1
|
Tressler CM, Ayyappan V, Nakuchima S, Yang E, Sonkar K, Tan Z, Glunde K. A multimodal pipeline using NMR spectroscopy and MALDI-TOF mass spectrometry imaging from the same tissue sample. NMR IN BIOMEDICINE 2023; 36:e4770. [PMID: 35538020 PMCID: PMC9867920 DOI: 10.1002/nbm.4770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 06/14/2023]
Abstract
NMR spectroscopy and matrix assisted laser desorption ionization mass spectrometry imaging (MALDI MSI) are both commonly used to detect large numbers of metabolites and lipids in metabolomic and lipidomic studies. We have demonstrated a new workflow, highlighting the benefits of both techniques to obtain metabolomic and lipidomic data, which has realized for the first time the combination of these two complementary and powerful technologies. NMR spectroscopy is frequently used to obtain quantitative metabolite information from cells and tissues. Lipid detection is also possible with NMR spectroscopy, with changes being visible across entire classes of molecules. Meanwhile, MALDI MSI provides relative measures of metabolite and lipid concentrations, mapping spatial information of many specific metabolite and lipid molecules across cells or tissues. We have used these two complementary techniques in combination to obtain metabolomic and lipidomic measurements from triple-negative human breast cancer cells and tumor xenograft models. We have emphasized critical experimental procedures that ensured the success of achieving NMR spectroscopy and MALDI MSI in a combined workflow from the same sample. Our data show that several phospholipid metabolite species were differentially distributed in viable and necrotic regions of breast tumor xenografts. This study emphasizes the power of combined NMR spectroscopy-MALDI imaging to advance metabolomic and lipidomic studies.
Collapse
Affiliation(s)
- Caitlin M. Tressler
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vinay Ayyappan
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sofia Nakuchima
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ethan Yang
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kanchan Sonkar
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zheqiong Tan
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristine Glunde
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Joseph N, Kolok AS. Assessment of Pediatric Cancer and Its Relationship to Environmental Contaminants: An Ecological Study in Idaho. GEOHEALTH 2022; 6:e2021GH000548. [PMID: 35310467 PMCID: PMC8917512 DOI: 10.1029/2021gh000548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 05/31/2023]
Abstract
The primary aim of this study was to determine the degree to which a multivariable principal component model based on several potentially carcinogenic metals and pesticides could explain the county-level pediatric cancer rates across Idaho. We contend that human exposure to environmental contaminants is one of the reasons for increased pediatric cancer incidence in the United States. Although several studies have been conducted to determine the relationship between environmental contaminants and carcinogenesis among children, research gaps exist in developing a meaningful association between them. For this study, pediatric cancer data was provided by the Cancer Data Registry of Idaho, concentrations of metals and metalloids in groundwater were collected from the Idaho Department of Water Resources, and pesticide use data were collected from the United States Geological Survey. Most environmental variables were significantly intercorrelated at an adjusted P-value <0.01 (97 out of 153 comparisons). Hence, a principal component analysis was employed to summarize those variables to a smaller number of components. An environmental burden index (EBI) was constructed using these principal components, which categorized the environmental burden profiles of counties into low, medium, and high. EBI was significantly associated with pediatric cancer incidence (P-value <0.05). The rate ratio of high EBI profile to low EBI profile for pediatric cancer incidence was estimated as 1.196, with lower and upper confidence intervals of 1.061 and 1.348, respectively. A model was also developed in the study using EBI to estimate the county-level pediatric cancer incidence in Idaho (Nash-Sutcliffe Efficiency = 0.97).
Collapse
Affiliation(s)
- Naveen Joseph
- Idaho Water Resources Research InstituteUniversity of IdahoMoscowIDUSA
| | - Alan S. Kolok
- Idaho Water Resources Research InstituteUniversity of IdahoMoscowIDUSA
| |
Collapse
|
3
|
Leong TKM, Lo WS, Lee WEZ, Tan B, Lee XZ, Lee LWJN, Lee JYJ, Suresh N, Loo LH, Szu E, Yeong J. Leveraging advances in immunopathology and artificial intelligence to analyze in vitro tumor models in composition and space. Adv Drug Deliv Rev 2021; 177:113959. [PMID: 34481035 DOI: 10.1016/j.addr.2021.113959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Cancer is the leading cause of death worldwide. Unfortunately, efforts to understand this disease are confounded by the complex, heterogenous tumor microenvironment (TME). Better understanding of the TME could lead to novel diagnostic, prognostic, and therapeutic discoveries. One way to achieve this involves in vitro tumor models that recapitulate the in vivo TME composition and spatial arrangement. Here, we review the potential of harnessing in vitro tumor models and artificial intelligence to delineate the TME. This includes (i) identification of novel features, (ii) investigation of higher-order relationships, and (iii) analysis and interpretation of multiomics data in a (iv) holistic, objective, reproducible, and efficient manner, which surpasses previous methods of TME analysis. We also discuss limitations of this approach, namely inadequate datasets, indeterminate biological correlations, ethical concerns, and logistical constraints; finally, we speculate on future avenues of research that could overcome these limitations, ultimately translating to improved clinical outcomes.
Collapse
|
4
|
Kumar P, Kumar V. Role of NMR Metabolomics and MR Imaging in Colon Cancer. COLON CANCER DIAGNOSIS AND THERAPY 2021:43-66. [DOI: 10.1007/978-3-030-63369-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Cell metabolic profiling of colorectal cancer via 1H NMR. Clin Chim Acta 2020; 510:291-297. [DOI: 10.1016/j.cca.2020.07.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 02/08/2023]
|
6
|
Bispo D, Fabris V, Lamb CA, Lanari C, Helguero LA, Gil AM. Hormone-Independent Mouse Mammary Adenocarcinomas with Different Metastatic Potential Exhibit Different Metabolic Signatures. Biomolecules 2020; 10:E1242. [PMID: 32867141 PMCID: PMC7563858 DOI: 10.3390/biom10091242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/13/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022] Open
Abstract
The metabolic characteristics of metastatic and non-metastatic breast carcinomas remain poorly studied. In this work, untargeted Nuclear Magnetic Resonance (NMR) metabolomics was used to compare two medroxyprogesterone acetate (MPA)-induced mammary carcinomas lines with different metastatic abilities. Different metabolic signatures distinguished the non-metastatic (59-2-HI) and the metastatic (C7-2-HI) lines, with glucose, amino acid metabolism, nucleotide metabolism and lipid metabolism as the major affected pathways. Non-metastatic tumours appeared to be characterised by: (a) reduced glycolysis and tricarboxylic acid cycle (TCA) activities, possibly resulting in slower NADH biosynthesis and reduced mitochondrial transport chain activity and ATP synthesis; (b) glutamate accumulation possibly related to reduced glutathione activity and reduced mTORC1 activity; and (c) a clear shift to lower phosphoscholine/glycerophosphocholine ratios and sphingomyelin levels. Within each tumour line, metabolic profiles also differed significantly between tumours (i.e., mice). Metastatic tumours exhibited marked inter-tumour changes in polar compounds, some suggesting different glycolytic capacities. Such tumours also showed larger intra-tumour variations in metabolites involved in nucleotide and cholesterol/fatty acid metabolism, in tandem with less changes in TCA and phospholipid metabolism, compared to non-metastatic tumours. This study shows the valuable contribution of untargeted NMR metabolomics to characterise tumour metabolism, thus opening enticing opportunities to find metabolic markers related to metastatic ability in endocrine breast cancer.
Collapse
Affiliation(s)
- Daniela Bispo
- Department of Chemistry and CICECO—Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal;
| | - Victoria Fabris
- IByME—Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina; (V.F.); (C.A.L.); (C.L.)
| | - Caroline A. Lamb
- IByME—Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina; (V.F.); (C.A.L.); (C.L.)
| | - Claudia Lanari
- IByME—Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina; (V.F.); (C.A.L.); (C.L.)
| | - Luisa A. Helguero
- iBIMED—Institute of Biomedicine, Department of Medical Sciences, Universidade de Aveiro, Agra do Crasto, 3810-193 Aveiro, Portugal;
| | - Ana M. Gil
- Department of Chemistry and CICECO—Aveiro Institute of Materials (CICECO/UA), University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal;
| |
Collapse
|
7
|
Zeinali-Rafsanjani B, Jalli R, Saeedi-Moghadam M, Pishdad P. Magnetic resonance spectroscopy and its application in colorectal cancer diagnosis and screening: A narrative review. J Med Imaging Radiat Sci 2020; 51:654-661. [PMID: 32718849 DOI: 10.1016/j.jmir.2020.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/28/2020] [Accepted: 07/10/2020] [Indexed: 12/01/2022]
Abstract
There are several slightly invasive methods to detect colorectal carcinoma (CRC) including colonoscopy and sigmoidoscopy; but there is no noninvasive, accurate screening test. It is recommended to initiate screening at the age of 50 for non-familial CRC. Laboratory tests are routinely suggested if internal observation and imaging are recommended for further evaluation. Spectroscopic-based imaging, such as magnetic resonance spectroscopy (MRS) is an interesting and promising tool with the potential to be an alternative to some minimally invasive procedures, such as biopsy. Accordingly, MRS might be a suitable substitution for invasive methods, such as colonoscopy. This article aimed to review the studies that have evaluated the MRS technique as a screening tool in CRC.
Collapse
Affiliation(s)
- Banafsheh Zeinali-Rafsanjani
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Nuclear Medicine and Molecular Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Jalli
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahdi Saeedi-Moghadam
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Parisa Pishdad
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Garza DR, Taddese R, Wirbel J, Zeller G, Boleij A, Huynen MA, Dutilh BE. Metabolic models predict bacterial passengers in colorectal cancer. Cancer Metab 2020; 8:3. [PMID: 32055399 PMCID: PMC7008539 DOI: 10.1186/s40170-020-0208-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a complex multifactorial disease. Increasing evidence suggests that the microbiome is involved in different stages of CRC initiation and progression. Beyond specific pro-oncogenic mechanisms found in pathogens, metagenomic studies indicate the existence of a microbiome signature, where particular bacterial taxa are enriched in the metagenomes of CRC patients. Here, we investigate to what extent the abundance of bacterial taxa in CRC metagenomes can be explained by the growth advantage resulting from the presence of specific CRC metabolites in the tumor microenvironment. METHODS We composed lists of metabolites and bacteria that are enriched on CRC samples by reviewing metabolomics experimental literature and integrating data from metagenomic case-control studies. We computationally evaluated the growth effect of CRC enriched metabolites on over 1500 genome-based metabolic models of human microbiome bacteria. We integrated the metabolomics data and the mechanistic models by using scores that quantify the response of bacterial biomass production to CRC-enriched metabolites and used these scores to rank bacteria as potential CRC passengers. RESULTS We found that metabolic networks of bacteria that are significantly enriched in CRC metagenomic samples either depend on metabolites that are more abundant in CRC samples or specifically benefit from these metabolites for biomass production. This suggests that metabolic alterations in the cancer environment are a major component shaping the CRC microbiome. CONCLUSION Here, we show with in sillico models that supplementing the intestinal environment with CRC metabolites specifically predicts the outgrowth of CRC-associated bacteria. We thus mechanistically explain why a range of CRC passenger bacteria are associated with CRC, enhancing our understanding of this disease. Our methods are applicable to other microbial communities, since it allows the systematic investigation of how shifts in the microbiome can be explained from changes in the metabolome.
Collapse
Affiliation(s)
- Daniel R. Garza
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Centre, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Rahwa Taddese
- Department of Pathology, Radboud University Medical Center, Postbus 9101, 6500 Nijmegen, HB Netherlands
| | - Jakob Wirbel
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany
| | - Georg Zeller
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117 Heidelberg, Germany
| | - Annemarie Boleij
- Department of Pathology, Radboud University Medical Center, Postbus 9101, 6500 Nijmegen, HB Netherlands
| | - Martijn A. Huynen
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Centre, Postbus 9101, 6500 HB Nijmegen, The Netherlands
| | - Bas E. Dutilh
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Centre, Postbus 9101, 6500 HB Nijmegen, The Netherlands
- Theoretical Biology and Bioinformatics, Sience4Life, Utrecht University, Hugo R. Kruytgebouw, Room Z-509, Padualaan 8, Utrecht, The Netherlands
| |
Collapse
|
9
|
Mass spectrometry-based metabolomics approach to reveal differential compounds in pufferfish soups: Flavor, nutrition, and safety. Food Chem 2019; 301:125261. [DOI: 10.1016/j.foodchem.2019.125261] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/05/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
|
10
|
Maraming P, Klaynongsruang S, Boonsiri P, Maijaroen S, Daduang S, Chung JG, Daduang J. Antitumor activity of RT2 peptide derived from crocodile leukocyte peptide on human colon cancer xenografts in nude mice. ENVIRONMENTAL TOXICOLOGY 2018; 33:972-977. [PMID: 30019842 DOI: 10.1002/tox.22584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 06/08/2023]
Abstract
RT2, derived from the leukocyte peptide of Crocodylus siamensis, can kill human cervical cancer cells via apoptosis induction, but no evidence has shown in vivo. In this study, we investigated the antitumor effect of RT2 on human colon cancer xenografts in nude mice. Twenty-four mice were injected subcutaneously with human colon cancer HCT 116 cells. Eleven days after cancer cell implantation, the mice were treated with intratumoral injections of phosphate buffered saline (PBS) or RT2 (0.01, 0.1, and 1 mg/mouse) once every 2 days for a total of 5 times. The effect of a 10-day intratumoral injection of RT2 on body weight, biochemical, and hematological parameters in BALB/c mice showed no significant difference between the groups. Tumor volume showed a significant decrease only in the treatment group with RT2 (1 mg/mouse) at day 6 (P < .05), day 8 (P < .01), and day 10 (P < .01) after the first treatment. The protein expression levels of cleaved poly (ADP-ribose) polymerase (PARP), apoptosis-inducing factor (AIF), and the p53 tumor suppressor protein (p53) in xenograft tumors increased after treatment with RT2 (1 mg/mouse) compared to those in the PBS-injected group. Moreover, RT2 increased the expression of Endo G and Bcl-2 family proteins. Therefore, the peptide RT2 can inhibit tumor growth via the induction of apoptosis in an in vivo xenograft model.
Collapse
Affiliation(s)
- Pornsuda Maraming
- Biomedical Sciences Program, Graduate School, Khon Kaen University, Khon Kaen, Thailand
| | - Sompong Klaynongsruang
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Patcharee Boonsiri
- Faculty of Medicine, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Surachai Maijaroen
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Sakda Daduang
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Science, Khon Kaen University, Khon Kaen, Thailand
| | - Jing-Gung Chung
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Jureerut Daduang
- Center for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
11
|
Gala de Pablo J, Armistead FJ, Peyman SA, Bonthron D, Lones M, Smith S, Evans SD. Biochemical fingerprint of colorectal cancer cell lines using label-free live single-cell Raman spectroscopy. JOURNAL OF RAMAN SPECTROSCOPY : JRS 2018; 49:1323-1332. [PMID: 31031517 PMCID: PMC6473482 DOI: 10.1002/jrs.5389] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/22/2018] [Accepted: 03/25/2018] [Indexed: 05/27/2023]
Abstract
Label-free live single-cell Raman spectroscopy was used to obtain a chemical fingerprint of colorectal cancer cells including the classification of the SW480 and SW620 cell line model system, derived from primary and secondary tumour cells from the same patient. High-quality Raman spectra were acquired from hundreds of live cells, showing high reproducibility between experiments. Principal component analysis with linear discriminant analysis yielded the best cell classification, with an accuracy of 98.7 ± 0.3% (standard error) when compared with discrimination trees or support vector machines. SW480 showed higher content of the disordered secondary protein structure Amide III band, whereas SW620 showed larger α-helix and β-sheet band content. The SW620 cell line also displayed higher nucleic acid, phosphates, saccharide, and CH2 content. HL60, HT29, HCT116, SW620, and SW480 live single-cell spectra were classified using principal component analysis or linear discriminant analysis with an accuracy of 92.4 ± 0.4% (standard error), showing differences mainly in the β-sheet content, the cytochrome C bands, the CH-stretching regions, the lactate contributions, and the DNA content. The lipids contributions above 2,900 cm-1 and the lactate contributions at 1,785 cm-1 appeared to be dependent on the colorectal adenocarcinoma stage, the advanced stage cell lines showing lower lipid, and higher lactate content. The results demonstrate that these cell lines can be distinguished with high confidence, suggesting that Raman spectroscopy on live cells can distinguish between different disease stages, and could play an important role clinically as a diagnostic tool for cell phenotyping.
Collapse
Affiliation(s)
- Julia Gala de Pablo
- Molecular and Nanoscale Physics Group, School of Physics and AstronomyUniversity of LeedsLeedsUK
| | - Fern J. Armistead
- Molecular and Nanoscale Physics Group, School of Physics and AstronomyUniversity of LeedsLeedsUK
| | - Sally A. Peyman
- Molecular and Nanoscale Physics Group, School of Physics and AstronomyUniversity of LeedsLeedsUK
- Welcome Trust Brenner Building, St James's University Hospital, Faculty of Medicine and HealthUniversity of LeedsLeedsUK
| | - David Bonthron
- Welcome Trust Brenner Building, St James's University Hospital, Faculty of Medicine and HealthUniversity of LeedsLeedsUK
| | - Michael Lones
- School of Mathematical and Computer SciencesHeriot‐Watt UniversityEdinburghUK
| | | | - Stephen D. Evans
- Molecular and Nanoscale Physics Group, School of Physics and AstronomyUniversity of LeedsLeedsUK
- Welcome Trust Brenner Building, St James's University Hospital, Faculty of Medicine and HealthUniversity of LeedsLeedsUK
| |
Collapse
|
12
|
Santana-Filho APD, Jacomasso T, Riter DS, Barison A, Iacomini M, Winnischofer SMB, Sassaki GL. NMR metabolic fingerprints of murine melanocyte and melanoma cell lines: application to biomarker discovery. Sci Rep 2017; 7:42324. [PMID: 28198377 PMCID: PMC5309734 DOI: 10.1038/srep42324] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 01/10/2017] [Indexed: 01/26/2023] Open
Abstract
Melanoma is the most aggressive type of skin cancer and efforts to improve the diagnosis of this neoplasia are largely based on the use of cell lines. Metabolomics is currently undergoing great advancements towards its use to screening for disease biomarkers. Although NMR metabolomics includes both 1D and 2D methodologies, there is a lack of data in the literature regarding heteronuclear 2D NMR assignments of the metabolome from eukaryotic cell lines. The present study applied NMR-based metabolomics strategies to characterize aqueous and lipid extracts from murine melanocytes and melanoma cell lines with distinct tumorigenic potential, successfully obtaining fingerprints of the metabolites from the extracts of the cell lines by means of 2D NMR HSQC correlation maps. Relative amounts of the identified metabolites were compared between the 4 cell lines. Multivariate analysis of 1H NMR data was able not only to differentiate the melanocyte cell line from the tumorigenic ones but also distinguish among the 3 tumorigenic cell lines. We also investigated the effects of mitogenic agents, and found that they can markedly influence the metabolome of the melanocyte cell line, resembling the pattern of most proliferative cell lines.
Collapse
Affiliation(s)
| | - Thiago Jacomasso
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Cx.P 19046, CEP 81531-990, Curitiba, PR, Brazil
| | - Daniel Suss Riter
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Cx.P 19046, CEP 81531-990, Curitiba, PR, Brazil
| | - Andersson Barison
- Departamento de Química, Universidade Federal do Paraná, Cx.P. 19081, CEP 81531-990, Curitiba, PR, Brazil
| | - Marcello Iacomini
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Cx.P 19046, CEP 81531-990, Curitiba, PR, Brazil
| | | | - Guilherme Lanzi Sassaki
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Cx.P 19046, CEP 81531-990, Curitiba, PR, Brazil
| |
Collapse
|
13
|
Tumor growth affects the metabonomic phenotypes of multiple mouse non-involved organs in an A549 lung cancer xenograft model. Sci Rep 2016; 6:28057. [PMID: 27329570 PMCID: PMC4916411 DOI: 10.1038/srep28057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 05/31/2016] [Indexed: 02/05/2023] Open
Abstract
The effects of tumorigenesis and tumor growth on the non-involved organs remain poorly understood although many research efforts have already been made for understanding the metabolic phenotypes of various tumors. To better the situation, we systematically analyzed the metabolic phenotypes of multiple non-involved mouse organ tissues (heart, liver, spleen, lung and kidney) in an A549 lung cancer xenograft model at two different tumor-growth stages using the NMR-based metabonomics approaches. We found that tumor growth caused significant metabonomic changes in multiple non-involved organ tissues involving numerous metabolic pathways, including glycolysis, TCA cycle and metabolisms of amino acids, fatty acids, choline and nucleic acids. Amongst these, the common effects are enhanced glycolysis and nucleoside/nucleotide metabolisms. These findings provided essential biochemistry information about the effects of tumor growth on the non-involved organs.
Collapse
|
14
|
Redalen KR, Sitter B, Bathen TF, Grøholt KK, Hole KH, Dueland S, Flatmark K, Ree AH, Seierstad T. High tumor glycine concentration is an adverse prognostic factor in locally advanced rectal cancer. Radiother Oncol 2015; 118:393-8. [PMID: 26705680 DOI: 10.1016/j.radonc.2015.11.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/23/2015] [Accepted: 11/28/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE Recognizing the link between altered tumor metabolism and disease aggressiveness, this study aimed to identify associations between tumor metabolic profiles and therapeutic outcome in locally advanced rectal cancer (LARC). MATERIALS AND METHODS Pretreatment tumor metabolic profiles from 54 LARC patients receiving combined-modality neoadjuvant treatment and surgery were acquired by high-resolution magic angle spinning magnetic resonance spectroscopy (HR MAS MRS). Metabolite concentrations were correlated to TNM and the presence of disseminated tumor cells (DTC) at diagnosis, ypTN and tumor regression grade (TRG) following neoadjuvant treatment, and progression-free survival (PFS). RESULTS Pretreatment tumor metabolite concentrations showed no significant associations to TNM, DTC, ypTN or TRG. In univariate regression analysis, high concentrations of glycine, creatine and myo-inositol were significantly associated with poor PFS, with metastasis as main PFS event. In multivariate analysis, high glycine concentration remained most significantly associated with poor PFS (hazard ratio=4.4, 95% confidence interval=1.4-14.3, p=0.008). CONCLUSIONS High tumor glycine concentration was identified as adverse prognostic factor for PFS in LARC. In a patient population treated with curative intent but with metastatic disease as main PFS event further investigations of glycine as early predictor of metastatic progression and therapeutic target are warranted.
Collapse
Affiliation(s)
| | - Beathe Sitter
- Department of Health Science, Sør-Trøndelag University College, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tone Frost Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Knut Håkon Hole
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Norway
| | - Svein Dueland
- Department of Oncology, Oslo University Hospital, Norway
| | - Kjersti Flatmark
- Department of Gastroenterological Surgery, Oslo University Hospital, Norway; Department of Tumor Biology, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway
| | - Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Norway
| | - Therese Seierstad
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Norway
| |
Collapse
|
15
|
Mustapha N, Pinon A, Limami Y, Simon A, Ghedira K, Hennebelle T, Chekir-Ghedira L. Crataegus azarolusLeaves Induce Antiproliferative Activity, Cell Cycle Arrest, and Apoptosis in Human HT-29 and HCT-116 Colorectal Cancer Cells. J Cell Biochem 2015; 117:1262-72. [DOI: 10.1002/jcb.25416] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 10/21/2015] [Indexed: 01/05/2023]
Affiliation(s)
- Nadia Mustapha
- Laboratoire de biologie cellulaire et moléculaire; Faculté de médecine dentaire; Université de Monastir, Rue Avicenne; 5000 Monastir Tunisie
- Unité de Substances naturelles bioactives et biotechnologie UR12ES12; Faculté de pharmacie de Monastir; Université de Monastir; Rue Avicenne 5000 Monastir Tunisie
| | - Aline Pinon
- Laboratoire de Chimie des Substances Naturelles; EA 1069; Faculté de Pharmacie; Université de Limoges; 2 rue du Dr marcland 87025 Limoges France
| | - Youness Limami
- Laboratoire de Chimie des Substances Naturelles; EA 1069; Faculté de Pharmacie; Université de Limoges; 2 rue du Dr marcland 87025 Limoges France
| | - Alain Simon
- Laboratoire de Chimie des Substances Naturelles; EA 1069; Faculté de Pharmacie; Université de Limoges; 2 rue du Dr marcland 87025 Limoges France
| | - Kamel Ghedira
- Unité de Substances naturelles bioactives et biotechnologie UR12ES12; Faculté de pharmacie de Monastir; Université de Monastir; Rue Avicenne 5000 Monastir Tunisie
| | - Thierry Hennebelle
- Laboratoire de Pharmacognosie; E.A. 4481; Faculté de Pharmacie B.P. 83; Université de Lille 2; 59006 Lille cedex France
| | - Leila Chekir-Ghedira
- Laboratoire de biologie cellulaire et moléculaire; Faculté de médecine dentaire; Université de Monastir, Rue Avicenne; 5000 Monastir Tunisie
- Unité de Substances naturelles bioactives et biotechnologie UR12ES12; Faculté de pharmacie de Monastir; Université de Monastir; Rue Avicenne 5000 Monastir Tunisie
| |
Collapse
|
16
|
Pacholczyk-Sienicka B, Fabiańska A, Pasz-Walczak G, Kordek R, Jankowski S. Prediction of survival for patients with advanced colorectal cancer using (1) H High-resolution magic angle spinning nuclear MR spectroscopy. J Magn Reson Imaging 2014; 41:1669-74. [PMID: 25146159 DOI: 10.1002/jmri.24734] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/08/2014] [Indexed: 12/28/2022] Open
Abstract
PURPOSE To evaluate whether the metabolic profiles of colorectal cancer specimens can be used for prediction of survival. MATERIALS AND METHODS The metabolic profiles of colorectal cancer tissues were determined using the high-resolution magic angle spinning (HR MAS) nuclear magnetic resonance (NMR) technique (16.4 T). HR MAS analysis was performed for 52 tissues taken from patients classified as survivors and nonsurvivors (30). Quantitative analysis was performed for each spectrum. Receiver operating characteristic (ROC) curves were used to evaluate the potential to predict patient survival over 5.5 years. RESULTS Analysis of (1) H NMR spectra led to the identification and quantitative analysis of 30 metabolites. A significant increase in the Tau/Gly and Tau/MI ratios were associated with long-term survival (P = 0.004 and P = 0.003, respectively). ROC analysis indicated that the Tau/MI ratio had the best predictive value for survival (sensitivity 64.7% and specificity 100%). Good predictive value of survival was found for Tau/Gly ratio (sensitivity 63.6% and specificity 96.3%). Moreover, the Glu/Gln metabolic ratio with a cutoff level of 1.74 was predictive of survival with a sensitivity of 83.3% and a specificity of 85.7%. CONCLUSION Our results indicate that HR MAS spectroscopy is potentially useful for survival prediction in advanced colorectal cancer.
Collapse
Affiliation(s)
| | - Anna Fabiańska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz, Lodz University of Technology, Poland
| | - Grażyna Pasz-Walczak
- Department of Pathology, Chair of Oncology, Lodz, Medical University of Lodz, Poland
| | - Radzisław Kordek
- Department of Pathology, Chair of Oncology, Lodz, Medical University of Lodz, Poland
| | - Stefan Jankowski
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz, Lodz University of Technology, Poland
| |
Collapse
|
17
|
Yang J, Song KD, Kim JH, Im GH, Yoon S, Namgung M, Hwang JH, Lee JH, Choi D. Characterization of brivanib therapy response in hepatocellular carcinoma xenografts using ¹H HR-MAS spectroscopy and histopathology. Mol Med Rep 2013; 8:1425-31. [PMID: 24064967 DOI: 10.3892/mmr.2013.1690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/21/2013] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis inhibition is an attractive therapeutic strategy in the management of solid tumors. Vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) are key factors in growth and neovascularization of hepatocellular carcinoma (HCC). Brivanib is a novel, orally available dual tyrosine kinase inhibitor that selectively targets the key angiogenesis receptors VEGF‑R2, FGF‑R1 and FGF‑R2. Recently, high‑resolution magic angle spinning magnetic resonance spectroscopy (HR‑MAS MRS) has provided the opportunity to investigate more detailed metabolic profiles from intact tissue specimens that are correlated with histopathology and is thus, a promising tool for monitoring changes induced by treatment. In the present study, 1H HR‑MAS MRS and immunohistochemistry were used to investigate the antitumor efficacy of brivanib in HCC xenograft models. Tumor growth was significantly suppressed in brivanib‑treated mice compared with the controls and treatment was associated with the inhibition of angiogenesis, increased apoptosis and inhibition of cell proliferation. Furthermore, HR‑MAS techniques showed altered metabolic profiles between the two groups. HR‑MAS spectra demonstrated a significant decrease in choline metabolite levels in the treated groups, concurrent with decreased cell proliferation and increased apoptosis. The results showed that 1H HR‑MAS MRS provides quantitative metabolite information that may be used to analyze the efficacy of brivanib treatment in Hep3B tumor xenografts. Thus, the HR‑MAS MRS technique may be a complementary method to support histopathological results and increase its potential for use in the clinic.
Collapse
Affiliation(s)
- Jehoon Yang
- Department of Medical Science, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Loftus NJ, Lai L, Wilkinson RW, Odedra R, Wilson ID, Barnes AJ. Global metabolite profiling of human colorectal cancer xenografts in mice using HPLC-MS/MS. J Proteome Res 2013; 12:2980-6. [PMID: 23631600 DOI: 10.1021/pr400260h] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Reversed-phase gradient LC-MS was used to perform untargeted metabonomic analysis on extracts of human colorectal cancer (CRC) cell lines (COLO 205, HT-29, HCT 116 and SW620) subcutaneously implanted into age-matched athymic nude male mice to study small molecule metabolic profiles and examine possible correlations with human cancer biopsies. Following high mass accuracy data analysis using MS and MS/MS, metabolites were identified by searching against major metabolite databases including METLIN, MASSBANK, The Human Metabolome Database, PubChem, Biospider, LipidMaps and KEGG. HT-29 and COLO 205 tumor xenografts showed a distribution of metabolites that differed from SW620 and HCT 116 xenografts (predominantly on the basis of relative differences in the amounts of amino acids and lipids detected). This finding is consistent with NMR-based analysis of human colorectal tissue, where the metabolite profiles of HT-29 tumors exhibit the greatest similarity to human rectal cancer tissue with respect to changes in the relative amounts of lipids and choline-containing compounds. As the metabolic signatures of cancer cells result from oncogene-directed metabolic reprogramming, the HT-29 xenografts in mice may prove to be a useful model to further study the tumor microenvironment and cancer biology.
Collapse
Affiliation(s)
- Neil J Loftus
- Mass Spectrometry Business Unit, Shimadzu, Manchester, United Kingdom.
| | | | | | | | | | | |
Collapse
|
19
|
Feasibility of MR metabolomics for immediate analysis of resection margins during breast cancer surgery. PLoS One 2013; 8:e61578. [PMID: 23613877 PMCID: PMC3629170 DOI: 10.1371/journal.pone.0061578] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 03/11/2013] [Indexed: 12/17/2022] Open
Abstract
In this study, the feasibility of high resolution magic angle spinning (HR MAS) magnetic resonance spectroscopy (MRS) of small tissue biopsies to distinguish between tumor and non-involved adjacent tissue was investigated. With the current methods, delineation of the tumor borders during breast cancer surgery is a challenging task for the surgeon, and a significant number of re-surgeries occur. We analyzed 328 tissue samples from 228 breast cancer patients using HR MAS MRS. Partial least squares discriminant analysis (PLS-DA) was applied to discriminate between tumor and non-involved adjacent tissue. Using proper double cross validation, high sensitivity and specificity of 91% and 93%, respectively was achieved. Analysis of the loading profiles from both principal component analysis (PCA) and PLS-DA showed the choline-containing metabolites as main biomarkers for tumor content, with phosphocholine being especially high in tumor tissue. Other indicative metabolites include glycine, taurine and glucose. We conclude that metabolic profiling by HR MAS MRS may be a potential method for on-line analysis of resection margins during breast cancer surgery to reduce the number of re-surgeries and risk of local recurrence.
Collapse
|
20
|
Metabolomics of colorectal cancer: past and current analytical platforms. Anal Bioanal Chem 2013; 405:5013-30. [PMID: 23494270 DOI: 10.1007/s00216-013-6777-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 01/18/2013] [Accepted: 01/22/2013] [Indexed: 02/07/2023]
Abstract
Metabolomics is coming of age as an important area of investigation which may help reveal answers to questions left unanswered or only partially understood from proteomic or genomic approaches. Increased knowledge of the relationship of genes and proteins to smaller biomolecules (metabolites) will advance our ability to diagnose, treat, and perhaps prevent cancer and other diseases that have eluded scientists for generations. Colorectal tumors are the second leading cause of cancer mortality in the USA, and the incidence is rising. Many patients present late, after the onset of symptoms, when the tumor has spread from the primary site. Once metastases have occurred, the prognosis is significantly worse. Understanding alterations in metabolic profiles that occur with tumor onset and progression could lead to better diagnostic tests as well as uncover new approaches to treat or even prevent colorectal cancer (CRC). In this review, we explore the various analytical technologies that have been applied in CRC metabolomics research and summarize all metabolites measured in CRC and integrate them into metabolic pathways. Early studies with nuclear magnetic resonance and gas-chromatographic mass spectrometry suggest that tumor cells are characterized by aerobic glycolysis, increased purine metabolism for DNA synthesis, and protein synthesis. Liquid chromatography, capillary electrophoresis, and ion mobility, each coupled with mass spectrometry, promise to advance the field and provide new insight into metabolic pathways used by cancer cells. Studies with improved technology are needed to identify better biomarkers and targets for treatment or prevention of CRC.
Collapse
|
21
|
Okushita K, Komatsu T, Chikayama E, Kikuchi J. Statistical approach for solid-state NMR spectra of cellulose derived from a series of variable parameters. Polym J 2012. [DOI: 10.1038/pj.2012.82] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
22
|
Chen W, Zhou X, Huang D, Chen F, Du X. Metabolic Profiling of Human Colorectal Cancer Using High Resolution 1H Nuclear Magnetic Resonance Spectroscopy. CHINESE J CHEM 2011. [DOI: 10.1002/cjoc.201180423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
23
|
Luo XJ, Zhang B, Li JG, Luo XA, Yang LF. Autofluorescence spectroscopy for evaluating dysplasia in colorectal tissues. Z Med Phys 2011; 22:40-7. [PMID: 22112637 DOI: 10.1016/j.zemedi.2011.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 09/29/2011] [Accepted: 10/20/2011] [Indexed: 01/23/2023]
Abstract
The aim of this study was to assess the applicability of autofluorescence (AF) spectroscopy as a method for the diagonosis of normal, benign and malignant of dysplasia in colorectal tissues experimentally. By improvement of optical design in laser pulse generator, wavelength-adjustable output was acquired and the optimal wavelength was defined as 380 nm. With 380-nm pulsed laser excitation, AF spectra of normal, benign and malignant colorectal tissues were recorded in the spectra region from 460-570 nm in vitro. The spectral analysis for discrimination among the different types of tissues was carried out using principal component analysis (PCA)-based Neural networks algorithm. The performance of analysis was pretty good with sensitivity, specificity and accuracy found to be 100%,90% and 96.7%, respectively. The AF spectroscopy may serve as an excellent tool for the evaluation of dysplasia in colorectal clinical diagnosis.
Collapse
Affiliation(s)
- Xiang-jian Luo
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | | | | | | | | |
Collapse
|
24
|
Wang H, Tso VK, Slupsky CM, Fedorak RN. Metabolomics and detection of colorectal cancer in humans: a systematic review. Future Oncol 2011; 6:1395-406. [PMID: 20919825 DOI: 10.2217/fon.10.107] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Metabolomics represents one of the new omics sciences and capitalizes on the unique presence and concentration of small molecules in tissues and body fluids to construct a 'fingerprint' that can be unique to the individual and, within that individual, unique to environmental influences, including health and disease states. As such, metabolomics has the potential to serve an important role in diagnosis and management of human conditions. Colorectal cancer is a major public health concern. Current population-based screening methods are suboptimal and whether metabolomics could represent a new tool of screening is under investigation. The purpose of this systematic review is to summarize existing literature on metabolomics and colorectal cancer, in terms of diagnostic accuracies and distinguishing metabolites. Eight studies are included. A total of 12 metabolites (taurine, lactate, choline, inositol, glycine, phosphocholine, proline, phenylalanine, alanine, threonine, valine and leucine) were found to be more prevalent in colorectal cancer and glucose was found to be in higher proportion in control specimens using tissue metabolomics. Serum and urine metabolomics identified several other differential metabolites between controls and colorectal cancer patients. This article highlights the novelty of the field of metabolomics in colorectal oncology.
Collapse
Affiliation(s)
- Haili Wang
- University of Alberta, 130 University Campus, 112th St & 85th Avenue, Edmonton, AB, Canada
| | | | | | | |
Collapse
|
25
|
Zhang L, Jia X, Peng X, Ou Q, Zhang Z, Qiu C, Yao Y, Shen F, Yang H, Ma F, Wang J, Yuan Z. Development and validation of a liquid chromatography-mass spectrometry metabonomic platform in human plasma of liver failure caused by hepatitis B virus. Acta Biochim Biophys Sin (Shanghai) 2010; 42:688-98. [PMID: 20810534 DOI: 10.1093/abbs/gmq078] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This paper presents an liquid chromatography (LC)/mass spectrometry (MS)-based metabonomic platform that combined the discovery of differential metabolites through principal component analysis (PCA) with the verification by selective multiple reaction monitoring (MRM). These methods were applied to analyze plasma samples from liver disease patients and healthy donors. LC-MS raw data (about 1000 compounds), from the plasma of liver failure patients (n = 26) and healthy controls (n = 16), were analyzed through the PCA method and a pattern recognition profile that had significant difference between liver failure patients and healthy controls (P < 0.05) was established. The profile was verified in 165 clinical subjects. The specificity and sensitivity of this model in predicting liver failure were 94.3 and 100.0%, respectively. The differential ions with m/z of 414.5, 432.0, 520.5, and 775.0 were verified to be consistent with the results from PCA by MRM mode in 40 clinical samples, and were proved not to be caused by the medicines taken by patients through rat model experiments. The compound with m/z of 520.5 was identified to be 1-Linoleoylglycerophosphocholine or 1-Linoleoylphosphatidylcholine through exact mass measurements performed using Ion Trap-Time-of-Flight MS and METLIN Metabolite Database search. In all, it was the first time to integrate metabonomic study and MRM relative quantification of differential peaks in a large number of clinical samples. Thereafter, a rat model was used to exclude drug effects on the abundance of differential ion peaks. 1-Linoleoylglycerophosphocholine or 1-Linoleoylphosphatidylcholine, a potential biomarker, was identified. The LC/MS-based metabonomic platform could be a powerful tool for the metabonomic screening of plasma biomarkers.
Collapse
Affiliation(s)
- Lijun Zhang
- Shanghai Public Health Clinical Center Affiliated to Fudan University, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tessem MB, Selnæs KM, Sjursen W, Tranø G, Giskeødegård GF, Bathen TF, Gribbestad IS, Hofsli E. Discrimination of Patients with Microsatellite Instability Colon Cancer using 1H HR MAS MR Spectroscopy and Chemometric Analysis. J Proteome Res 2010; 9:3664-70. [DOI: 10.1021/pr100176g] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- May-Britt Tessem
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway, Department of Laboratory Medicine Children’s and Women’s Health, NTNU, Trondheim, Norway, Department of Pathology and Medical Genetics, St. Olavs University Hospital, Trondheim, Norway, Department of Surgery, Levanger Hospital, Sykehuset Innherred, Levanger, Norway, and Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Kirsten M. Selnæs
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway, Department of Laboratory Medicine Children’s and Women’s Health, NTNU, Trondheim, Norway, Department of Pathology and Medical Genetics, St. Olavs University Hospital, Trondheim, Norway, Department of Surgery, Levanger Hospital, Sykehuset Innherred, Levanger, Norway, and Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Wenche Sjursen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway, Department of Laboratory Medicine Children’s and Women’s Health, NTNU, Trondheim, Norway, Department of Pathology and Medical Genetics, St. Olavs University Hospital, Trondheim, Norway, Department of Surgery, Levanger Hospital, Sykehuset Innherred, Levanger, Norway, and Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Gerd Tranø
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway, Department of Laboratory Medicine Children’s and Women’s Health, NTNU, Trondheim, Norway, Department of Pathology and Medical Genetics, St. Olavs University Hospital, Trondheim, Norway, Department of Surgery, Levanger Hospital, Sykehuset Innherred, Levanger, Norway, and Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Guro F. Giskeødegård
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway, Department of Laboratory Medicine Children’s and Women’s Health, NTNU, Trondheim, Norway, Department of Pathology and Medical Genetics, St. Olavs University Hospital, Trondheim, Norway, Department of Surgery, Levanger Hospital, Sykehuset Innherred, Levanger, Norway, and Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Tone F. Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway, Department of Laboratory Medicine Children’s and Women’s Health, NTNU, Trondheim, Norway, Department of Pathology and Medical Genetics, St. Olavs University Hospital, Trondheim, Norway, Department of Surgery, Levanger Hospital, Sykehuset Innherred, Levanger, Norway, and Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Ingrid S. Gribbestad
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway, Department of Laboratory Medicine Children’s and Women’s Health, NTNU, Trondheim, Norway, Department of Pathology and Medical Genetics, St. Olavs University Hospital, Trondheim, Norway, Department of Surgery, Levanger Hospital, Sykehuset Innherred, Levanger, Norway, and Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Eva Hofsli
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway, Department of Laboratory Medicine Children’s and Women’s Health, NTNU, Trondheim, Norway, Department of Pathology and Medical Genetics, St. Olavs University Hospital, Trondheim, Norway, Department of Surgery, Levanger Hospital, Sykehuset Innherred, Levanger, Norway, and Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| |
Collapse
|
27
|
Ma Y, Liu W, Peng J, Huang L, Zhang P, Zhao X, Cheng Y, Qin H. A pilot study of gas chromatograph/mass spectrometry-based serum metabolic profiling of colorectal cancer after operation. Mol Biol Rep 2009; 37:1403-11. [DOI: 10.1007/s11033-009-9524-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 03/20/2009] [Indexed: 01/09/2023]
|
28
|
Righi V, Durante C, Cocchi M, Calabrese C, Di Febo G, Lecce F, Pisi A, Tugnoli V, Mucci A, Schenetti L. Discrimination of Healthy and Neoplastic Human Colon Tissues by ex Vivo HR-MAS NMR Spectroscopy and Chemometric Analyses. J Proteome Res 2009; 8:1859-69. [DOI: 10.1021/pr801094b] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Valeria Righi
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| | - Caterina Durante
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| | - Marina Cocchi
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| | - Carlo Calabrese
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| | - Giulio Di Febo
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| | - Ferdinando Lecce
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| | - Annamaria Pisi
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| | - Vitaliano Tugnoli
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| | - Adele Mucci
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| | - Luisa Schenetti
- Dipartimento di Biochimica “G. Moruzzi”, Università di Bologna, Via Belmeloro 8/2, 40126 Bologna, Italy, Dipartimento di Chimica, Università di Modena e Reggio Emilia, Via G. Campi 183, 41100 Modena, Italy, Dipartimento di Medicina Interna e Gastroenterologia, Università di Bologna, Via G. Massarenti 9, 40138, Bologna, Italy, Dipartimento Emergenza/Urgenza, Chirurgia Generale e dei Trapianti, Università di Bologna, Via G. Massarenti 9, 40138 Bologna, Italy, and DiSTA, Università di Bologna, Viale Fanin
| |
Collapse
|
29
|
Mal M, Koh PK, Cheah PY, Chan ECY. Development and validation of a gas chromatography/mass spectrometry method for the metabolic profiling of human colon tissue. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2009; 23:487-494. [PMID: 19140133 DOI: 10.1002/rcm.3898] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
In this study, a gas chromatography/mass spectrometry (GC/MS) method was developed and validated for the metabolic profiling of human colon tissue. Each colon tissue sample (20 mg) was ultra-sonicated with 1 mL of a mixture of chloroform/methanol/water in the ratio of 20:50:20 (v/v/v), followed by centrifugation, collection of supernatant, drying, removal of moisture using anhydrous toluene and finally derivatization using N-methyl-N-trifluoroacetamide (MSTFA) with 1% trimethylchlorosilane (TMCS). A volume of 1 microL of the derivatized mixture was injected into the GC/MS system. A total of 53 endogenous metabolites were separated and identified in the GC/MS chromatogram, all of which were selected to evaluate the sample stability and precision of the method. Of the identified endogenous metabolites 19 belonging to diverse chemical classes and covering a wide range of the GC retention times (Rt) were selected to investigate the quantitative linearity of the method. The developed GC/MS method demonstrated good reproducibility with intra- and inter-day precision within relative standard deviation (RSD) of +/-15%. The metabolic profiles of the intact tissue were determined to be stable (100 +/- 15%) for up to 90 days at -80 degrees C. Satisfactory results were also obtained in the case of other stability-indicating studies such as freeze/thaw cycle stability, bench-top stability and autosampler stability. The developed method showed a good linear response for each of the 19 analytes tested (r(2) > 0.99). Our GC/MS metabolic profiling method was successfully applied to discriminate biopsied colorectal cancer (CRC) tissue from their matched normal tissue obtained from six CRC patients using orthogonal partial least-squares discriminant analysis [two latent variables, R(2)Y = 0.977 and Q(2) (cumulative) = 0.877].
Collapse
Affiliation(s)
- Mainak Mal
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543
| | | | | | | |
Collapse
|