15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Spatially resolved metabolomics to discover tumor-associated metabolic alterations

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Significance

          Tumor cells reprogram their metabolism to support cell growth, proliferation, and differentiation, thus driving cancer progression. Profiling of the metabolic signatures in heterogeneous tumors facilitates the understanding of tumor metabolism and introduces potential metabolic vulnerabilities that might be targeted therapeutically. We proposed a spatially resolved metabolomics method for high-throughput discovery of tumor-associated metabolite and enzyme alterations using ambient mass spectrometry imaging. Metabolic pathway-related metabolites and metabolic enzymes that are associated with tumor metabolism were efficiently discovered and visualized in heterogeneous esophageal cancer tissues. Spatially resolved metabolic alterations hold the key to defining the dependencies of metabolism that are most limiting for cancer growth and exploring metabolic targeted strategies for better cancer treatment.

          Abstract

          Characterization of tumor metabolism with spatial information contributes to our understanding of complex cancer metabolic reprogramming, facilitating the discovery of potential metabolic vulnerabilities that might be targeted for tumor therapy. However, given the metabolic variability and flexibility of tumors, it is still challenging to characterize global metabolic alterations in heterogeneous cancer. Here, we propose a spatially resolved metabolomics approach to discover tumor-associated metabolites and metabolic enzymes directly in their native state. A variety of metabolites localized in different metabolic pathways were mapped by airflow-assisted desorption electrospray ionization mass spectrometry imaging (AFADESI-MSI) in tissues from 256 esophageal cancer patients. In combination with in situ metabolomics analysis, this method provided clues into tumor-associated metabolic pathways, including proline biosynthesis, glutamine metabolism, uridine metabolism, histidine metabolism, fatty acid biosynthesis, and polyamine biosynthesis. Six abnormally expressed metabolic enzymes that are closely associated with the altered metabolic pathways were further discovered in esophageal squamous cell carcinoma (ESCC). Notably, pyrroline-5-carboxylate reductase 2 (PYCR2) and uridine phosphorylase 1 (UPase1) were found to be altered in ESCC. The spatially resolved metabolomics reveal what occurs in cancer at the molecular level, from metabolites to enzymes, and thus provide insights into the understanding of cancer metabolic reprogramming.

          Related collections

          Most cited references28

          • Record: found
          • Abstract: found
          • Article: not found

          Laser ablation electrospray ionization for atmospheric pressure, in vivo, and imaging mass spectrometry.

          Mass spectrometric analysis of biomolecules under ambient conditions promises to enable the in vivo investigation of diverse biochemical changes in organisms with high specificity. Here we report on a novel combination of infrared laser ablation with electrospray ionization (LAESI) as an ambient ion source for mass spectrometry. As a result of the interactions between the ablation plume and the spray, LAESI accomplishes electrospray-like ionization. Without any sample preparation or pretreatment, this technique was capable of detecting a variety of molecular classes and size ranges (up to 66 kDa) with a detection limit of 8 and 25 fmol for verapamil and reserpine, respectively, and quantitation capabilities with a four-decade dynamic range. We demonstrated the utility of LAESI in a broad variety of applications ranging from plant biology to clinical analysis. Proteins, lipids, and metabolites were identified, and antihistamine excretion was followed via the direct analysis of bodily fluids (urine, blood, and serum). We also performed in vivo spatial profiling (on leaf, stem, and root) of metabolites in a French marigold (Tagetes patula) seedling.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Atmospheric pressure MALDI mass spectrometry imaging of tissues and cells at 1.4-μm lateral resolution

            An instrumental setup for atmospheric pressure MALDI-based mass spectrometry imaging with improved lateral resolution enables subcellular-level details to be resolved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              FDR-controlled metabolite annotation for high-resolution imaging mass spectrometry

              The authors present a computational framework for false-discovery-rate-controlled metabolite annotation from high-resolution imaging mass spectrometry data.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                2 January 2019
                17 December 2018
                17 December 2018
                : 116
                : 1
                : 52-57
                Affiliations
                [1] aState Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , 100050 Beijing, China;
                [2] bDepartment of Radiation Oncology, Cancer Institute, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , 100021 Beijing, China;
                [3] cDepartment of Pathology and Thoracic Surgery, Linzhou Esophageal Cancer Hospital , 456500 Linzhou, China;
                [4] dCenter for Imaging and Systems Biology, Minzu University of China , 100081 Beijing, China;
                [5] eState Key Laboratory of Molecular Oncology, Cancer Institute, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , 100021 Beijing, China
                Author notes
                1To whom correspondence may be addressed. Email: hejiuming@ 123456imm.ac.cn or zeper@ 123456imm.ac.cn .

                Edited by Matthew G. Vander Heiden, Koch Institute at Massachusetts Institute of Technology, and accepted by Editorial Board Member Rakesh K. Jain November 27, 2018 (received for review May 24, 2018)

                Author contributions: J.H. and Z.A. designed research; C.S., L.H., Q.Z., N.B., G.J., Y.H., R.Z., Z.L., and L.W. performed research; C.S., T.L., X.S., J.X., Y.C., X.L., Z.W., Y.S., J.H., and Z.A. analyzed data; and C.S., J.H., and Z.A. wrote the paper.

                Article
                201808950
                10.1073/pnas.1808950116
                6320512
                30559182
                44921fc7-943f-4601-8329-23e86af1d296
                Copyright © 2019 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 6
                Funding
                Funded by: National Natural Science Foundation of China (NSFC) 501100001809
                Award ID: 21335007
                Award Recipient : Qingce Zang Award Recipient : Jing Xu Award Recipient : Yanhua Chen Award Recipient : Ruiping Zhang Award Recipient : Zhigang Luo Award Recipient : Jiuming He Award Recipient : Zeper Abliz
                Funded by: National Natural Science Foundation of China (NSFC) 501100001809
                Award ID: 81773678
                Award Recipient : Qingce Zang Award Recipient : Jing Xu Award Recipient : Yanhua Chen Award Recipient : Ruiping Zhang Award Recipient : Zhigang Luo Award Recipient : Jiuming He Award Recipient : Zeper Abliz
                Funded by: The Leading Talent Support Program of State Ethnic Affairs Commission, and Program of Collaborative Innovation Center for Ethnic Minority Development of Minzu University of China
                Award ID: 017004040501
                Award Recipient : Zhigang Luo Award Recipient : Zhonghua Wang Award Recipient : Jiuming He Award Recipient : Zeper Abliz
                Funded by: The National Instrumentation Program
                Award ID: 2016YFF0100304
                Award Recipient : Zhigang Luo Award Recipient : Zhonghua Wang Award Recipient : Jiuming He Award Recipient : Zeper Abliz
                Categories
                Physical Sciences
                Chemistry

                mass spectrometry imaging,metabolomics,esophageal cancer,metabolic alterations,airflow-assisted ionization

                Comments

                Comment on this article