1
|
Fatfat Z, Hussein M, Fatfat M, Gali-Muhtasib H. Omics technologies as powerful approaches to unravel colorectal cancer complexity and improve its management. Mol Cells 2025; 48:100200. [PMID: 40024318 PMCID: PMC11976254 DOI: 10.1016/j.mocell.2025.100200] [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: 10/25/2024] [Revised: 01/31/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025] Open
Abstract
Colorectal cancer (CRC) continues to rank among the deadliest and most prevalent cancers worldwide, necessitating an innovative and comprehensive approach that addresses this serious health challenge at various stages, from screening and diagnosis to treatment and prognosis. As CRC research progresses, the adoption of an omics-centered approach holds transformative potential to revolutionize the management of this disease. Advances in omics technologies encompassing genomics, transcriptomics, proteomics, metabolomics, and epigenomics allow to unravel the oncogenic alterations at these levels, elucidating the intricacies and the heterogeneous nature of CRC. By providing a comprehensive molecular landscape of CRC, omics technologies enable the discovery of potential biomarkers for early non-invasive detection of CRC, definition of CRC subtypes, prediction of its staging, prognosis, and overall survival of CRC patients. They also allow the identification of potential therapeutic targets, prediction of drug response, tracking treatment efficacy, detection of residual disease and cancer relapse, and deciphering the mechanisms of drug resistance. Moreover, they allow the distinction of non-metastatic CRC patients from metastatic ones as well as the stratification of metastatic risk. Importantly, omics technologies open up new opportunities to establish molecular-based criteria to guide the selection of effective treatment paving the way for the personalization of therapy for CRC patients. This review consolidates current knowledge on the omics-based preclinical discoveries in CRC research emphasizing the significant potential of these technologies to improve CRC screening, diagnosis, and prognosis and promote the implementation of personalized medicine to ultimately reduce CRC prevalence and mortality.
Collapse
Affiliation(s)
- Zaynab Fatfat
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Marwa Hussein
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, Lebanon
| | - Maamoun Fatfat
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
2
|
Hou Y, Lin J, Yao H, Wu Z, Lin Y, Lin J. Linking Metastatic Behavior and Metabolic Heterogeneity of Circulating Tumor Cells at Single-Cell Level Using an Integrative Microfluidic System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413978. [PMID: 39960842 PMCID: PMC11984876 DOI: 10.1002/advs.202413978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/28/2024] [Indexed: 04/12/2025]
Abstract
Circulating tumor cells (CTCs) are pivotal biomarkers in tumor metastasis, however, the underlying molecular mechanism of CTCs behavioral heterogeneity during metastasis remains unexplored. Here, an integrative workflow is developed to link behavior characteristics to metabolic profiling within individual CTCs, which simulates the metastatic process on a microfluidic system and combined with single-cell mass spectrometry (MS) detection. Spheroid-derived HCT116 cells are tracked and extracted via a temporary vascular system, revealing various arrest patterns under biomimetic vascular shear flow. Downstream MS analysis characterizes 17 cellular metabolites and associates metabolic profiles with de-adhesion behaviors of the same CTCs, identifying a potential high-metastatic subpopulation with enhanced arrest ability and evaluating critical metabolites involved in metastasis pathways. Additionally, the metastasis-inhibiting effect of anti-tumor drug 5-fluorouracil by reducing high-metastatic cells in spheroids is elucidated. This approach offers a valuable opportunity to dissect the interplay of the metastatic behavior and metabolic profiles of CTCs and foster insights into the molecular mechanisms underlying behavioral phenotypes in the tumor metastasis process.
Collapse
Affiliation(s)
- Ying Hou
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Jiaxu Lin
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Hongren Yao
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Zengnan Wu
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Yongning Lin
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Jin‐Ming Lin
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| |
Collapse
|
3
|
Sinha A, Griffith L, Acharjee A. Systematic Review and Meta-Analysis: Taurine and Its Association With Colorectal Carcinoma. Cancer Med 2024; 13:e70424. [PMID: 39632512 PMCID: PMC11617591 DOI: 10.1002/cam4.70424] [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: 08/19/2024] [Revised: 09/30/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common cancers. Various options are available for treatment, but prognosis is still poor in the more advanced stages. Current screening methods are not as accurate for distinguishing between benign and malignant growths, resulting in unnecessary invasive procedures. Recently a focus has been placed on identifying metabolites. Of these, taurine has frequently been detected, and this particular compound has a multifactorial role in human physiology. METHODS We conducted a systematic review of studies up till November 2023. Searches were done in three databases- MEDLINE, CINAHL-Ebsco, and PubMed. Three independent reviewers filter titles, abstracts, and full-texts according to selection criteria. Ten studies (samples = 1714) were identified showing a differential level of taurine in CRC patient samples. Quality assessment accounted for the risk of bias of each study using the 'robvis' tool. Where meaningful comparisons could be made, meta-analyses were carried out using the 'R' program for precalculated effect sizes with 'metagen' in R. The 'meta' package was utilised for creation of forest plots. FINDINGS Taurine was shown to significantly increase odds of CRC. It was also significantly associated with being a discriminator for CRC as a diagnostic metabolite. This was maintained at various stages of CRC. Taurine had increased expression in CRC patients, especially when the matrix utilised was blood. Nevertheless, there was significant heterogeneity for some outcomes. INTERPRETATION In conclusion, these findings highlight the potential of using taurine as well as other bile acid metabolites (lithocholic and ursodeoxycholic acid) to diagnose CRC and illustrate the link with microbiome interactions. Overall increased taurine concentration are associated with significantly increased odds for CRC. There was mostly an increase in relative expression of taurine in CRC samples, excluding results from Wang et al.
Collapse
Affiliation(s)
- Akshat Sinha
- Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
| | - Liam Griffith
- Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
| | - Animesh Acharjee
- Institute of Cancer and Genomic SciencesUniversity of BirminghamBirminghamUK
- MRC Health Data Research UK (HDR UK)BirminghamUK
- Institute of Translational MedicineUniversity Hospitals Birmingham NHS, Foundation TrustBirminghamUK
- Centre for Health Data ResearchUniversity of BirminghamBirminghamUK
| |
Collapse
|
4
|
Yang R, Tsigelny IF, Kesari S, Kouznetsova VL. Colorectal Cancer Detection via Metabolites and Machine Learning. Curr Issues Mol Biol 2024; 46:4133-4146. [PMID: 38785522 PMCID: PMC11119033 DOI: 10.3390/cimb46050254] [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: 03/21/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Today, colorectal cancer (CRC) diagnosis is performed using colonoscopy, which is the current, most effective screening method. However, colonoscopy poses risks of harm to the patient and is an invasive process. Recent research has proven metabolomics as a potential, non-invasive detection method, which can use identified biomarkers to detect potential cancer in a patient's body. The aim of this study is to develop a machine-learning (ML) model based on chemical descriptors that will recognize CRC-associated metabolites. We selected a set of metabolites found as the biomarkers of CRC, confirmed that they participate in cancer-related pathways, and used them for training a machine-learning model for the diagnostics of CRC. Using a set of selective metabolites and random compounds, we developed a range of ML models. The best performing ML model trained on Stage 0-2 CRC metabolite data predicted a metabolite class with 89.55% accuracy. The best performing ML model trained on Stage 3-4 CRC metabolite data predicted a metabolite class with 95.21% accuracy. Lastly, the best-performing ML model trained on Stage 0-4 CRC metabolite data predicted a metabolite class with 93.04% accuracy. These models were then tested on independent datasets, including random and unrelated-disease metabolites. In addition, six pathways related to these CRC metabolites were also distinguished: aminoacyl-tRNA biosynthesis; glyoxylate and dicarboxylate metabolism; glycine, serine, and threonine metabolism; phenylalanine, tyrosine, and tryptophan biosynthesis; arginine biosynthesis; and alanine, aspartate, and glutamate metabolism. Thus, in this research study, we created machine-learning models based on metabolite-related descriptors that may be helpful in developing a non-invasive diagnosis method for CRC.
Collapse
Affiliation(s)
- Rachel Yang
- REHS Program, San Diego Supercomputer Center, University of California San Diego, MC 0505, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Igor F. Tsigelny
- San Diego Supercomputer Center, University of California San Diego, MC 0505, 9500 Gilman Drive, La Jolla, CA 92093, USA;
- BiAna, P.O. Box 2525, La Jolla, CA 92038, USA
- Department of Neurosciences, University of California San Diego, MC00505, 9500 Gilman Drive, La Jolla, CA 92093, USA
- CureScience Institute, 5820 Oberlin Drive, STE 202, San Diego, CA 92121, USA
| | - Santosh Kesari
- Pacific Neuroscience Institute, 2125 Arizona Avenue, Santa Monica, CA 90404, USA;
| | - Valentina L. Kouznetsova
- San Diego Supercomputer Center, University of California San Diego, MC 0505, 9500 Gilman Drive, La Jolla, CA 92093, USA;
- BiAna, P.O. Box 2525, La Jolla, CA 92038, USA
- CureScience Institute, 5820 Oberlin Drive, STE 202, San Diego, CA 92121, USA
| |
Collapse
|
5
|
Futami M, Naito H, Ninomiya S, Chen LC, Iwano T, Yoshimura K, Ukita Y. Automated sample preparation for electrospray ionization mass spectrometry based on CLOCK-controlled autonomous centrifugal microfluidics. Biomed Microdevices 2024; 26:22. [PMID: 38592604 PMCID: PMC11003918 DOI: 10.1007/s10544-024-00703-4] [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] [Accepted: 03/20/2024] [Indexed: 04/10/2024]
Abstract
We report a centrifugal microfluidic device that automatically performs sample preparation under steady-state rotation for clinical applications using mass spectrometry. The autonomous microfluidic device was designed for the control of liquid operation on centrifugal hydrokinetics (CLOCK) paradigm. The reported device was highly stable, with less than 7% variation with respect to the time of each unit operation (sample extraction, mixing, and supernatant extraction) in the preparation process. An agitation mechanism with bubbling was used to mix the sample and organic solvent in this device. We confirmed that the device effectively removed the protein aggregates from the sample, and the performance was comparable to those of conventional manual sample preparation procedures that use high-speed centrifugation. In addition, probe electrospray ionization mass spectrometry (PESI-MS) was performed to compare the device-treated and manually treated samples. The obtained PESI-MS spectra were analyzed by partial least squares discriminant analysis, and the preparation capability of the device was found to be equivalent to that of the conventional method.
Collapse
Affiliation(s)
- Masahiro Futami
- Department of Engineering, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, Graduate School of University of Yamanashi, 4-3-11 Takeda, Kofu, 400-8510, Japan
| | - Hiroki Naito
- Department of Engineering, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, Graduate School of University of Yamanashi, 4-3-11 Takeda, Kofu, 400-8510, Japan
| | - Satoshi Ninomiya
- Graduate Faculty of Interdisciplinary Research, University of Yamanashi, 4-3-11 Takeda, Kofu, 400-8510, Japan
| | - Lee Chuin Chen
- Graduate Faculty of Interdisciplinary Research, University of Yamanashi, 4-3-11 Takeda, Kofu, 400-8510, Japan
| | - Tomohiko Iwano
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, 409-3898, Japan
| | - Kentaro Yoshimura
- Division of Molecular Biology, Center for Medical Education and Sciences, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, 409-3898, Japan
| | - Yoshiaki Ukita
- Graduate Faculty of Interdisciplinary Research, University of Yamanashi, 4-3-11 Takeda, Kofu, 400-8510, Japan.
| |
Collapse
|
6
|
Tadaka S, Kawashima J, Hishinuma E, Saito S, Okamura Y, Otsuki A, Kojima K, Komaki S, Aoki Y, Kanno T, Saigusa D, Inoue J, Shirota M, Takayama J, Katsuoka F, Shimizu A, Tamiya G, Shimizu R, Hiratsuka M, Motoike I, Koshiba S, Sasaki M, Yamamoto M, Kinoshita K. jMorp: Japanese Multi-Omics Reference Panel update report 2023. Nucleic Acids Res 2024; 52:D622-D632. [PMID: 37930845 PMCID: PMC10767895 DOI: 10.1093/nar/gkad978] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023] Open
Abstract
Modern medicine is increasingly focused on personalized medicine, and multi-omics data is crucial in understanding biological phenomena and disease mechanisms. Each ethnic group has its unique genetic background with specific genomic variations influencing disease risk and drug response. Therefore, multi-omics data from specific ethnic populations are essential for the effective implementation of personalized medicine. Various prospective cohort studies, such as the UK Biobank, All of Us and Lifelines, have been conducted worldwide. The Tohoku Medical Megabank project was initiated after the Great East Japan Earthquake in 2011. It collects biological specimens and conducts genome and omics analyses to build a basis for personalized medicine. Summary statistical data from these analyses are available in the jMorp web database (https://jmorp.megabank.tohoku.ac.jp), which provides a multidimensional approach to the diversity of the Japanese population. jMorp was launched in 2015 as a public database for plasma metabolome and proteome analyses and has been continuously updated. The current update will significantly expand the scale of the data (metabolome, genome, transcriptome, and metagenome). In addition, the user interface and backend server implementations were rewritten to improve the connectivity between the items stored in jMorp. This paper provides an overview of the new version of the jMorp.
Collapse
Affiliation(s)
- Shu Tadaka
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Junko Kawashima
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Eiji Hishinuma
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Sakae Saito
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Yasunobu Okamura
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Akihito Otsuki
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Kaname Kojima
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Shohei Komaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Shiwa-gun, Iwate 028-3609, Japan
| | - Yuichi Aoki
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, Sendai, Miyagi 980-8579, Japan
| | - Takanari Kanno
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Daisuke Saigusa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Faculty of Pharma-Science, Teikyo University, Tokyo 173-8605, Japan
| | - Jin Inoue
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Matsuyuki Shirota
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Jun Takayama
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
- RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Fumiki Katsuoka
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Atsushi Shimizu
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Shiwa-gun, Iwate 028-3609, Japan
| | - Gen Tamiya
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
- RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Ritsuko Shimizu
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Masahiro Hiratsuka
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Ikuko N Motoike
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, Sendai, Miyagi 980-8579, Japan
| | - Seizo Koshiba
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Shiwa-gun, Iwate 028-3609, Japan
| | - Masayuki Yamamoto
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
| | - Kengo Kinoshita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, Sendai, Miyagi 980-8579, Japan
| |
Collapse
|
7
|
Bhalla M, Mittal R, Kumar M, Bhatia R, Kushwah AS. Metabolomics: A Tool to Envisage Biomarkers in Clinical Interpretation of Cancer. Curr Drug Res Rev 2024; 16:333-348. [PMID: 37702236 DOI: 10.2174/2589977516666230912120412] [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: 05/19/2023] [Revised: 06/22/2023] [Accepted: 07/20/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Cancer is amongst the most dreadful ailments of modern times, and its impact continuously worsens global health systems. Early diagnosis and suitable therapeutic agents are the prime keys to managing this disease. Metabolomics deals with the complete profiling of cells and physiological phenomena in their organelles, thus helping in keen knowledge of the pathological status of the disease. It has been proven to be one of the best strategies in the early screening of cancer. OBJECTIVE This review has covered the recent updates on the promising role of metabolomics in the identification of significant biochemical markers in cancer-prone individuals that could lead to the identification of cancer in the early stages. METHODS The literature was collected through various databases, like Scopus, PubMed, and Google Scholar, with stress laid on the last ten years' publications. CONCLUSION It was assessed in this review that early recognition of cancerous growth could be achieved via complete metabolic profiling in association with transcriptomics and proteomics. The outcomes are rooted in various clinical studies that anticipated various biomarkers like tryptophan, phenylalanine, lactates, and different metabolic pathways associated with the Warburg effect. This metabolite imaging has been a fundamental step for the target acquisition, evaluation of predictive cancer biomarkers for early detection, and outlooks into cancer therapy along with critical evaluation. Significant efforts should be made to make this technique most reliable and easy.
Collapse
Affiliation(s)
- Medha Bhalla
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Ropar, 140111, India
| | - Roopal Mittal
- Department of Pharmacology, IKG Punjab Technical University, Jalandhar, 144601, India
- Department of Pharmacology, R.K.S.D. College of Pharmacy, Kaithal, 136027, India
| | - Manish Kumar
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Rohit Bhatia
- Department of Pharmaceutical Chemistry, Indo Soviet Friendship College of Pharmacy, Moga, 142001, India
| | - Ajay Singh Kushwah
- Department of Pharmacology, Amar Shaheed Baba Ajit Singh Jujhar Singh Memorial College of Pharmacy, Ropar, 140111, India
| |
Collapse
|
8
|
Qiu X, Lu R, He Q, Chen S, Huang C, Lin D. Metabolic signatures and potential biomarkers for the diagnosis and treatment of colon cancer cachexia. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1913-1924. [PMID: 37705348 PMCID: PMC11294056 DOI: 10.3724/abbs.2023151] [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: 05/24/2023] [Accepted: 06/29/2023] [Indexed: 09/15/2023] Open
Abstract
Cancer cachexia (CAC) is a debilitating condition that often arises from noncachexia cancer (NCAC), with distinct metabolic characteristics and medical treatments. However, the metabolic changes and underlying molecular mechanisms during cachexia progression remain poorly understood. Understanding the progression of CAC is crucial for developing diagnostic approaches to distinguish between CAC and NCAC stages, facilitating appropriate treatment for cancer patients. In this study, we establish a mouse model of colon CAC and categorize the mice into three groups: CAC, NCAC and normal control (NOR). By performing nuclear magnetic resonance (NMR)-based metabolomic profiling on mouse sera, we elucidate the metabolic properties of these groups. Our findings unveil significant differences in the metabolic profiles among the CAC, NCAC and NOR groups, highlighting significant impairments in energy metabolism and amino acid metabolism during cachexia progression. Additionally, we observe the elevated serum levels of lysine and acetate during the transition from the NCAC to CAC stages. Using multivariate ROC analysis, we identify lysine and acetate as potential biomarkers for distinguishing between CAC and NCAC stages. These biomarkers hold promise for the diagnosis of CAC from noncachexia cancer. Our study provides novel insights into the metabolic mechanisms underlying cachexia progression and offers valuable avenues for the diagnosis and treatment of CAC in clinical settings.
Collapse
Affiliation(s)
- Xu Qiu
- Key Laboratory for Chemical Biology of Fujian ProvinceMOE Key Laboratory of Spectrochemical Analysis and InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Ruohan Lu
- Key Laboratory for Chemical Biology of Fujian ProvinceMOE Key Laboratory of Spectrochemical Analysis and InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Qiqing He
- Key Laboratory for Chemical Biology of Fujian ProvinceMOE Key Laboratory of Spectrochemical Analysis and InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Shu Chen
- Key Laboratory for Chemical Biology of Fujian ProvinceMOE Key Laboratory of Spectrochemical Analysis and InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Caihua Huang
- Research and Communication Center of Exercise and HealthXiamen University of TechnologyXiamen361005China
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian ProvinceMOE Key Laboratory of Spectrochemical Analysis and InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| |
Collapse
|
9
|
Santos MD, Barros I, Brandão P, Lacerda L. Amino Acid Profiles in the Biological Fluids and Tumor Tissue of CRC Patients. Cancers (Basel) 2023; 16:69. [PMID: 38201497 PMCID: PMC10778074 DOI: 10.3390/cancers16010069] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Amino acids are the building blocks of proteins and essential players in pathways such as the citric acid and urea cycle, purine and pyrimidine biosynthesis, and redox cell signaling. Therefore, it is unsurprising that these molecules have a significant role in cancer metabolism and its metabolic plasticity. As one of the most prevalent malign diseases, colorectal cancer needs biomarkers for its early detection, prognostic, and prediction of response to therapy. However, the available biomarkers for this disease must be more powerful and present several drawbacks, such as high costs and complex laboratory procedures. Metabolomics has gathered substantial attention in the past two decades as a screening platform to study new metabolites, partly due to the development of techniques, such as mass spectrometry or liquid chromatography, which have become standard practice in diagnostic procedures for other diseases. Extensive metabolomic studies have been performed in colorectal cancer (CRC) patients in the past years, and several exciting results concerning amino acid metabolism have been found. This review aims to gather and present findings concerning alterations in the amino acid plasma pool of colorectal cancer patients.
Collapse
Affiliation(s)
- Marisa Domingues Santos
- Colorectal Unit, Hospital de Santo António, Centro Hospitalar Universitário de Santo António, 4050-651 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Ivo Barros
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
| | - Pedro Brandão
- Colorectal Unit, Hospital de Santo António, Centro Hospitalar Universitário de Santo António, 4050-651 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
| | - Lúcia Lacerda
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal; (I.B.); (L.L.)
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-313 Porto, Portugal
- Genetic Laboratory Service, Centro de Genética Médica Jacinto de Magalhães, Centro Hospitalar Universitário de Santo António, 4050-651 Porto, Portugal
| |
Collapse
|
10
|
Bull C, Hazelwood E, Bell JA, Tan V, Constantinescu AE, Borges C, Legge D, Burrows K, Huyghe JR, Brenner H, Castellvi-Bel S, Chan AT, Kweon SS, Le Marchand L, Li L, Cheng I, Pai RK, Figueiredo JC, Murphy N, Gunter MJ, Timpson NJ, Vincent EE. Identifying metabolic features of colorectal cancer liability using Mendelian randomization. eLife 2023; 12:RP87894. [PMID: 38127078 PMCID: PMC10735227 DOI: 10.7554/elife.87894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Background Recognizing the early signs of cancer risk is vital for informing prevention, early detection, and survival. Methods To investigate whether changes in circulating metabolites characterize the early stages of colorectal cancer (CRC) development, we examined the associations between a genetic risk score (GRS) associated with CRC liability (72 single-nucleotide polymorphisms) and 231 circulating metabolites measured by nuclear magnetic resonance spectroscopy in the Avon Longitudinal Study of Parents and Children (N = 6221). Linear regression models were applied to examine the associations between genetic liability to CRC and circulating metabolites measured in the same individuals at age 8 y, 16 y, 18 y, and 25 y. Results The GRS for CRC was associated with up to 28% of the circulating metabolites at FDR-P < 0.05 across all time points, particularly with higher fatty acids and very-low- and low-density lipoprotein subclass lipids. Two-sample reverse Mendelian randomization (MR) analyses investigating CRC liability (52,775 cases, 45,940 controls) and metabolites measured in a random subset of UK Biobank participants (N = 118,466, median age 58 y) revealed broadly consistent effect estimates with the GRS analysis. In conventional (forward) MR analyses, genetically predicted polyunsaturated fatty acid concentrations were most strongly associated with higher CRC risk. Conclusions These analyses suggest that higher genetic liability to CRC can cause early alterations in systemic metabolism and suggest that fatty acids may play an important role in CRC development. Funding This work was supported by the Elizabeth Blackwell Institute for Health Research, University of Bristol, the Wellcome Trust, the Medical Research Council, Diabetes UK, the University of Bristol NIHR Biomedical Research Centre, and Cancer Research UK. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. This work used the computational facilities of the Advanced Computing Research Centre, University of Bristol - http://www.bristol.ac.uk/acrc/.
Collapse
Affiliation(s)
- Caroline Bull
- MRC Integrative Epidemiology Unit at the University of BristolBristolUnited Kingdom
- Population Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
- Translational Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| | - Emma Hazelwood
- MRC Integrative Epidemiology Unit at the University of BristolBristolUnited Kingdom
- Population Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| | - Joshua A Bell
- MRC Integrative Epidemiology Unit at the University of BristolBristolUnited Kingdom
- Population Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| | - Vanessa Tan
- MRC Integrative Epidemiology Unit at the University of BristolBristolUnited Kingdom
- Population Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| | - Andrei-Emil Constantinescu
- MRC Integrative Epidemiology Unit at the University of BristolBristolUnited Kingdom
- Population Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| | - Carolina Borges
- MRC Integrative Epidemiology Unit at the University of BristolBristolUnited Kingdom
- Population Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| | - Danny Legge
- Translational Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| | - Kimberley Burrows
- MRC Integrative Epidemiology Unit at the University of BristolBristolUnited Kingdom
- Population Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| | - Jeroen R Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ)HeidelbergGermany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT)HeidelbergGermany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Sergi Castellvi-Bel
- Gastroenterology Department, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard UniversityBostonUnited States
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard UniversityBostonUnited States
| | - Sun-Seog Kweon
- Department of Preventive Medicine, Chonnam National University Medical SchoolGwangjuRepublic of Korea
- Jeonnam Regional Cancer Center, Chonnam National University Hwasun HospitalHwasunRepublic of Korea
| | | | - Li Li
- Department of Family Medicine, University of VirginiaCharlottesvilleUnited States
| | - Iona Cheng
- Department of Epidemiology and Biostatistics, University of California, San FranciscoSan FranciscoUnited States
- University of California, San Francisco Helen Diller Family Comprehensive Cancer Center, San FranciscoSan FranciscoUnited States
| | - Rish K Pai
- Department of Pathology and Laboratory Medicine, Mayo ClinicScottsdaleUnited States
| | - Jane C Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical CenterLos AngelesUnited States
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on CancerLyonFrance
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on CancerLyonFrance
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit at the University of BristolBristolUnited Kingdom
- Population Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| | - Emma E Vincent
- MRC Integrative Epidemiology Unit at the University of BristolBristolUnited Kingdom
- Population Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
- Translational Health Sciences, Bristol Medical School, University of BristolBristolUnited Kingdom
| |
Collapse
|
11
|
Yang Y, Wang Z, Li X, Lv J, Zhong R, Gao S, Zhang F, Chen W. Profiling the metabolic disorder and detection of colorectal cancer based on targeted amino acids metabolomics. J Transl Med 2023; 21:824. [PMID: 37978537 PMCID: PMC10655464 DOI: 10.1186/s12967-023-04604-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/06/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND The morbidity of cancer keeps growing worldwide, and among that, the colorectal cancer (CRC) has jumped to third. Existing early screening tests for CRC are limited. The aim of this study was to develop a diagnostic strategy for CRC by plasma metabolomics. METHODS A targeted amino acids metabolomics method was developed to quantify 32 plasma amino acids in 130 CRC patients and 216 healthy volunteers, to identify potential biomarkers for CRC, and an independent sample cohort comprising 116 CRC subjects, 33 precancerosiss patients and 195 healthy volunteers was further used to validate the diagnostic model. Amino acids-related genes were retrieved from Gene Expression Omnibus and Molecular Signatures Database and analyzed. RESULTS Three were chosen out of the 32 plasma amino acids examined. The tryptophan / sarcosine / glutamic acid -based receiver operating characteristic (ROC) curve showed the area under the curve (AUC) of 0.955 (specificity 83.3% and sensitivity 96.8%) for all participants, and the logistic regression model were used to distinguish between early stage (I and II) of CRC and precancerosiss patients, which showed superiority to the commonly used carcinoembryonic antigen. The GO and KEGG enrichment analysis proved many alterations in amino acids metabolic pathways in tumorigenesis. CONCLUSION This altered plasma amino acid profile could effectively distinguish CRC patients from precancerosiss patients and healthy volunteers with high accuracy. Prognostic tests based on the tryptophan/sarcosine/glutamic acid biomarkers in the large population could assess the clinical significance of CRC early detection and intervention.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
- Department of Pharmacy, the Affiliated Huaihai Hospital of Xuzhou Medical University / the 71st Group Army Hospital of CPLA Army, Xuzhou, 221004, Jiangsu, China
- Department of Laboratory Medicine, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Zhipeng Wang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Xinxing Li
- Department of General Surgery, Tongji Hospital, Tongji University, Shanghai, 200092, China
| | - Jianfeng Lv
- Department of Pharmacy, Taixing People's Hospital, Taixing, 225400, Jiangsu, China
| | - Renqian Zhong
- Department of Laboratory Medicine, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Shouhong Gao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| | - Feng Zhang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| | - Wansheng Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| |
Collapse
|
12
|
Bull CJ, Hazelwood E, Bell JA, Tan VY, Constantinescu AE, Borges MC, Legge DN, Burrows K, Huyghe JR, Brenner H, Castellví-Bel S, Chan AT, Kweon SS, Marchand LL, Li L, Cheng I, Pai RK, Figueiredo JC, Murphy N, Gunter MJ, Timpson NJ, Vincent EE. Identifying metabolic features of colorectal cancer liability using Mendelian randomization. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.10.23287084. [PMID: 36945480 PMCID: PMC10029059 DOI: 10.1101/2023.03.10.23287084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Background Recognizing the early signs of cancer risk is vital for informing prevention, early detection, and survival. Methods To investigate whether changes in circulating metabolites characterise the early stages of colorectal cancer (CRC) development, we examined associations between a genetic risk score (GRS) associated with CRC liability (72 single nucleotide polymorphisms) and 231 circulating metabolites measured by nuclear magnetic resonance spectroscopy in the Avon Longitudinal Study of Parents and Children (N=6,221). Linear regression models were applied to examine associations between genetic liability to colorectal cancer and circulating metabolites measured in the same individuals at age 8, 16, 18 and 25 years. Results The GRS for CRC was associated with up to 28% of the circulating metabolites at FDR-P<0.05 across all time points, particularly with higher fatty acids and very-low- and low-density lipoprotein subclass lipids. Two-sample reverse Mendelian randomization (MR) analyses investigating CRC liability (52,775 cases, 45,940 controls) and metabolites measured in a random subset of UK Biobank participants (N=118,466, median age 58y) revealed broadly consistent effect estimates with the GRS analysis. In conventional (forward) MR analyses, genetically predicted polyunsaturated fatty acid concentrations were most strongly associated with higher CRC risk. Conclusions These analyses suggest that higher genetic liability to CRC can cause early alterations in systemic metabolism, and suggest that fatty acids may play an important role in CRC development. Funding This work was supported by the Elizabeth Blackwell Institute for Health Research, University of Bristol, the Wellcome Trust, the Medical Research Council, Diabetes UK, the University of Bristol NIHR Biomedical Research Centre, and Cancer Research UK. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. This work used the computational facilities of the Advanced Computing Research Centre, University of Bristol - http://www.bristol.ac.uk/acrc/.
Collapse
Affiliation(s)
- Caroline J. Bull
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| | - Emma Hazelwood
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Joshua A. Bell
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Vanessa Y. Tan
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Andrei-Emil Constantinescu
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Danny N. Legge
- Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| | - Kimberly Burrows
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jeroen R. Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sergi Castellví-Bel
- Gastroenterology Department, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of Barcelona, Barcelona, Spain
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Sun-Seog Kweon
- Department of Preventive Medicine, Chonnam National University Medical School, Gwangju, Korea
- Jeonnam Regional Cancer Center, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | | | - Li Li
- Department of Family Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Iona Cheng
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
- University of California, San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, San Francisco, California, USA
| | - Rish K. Pai
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Arizona, Scottsdale, Arizona, USA
| | - Jane C. Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Emma E. Vincent
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Translational Health Sciences, Bristol Medical School, University of Bristol, UK
| |
Collapse
|
13
|
Watanabe K, Sato E, Mishima E, Moriya S, Sakabe T, Sato A, Fujiwara M, Fujimaru T, Ito Y, Taki F, Nagahama M, Tanaka K, Kazama JJ, Nakayama M. Changes in Metabolomic Profiles Induced by Switching from an Erythropoiesis-Stimulating Agent to a Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitor in Hemodialysis Patients: A Pilot Study. Int J Mol Sci 2023; 24:12752. [PMID: 37628932 PMCID: PMC10454178 DOI: 10.3390/ijms241612752] [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: 07/26/2023] [Revised: 08/07/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF-PHIs) are a new class of medications for managing renal anemia in patients with chronic kidney disease (CKD). In addition to their erythropoietic activity, HIF-PHIs exhibit multifaceted effects on iron and glucose metabolism, mitochondrial metabolism, and angiogenesis through the regulation of a wide range of HIF-responsive gene expressions. However, the systemic biological effects of HIF-PHIs in CKD patients have not been fully explored. In this prospective, single-center study, we comprehensively investigated changes in plasma metabolomic profiles following the switch from an erythropoiesis-stimulating agent (ESA) to an HIF-PHI, daprodustat, in 10 maintenance hemodialysis patients. Plasma metabolites were measured before and three months after the switch from an ESA to an HIF-PHI. Among 106 individual markers detected in plasma, significant changes were found in four compounds (erythrulose, n-butyrylglycine, threonine, and leucine), and notable but non-significant changes were found in another five compounds (inositol, phosphoric acid, lyxose, arabinose, and hydroxylamine). Pathway analysis indicated decreased levels of plasma metabolites, particularly those involved in phosphatidylinositol signaling, ascorbate and aldarate metabolism, and inositol phosphate metabolism. Our results provide detailed insights into the systemic biological effects of HIF-PHIs in hemodialysis patients and are expected to contribute to an evaluation of the potential side effects that may result from long-term use of this class of drugs.
Collapse
Affiliation(s)
- Kimio Watanabe
- Division of Nephrology and Hypertension, Fukushima Medical University, Fukushima 960-1295, Japan; (T.S.); (A.S.); (M.F.); (K.T.); (J.J.K.)
- Kidney Center, St Luke’s International Hospital, Tokyo 104-8560, Japan; (T.F.); (Y.I.); (F.T.); (M.N.); (M.N.)
| | - Emiko Sato
- Division of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan;
| | - Eikan Mishima
- Division of Nephrology, Rheumatology and Endocrinology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan;
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Shinobu Moriya
- Clinical Engineering Center, St Luke’s International Hospital, Tokyo 104-8560, Japan;
| | - Takuma Sakabe
- Division of Nephrology and Hypertension, Fukushima Medical University, Fukushima 960-1295, Japan; (T.S.); (A.S.); (M.F.); (K.T.); (J.J.K.)
| | - Atsuya Sato
- Division of Nephrology and Hypertension, Fukushima Medical University, Fukushima 960-1295, Japan; (T.S.); (A.S.); (M.F.); (K.T.); (J.J.K.)
| | - Momoko Fujiwara
- Division of Nephrology and Hypertension, Fukushima Medical University, Fukushima 960-1295, Japan; (T.S.); (A.S.); (M.F.); (K.T.); (J.J.K.)
| | - Takuya Fujimaru
- Kidney Center, St Luke’s International Hospital, Tokyo 104-8560, Japan; (T.F.); (Y.I.); (F.T.); (M.N.); (M.N.)
| | - Yugo Ito
- Kidney Center, St Luke’s International Hospital, Tokyo 104-8560, Japan; (T.F.); (Y.I.); (F.T.); (M.N.); (M.N.)
| | - Fumika Taki
- Kidney Center, St Luke’s International Hospital, Tokyo 104-8560, Japan; (T.F.); (Y.I.); (F.T.); (M.N.); (M.N.)
| | - Masahiko Nagahama
- Kidney Center, St Luke’s International Hospital, Tokyo 104-8560, Japan; (T.F.); (Y.I.); (F.T.); (M.N.); (M.N.)
| | - Kenichi Tanaka
- Division of Nephrology and Hypertension, Fukushima Medical University, Fukushima 960-1295, Japan; (T.S.); (A.S.); (M.F.); (K.T.); (J.J.K.)
| | - Junichiro James Kazama
- Division of Nephrology and Hypertension, Fukushima Medical University, Fukushima 960-1295, Japan; (T.S.); (A.S.); (M.F.); (K.T.); (J.J.K.)
| | - Masaaki Nakayama
- Kidney Center, St Luke’s International Hospital, Tokyo 104-8560, Japan; (T.F.); (Y.I.); (F.T.); (M.N.); (M.N.)
| |
Collapse
|
14
|
Ose J, Gigic B, Brezina S, Lin T, Peoples AR, Schobert PP, Baierl A, van Roekel E, Robinot N, Gicquiau A, Achaintre D, Scalbert A, van Duijnhoven FJB, Holowatyj AN, Gumpenberger T, Schrotz-King P, Ulrich AB, Ulvik A, Ueland PM, Weijenberg MP, Habermann N, Keski-Rahkonen P, Gsur A, Kok DE, Ulrich CM. Higher Plasma Creatinine Is Associated with an Increased Risk of Death in Patients with Non-Metastatic Rectal but Not Colon Cancer: Results from an International Cohort Consortium. Cancers (Basel) 2023; 15:3391. [PMID: 37444500 PMCID: PMC10340258 DOI: 10.3390/cancers15133391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Colorectal cancer (CRC) is increasingly recognized as a heterogeneous disease. No studies have prospectively examined associations of blood metabolite concentrations with all-cause mortality in patients with colon and rectal cancer separately. Targeted metabolomics (Biocrates AbsoluteIDQ p180) and pathway analyses (MetaboAnalyst 4.0) were performed on pre-surgery collected plasma from 674 patients with non-metastasized (stage I-III) colon (n = 394) or rectal cancer (n = 283). Metabolomics data and covariate information were received from the international cohort consortium MetaboCCC. Cox proportional hazards models were computed to investigate associations of 148 metabolite levels with all-cause mortality adjusted for age, sex, tumor stage, tumor site (whenever applicable), and cohort; the false discovery rate (FDR) was used to account for multiple testing. A total of 93 patients (14%) were deceased after an average follow-up time of 4.4 years (60 patients with colon cancer and 33 patients with rectal cancer). After FDR adjustment, higher plasma creatinine was associated with a 39% increase in all-cause mortality in patients with rectal cancer. HR: 1.39, 95% CI 1.23-1.72, pFDR = 0.03; but not colon cancer: pFDR = 0.96. Creatinine is a breakdown product of creatine phosphate in muscle and may reflect changes in skeletal muscle mass. The starch and sucrose metabolisms were associated with increased all-cause mortality in colon cancer but not in rectal cancer. Genes in the starch and sucrose metabolism pathways were previously linked to worse clinical outcomes in CRC. In summary, our findings support the hypothesis that colon and rectal cancer have different etiological and clinical outcomes that need to be considered for targeted treatments.
Collapse
Affiliation(s)
- Jennifer Ose
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, 69117 Heidelberg, Germany; (B.G.)
| | - Stefanie Brezina
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 23, 1090 Vienna, Austria; (S.B.)
| | - Tengda Lin
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Anita R. Peoples
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Pauline P. Schobert
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84112, USA
- School of Medicine, Ludwig-Maximilians University, 80539 Munich, Germany
- School of Medicine, Technical University of Munich, 80333 Munich, Germany
| | - Andreas Baierl
- Department of Statistics and Operations Research, University of Vienna, 1, 1010 Wien, Austria
| | - Eline van Roekel
- Department of Epidemiology, GROW-School of Oncology and Developmental Biology, Maastricht University, 30, 6229 Maastricht, The Netherlands
| | - Nivonirina Robinot
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, WHO, 69366 Lyon, France
| | - Audrey Gicquiau
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, WHO, 69366 Lyon, France
| | - David Achaintre
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, WHO, 69366 Lyon, France
| | - Augustin Scalbert
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, WHO, 69366 Lyon, France
| | | | - Andreana N. Holowatyj
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84112, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA
| | - Tanja Gumpenberger
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 23, 1090 Vienna, Austria; (S.B.)
| | - Petra Schrotz-King
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Alexis B. Ulrich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, 69117 Heidelberg, Germany; (B.G.)
- Klinik für Allgemein-, Viszeral-, Thorax- und Gefäßchirurgie, Städtische Kliniken Neuss, 84, 41464 Neuss, Germany
| | | | | | - Matty P. Weijenberg
- Department of Epidemiology, GROW-School of Oncology and Developmental Biology, Maastricht University, 30, 6229 Maastricht, The Netherlands
| | - Nina Habermann
- Genome Biology, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, WHO, 69366 Lyon, France
| | - Andrea Gsur
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 23, 1090 Vienna, Austria; (S.B.)
| | - Dieuwertje E. Kok
- Division of Human Nutrition and Health, Wageningen University & Research, 6708 Wageningen, The Netherlands
| | - Cornelia M. Ulrich
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
15
|
Yi Y, Wang J, Liang C, Ren C, Lian X, Han C, Sun W. LC-MS-based serum metabolomics analysis for the screening and monitoring of colorectal cancer. Front Oncol 2023; 13:1173424. [PMID: 37448516 PMCID: PMC10338013 DOI: 10.3389/fonc.2023.1173424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
Background Colorectal Cancer (CRC) is a prevalent digestive system tumour with significant mortality and recurrence rates. Serum metabolomics, with its high sensitivity and high throughput, has shown potential as a tool to discover biomarkers for clinical screening and monitoring of the CRC patients. Methods Serum metabolites of 61 sex and age-matched healthy controls and 62 CRC patients (before and after surgical intervention) were analyzed using a ultra-performance liquid chromatography-high resolution mass spectrometer (UPLC-MS). Statistical methods and pathway enrichment analysis were used to identify potential biomarkers and altered metabolic pathways. Results Our analysis revealed a clear distinction in the serum metabolic profile between CRC patients and healthy controls (HCs). Pathway analysis indicated a significant association with arginine biosynthesis, pyrimidine metabolism, pantothenate, and CoA biosynthesis. Univariate and multivariate statistical analysis showed that 9 metabolites had significant diagnostic value for CRC, among them, Guanosine with Area Under the Curve (AUC) values of 0.951 for the training group and0.998 for the validation group. Furthermore, analysis of four specific metabolites (N-Phenylacetylasparticacid, Tyrosyl-Gamma-glutamate, Tyr-Ser and Sphingosine) in serum samples of CRC patients before and after surgery indicated a return to healthy levels after an intervention. Conclusion Our results suggest that serum metabolomics may be a valuable tool for the screening and monitoring of CRC patients.
Collapse
Affiliation(s)
- Yanan Yi
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Jianjian Wang
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Chengtong Liang
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Chuanli Ren
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Xu Lian
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Chongxu Han
- Department of Laboratory Medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Wei Sun
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
2 Hydroxybutyric Acid-Producing Bacteria in Gut Microbiome and Fusobacterium nucleatum Regulates 2 Hydroxybutyric Acid Level In Vivo. Metabolites 2023; 13:metabo13030451. [PMID: 36984891 PMCID: PMC10059959 DOI: 10.3390/metabo13030451] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/03/2023] [Accepted: 03/16/2023] [Indexed: 03/22/2023] Open
Abstract
2-hydroxybutyric acid (2HB) serves as an important regulatory factor in a variety of diseases. The circulating level of 2HB in serum is significantly higher in multiple diseases, such as cancer and type 2 diabetes (T2D). However, there is currently no systematic study on 2HB-producing bacteria that demonstrates whether gut bacteria contribute to the circulating 2HB pool. To address this question, we used BLASTP to reveal the taxonomic profiling of 2HB-producing bacteria in the human microbiome, which are mainly distributed in the phylum Proteobacteria and Firmicutes. In vitro experiments showed that most gut bacteria (21/32) have at least one path to produce 2HB, which includes Aspartic acid, methionine, threonine, and 2-aminobutyric acid. Particularly, Fusobacterium nucleatum has the strongest ability to synthesize 2HB, which is sufficient to alter colon 2HB concentration in mice. Nevertheless, neither antibiotic (ABX) nor Fusobacterium nucleatum gavage significantly affected mouse serum 2HB levels during the time course of this study. Taken together, our study presents the profiles of 2HB-producing bacteria and demonstrates that gut microbiota was a major contributor to 2HB concentration in the intestinal lumen but a relatively minor contributor to serum 2HB concentration.
Collapse
|
17
|
Akiyama M, Akiyama T, Saigusa D, Hishinuma E, Matsukawa N, Shibata T, Tsuchiya H, Mori A, Fujii Y, Mogami Y, Tokorodani C, Kuwahara K, Numata-Uematsu Y, Inoue K, Kobayashi K. Comprehensive study of metabolic changes induced by a ketogenic diet therapy using GC/MS- and LC/MS-based metabolomics. Seizure 2023; 107:52-59. [PMID: 36958064 DOI: 10.1016/j.seizure.2023.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/17/2023] Open
Abstract
OBJECTIVE The ketogenic diet (KD), a high-fat and low-carbohydrate diet, is effective for a subset of patients with drug-resistant epilepsy, although the mechanisms of the KD have not been fully elucidated. The aims of this observational study were to investigate comprehensive short-term metabolic changes induced by the KD and to explore candidate metabolites or pathways for potential new therapeutic targets. METHODS Subjects included patients with intractable epilepsy who had undergone the KD therapy (the medium-chain triglyceride [MCT] KD or the modified Atkins diet using MCT oil). Plasma and urine samples were obtained before and at 2-4 weeks after initiation of the KD. Targeted metabolome analyses of these samples were performed using gas chromatography-tandem mass spectrometry (GC/MS/MS) and liquid chromatography-tandem mass spectrometry (LC/MS/MS). RESULTS Samples from 10 and 11 patients were analysed using GC/MS/MS and LC/MS/MS, respectively. The KD increased ketone bodies, various fatty acids, lipids, and their conjugates. In addition, levels of metabolites located upstream of acetyl-CoA and propionyl-CoA, including catabolites of branched-chain amino acids and structural analogues of γ-aminobutyric acid and lactic acid, were elevated. CONCLUSIONS The metabolites that were significantly changed after the initiation of the KD and related metabolites may be candidates for further studies for neuronal actions to develop new anti-seizure medications.
Collapse
Affiliation(s)
- Mari Akiyama
- Department of Child Neurology, Okayama University Hospital, Okayama, Japan
| | - Tomoyuki Akiyama
- Department of Paediatrics (Child Neurology), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | - Daisuke Saigusa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Eiji Hishinuma
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Advanced Research Centre for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Japan
| | - Naomi Matsukawa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Takashi Shibata
- Department of Child Neurology, Okayama University Hospital, Okayama, Japan
| | - Hiroki Tsuchiya
- Department of Child Neurology, Okayama University Hospital, Okayama, Japan
| | - Atsushi Mori
- Department of Neurology, Shiga Medical Centre for Children, Moriyama, Japan
| | - Yuji Fujii
- Department of Paediatrics, Hiroshima City Funairi Citizens Hospital, Hiroshima, Japan
| | - Yukiko Mogami
- Department of Paediatric Neurology, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Chiho Tokorodani
- Department of Paediatrics, Kochi Health Sciences Centre, Kochi, Japan
| | - Kozue Kuwahara
- Department of Paediatrics, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | | | - Kenji Inoue
- Department of Neurology, Shiga Medical Centre for Children, Moriyama, Japan
| | - Katsuhiro Kobayashi
- Department of Paediatrics (Child Neurology), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
18
|
Rothwell JA, Bešević J, Dimou N, Breeur M, Murphy N, Jenab M, Wedekind R, Viallon V, Ferrari P, Achaintre D, Gicquiau A, Rinaldi S, Scalbert A, Huybrechts I, Prehn C, Adamski J, Cross AJ, Keun H, Chadeau-Hyam M, Boutron-Ruault MC, Overvad K, Dahm CC, Nøst TH, Sandanger TM, Skeie G, Zamora-Ros R, Tsilidis KK, Eichelmann F, Schulze MB, van Guelpen B, Vidman L, Sánchez MJ, Amiano P, Ardanaz E, Smith-Byrne K, Travis R, Katzke V, Kaaks R, Derksen JWG, Colorado-Yohar S, Tumino R, Bueno-de-Mesquita B, Vineis P, Palli D, Pasanisi F, Eriksen AK, Tjønneland A, Severi G, Gunter MJ. Circulating amino acid levels and colorectal cancer risk in the European Prospective Investigation into Cancer and Nutrition and UK Biobank cohorts. BMC Med 2023; 21:80. [PMID: 36855092 PMCID: PMC9976469 DOI: 10.1186/s12916-023-02739-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/16/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Amino acid metabolism is dysregulated in colorectal cancer patients; however, it is not clear whether pre-diagnostic levels of amino acids are associated with subsequent risk of colorectal cancer. We investigated circulating levels of amino acids in relation to colorectal cancer risk in the European Prospective Investigation into Cancer and Nutrition (EPIC) and UK Biobank cohorts. METHODS Concentrations of 13-21 amino acids were determined in baseline fasting plasma or serum samples in 654 incident colorectal cancer cases and 654 matched controls in EPIC. Amino acids associated with colorectal cancer risk following adjustment for the false discovery rate (FDR) were then tested for associations in the UK Biobank, for which measurements of 9 amino acids were available in 111,323 participants, of which 1221 were incident colorectal cancer cases. RESULTS Histidine levels were inversely associated with colorectal cancer risk in EPIC (odds ratio [OR] 0.80 per standard deviation [SD], 95% confidence interval [CI] 0.69-0.92, FDR P-value=0.03) and in UK Biobank (HR 0.93 per SD, 95% CI 0.87-0.99, P-value=0.03). Glutamine levels were borderline inversely associated with colorectal cancer risk in EPIC (OR 0.85 per SD, 95% CI 0.75-0.97, FDR P-value=0.08) and similarly in UK Biobank (HR 0.95, 95% CI 0.89-1.01, P=0.09) In both cohorts, associations changed only minimally when cases diagnosed within 2 or 5 years of follow-up were excluded. CONCLUSIONS Higher circulating levels of histidine were associated with a lower risk of colorectal cancer in two large prospective cohorts. Further research to ascertain the role of histidine metabolism and potentially that of glutamine in colorectal cancer development is warranted.
Collapse
Affiliation(s)
- Joseph A Rothwell
- Centre for Epidemiology and Population Health (Inserm U1018), Exposome and Heredity team, Faculté de Médecine, Université Paris-Saclay, UVSQ, Gustave Roussy, F-94805, Villejuif, France.
| | - Jelena Bešević
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Niki Dimou
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Marie Breeur
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Neil Murphy
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Mazda Jenab
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Roland Wedekind
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Vivian Viallon
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Pietro Ferrari
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - David Achaintre
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Audrey Gicquiau
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Sabina Rinaldi
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Augustin Scalbert
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Inge Huybrechts
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Cornelia Prehn
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Jerzy Adamski
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore, 117597, Singapore
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - Amanda J Cross
- School of Public Health, Imperial College London, London, UK
| | - Hector Keun
- Department of Surgery & Cancer, Imperial College London, London, UK
| | | | - Marie-Christine Boutron-Ruault
- Centre for Epidemiology and Population Health (Inserm U1018), Exposome and Heredity team, Faculté de Médecine, Université Paris-Saclay, UVSQ, Gustave Roussy, F-94805, Villejuif, France
| | - Kim Overvad
- Department of Public Health, Aarhus University, Bartholins Allé 2, DK-8000, Aarhus, Denmark
| | - Christina C Dahm
- Department of Public Health, Aarhus University, Bartholins Allé 2, DK-8000, Aarhus, Denmark
| | - Therese Haugdahl Nøst
- Faculty of Health Sciences, Department of Community Medicine, UiT the Arctic University of Norway, N-9037, Tromsø, Norway
| | - Torkjel M Sandanger
- Faculty of Health Sciences, Department of Community Medicine, UiT the Arctic University of Norway, N-9037, Tromsø, Norway
| | - Guri Skeie
- Faculty of Health Sciences, Department of Community Medicine, UiT the Arctic University of Norway, N-9037, Tromsø, Norway
| | - Raul Zamora-Ros
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Kostas K Tsilidis
- School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Fabian Eichelmann
- German Center for Diabetes Research (DZD), Munchen-Neuherberg, Germany
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Bethany van Guelpen
- Department of Radiation Sciences, Oncology Unit, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Linda Vidman
- Department of Radiation Sciences, Oncology Unit, Umeå University, Umeå, Sweden
| | - Maria-José Sánchez
- Escuela Andaluza de Salud Pública (EASP), 18011, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012, Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029, Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, 18071, Granada, Spain
| | - Pilar Amiano
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, San Sebastián, Spain
- Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, San Sebastián, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Eva Ardanaz
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029, Madrid, Spain
- Navarra Public Health Institute, Leyre 15, 31003, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Karl Smith-Byrne
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
| | - Ruth Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Verena Katzke
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Rudolf Kaaks
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Jeroen W G Derksen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sandra Colorado-Yohar
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029, Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
- Research Group on Demography and Health, National Faculty of Public Health, University of Antioquia, Medellín, Colombia
| | - Rosario Tumino
- Cancer Registry and Histopathology Department, Provincial Health Authority (ASP), Ragusa, Italy
| | - Bas Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720, BA, Bilthoven, The Netherlands
| | - Paolo Vineis
- School of Public Health, Imperial College London, London, UK
- Italian Institute of Technology, Genova, Italy
| | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network - ISPRO, Florence, Italy
| | - Fabrizio Pasanisi
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Anne Kirstine Eriksen
- Danish Cancer Society Research Center, Diet, Genes and Environment, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Diet, Genes and Environment, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Gianluca Severi
- Centre for Epidemiology and Population Health (Inserm U1018), Exposome and Heredity team, Faculté de Médecine, Université Paris-Saclay, UVSQ, Gustave Roussy, F-94805, Villejuif, France
- Department of Statistics, Computer Science, Applications "G. Parenti" University of Florence, Florence, Italy
| | - Marc J Gunter
- International Agency for Research on Cancer (IARC), 150 cours Albert Thomas, 69008, Lyon, France
- School of Public Health, Imperial College London, London, UK
| |
Collapse
|
19
|
Shekher A, Puneet, Awasthee N, Kumar U, Raj R, Kumar D, Gupta SC. Association of altered metabolic profiles and long non-coding RNAs expression with disease severity in breast cancer patients: analysis by 1H NMR spectroscopy and RT-q-PCR. Metabolomics 2023; 19:8. [PMID: 36710275 DOI: 10.1007/s11306-023-01972-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Globally, one of the major causes of cancer related deaths in women is breast cancer. Although metabolic pattern is altered in cancer patients, robust metabolic biomarkers with a potential to improve the screening and disease monitoring are lacking. A complete metabolome profiling of breast cancer patients may lead to the identification of diagnostic/prognostic markers and potential targets. OBJECTIVES The aim of this study was to analyze the metabolic profile in the serum from 43 breast cancer patients and 13 healthy individuals. MATERIALS & METHODS We used 1H NMR spectroscopy for the identification and quantification of metabolites. q-RT-PCR was used to examine the relative expression of lncRNAs. RESULTS Metabolites such as amino acids, lipids, membrane metabolites, lipoproteins, and energy metabolites were observed in the serum from both patients and healthy individuals. Using unsupervised PCA, supervised PLS-DA, supervised OPLS-DA, and random forest classification, we observed that more than 25 metabolites were altered in the breast cancer patients. Metabolites with AUC value > 0.9 were selected for further analysis that revealed significant elevation of lactate, LPR and glycerol, while the level of glucose, succinate, and isobutyrate was reduced in breast cancer patients in comparison to healthy control. The level of these metabolites (except LPR) was altered in advanced-stage breast cancer patients in comparison to early-stage breast cancer patients. The altered metabolites were also associated with over 25 signaling pathways related to metabolism. Further, lncRNAs such as H19, MEG3 and GAS5 were dysregulated in the breast tumor tissue in comparison to normal adjacent tissue. CONCLUSION The study provides insights into metabolic alteration in breast cancer patients. It also provides an avenue to examine the association of lncRNAs with metabolic patterns in patients.
Collapse
Affiliation(s)
- Anusmita Shekher
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221 005, India
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221 005, India
| | - Puneet
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221 005, India
| | - Nikee Awasthee
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221 005, India
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Umesh Kumar
- Centre of Biomedical Research (CBMR), SGPGIMS, Lucknow, Uttar Pradesh, 226 014, India
| | - Ritu Raj
- Centre of Biomedical Research (CBMR), SGPGIMS, Lucknow, Uttar Pradesh, 226 014, India
| | - Dinesh Kumar
- Centre of Biomedical Research (CBMR), SGPGIMS, Lucknow, Uttar Pradesh, 226 014, India.
| | - Subash Chandra Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221 005, India.
- Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, Assam, 781101, India.
| |
Collapse
|
20
|
Costantini S, Di Gennaro E, Capone F, De Stefano A, Nasti G, Vitagliano C, Setola SV, Tatangelo F, Delrio P, Izzo F, Avallone A, Budillon A. Plasma metabolomics, lipidomics and cytokinomics profiling predict disease recurrence in metastatic colorectal cancer patients undergoing liver resection. Front Oncol 2023; 12:1110104. [PMID: 36713567 PMCID: PMC9875807 DOI: 10.3389/fonc.2022.1110104] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Purpose In metastatic colorectal cancer (mCRC) patients (pts), treatment strategies integrating liver resection with induction chemotherapy offer better 5-year survival rates than chemotherapy alone. However, liver resection is a complex and costly procedure, and recurrence occurs in almost 2/3rds of pts, suggesting the need to identify those at higher risk. The aim of this work was to evaluate whether the integration of plasma metabolomics and lipidomics combined with the multiplex analysis of a large panel of plasma cytokines can be used to predict the risk of relapse and other patient outcomes after liver surgery, beyond or in combination with clinical morphovolumetric criteria. Experimental design Peripheral blood metabolomics and lipidomics were performed by 600 MHz NMR spectroscopy on plasma from 30 unresectable mCRC pts treated with bevacizumab plus oxaliplatin-based regimens within the Obelics trial (NCT01718873) and subdivided into responder (R) and non-R (NR) according to 1-year disease-free survival (DFS): ≥ 1-year (R, n = 12) and < 1-year (NR, n = 18). A large panel of cytokines, chemokines, and growth factors was evaluated on the same plasma using Luminex xMAP-based multiplex bead-based immunoassay technology. A multiple biomarkers model was built using a support vector machine (SVM) classifier. Results Sparse partial least squares discriminant analysis (sPLS-DA) and loading plots obtained by analyzing metabolomics profiles of samples collected at the time of response evaluation when resectability was established showed significantly different levels of metabolites between the two groups. Two metabolites, 3-hydroxybutyrate and histidine, significantly predicted DFS and overall survival. Lipidomics analysis confirmed clear differences between the R and NR pts, indicating a statistically significant increase in lipids (cholesterol, triglycerides and phospholipids) in NR pts, reflecting a nonspecific inflammatory response. Indeed, a significant increase in proinflammatory cytokines was demonstrated in NR pts plasma. Finally, a multiple biomarkers model based on the combination of presurgery plasma levels of 3-hydroxybutyrate, cholesterol, phospholipids, triglycerides and IL-6 was able to correctly classify patients by their DFS with good accuracy. Conclusion Overall, this exploratory study suggests the potential of these combined biomarker approaches to predict outcomes in mCRC patients who are candidates for liver metastasis resection after induction treatment for defining personalized management and treatment strategies.
Collapse
Affiliation(s)
- Susan Costantini
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Francesca Capone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Alfonso De Stefano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Guglielmo Nasti
- Innovative Therapy for Abdominal Metastases Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Carlo Vitagliano
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Sergio Venanzio Setola
- Radiology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Fabiana Tatangelo
- Pathology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Paolo Delrio
- Colorectal Oncological Surgery Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Francesco Izzo
- Hepatobiliary Surgery Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy,*Correspondence: Alfredo Budillon,
| |
Collapse
|
21
|
Relationship between 4-Hydroxynonenal (4-HNE) as Systemic Biomarker of Lipid Peroxidation and Metabolomic Profiling of Patients with Prostate Cancer. Biomolecules 2023; 13:biom13010145. [PMID: 36671530 PMCID: PMC9855859 DOI: 10.3390/biom13010145] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
An oxidative degradation product of the polyunsaturated fatty acids, 4-hydroxynonenal (4-HNE), is of particular interest in cancer research due to its concentration-dependent pleiotropic activities affecting cellular antioxidants, metabolism, and growth control. Although an increase in oxidative stress and lipid peroxidation was already associated with prostate cancer progression a few decades ago, the knowledge of the involvement of 4-HNE in prostate cancer tumorigenesis is limited. This study investigated the appearance of 4-HNE-protein adducts in prostate cancer tissue by immunohistochemistry using a genuine 4-HNE monoclonal antibody. Plasma samples of the same patients and samples of the healthy controls were also analyzed for the presence of 4-HNE-protein adducts, followed by metabolic profiling using LC-ESI-QTOF-MS and GC-EI-Q-MS. Finally, the analysis of the metabolic pathways affected by 4-HNE was performed. The obtained results revealed the absence of 4-HNE-protein adducts in prostate carcinoma tissue but increased 4-HNE-protein levels in the plasma of these patients. Metabolomics revealed a positive association of different long-chain and medium-chain fatty acids with the presence of prostate cancer. Furthermore, while linoleic acid positively correlated with the levels of 4-HNE-protein adducts in the blood of healthy men, no correlation was obtained for cancer patients indicating altered lipid metabolism in this case. The metabolic pathway of unsaturated fatty acids biosynthesis emerged as significantly affected by 4-HNE. Overall, this is the first study linking 4-HNE adduction to plasma proteins with specific alterations in the plasma metabolome of prostate cancer patients. This study revealed that increased 4-HNE plasma protein adducts could modulate the unsaturated fatty acids biosynthesis pathway. It is yet to be determined if this is a direct result of 4-HNE or whether they are produced by the same underlying mechanisms. Further mechanistic studies are needed to grasp the biological significance of the observed changes in prostate cancer tumorigenesis.
Collapse
|
22
|
Systematic Review: Contribution of the Gut Microbiome to the Volatile Metabolic Fingerprint of Colorectal Neoplasia. Metabolites 2022; 13:metabo13010055. [PMID: 36676980 PMCID: PMC9865897 DOI: 10.3390/metabo13010055] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Colorectal cancer (CRC) has been associated with changes in volatile metabolic profiles in several human biological matrices. This enables its non-invasive detection, but the origin of these volatile organic compounds (VOCs) and their relation to the gut microbiome are not yet fully understood. This systematic review provides an overview of the current understanding of this topic. A systematic search using PubMed, Embase, Medline, Cochrane Library, and the Web of Science according to PRISMA guidelines resulted in seventy-one included studies. In addition, a systematic search was conducted that identified five systematic reviews from which CRC-associated gut microbiota data were extracted. The included studies analyzed VOCs in feces, urine, breath, blood, tissue, and saliva. Eight studies performed microbiota analysis in addition to VOC analysis. The most frequently reported dysregulations over all matrices included short-chain fatty acids, amino acids, proteolytic fermentation products, and products related to the tricarboxylic acid cycle and Warburg metabolism. Many of these dysregulations could be related to the shifts in CRC-associated microbiota, and thus the gut microbiota presumably contributes to the metabolic fingerprint of VOC in CRC. Future research involving VOCs analysis should include simultaneous gut microbiota analysis.
Collapse
|
23
|
Yu Z, Matsukawa N, Saigusa D, Motoike IN, Ono C, Okamura Y, Onuma T, Takahashi Y, Sakai M, Kudo H, Obara T, Murakami K, Shirota M, Kikuchi S, Kobayashi N, Kikuchi Y, Sugawara J, Minegishi N, Ogishima S, Kinoshita K, Yamamoto M, Yaegashi N, Kuriyama S, Koshiba S, Tomita H. Plasma metabolic disturbances during pregnancy and postpartum in women with depression. iScience 2022; 25:105666. [PMID: 36505921 PMCID: PMC9732390 DOI: 10.1016/j.isci.2022.105666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/17/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Examining plasma metabolic profiling during pregnancy and postpartum could help clinicians understand the risk factors for postpartum depression (PPD) development. This analysis targeted paired plasma metabolites in mid-late gestational and 1 month postpartum periods in women with (n = 209) or without (n = 222) PPD. Gas chromatogram-mass spectrometry was used to analyze plasma metabolites at these two time points. Among the 170 objected plasma metabolites, principal component analysis distinguished pregnancy and postpartum metabolites but failed to discriminate women with and without PPD. Compared to women without PPD, those with PPD exhibited 37 metabolites with disparate changes during pregnancy and the 1-month postpartum period and an enriched citrate cycle. Machine learning and multivariate statistical analysis identified two or three compounds that could be potential biomarkers for PPD prediction during pregnancy. Our findings suggest metabolic disturbances in women with depression and may help to elucidate metabolic processes associated with PPD development.
Collapse
Affiliation(s)
- Zhiqian Yu
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan,Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Corresponding author
| | - Naomi Matsukawa
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University
| | - Ikuko N. Motoike
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Department of System Bioinformatics, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Chiaki Ono
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yasunobu Okamura
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Innovations in Next-Generation Medicine, Advanced Research Center, Tohoku University, Sendai, Japan
| | - Tomomi Onuma
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Yuta Takahashi
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Mai Sakai
- Department of Disaster Psychiatry, International Research Institute for Disaster Science, Tohoku University, Sendai, Japan
| | - Hisaaki Kudo
- Department of Biobank Life Science, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Taku Obara
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Keiko Murakami
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Matusyuki Shirota
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Saya Kikuchi
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Natsuko Kobayashi
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yoshie Kikuchi
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Junichi Sugawara
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Naoko Minegishi
- Department of Biobank Life Science, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Soichi Ogishima
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Kengo Kinoshita
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Department of System Bioinformatics, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuo Yaegashi
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Department of Gynecology and Obstetrics, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Shinichi Kuriyama
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Division of Disaster Public Health, International Research Institute for Disaster Science, Tohoku University, Sendai, Japan
| | - Seizo Koshiba
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Hiroaki Tomita
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan,Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan,Department of Disaster Psychiatry, International Research Institute for Disaster Science, Tohoku University, Sendai, Japan
| |
Collapse
|
24
|
A Comprehensive Mass Spectrometry-Based Workflow for Clinical Metabolomics Cohort Studies. Metabolites 2022; 12:metabo12121168. [PMID: 36557207 PMCID: PMC9782571 DOI: 10.3390/metabo12121168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022] Open
Abstract
As a comprehensive analysis of all metabolites in a biological system, metabolomics is being widely applied in various clinical/health areas for disease prediction, diagnosis, and prognosis. However, challenges remain in dealing with the metabolomic complexity, massive data, metabolite identification, intra- and inter-individual variation, and reproducibility, which largely limit its widespread implementation. This study provided a comprehensive workflow for clinical metabolomics, including sample collection and preparation, mass spectrometry (MS) data acquisition, and data processing and analysis. Sample collection from multiple clinical sites was strictly carried out with standardized operation procedures (SOP). During data acquisition, three types of quality control (QC) samples were set for respective MS platforms (GC-MS, LC-MS polar, and LC-MS lipid) to assess the MS performance, facilitate metabolite identification, and eliminate contamination. Compounds annotation and identification were implemented with commercial software and in-house-developed PAppLineTM and UlibMS library. The batch effects were removed using a deep learning model method (NormAE). Potential biomarkers identification was performed with tree-based modeling algorithms including random forest, AdaBoost, and XGBoost. The modeling performance was evaluated using the F1 score based on a 10-times repeated trial for each. Finally, a sub-cohort case study validated the reliability of the entire workflow.
Collapse
|
25
|
Hu L, Brito LF, Zhang H, Zhao M, Liu H, Chai H, Wang D, Wu H, Cui J, Liu A, Xu Q, Wang Y. Metabolome profiling of plasma reveals different metabolic responses to acute cold challenge between Inner-Mongolia Sanhe and Holstein cattle. J Dairy Sci 2022; 105:9162-9178. [PMID: 36175226 DOI: 10.3168/jds.2022-21996] [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: 02/21/2022] [Accepted: 06/27/2022] [Indexed: 11/19/2022]
Abstract
Low-temperature conditions influence cattle productivity and survivability. Understanding the metabolic regulations of specific cattle breeds and identifying potential biomarkers related to cold challenges are important for cattle management and optimization of genetic improvement programs. In this study, 28 Inner-Mongolia Sanhe and 22 Holstein heifers were exposed to -25°C for 1 h to evaluate the differences in metabolic mechanisms of thermoregulation. In response to this acute cold challenge, altered rectal temperature was only observed in Holstein cattle. Further metabolome analyses showed a greater baseline of glycolytic activity and mobilization of AA in Sanhe cattle during normal conditions. Both breeds responded to the acute cold challenge by altering their metabolism of volatile fatty acids and AA for gluconeogenesis, which resulted in increased glucose levels. Furthermore, Sanhe cattle mobilized the citric acid cycle activity, and creatine and creatine phosphate metabolism to supply energy, whereas Holstein cattle used greater AA metabolism for this purpose. Altogether, we found that propionate and methanol are potential biomarkers of acute cold challenge response in cattle. Our findings provide novel insights into the biological mechanisms of acute cold response and climatic resilience, and will be used as the basis when developing breeding tools for genetically selecting for improved cold adaptation in cattle.
Collapse
Affiliation(s)
- Lirong Hu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, National Engineering Laboratory for Animal Breeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Haidian District, Beijing, 100193, China; College of Life Sciences and Bioengineering, Beijing Jiaotong University, Haidian District, Beijing, 100044, China; Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Hailiang Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, National Engineering Laboratory for Animal Breeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Man Zhao
- Xiertala Cattle Breeding Farm, Hailaer Farm Buro, Hailaer, Inner Mongolia, 021012, China
| | - Huazhu Liu
- Xiertala Cattle Breeding Farm, Hailaer Farm Buro, Hailaer, Inner Mongolia, 021012, China
| | - He Chai
- Xiertala Cattle Breeding Farm, Hailaer Farm Buro, Hailaer, Inner Mongolia, 021012, China
| | - Dongsheng Wang
- Xiertala Cattle Breeding Farm, Hailaer Farm Buro, Hailaer, Inner Mongolia, 021012, China
| | - Hongjun Wu
- Xiertala Cattle Breeding Farm, Hailaer Farm Buro, Hailaer, Inner Mongolia, 021012, China
| | - Jiuhui Cui
- Xiertala Cattle Breeding Farm, Hailaer Farm Buro, Hailaer, Inner Mongolia, 021012, China
| | - Airong Liu
- Xiertala Cattle Breeding Farm, Hailaer Farm Buro, Hailaer, Inner Mongolia, 021012, China
| | - Qing Xu
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Haidian District, Beijing, 100044, China.
| | - Yachun Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, National Engineering Laboratory for Animal Breeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Haidian District, Beijing, 100193, China.
| |
Collapse
|
26
|
Chen F, Dai X, Zhou CC, Li KX, Zhang YJ, Lou XY, Zhu YM, Sun YL, Peng BX, Cui W. Integrated analysis of the faecal metagenome and serum metabolome reveals the role of gut microbiome-associated metabolites in the detection of colorectal cancer and adenoma. Gut 2022; 71:1315-1325. [PMID: 34462336 PMCID: PMC9185821 DOI: 10.1136/gutjnl-2020-323476] [Citation(s) in RCA: 160] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 08/12/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To profile gut microbiome-associated metabolites in serum and investigate whether these metabolites could distinguish individuals with colorectal cancer (CRC) or adenoma from normal healthy individuals. DESIGN Integrated analysis of untargeted serum metabolomics by liquid chromatography-mass spectrometry and metagenome sequencing of paired faecal samples was applied to identify gut microbiome-associated metabolites with significantly altered abundance in patients with CRC and adenoma. The ability of these metabolites to discriminate between CRC and colorectal adenoma was tested by targeted metabolomic analysis. A model based on gut microbiome-associated metabolites was established and evaluated in an independent validation cohort. RESULTS In total, 885 serum metabolites were significantly altered in both CRC and adenoma, including eight gut microbiome-associated serum metabolites (GMSM panel) that were reproducibly detected by both targeted and untargeted metabolomics analysis and accurately discriminated CRC and adenoma from normal samples. A GMSM panel-based model to predict CRC and colorectal adenoma yielded an area under the curve (AUC) of 0.98 (95% CI 0.94 to 1.00) in the modelling cohort and an AUC of 0.92 (83.5% sensitivity, 84.9% specificity) in the validation cohort. The GMSM model was significantly superior to the clinical marker carcinoembryonic antigen among samples within the validation cohort (AUC 0.92 vs 0.72) and also showed promising diagnostic accuracy for adenomas (AUC=0.84) and early-stage CRC (AUC=0.93). CONCLUSION Gut microbiome reprogramming in patients with CRC is associated with alterations of the serum metabolome, and GMSMs have potential applications for CRC and adenoma detection.
Collapse
Affiliation(s)
- Feng Chen
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xudong Dai
- Dept of Clinical Research, Precogify Pharmaceutical Co, Ltd, Beijing, China
| | - Chang-Chun Zhou
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ke-Xin Li
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu-Juan Zhang
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiao-Ying Lou
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuan-Min Zhu
- Department of Gastroenterology, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Yan-Lai Sun
- Department of Gastrointestinal Cancer Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Bao-Xiang Peng
- Clinical Laboratory, Linyi Cancer Hospital, Linyi, China
| | - Wei Cui
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
27
|
Telleria O, Alboniga OE, Clos-Garcia M, Nafría-Jimenez B, Cubiella J, Bujanda L, Falcón-Pérez JM. A Comprehensive Metabolomics Analysis of Fecal Samples from Advanced Adenoma and Colorectal Cancer Patients. Metabolites 2022; 12:550. [PMID: 35736483 PMCID: PMC9229737 DOI: 10.3390/metabo12060550] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 12/14/2022] Open
Abstract
Accurate diagnosis of colorectal cancer (CRC) still relies on invasive colonoscopy. Noninvasive methods are less sensitive in detecting the disease, particularly in the early stage. In the current work, a metabolomics analysis of fecal samples was carried out by ultra-high-performance liquid chromatography-tandem mass spectroscopy (UPLC-MS/MS). A total of 1380 metabolites were analyzed in a cohort of 120 fecal samples from patients with normal colonoscopy, advanced adenoma (AA) and CRC. Multivariate analysis revealed that metabolic profiles of CRC and AA patients were similar and could be clearly separated from control individuals. Among the 25 significant metabolites, sphingomyelins (SM), lactosylceramides (LacCer), secondary bile acids, polypeptides, formiminoglutamate, heme and cytidine-containing pyrimidines were found to be dysregulated in CRC patients. Supervised random forest (RF) and logistic regression algorithms were employed to build a CRC accurate predicted model consisting of the combination of hemoglobin (Hgb) and bilirubin E,E, lactosyl-N-palmitoyl-sphingosine, glycocholenate sulfate and STLVT with an accuracy, sensitivity and specificity of 91.67% (95% Confidence Interval (CI) 0.7753-0.9825), 0.7 and 1, respectively.
Collapse
Affiliation(s)
- Oiana Telleria
- Exosomes Laboratory, CIC bioGUNE-BRTA, CIBERehd, Bizkaia Technology Park, 48160 Bilbao, Spain
| | - Oihane E. Alboniga
- Metabolomics Platform, CIC bioGUNE-BRTA, CIBERehd, Bizkaia Technology Park, 48160 Bilbao, Spain;
| | - Marc Clos-Garcia
- LEITAT Technological Center, C/de la Innovació, 2, 08225 Terrassa, Spain;
| | - Beatriz Nafría-Jimenez
- Osakidetza Basque Health Service, Donostialdea Integrated Health Organisation, Clinical Biochemistry Department, 20014 San Sebastian, Spain;
| | - Joaquin Cubiella
- Department of Gastroenterology, Complexo Hospitalario Universitario de Ourense, Instituto de Investigación Sanitaria Galicia Sur, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 32005 Ourense, Spain;
| | - Luis Bujanda
- Department of Gastroenterology, Hospital Donostia/Instituto Biodonostia, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco (UPV/EHU), 20014 San Sebastián, Spain;
| | - Juan Manuel Falcón-Pérez
- Exosomes Laboratory, CIC bioGUNE-BRTA, CIBERehd, Bizkaia Technology Park, 48160 Bilbao, Spain
- Metabolomics Platform, CIC bioGUNE-BRTA, CIBERehd, Bizkaia Technology Park, 48160 Bilbao, Spain;
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| |
Collapse
|
28
|
Nava GM, Madrigal Perez LA. Metabolic profile of the Warburg effect as a tool for molecular prognosis and diagnosis of cancer. Expert Rev Mol Diagn 2022; 22:439-447. [PMID: 35395916 DOI: 10.1080/14737159.2022.2065196] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Adaptations of eukaryotic cells to environmental changes are important for their survival. However, under some circumstances, microenvironmental changes promote that eukaryotic cells utilize a metabolic signature resembling a unicellular organism named the Warburg effect. Most cancer cells share the Warburg effect displaying lactic fermentation and high glucose uptake. The Warburg effect also induces a metabolic rewiring stimulating glutamine consumption and lipid synthesis, also considered cancer hallmarks. Amino acid metabolism alteration due to the Warburg effect increases plasma levels of proline and branched-chain amino acids in several cancer types. Proline and lipids are probably used as electron transfer molecules in carcinogenic cells. In addition, branched-chain amino acids fuel the Krebs cycle, protein synthesis, and signaling in cancer cells. AREAS COVERED This review covers how metabolomics studies describe changes in some metabolites and proteins associated with the Warburg effect and related metabolic pathways. EXPERT OPINION In this review, we analyze the metabolic signature of the Warburg effect and related phenotypes and propose some Warburg effect-related metabolites and proteins (lactate, glucose uptake, glucose transporters, glutamine, branched-chain amino acids, proline, and some lipogenic enzymes) as promising cancer biomarkers.
Collapse
Affiliation(s)
- Gerardo M Nava
- Universidad Autónoma de Querétaro, Cerro de las Campanas, Santiago de Querétaro, Qro, 76010, México
| | - Luis Alberto Madrigal Perez
- Tecnológico Nacional de México/ Instituto Tecnológico Superior de Ciudad Hidalgo, Av. Ing. Carlos Rojas Gutiérrez #2120, Ciudad Hidalgo, Michoacán, 61100, México
| |
Collapse
|
29
|
Su Y, Lai W, Liang Y, Zhang C. Novel cloth-based closed bipolar solid-state electrochemiluminescence (CBP-SS-ECL) aptasensor for detecting carcinoembryonic antigen. Anal Chim Acta 2022; 1206:339789. [DOI: 10.1016/j.aca.2022.339789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/29/2022]
|
30
|
The Application of Metabolomics in Recent Colorectal Cancer Studies: A State-of-the-Art Review. Cancers (Basel) 2022; 14:cancers14030725. [PMID: 35158992 PMCID: PMC8833341 DOI: 10.3390/cancers14030725] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/16/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Colorectal Cancer (CRC) is one of the leading causes of cancer-related death in the United States. Current diagnosis techniques are either highly invasive or lack sensitivity, suggesting the need for alternative techniques for biomarker detection. Metabolomics represents one such technique with great promise in identifying CRC biomarkers with high sensitivity and specificity, but thus far is rarely employed in a clinical setting. In order to provide a framework for future clinical usage, we characterized dysregulated metabolites across recent literature, identifying metabolites dysregulated across a variety of biospecimens. We additionally put special focus on the interplay of the gut microbiome and perturbed metabolites in CRC. We were able to identify many metabolites showing consistent dysregulation in CRC, demonstrating the value of metabolomics as a promising diagnostic technique. Abstract Colorectal cancer (CRC) is a highly prevalent disease with poor prognostic outcomes if not diagnosed in early stages. Current diagnosis techniques are either highly invasive or lack sufficient sensitivity. Thus, identifying diagnostic biomarkers of CRC with high sensitivity and specificity is desirable. Metabolomics represents an analytical profiling technique with great promise in identifying such biomarkers and typically represents a close tie with the phenotype of a specific disease. We thus conducted a systematic review of studies reported from January 2012 to July 2021 relating to the detection of CRC biomarkers through metabolomics to provide a collection of knowledge for future diagnostic development. We identified thirty-seven metabolomics studies characterizing CRC, many of which provided metabolites/metabolic profile-based diagnostic models with high sensitivity and specificity. These studies demonstrated that a great number of metabolites can be differentially regulated in CRC patients compared to healthy controls, adenomatous polyps, or across stages of CRC. Among these metabolite biomarkers, especially dysregulated were certain amino acids, fatty acids, and lysophosphatidylcholines. Additionally, we discussed the contribution of the gut bacterial population to pathogenesis of CRC through their modulation to fecal metabolite pools and summarized the established links in the literature between certain microbial genera and altered metabolite levels in CRC patients. Taken together, we conclude that metabolomics presents itself as a promising and effective method of CRC biomarker detection.
Collapse
|
31
|
Troisi J, Tafuro M, Lombardi M, Scala G, Richards SM, Symes SJK, Ascierto PA, Delrio P, Tatangelo F, Buonerba C, Pierri B, Cerino P. A Metabolomics-Based Screening Proposal for Colorectal Cancer. Metabolites 2022; 12:110. [PMID: 35208185 PMCID: PMC8878838 DOI: 10.3390/metabo12020110] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/17/2022] [Accepted: 01/22/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a high incidence disease, characterized by high morbidity and mortality rates. Early diagnosis remains challenging because fecal occult blood screening tests have performed sub-optimally, especially due to hemorrhoidal, inflammatory, and vascular diseases, while colonoscopy is invasive and requires a medical setting to be performed. The objective of the present study was to determine if serum metabolomic profiles could be used to develop a novel screening approach for colorectal cancer. Furthermore, the study evaluated the metabolic alterations associated with the disease. Untargeted serum metabolomic profiles were collected from 100 CRC subjects, 50 healthy controls, and 50 individuals with benign colorectal disease. Different machine learning models, as well as an ensemble model based on a voting scheme, were built to discern CRC patients from CTRLs. The ensemble model correctly classified all CRC and CTRL subjects (accuracy = 100%) using a random subset of the cohort as a test set. Relevant metabolites were examined in a metabolite-set enrichment analysis, revealing differences in patients and controls primarily associated with cell glucose metabolism. These results support a potential use of the metabolomic signature as a non-invasive screening tool for CRC. Moreover, metabolic pathway analysis can provide valuable information to enhance understanding of the pathophysiological mechanisms underlying cancer. Further studies with larger cohorts, including blind trials, could potentially validate the reported results.
Collapse
Affiliation(s)
- Jacopo Troisi
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
- Theoreo srl, Via degli Ulivi 3, 84090 Montecorvino Pugliano, Italy; (M.L.); (G.S.)
| | - Maria Tafuro
- Centro di Referenza Nazionale per l’Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (M.T.); (C.B.); (P.C.)
| | - Martina Lombardi
- Theoreo srl, Via degli Ulivi 3, 84090 Montecorvino Pugliano, Italy; (M.L.); (G.S.)
| | - Giovanni Scala
- Theoreo srl, Via degli Ulivi 3, 84090 Montecorvino Pugliano, Italy; (M.L.); (G.S.)
- Hosmotic srl, Via R. Bosco 178, 80069 Vico Equense, Italy
| | - Sean M. Richards
- Department of Obstetrics and Gynecology, Section on Maternal-Fetal Medicine, University of Tennessee College of Medicine, 960 East Third Street, Suite 100, 902 McCallie Avenue, Chattanooga, TN 37403, USA; (S.M.R.); (S.J.K.S.)
- Department of Biology, Geology and Environmental Sciences, University of Tennessee at Chattanooga, 615 McCallie Ave., Chattanooga, TN 37403, USA
| | - Steven J. K. Symes
- Department of Obstetrics and Gynecology, Section on Maternal-Fetal Medicine, University of Tennessee College of Medicine, 960 East Third Street, Suite 100, 902 McCallie Avenue, Chattanooga, TN 37403, USA; (S.M.R.); (S.J.K.S.)
- Department of Chemistry and Physics, University of Tennessee at Chattanooga, 615 McCallie Ave., Chattanooga, TN 37403, USA
| | - Paolo Antonio Ascierto
- Istituto Nazionale Tumori Fondazione Pascale IRCCS, 80131 Napoli, Italy; (P.A.A.); (P.D.); (F.T.)
| | - Paolo Delrio
- Istituto Nazionale Tumori Fondazione Pascale IRCCS, 80131 Napoli, Italy; (P.A.A.); (P.D.); (F.T.)
| | - Fabiana Tatangelo
- Istituto Nazionale Tumori Fondazione Pascale IRCCS, 80131 Napoli, Italy; (P.A.A.); (P.D.); (F.T.)
| | - Carlo Buonerba
- Centro di Referenza Nazionale per l’Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (M.T.); (C.B.); (P.C.)
| | - Biancamaria Pierri
- Centro di Referenza Nazionale per l’Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (M.T.); (C.B.); (P.C.)
| | - Pellegrino Cerino
- Centro di Referenza Nazionale per l’Analisi e Studio di Correlazione tra Ambiente, Animale e Uomo, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy; (M.T.); (C.B.); (P.C.)
| |
Collapse
|
32
|
Biomarker profiling of postmortem blood for diabetes mellitus and discussion of possible applications of metabolomics for forensic casework. Int J Legal Med 2022; 136:1075-1090. [DOI: 10.1007/s00414-021-02767-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/14/2021] [Indexed: 10/19/2022]
|
33
|
Exploring Serum NMR-Based Metabolomic Fingerprint of Colorectal Cancer Patients: Effects of Surgery and Possible Associations with Cancer Relapse. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112311120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: Colorectal cancer (CRC) is the fourth most commonly diagnosed and third most deadly cancer worldwide. Surgery is the main treatment option for early disease; however, a relevant proportion of CRC patients relapse. Here, variations among preoperative and postoperative serum metabolomic fingerprint of CRC patients were studied, and possible associations between metabolic variations and cancer relapse were explored. Methods: A total of 41 patients with stage I-III CRC, planned for radical resection, were enrolled. Serum samples, collected preoperatively (t0) and 4–6 weeks after surgery before the start of any treatment (t1), were analyzed via NMR spectroscopy. NMR data were analyzed using multivariate and univariate statistical approaches. Results: Serum metabolomic fingerprints show differential clustering between t0 and t1 (82–85% accuracy). Pyruvate, HDL-related parameters, acetone, and 3-hydroxybutyrate appear to be the major players in this discrimination. Eight out of the 41 CRC patients enrolled developed cancer relapse. Postoperative, relapsed patients show an increase of pyruvate and HDL-related parameters, and a decrease of Apo-A1 Apo-B100 ratio and VLDL-related parameters. Conclusions: Surgery significantly alters the metabolomic fingerprint of CRC patients. Some metabolic changes seem to be associated with the development of cancer relapse. These data, if validated in a larger cohort, open new possibilities for risk stratification in patients with early-stage CRC.
Collapse
|
34
|
Beklen H, Yildirim E, Kori M, Turanli B, Arga KY. Systems-level biomarkers identification and drug repositioning in colorectal cancer. World J Gastrointest Oncol 2021. [DOI: 10.4251/wjgo.v13.i7.463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
35
|
Beklen H, Yildirim E, Kori M, Turanli B, Arga KY. Systems-level biomarkers identification and drug repositioning in colorectal cancer. World J Gastrointest Oncol 2021; 13:638-661. [PMID: 34322194 PMCID: PMC8299930 DOI: 10.4251/wjgo.v13.i7.638] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/20/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the most commonly diagnosed fatal cancer in both women and men worldwide. CRC ranked second in mortality and third in incidence in 2020. It is difficult to diagnose CRC at an early stage as there are no clinical symptoms. Despite advances in molecular biology, only a limited number of biomarkers have been translated into routine clinical practice to predict risk, prognosis and response to treatment. In the last decades, systems biology approaches at the omics level have gained importance. Over the years, several biomarkers for CRC have been discovered in terms of disease diagnosis and prognosis. On the other hand, a few drugs are being developed and used in clinics for the treatment of CRC. However, the development of new drugs is very costly and time-consuming as the research and development takes about 10 years and more than $1 billion. Therefore, drug repositioning (DR) could save time and money by establishing new indications for existing drugs. In this review, we aim to provide an overview of biomarkers for the diagnosis and prognosis of CRC from the systems biology perspective and insights into DR approaches for the prevention or treatment of CRC.
Collapse
Affiliation(s)
- Hande Beklen
- Department of Bioengineering, Marmara University, Istanbul 34722, Turkey
| | - Esra Yildirim
- Department of Bioengineering, Marmara University, Istanbul 34722, Turkey
| | - Medi Kori
- Department of Bioengineering, Marmara University, Istanbul 34722, Turkey
| | - Beste Turanli
- Department of Bioengineering, Marmara University, Istanbul 34722, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Marmara University, Istanbul 34722, Turkey
| |
Collapse
|
36
|
Stefan-van Staden RI, Gheorghe DC, Ilie-Mihai RM, Tudoran LB, Pruneanu SM. Stochastic biosensors based on N- and S-doped graphene for the enantioanalysis of aspartic acid in biological samples. RSC Adv 2021; 11:23301-23309. [PMID: 35479820 PMCID: PMC9036598 DOI: 10.1039/d1ra02066h] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/22/2021] [Indexed: 01/15/2023] Open
Abstract
Metabolomics plays a very important role in cancer mechanism and also in the early diagnosis of cancer. The enantioanalysis of aminoacids found in biological samples may favor the development of new screening tests for the early diagnosis of cancer. Two stochastic biosensors, based on the immobilization of α-hemolysin and hemin in exfoliated reduced graphene modified with nitrogen and sulphur, were proposed for the enantioanalysis of aspartic acid in biological samples (whole blood, urine, saliva, and tissue). The proposed biosensors showed low limits of quantification, high sensitivity, and large linear concentration ranges. The recoveries of d- and l-aspartic acid in biological samples were higher than 95.00%, with a relative standard deviation lower than 1.00%. Stochastic biosensors based on N- and S-doped graphene modified with hemin or α-hemolysin contributed to establishing the metabolomics of gastric cancer by performing the enantioanalysis of aspartic acid in different biological samples.![]()
Collapse
Affiliation(s)
- Raluca-Ioana Stefan-van Staden
- Laboratory of Electrochemistry and PATLAB, National Institute of Research for Electrochemistry and Condensed Matter 202 Splaiul Independentei Str. 060021 Bucharest-6 Romania .,Faculty of Applied Chemistry and Material Science, Politehnica University of Bucharest Bucharest Romania
| | - Damaris-Cristina Gheorghe
- Laboratory of Electrochemistry and PATLAB, National Institute of Research for Electrochemistry and Condensed Matter 202 Splaiul Independentei Str. 060021 Bucharest-6 Romania .,Faculty of Applied Chemistry and Material Science, Politehnica University of Bucharest Bucharest Romania
| | - Ruxandra-Maria Ilie-Mihai
- Laboratory of Electrochemistry and PATLAB, National Institute of Research for Electrochemistry and Condensed Matter 202 Splaiul Independentei Str. 060021 Bucharest-6 Romania
| | - Lucian-Barbu Tudoran
- National Institute for Research and Development of Isotopic and Molecular Technologies Donat Street 67-103 Cluj-Napoca Romania
| | - Stela Maria Pruneanu
- National Institute for Research and Development of Isotopic and Molecular Technologies Donat Street 67-103 Cluj-Napoca Romania
| |
Collapse
|
37
|
Furukawa H, Oka S, Higuchi T, Shimada K, Hashimoto A, Matsui T, Tohma S. Biomarkers for interstitial lung disease and acute-onset diffuse interstitial lung disease in rheumatoid arthritis. Ther Adv Musculoskelet Dis 2021; 13:1759720X211022506. [PMID: 34211592 PMCID: PMC8216360 DOI: 10.1177/1759720x211022506] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/11/2021] [Indexed: 12/31/2022] Open
Abstract
Interstitial lung disease (ILD) is frequently a complication of rheumatoid arthritis (RA) as an extra-articular manifestation which has a poor prognosis. Acute-onset diffuse ILD (AoDILD) occasionally occurs in RA and includes acute exacerbation of ILD, drug-induced ILD, and Pneumocystis pneumonia. AoDILD also confers a poor prognosis in RA. Previously-established biomarkers for ILD include Krebs von den lungen-6 and surfactant protein-D originally defined in patients with idiopathic pulmonary fibrosis; the sensitivity of these markers for RA-associated ILD (RA-ILD) is low. Although many studies on ILD markers have been performed in idiopathic pulmonary fibrosis, only a few validation studies in RA-ILD or AoDILD have been reported. Biomarkers for RA-ILD and AoDILD are thus still required. Recently, genomic, cytokine, antibody, and metabolomic profiles of RA-ILD or AoDILD have been investigated with the aim of improving biomarkers. In this review, we summarize current preliminary data on these potential biomarkers for RA-ILD or AoDILD. The development of biomarkers on RA-ILD has only just begun. When validated, such candidate biomarkers will provide valuable information on pathogenesis, prognosis, and drug responses in RA-ILD in future.
Collapse
Affiliation(s)
- Hiroshi Furukawa
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose 204-8585, Japan
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
| | - Shomi Oka
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
| | - Takashi Higuchi
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
- Department of Nephrology, Ushiku Aiwa General Hospital, Ushiku, Japan
| | - Kota Shimada
- Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan
| | - Atsushi Hashimoto
- Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
- Department of Internal Medicine, Sagami Seikyou Hospital, Minami-ku, Sagamihara, Japan
| | - Toshihiro Matsui
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
- Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
| | - Shigeto Tohma
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
| |
Collapse
|
38
|
Di Donato S, Vignoli A, Biagioni C, Malorni L, Mori E, Tenori L, Calamai V, Parnofiello A, Di Pierro G, Migliaccio I, Cantafio S, Baraghini M, Mottino G, Becheri D, Del Monte F, Miceli E, McCartney A, Di Leo A, Luchinat C, Biganzoli L. A Serum Metabolomics Classifier Derived from Elderly Patients with Metastatic Colorectal Cancer Predicts Relapse in the Adjuvant Setting. Cancers (Basel) 2021; 13:cancers13112762. [PMID: 34199435 PMCID: PMC8199587 DOI: 10.3390/cancers13112762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/14/2021] [Accepted: 05/29/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Around 30–40% of patients with early stage colorectal cancer (eCRC) experience relapse after surgery. Current recommendations for adjuvant therapy are based on suboptimal risk-stratification tools. In elderly patients, risk of relapse assessment is particularly important to ultimately avoid unnecessary chemotherapy-related toxicity in this frailer population. Serum metabolomics via NMR spectroscopy may improve risk stratification by identifying patients with residual micrometastases after surgery and thus at higher risk of relapse. We evaluated the serum metabolomic fingerprints of 94 elderly patients with eCRC (65 relapse free and 29 relapsed), and of 75 elderly patients with metastatic disease. Metabolomics efficiently discriminated patients with relapse-free eCRC from those with metastatic disease, correctly predicting relapse in 69% of relapsed eCRC patients. The metabolomic score was strongly and independently associated with prognosis. Our data suggest metabolomics as a valid addition to standard tools to refine risk stratification for eCRC and warrant further investigation. Abstract Adjuvant treatment for patients with early stage colorectal cancer (eCRC) is currently based on suboptimal risk stratification, especially for elderly patients. Metabolomics may improve the identification of patients with residual micrometastases after surgery. In this retrospective study, we hypothesized that metabolomic fingerprinting could improve risk stratification in patients with eCRC. Serum samples obtained after surgery from 94 elderly patients with eCRC (65 relapse free and 29 relapsed, after 5-years median follow up), and from 75 elderly patients with metastatic colorectal cancer (mCRC) obtained before a new line of chemotherapy, were retrospectively analyzed via proton nuclear magnetic resonance spectroscopy. The prognostic role of metabolomics in patients with eCRC was assessed using Kaplan–Meier curves. PCA-CA-kNN could discriminate the metabolomic fingerprint of patients with relapse-free eCRC and mCRC (70.0% accuracy using NOESY spectra). This model was used to classify the samples of patients with relapsed eCRC: 69% of eCRC patients with relapse were predicted as metastatic. The metabolomic classification was strongly associated with prognosis (p-value 0.0005, HR 3.64), independently of tumor stage. In conclusion, metabolomics could be an innovative tool to refine risk stratification in elderly patients with eCRC. Based on these results, a prospective trial aimed at improving risk stratification by metabolomic fingerprinting (LIBIMET) is ongoing.
Collapse
Affiliation(s)
- Samantha Di Donato
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
- Correspondence: ; Tel.: +39-057-480-2520
| | - Alessia Vignoli
- Magnetic Resonance Center, University of Florence, 50019 Sesto Fiorentino, Italy; (A.V.); (L.T.); (C.L.)
- Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Chiara Biagioni
- Bioinformatics Unit, Medical Oncology Department, New Hospital of Prato S. Stefano, 59100 Prato, Italy;
| | - Luca Malorni
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
- “Sandro Pitigliani” Translational Research Unit, New Hospital of Prato, Stefano, 59100 Prato, Italy;
| | - Elena Mori
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
| | - Leonardo Tenori
- Magnetic Resonance Center, University of Florence, 50019 Sesto Fiorentino, Italy; (A.V.); (L.T.); (C.L.)
- Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Vanessa Calamai
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
| | - Annamaria Parnofiello
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Giulia Di Pierro
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
| | - Ilenia Migliaccio
- “Sandro Pitigliani” Translational Research Unit, New Hospital of Prato, Stefano, 59100 Prato, Italy;
| | - Stefano Cantafio
- Department of Surgery, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (S.C.); (M.B.)
| | - Maddalena Baraghini
- Department of Surgery, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (S.C.); (M.B.)
| | - Giuseppe Mottino
- Department of Geriatrics, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (G.M.); (D.B.)
| | - Dimitri Becheri
- Department of Geriatrics, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (G.M.); (D.B.)
| | - Francesca Del Monte
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
| | - Elisangela Miceli
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
| | - Amelia McCartney
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
- School of Clinical Sciences, Monash University, 3168 Clayton, Australia
| | - Angelo Di Leo
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
| | - Claudio Luchinat
- Magnetic Resonance Center, University of Florence, 50019 Sesto Fiorentino, Italy; (A.V.); (L.T.); (C.L.)
- Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (C.I.R.M.M.P.), 50019 Sesto Fiorentino, Italy
| | - Laura Biganzoli
- Department of Medical Oncology, New Hospital of Prato S. Stefano, 59100 Prato, Italy; (L.M.); (E.M.); (V.C.); (A.P.); (G.D.P.); (F.D.M.); (E.M.); (A.M.); (A.D.L.); (L.B.)
| |
Collapse
|
39
|
Druce P, Calanzani N, Snudden C, Milley K, Boscott R, Behiyat D, Martinez-Gutierrez J, Saji S, Oberoi J, Funston G, Messenger M, Walter FM, Emery J. Identifying Novel Biomarkers Ready for Evaluation in Low-Prevalence Populations for the Early Detection of Lower Gastrointestinal Cancers: A Systematic Review and Meta-Analysis. Adv Ther 2021; 38:3032-3065. [PMID: 33907946 PMCID: PMC8078393 DOI: 10.1007/s12325-021-01645-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/30/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Lower gastrointestinal (GI) cancers are a major cause of cancer deaths worldwide. Prognosis improves with earlier diagnosis, and non-invasive biomarkers have the potential to aid with early detection. Substantial investment has been made into the development of biomarkers; however, studies are often carried out in specialist settings and few have been evaluated for low-prevalence populations. METHODS We aimed to identify novel biomarkers for the detection of lower GI cancers that have the potential to be evaluated for use in primary care. MEDLINE, Embase, Emcare and Web of Science were systematically searched for studies published in English from January 2000 to October 2019. Reference lists of included studies were also assessed. Studies had to report on measures of diagnostic performance for biomarkers (single or in panels) used to detect colorectal or anal cancers. We included all designs and excluded studies with fewer than 50 cases/controls. Data were extracted from published studies on types of biomarkers, populations and outcomes. Narrative synthesis was used, and measures of specificity and sensitivity were meta-analysed where possible. RESULTS We identified 142 studies reporting on biomarkers for lower GI cancers, for 24,844 cases and 45,374 controls. A total of 378 unique biomarkers were identified. Heterogeneity of study design, population type and sample source precluded meta-analysis for all markers except methylated septin 9 (mSEPT9) and pyruvate kinase type tumour M2 (TuM2-PK). The estimated sensitivity and specificity of mSEPT9 was 80.6% (95% CI 76.6-84.0%) and 88.0% (95% CI 79.1-93.4%) respectively; TuM2-PK had an estimated sensitivity of 81.6% (95% CI 75.2-86.6%) and specificity of 80.1% (95% CI 76.7-83.0%). CONCLUSION Two novel biomarkers (mSEPT9 and TuM2-PK) were identified from the literature with potential for use in lower-prevalence populations. Further research is needed to validate these biomarkers in primary care for screening and assessment of symptomatic patients.
Collapse
Affiliation(s)
- Paige Druce
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Melbourne, VIC, Australia.
| | - Natalia Calanzani
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Claudia Snudden
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Kristi Milley
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Melbourne, VIC, Australia
| | - Rachel Boscott
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Dawnya Behiyat
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Javiera Martinez-Gutierrez
- Department of Family Medicine, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Smiji Saji
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Jasmeen Oberoi
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Melbourne, VIC, Australia
| | - Garth Funston
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Mike Messenger
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Fiona M Walter
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Melbourne, VIC, Australia
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Jon Emery
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Melbourne, VIC, Australia
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
40
|
Zhou D, Zhu W, Sun T, Wang Y, Chi Y, Chen T, Lin J. iMAP: A Web Server for Metabolomics Data Integrative Analysis. Front Chem 2021; 9:659656. [PMID: 34026726 PMCID: PMC8133432 DOI: 10.3389/fchem.2021.659656] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Metabolomics data analysis depends on the utilization of bioinformatics tools. To meet the evolving needs of metabolomics research, several integrated platforms have been developed. Our group has developed a desktop platform IP4M (integrated Platform for Metabolomics Data Analysis) which allows users to perform a nearly complete metabolomics data analysis in one-stop. With the extensive usage of IP4M, more and more demands were raised from users worldwide for a web version and a more customized workflow. Thus, iMAP (integrated Metabolomics Analysis Platform) was developed with extended functions, improved performances, and redesigned structures. Compared with existing platforms, iMAP has more methods and usage modes. A new module was developed with an automatic pipeline for train-test set separation, feature selection, and predictive model construction and validation. A new module was incorporated with sufficient editable parameters for network construction, visualization, and analysis. Moreover, plenty of plotting tools have been upgraded for highly customized publication-ready figures. Overall, iMAP is a good alternative tool with complementary functions to existing metabolomics data analysis platforms. iMAP is freely available for academic usage at https://imap.metaboprofile.cloud/ (License MPL 2.0).
Collapse
Affiliation(s)
- Di Zhou
- Metabo-Profile Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Wenjia Zhu
- Metabo-Profile Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Tao Sun
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yang Wang
- Metabo-Profile Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Yi Chi
- Metabo-Profile Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| | - Tianlu Chen
- Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jingchao Lin
- Metabo-Profile Biotechnology (Shanghai) Co. Ltd., Shanghai, China
| |
Collapse
|
41
|
Abstract
Nuclear magnetic resonance (NMR) spectroscopy offers reproducible quantitative analysis and structural identification of metabolites in various complex biological samples, such as biofluids (plasma, serum, and urine), cells, tissue extracts, and even intact organs. Therefore, NMR-based metabolomics, a mainstream metabolomic platform, has been extensively applied in many research fields, including pharmacology, toxicology, pathophysiology, nutritional intervention, disease diagnosis/prognosis, and microbiology. In particular, NMR-based metabolomics has been successfully used for cancer research to investigate cancer metabolism and identify biomarker and therapeutic targets. This chapter highlights the innovations and challenges of NMR-based metabolomics platform and its applications in cancer research.
Collapse
|
42
|
Aftabi Y, Soleymani J, Jouyban A. Efficacy of Analytical Technologies in Metabolomics Studies of the Gastrointestinal Cancers. Crit Rev Anal Chem 2021; 52:1593-1605. [PMID: 33757389 DOI: 10.1080/10408347.2021.1901646] [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] [Indexed: 01/02/2023]
Abstract
According to the reports of the World Health Organization and the International Agency for Research on Cancer, cancer is the second leading cause of human death worldwide. However, early-stage detection of cancers can efficiently enhance the chance of therapy and saving lives. Metabolomics strategies apply a variety of approaches to discover new potential diagnoses, prognoses, and/or therapeutic biomarkers of various diseases. Metabolomics aims to identify and measure different low-molecular-weight biomolecules in physiological environments. In these studies, special metabolites are extracted from biological samples and identified using analytical techniques. Afterward, using data processing programs discovering significantly associated biomarkers is pursued. In the present review, we aimed to discuss recently reported analytical approaches on the metabolomics studies of gastrointestinal cancers including gastric, colorectal, and esophageal cancers. The gas- and liquid-chromatography with different detectors have been shown that are the main analytical techniques and for metabolites quantification, nuclear magnetic resonance has been utilized as a master method.
Collapse
Affiliation(s)
- Younes Aftabi
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleymani
- Pharmaceutical Analysis Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolghasem Jouyban
- Pharmaceutical Analysis Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
43
|
Răchieriu C, Eniu DT, Moiş E, Graur F, Socaciu C, Socaciu MA, Hajjar NA. Lipidomic Signatures for Colorectal Cancer Diagnosis and Progression Using UPLC-QTOF-ESI +MS. Biomolecules 2021; 11:biom11030417. [PMID: 33799830 PMCID: PMC8035671 DOI: 10.3390/biom11030417] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022] Open
Abstract
Metabolomics coupled with bioinformatics may identify relevant biomolecules such as putative biomarkers of specific metabolic pathways related to colorectal diagnosis, classification and prognosis. This study performed an integrated metabolomic profiling of blood serum from 25 colorectal cancer (CRC) cases previously classified (Stage I to IV) compared with 16 controls (disease-free, non-CRC patients), using high-performance liquid chromatography and mass spectrometry (UPLC-QTOF-ESI+ MS). More than 400 metabolites were separated and identified, then all data were processed by the advanced Metaboanalyst 5.0 online software, using multi- and univariate analysis, including specificity/sensitivity relationships (area under the curve (AUC) values), enrichment and pathway analysis, identifying the specific pathways affected by cancer progression in the different stages. Several sub-classes of lipids including phosphatidylglycerols (phosphatidylcholines (PCs), phosphatidylethanolamines (PEs) and PAs), fatty acids and sterol esters as well as ceramides confirmed the “lipogenic phenotype” specific to CRC development, namely the upregulated lipogenesis associated with tumor progression. Both multivariate and univariate bioinformatics confirmed the relevance of some putative lipid biomarkers to be responsible for the altered metabolic pathways in colorectal cancer.
Collapse
Affiliation(s)
- Claudiu Răchieriu
- Surgery Department, County Hospital Alba, 510118 Alba Iulia, Romania;
- Iuliu Hatieganu University of Medicine and Pharmacy, Regional Institute of Gastroenterology and Hepatology “Octavian Fodor”, 400015 Cluj-Napoca, Romania; (E.M.); (F.G.); (N.A.H.)
| | - Dan Tudor Eniu
- Oncology Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania;
| | - Emil Moiş
- Iuliu Hatieganu University of Medicine and Pharmacy, Regional Institute of Gastroenterology and Hepatology “Octavian Fodor”, 400015 Cluj-Napoca, Romania; (E.M.); (F.G.); (N.A.H.)
| | - Florin Graur
- Iuliu Hatieganu University of Medicine and Pharmacy, Regional Institute of Gastroenterology and Hepatology “Octavian Fodor”, 400015 Cluj-Napoca, Romania; (E.M.); (F.G.); (N.A.H.)
| | - Carmen Socaciu
- University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
- Research Center for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
- Correspondence: (C.S.); (M.A.S.)
| | - Mihai Adrian Socaciu
- Iuliu Hatieganu University of Medicine and Pharmacy, Regional Institute of Gastroenterology and Hepatology “Octavian Fodor”, 400015 Cluj-Napoca, Romania; (E.M.); (F.G.); (N.A.H.)
- Correspondence: (C.S.); (M.A.S.)
| | - Nadim Al Hajjar
- Iuliu Hatieganu University of Medicine and Pharmacy, Regional Institute of Gastroenterology and Hepatology “Octavian Fodor”, 400015 Cluj-Napoca, Romania; (E.M.); (F.G.); (N.A.H.)
| |
Collapse
|
44
|
Gumpenberger T, Brezina S, Keski-Rahkonen P, Baierl A, Robinot N, Leeb G, Habermann N, Kok DEG, Scalbert A, Ueland PM, Ulrich CM, Gsur A. Untargeted Metabolomics Reveals Major Differences in the Plasma Metabolome between Colorectal Cancer and Colorectal Adenomas. Metabolites 2021; 11:119. [PMID: 33669644 PMCID: PMC7922413 DOI: 10.3390/metabo11020119] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/09/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Sporadic colorectal cancer is characterized by a multistep progression from normal epithelium to precancerous low-risk and high-risk adenomas to invasive cancer. Yet, the underlying molecular mechanisms of colorectal carcinogenesis are not completely understood. Within the "Metabolomic profiles throughout the continuum of colorectal cancer" (MetaboCCC) consortium we analyzed data generated by untargeted, mass spectrometry-based metabolomics using plasma from 88 colorectal cancer patients, 200 patients with high-risk adenomas and 200 patients with low-risk adenomas recruited within the "Colorectal Cancer Study of Austria" (CORSA). Univariate logistic regression models comparing colorectal cancer to adenomas resulted in 442 statistically significant molecular features. Metabolites discriminating colorectal cancer patients from those with adenomas in our dataset included acylcarnitines, caffeine, amino acids, glycerophospholipids, fatty acids, bilirubin, bile acids and bacterial metabolites of tryptophan. The data obtained discovers metabolite profiles reflecting metabolic differences between colorectal cancer and colorectal adenomas and delineates a potentially underlying biological interpretation.
Collapse
Affiliation(s)
- Tanja Gumpenberger
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (T.G.); (S.B.)
| | - Stefanie Brezina
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (T.G.); (S.B.)
| | - Pekka Keski-Rahkonen
- International Agency for Research on Cancer, 69372 Lyon, France; (P.K.-R.); (N.R.); (A.S.)
| | - Andreas Baierl
- Department of Statistics and Operations Research, University of Vienna, 1090 Vienna, Austria;
| | - Nivonirina Robinot
- International Agency for Research on Cancer, 69372 Lyon, France; (P.K.-R.); (N.R.); (A.S.)
| | - Gernot Leeb
- Department of Internal Medicine, Hospital Oberpullendorf, 7350 Oberpullendorf, Austria;
| | - Nina Habermann
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Genome Biology, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Dieuwertje E G Kok
- Division of Human Nutrition and Health, Wageningen University & Research, 6708 Wageningen, The Netherlands;
| | - Augustin Scalbert
- International Agency for Research on Cancer, 69372 Lyon, France; (P.K.-R.); (N.R.); (A.S.)
| | | | - Cornelia M Ulrich
- Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT 84112, USA;
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84108, USA
| | - Andrea Gsur
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (T.G.); (S.B.)
| |
Collapse
|
45
|
Tadaka S, Hishinuma E, Komaki S, Motoike IN, Kawashima J, Saigusa D, Inoue J, Takayama J, Okamura Y, Aoki Y, Shirota M, Otsuki A, Katsuoka F, Shimizu A, Tamiya G, Koshiba S, Sasaki M, Yamamoto M, Kinoshita K. jMorp updates in 2020: large enhancement of multi-omics data resources on the general Japanese population. Nucleic Acids Res 2021; 49:D536-D544. [PMID: 33179747 PMCID: PMC7779038 DOI: 10.1093/nar/gkaa1034] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 12/30/2022] Open
Abstract
In the Tohoku Medical Megabank project, genome and omics analyses of participants in two cohort studies were performed. A part of the data is available at the Japanese Multi Omics Reference Panel (jMorp; https://jmorp.megabank.tohoku.ac.jp) as a web-based database, as reported in our previous manuscript published in Nucleic Acid Research in 2018. At that time, jMorp mainly consisted of metabolome data; however, now genome, methylome, and transcriptome data have been integrated in addition to the enhancement of the number of samples for the metabolome data. For genomic data, jMorp provides a Japanese reference sequence obtained using de novo assembly of sequences from three Japanese individuals and allele frequencies obtained using whole-genome sequencing of 8,380 Japanese individuals. In addition, the omics data include methylome and transcriptome data from ∼300 samples and distribution of concentrations of more than 755 metabolites obtained using high-throughput nuclear magnetic resonance and high-sensitivity mass spectrometry. In summary, jMorp now provides four different kinds of omics data (genome, methylome, transcriptome, and metabolome), with a user-friendly web interface. This will be a useful scientific data resource on the general population for the discovery of disease biomarkers and personalized disease prevention and early diagnosis.
Collapse
Affiliation(s)
- Shu Tadaka
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Eiji Hishinuma
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Tohoku University Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Shohei Komaki
- Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Ikuko N Motoike
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, 6-3-09 Aramaki aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| | - Junko Kawashima
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Daisuke Saigusa
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Jin Inoue
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Jun Takayama
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Tohoku University Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Yasunobu Okamura
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Tohoku University Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Yuichi Aoki
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, 6-3-09 Aramaki aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| | - Matsuyuki Shirota
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Tohoku University Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Graduate School of Information Sciences, Tohoku University, 6-3-09 Aramaki aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| | - Akihito Otsuki
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Fumiki Katsuoka
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Atsushi Shimizu
- Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
- Institute for Biomedical Sciences, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Gen Tamiya
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Seizo Koshiba
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Tohoku University Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
- Institute for Biomedical Sciences, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Masayuki Yamamoto
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Tohoku University Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Kengo Kinoshita
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Tohoku University Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
- Graduate School of Information Sciences, Tohoku University, 6-3-09 Aramaki aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| |
Collapse
|
46
|
Maeyama M, Tanaka K, Nishihara M, Irino Y, Shinohara M, Nagashima H, Tanaka H, Nakamizo S, Hashiguchi M, Fujita Y, Kohta M, Kohmura E, Sasayama T. Metabolic changes and anti-tumor effects of a ketogenic diet combined with anti-angiogenic therapy in a glioblastoma mouse model. Sci Rep 2021; 11:79. [PMID: 33420169 PMCID: PMC7794443 DOI: 10.1038/s41598-020-79465-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
The ketogenic diet (KD) is a high fat and low carbohydrate diet that produces ketone bodies through imitation of starvation. The combination of KD and Bevacizumab (Bev), a VEGF inhibitor, is considered to further reduce the supply of glucose to the tumor. The metabolite changes in U87 glioblastoma mouse models treated with KD and/or Bev were examined using gas chromatography-mass spectrometry. The combination therapy of KD and Bev showed a decrease in the rate of tumor growth and an increase in the survival time of mice, although KD alone did not have survival benefit. In the metabolome analysis, the pattern of changes for most amino acids are similar between tumor and brain tissues, however, some amino acids such as aspartic acid and glutamic acid were different between tumors and brain tissues. The KD enhanced the anti-tumor efficacy of Bev in a glioblastoma intracranial implantation mouse model, based on lowest levels of microvascular density (CD31) and cellular proliferation markers (Ki-67 and CCND1) in KD + Bev tumors compared to the other groups. These results suggested that KD combined with Bev may be a useful treatment strategy for patients with GBM.
Collapse
Affiliation(s)
- Masahiro Maeyama
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kazuhiro Tanaka
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | | | - Yasuhiro Irino
- Division of Evidence-Based Laboratory Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masakazu Shinohara
- Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Epidemiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroaki Nagashima
- Department of Neurosurgery, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Hirotomo Tanaka
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Satoshi Nakamizo
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Mitsuru Hashiguchi
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yuichi Fujita
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masaaki Kohta
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Eiji Kohmura
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takashi Sasayama
- Department of Neurosurgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
47
|
Saigusa D, Matsukawa N, Hishinuma E, Koshiba S. Identification of biomarkers to diagnose diseases and find adverse drug reactions by metabolomics. Drug Metab Pharmacokinet 2020; 37:100373. [PMID: 33631535 DOI: 10.1016/j.dmpk.2020.11.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Metabolomics has been widely used for investigating the biological functions of disease expression and has the potential to discover biomarkers in circulating biofluids or tissue extracts that reflect in phenotypic changes. Metabolic profiling has advantages because of the use of unbiased techniques, including multivariate analysis, and has been applied in pharmacological studies to predict therapeutic and adverse reactions of drugs, which is called pharmacometabolomics (PMx). Nuclear magnetic resonance (NMR)- and mass spectrometry (MS)-based metabolomics has contributed to the discovery of recent disease biomarkers; however, the optimal strategy for the study purpose must be selected from many established protocols, methodologies and analytical platforms. Additionally, information on molecular localization in tissue is essential for further functional analyses related to therapeutic and adverse effects of drugs in the process of drug development. MS imaging (MSI) is a promising technology that can visualize molecules on tissue surfaces without labeling and thus provide localized information. This review summarizes recent uses of MS-based global and wide-targeted metabolomics technologies and the advantages of the MSI approach for PMx and highlights the PMx technique for the biomarker discovery of adverse drug effects.
Collapse
Affiliation(s)
- Daisuke Saigusa
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Naomi Matsukawa
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Eiji Hishinuma
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan.
| | - Seizo Koshiba
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan; Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan; Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan.
| |
Collapse
|
48
|
Furukawa H, Oka S, Shimada K, Okamoto A, Hashimoto A, Komiya A, Saisho K, Yoshikawa N, Katayama M, Matsui T, Fukui N, Migita K, Tohma S. Serum Metabolomic Profiling in Rheumatoid Arthritis Patients With Interstitial Lung Disease: A Case-Control Study. Front Med (Lausanne) 2020; 7:599794. [PMID: 33392224 PMCID: PMC7773768 DOI: 10.3389/fmed.2020.599794] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Objectives: Interstitial lung disease (ILD) is an extra-articular manifestation in rheumatoid arthritis (RA), detected in 10.7% of patients, and causing a poor prognosis. Hence, biomarkers for ILD are urgently required in RA. Low molecular weight metabolites can be assessed by metabolomic analyses, and although these have been conducted in RA and in idiopathic pulmonary fibrosis, few have been carried out for ILD in the context of RA. Therefore, we analyzed serum metabolomic profiles of ILD in RA to identify novel biomarkers. Methods: Serum samples from 100 RA patients with ILD and 100 matched RA patients without chronic lung disease (CLD) were collected. These samples were subjected to metabolomic analyses using capillary electrophoresis time-of-flight mass spectrometry. Results: A total of 299 metabolites were detected in the metabolomic analysis. By univariate analysis, serum levels of decanoic acid and morpholine were lower in RA with ILD (false discovery rate Q = 1.87 × 10−11 and 7.09 × 10−6, respectively), and glycerol was higher (Q = 1.20 × 10−6), relative to RA without CLD. Serum levels of these metabolites in RA with usual interstitial pneumonia or RA with non-specific interstitial pneumonia were also altered. The partial least squares-discriminant analysis model generated from these three metabolites could successfully discriminate ILD in RA (area under the curve: 0.919, 95% confidence interval: 0.867–0.968, sensitivity 0.880, specificity 0.780). Conclusions: Serum levels of some metabolites were significantly different in RA with ILD compared with RA without CLD. It is concluded that metabolomic profiling will be useful for discovering candidate screening biomarkers for ILD in RA.
Collapse
Affiliation(s)
- Hiroshi Furukawa
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan.,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Shomi Oka
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan.,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Kota Shimada
- Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan.,Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan
| | - Akira Okamoto
- Department of Rheumatology, National Hospital Organization Himeji Medical Center, Himeji, Japan
| | - Atsushi Hashimoto
- Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan.,Department of Internal Medicine, Sagami Seikyou Hospital, Sagamihara, Japan
| | - Akiko Komiya
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan.,Department of Clinical Laboratory, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Koichiro Saisho
- Department of Orthopedics/Rheumatology, National Hospital Organization Miyakonojo Medical Center, Miyakonojo, Japan.,Tanimura Hospital, Nobeoka, Japan
| | - Norie Yoshikawa
- Department of Orthopedics/Rheumatology, National Hospital Organization Miyakonojo Medical Center, Miyakonojo, Japan
| | - Masao Katayama
- Department of Internal Medicine, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Toshihiro Matsui
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan.,Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Naoshi Fukui
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Kiyoshi Migita
- Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Ōmura, Japan.,Department of Gastroenterology and Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shigeto Tohma
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan.,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| |
Collapse
|
49
|
Lee YR, An KY, Jeon J, Kim NK, Lee JW, Hong J, Chung BC. Untargeted Metabolomics and Polyamine Profiling in Serum before and after Surgery in Colorectal Cancer Patients. Metabolites 2020; 10:metabo10120487. [PMID: 33260822 PMCID: PMC7760053 DOI: 10.3390/metabo10120487] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer is one of the most prevalent cancers in Korea and globally. In this study, we aimed to characterize the differential serum metabolomic profiles between pre-operative and post-operative patients with colorectal cancer. To investigate the significant metabolites and metabolic pathways associated with colorectal cancer, we analyzed serum samples from 68 patients (aged 20–71, mean 57.57 years). Untargeted and targeted metabolomics profiling in patients with colorectal cancer were performed using liquid chromatography-mass spectrometry. Untargeted analysis identified differences in sphingolipid metabolism, steroid biosynthesis, and arginine and proline metabolism in pre- and post-operative patients with colorectal cancer. We then performed quantitative target profiling of polyamines, synthesized from arginine and proline metabolism, to identify potential polyamines that may serve as effective biomarkers for colorectal cancer. Results indicate a significantly reduced serum concentration of putrescine in post-operative patients compared to pre-operative patients. Our metabolomics approach provided insights into the physiological alterations in patients with colorectal cancer after surgery.
Collapse
Affiliation(s)
- Yu Ra Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea;
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea
| | - Ki-Yong An
- Faculty of Kinesiology, Sport and Recreation, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Justin Jeon
- Department of Sport Industry, Yonsei University, Seoul 03722, Korea;
- Exercise Medicine Center for Diabetes and Cancer Patients, ICONS, Yonsei University, Seoul 03722, Korea
| | - Nam Kyu Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Ji Won Lee
- Department of Family Medicine, Yonsei University College of Medicine, Seoul 06273, Korea;
| | - Jongki Hong
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea
- College of Pharmacy, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (J.H.); (B.C.C.); Tel.: +82-2-961-9255 (J.H.); +82-2-958-5067 (B.C.C.)
| | - Bong Chul Chung
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea;
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (J.H.); (B.C.C.); Tel.: +82-2-961-9255 (J.H.); +82-2-958-5067 (B.C.C.)
| |
Collapse
|
50
|
Dalal N, Jalandra R, Sharma M, Prakash H, Makharia GK, Solanki PR, Singh R, Kumar A. Omics technologies for improved diagnosis and treatment of colorectal cancer: Technical advancement and major perspectives. Biomed Pharmacother 2020; 131:110648. [PMID: 33152902 DOI: 10.1016/j.biopha.2020.110648] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/09/2020] [Accepted: 08/16/2020] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) ranks third among the most commonly occurring cancers worldwide, and it causes half a million deaths annually. Alongside mechanistic study for CRC detection and treatment by conventional techniques, new technologies have been developed to study CRC. These technologies include genomics, transcriptomics, proteomics, and metabolomics which elucidate DNA markers, RNA transcripts, protein and, metabolites produced inside the colon and rectum part of the gut. All these approaches form the omics arena, which presents a remarkable opportunity for the discovery of novel prognostic, diagnostic and therapeutic biomarkers and also delineate the underlying mechanism of CRC causation, which may further help in devising treatment strategies. This review also mentions the latest developments in metagenomics and culturomics as emerging evidence suggests that metagenomics of gut microbiota has profound implications in the causation, prognosis, and treatment of CRC. A majority of bacteria cannot be studied as they remain unculturable, so culturomics has also been strengthened to develop culture conditions suitable for the growth of unculturable bacteria and identify unknown bacteria. The overall purpose of this review is to succinctly evaluate the application of omics technologies in colorectal cancer research for improving the diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Nishu Dalal
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India; Department of Environmental Science, Satyawati College, Delhi University, Delhi 110052, India
| | - Rekha Jalandra
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India; Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India
| | - Minakshi Sharma
- Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India
| | - Hridayesh Prakash
- Amity Institute of Virology and Immunology, Amity University, Sector 125, Noida 201313, Uttar Pradesh, India
| | - Govind K Makharia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Pratima R Solanki
- Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajeev Singh
- Department of Environmental Science, Satyawati College, Delhi University, Delhi 110052, India.
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India.
| |
Collapse
|