocandins are an important class of antifungal agents in the treatment of invasive candidiasis. However, little is known about the metabolomic effects of echinocandins on Candida . We therefore performed LC-high-resolution MS (LC-HRMS)-based metabolomics profiling of the response of Candida albicans cells to increasing concentrations of micafungin to determine the metabolic response of Candida to micafungin subinhibitory injury. Methods Isolates of C. albicans were cultured on nitrocellulose filters to mid-logarithmic phase of growth and micafungin (0-0.25 mg/L) was added. At mid-logarithmic phase, replicates were metabolically quenched. Intracellular metabolites were analysed by LC-HRMS. Changes in pool sizes of individual metabolites were analysed by Student's t -test adjusted for multiple hypothesis testing by Benjamini-Hochberg correction. Metabolites were ascribed by the Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic pathways database. Results Among 3446 detected metabolites, 204 were identified by comparison against pure standard or comparison against a library of mass-retention-time pairs. Fifty had significantly altered abundances in response to increasing micafungin concentrations. Pool sizes of amino acids, nucleic acids and polyamine metabolism were significantly increased at subinhibitory concentrations, while exposure to inhibitory concentrations resulted in a precipitous decrease consistent with fungicidal activity. Conclusions Micafungin induces a re-routing of metabolic pathways inhibiting protein synthesis and cell replication. These results shed light on new mechanisms of action of echinocandins.
Collapse
Affiliation(s)
- Aspasia Katragkou
- Division of Infectious Diseases, Department of Medicine, Transplantation-Oncology Infectious Diseases Program, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Michael Williams
- University of Florida, Southeast Center for Integrated Metabolomics, Gainesville, FL, USA
| | - Sandi Sternberg
- University of Florida, Southeast Center for Integrated Metabolomics, Gainesville, FL, USA
| | - Dennis Pantazatos
- Division of Infectious Diseases, Department of Medicine, Transplantation-Oncology Infectious Diseases Program, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Emmanuel Roilides
- Infectious Disease Unit, 3rd Department of Pediatrics, Faculty of Medicine, Aristotle University School of Health Sciences, Hippokration Hospital, Thessaloniki, Greece
| | - Thomas J. Walsh
- Division of Infectious Diseases, Department of Medicine, Transplantation-Oncology Infectious Diseases Program, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Departments of Pediatrics, Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
24
|
Ulmer CZ, Koelmel JP, Ragland JM, Garrett TJ, Bowden JA. LipidPioneer : A Comprehensive User-Generated Exact Mass Template for Lipidomics. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2017; 28:562-565. [PMID: 28074328 PMCID: PMC5557379 DOI: 10.1007/s13361-016-1579-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 05/21/2023]
Abstract
Lipidomics, the comprehensive measurement of lipid species in a biological system, has promising potential in biomarker discovery and disease etiology elucidation. Advances in chromatographic separation, mass spectrometric techniques, and novel substrate applications continue to expand the number of lipid species observed. The total number and type of lipid species detected in a given sample are generally indicative of the sample matrix examined (e.g., serum, plasma, cells, bacteria, tissue, etc.). Current exact mass lipid libraries are static and represent the most commonly analyzed matrices. It is common practice for users to manually curate their own lists of lipid species and adduct masses; however, this process is time-consuming. LipidPioneer, an interactive template, can be used to generate exact masses and molecular formulas of lipid species that may be encountered in the mass spectrometric analysis of lipid profiles. Over 60 lipid classes are present in the LipidPioneer template and include several unique lipid species, such as ether-linked lipids and lipid oxidation products. In the template, users can add any fatty acyl constituents without limitation in the number of carbons or degrees of unsaturation. LipidPioneer accepts naming using the lipid class level (sum composition) and the LIPID MAPS notation for fatty acyl structure level. In addition to lipid identification, user-generated lipid m/z values can be used to develop inclusion lists for targeted fragmentation experiments. Resulting lipid names and m/z values can be imported into software such as MZmine or Compound Discoverer to automate exact mass searching and isotopic pattern matching across experimental data. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Candice Z Ulmer
- National Institute of Standards and Technology, Hollings Marine Laboratory, 331 Ft. Johnson Road, Charleston, SC, 29412, USA
| | - Jeremy P Koelmel
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL, 32611, USA
| | - Jared M Ragland
- National Institute of Standards and Technology, Hollings Marine Laboratory, 331 Ft. Johnson Road, Charleston, SC, 29412, USA
| | - Timothy J Garrett
- Department of Pathology, University of Florida, 1395 Center Dr, Gainesville, FL, 32610, USA
| | - John A Bowden
- National Institute of Standards and Technology, Hollings Marine Laboratory, 331 Ft. Johnson Road, Charleston, SC, 29412, USA.
| |
Collapse
|
25
|
A Robust Lipidomics Workflow for Mammalian Cells, Plasma, and Tissue Using Liquid-Chromatography High-Resolution Tandem Mass Spectrometry. Methods Mol Biol 2017; 1609:91-106. [PMID: 28660577 DOI: 10.1007/978-1-4939-6996-8_10] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lipids have been analyzed in applications including drug discovery, disease etiology elucidation, and natural products. The chemical and structural diversity of lipids requires a tailored lipidomics workflow for each sample type. Therefore, every protocol in the lipidomics workflow, especially those involving sample preparation, should be optimized to avoid the introduction of bias. The coupling of ultra-high-performance liquid chromatography (UHPLC) with high-resolution mass spectrometry (HRMS) allows for the separation and identification of lipids based on class and fatty acid acyl chain. This work provides a comprehensive untargeted lipidomics workflow that was optimized for various sample types (mammalian cells, plasma, and tissue) to balance extensive lipid coverage and specificity with high sample throughput. For identification purposes, both data-dependent and data-independent tandem mass spectrometric approaches were incorporated, providing more extensive lipid coverage. Popular open-source feature detection, data processing, and identification software are also outlined.
Collapse
|
26
|
Ortiz-Urquiza A, Fan Y, Garrett T, Keyhani NO. Growth substrates and caleosin-mediated functions affect conidial virulence in the insect pathogenic fungus Beauveria bassiana. Microbiology (Reading) 2016; 162:1913-1921. [DOI: 10.1099/mic.0.000375] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Almudena Ortiz-Urquiza
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Yanhua Fan
- Biotechnology Research Center, Southwest University, Beibei, Chongqing, PR China
| | - Timothy Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Nemat O. Keyhani
- Genetic Engineering Research Center, Chongqing University, School of Life Sciences, Chongqing 40045, PR China
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|