51
|
Schäfermann J, Kliewer G, Lösch J, Bednarz H, Giampà M, Niehaus K. Immersion by rotation-based application of the matrix for fast and reproducible sample preparations and robust results in mass spectrometry imaging. JOURNAL OF MASS SPECTROMETRY : JMS 2020; 55:e4488. [PMID: 31826308 DOI: 10.1002/jms.4488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 11/22/2019] [Accepted: 12/08/2019] [Indexed: 06/10/2023]
Abstract
Automated matrix deposition for matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) is crucial for producing reproducible analyte ion signals. Here we report an innovative method employing an automated immersion apparatus, which enables a robust matrix deposition within 5 minutes and with scalable throughput by using MAPS matrix and non-polar solvents. MSI results received from mouse heart and rat brain tissues were qualitatively similar to those from nozzle sprayed samples with respect to peak number and quality of the ion images. Overall, the immersion-method enables a fast and careful matrix deposition and has the future potential for implementation in clinical tissue diagnostics.
Collapse
Affiliation(s)
- Johanna Schäfermann
- MSI Diagnostics GmbH, Bielefeld, Germany
- Proteome and Metabolome Research, Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Georg Kliewer
- MSI Diagnostics GmbH, Bielefeld, Germany
- Proteome and Metabolome Research, Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | | | - Hanna Bednarz
- Center for Biotechnology, Bielefeld University, Bielefeld, Germany
- Proteome and Metabolome Research, Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - Marco Giampà
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Karsten Niehaus
- Center for Biotechnology, Bielefeld University, Bielefeld, Germany
- Proteome and Metabolome Research, Faculty of Biology, Bielefeld University, Bielefeld, Germany
| |
Collapse
|
52
|
Josenhans C, Müthing J, Elling L, Bartfeld S, Schmidt H. How bacterial pathogens of the gastrointestinal tract use the mucosal glyco-code to harness mucus and microbiota: New ways to study an ancient bag of tricks. Int J Med Microbiol 2020; 310:151392. [DOI: 10.1016/j.ijmm.2020.151392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/28/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
|
53
|
Li N, Wang P, Liu X, Han C, Ren W, Li T, Li X, Tao F, Zhao Z. Developing IR-780 as a Novel Matrix for Enhanced MALDI MS Imaging of Endogenous High-Molecular-Weight Lipids in Brain Tissues. Anal Chem 2019; 91:15873-15882. [PMID: 31718156 DOI: 10.1021/acs.analchem.9b04315] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The matrix plays a prominent role in expanding the ability of matrix assisted laser desorption/ionization mass spectrometry (MALDI MS). However, on account of the unclarity of necessary properties of the matrix in MALDI MS, development of a new matrix is still in the exploratory stage and lacks systematic theoretical guidance. Meanwhile, most of the existing matrices are unable to simultaneously detect various high-molecular-weight (high-MW) lipids including (poly-)phosphoinositides, cardiolipins, and gangliosides. In this study, we have successfully screened and optimized the application of commercially available IR-780 as a novel matrix for simultaneously profiling and imaging high-MW lipids in brain tissues by MALDI MS for the first time. The properties of IR-780 related to the matrix of MALDI MS, mainly including the optical properties (UV absorption, fluorescence emission, and photothermal efficiency), proton affinity, collision cross-sections (CCSs), salt-tolerance ability, and homogeneity, were comprehensively characterized, which demonstrated that high photothermal ability and large CCSs might guarantee the superior performance of IR-780 as matrix for the analysis of high-MW lipids in biological samples. This work provided some references for the development of a novel matrix, and especially, the concept of CCS was first introduced as a parameter for the development of a matrix. In addition, the simultaneous identification and imaging of endogenous high-MW lipids in rat brain tissues subjected to traumatic brain injury were successfully performed.
Collapse
Affiliation(s)
- Na Li
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing Mass Spectrum Center, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China
- Graduate School , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Peng Wang
- College of Biochemistry Engineering , Beijing Union University , Beijing 100023 , China
| | - Xiaolong Liu
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing Mass Spectrum Center, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China
| | - Chao Han
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing Mass Spectrum Center, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China
- Graduate School , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Wei Ren
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing Mass Spectrum Center, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China
- Graduate School , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Tuo Li
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing Mass Spectrum Center, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China
- Graduate School , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Xing Li
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing Mass Spectrum Center, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China
- Graduate School , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Fengyun Tao
- College of Biochemistry Engineering , Beijing Union University , Beijing 100023 , China
| | - Zhenwen Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing Mass Spectrum Center, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China
- Graduate School , University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
54
|
Confirmation of sub-cellular resolution using oversampling imaging mass spectrometry. Anal Bioanal Chem 2019; 411:7935-7941. [DOI: 10.1007/s00216-019-02212-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/04/2019] [Accepted: 10/11/2019] [Indexed: 01/26/2023]
|
55
|
Fung AWS, Sugumar V, Ren AH, Kulasingam V. Emerging role of clinical mass spectrometry in pathology. J Clin Pathol 2019; 73:61-69. [PMID: 31690564 DOI: 10.1136/jclinpath-2019-206269] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 10/14/2019] [Indexed: 12/20/2022]
Abstract
Mass spectrometry-based assays have been increasingly implemented in various disciplines in clinical diagnostic laboratories for their combined advantages in multiplexing capacity and high analytical specificity and sensitivity. It is now routinely used in areas including reference methods development, therapeutic drug monitoring, toxicology, endocrinology, paediatrics, immunology and microbiology to identify and quantify biomolecules in a variety of biological specimens. As new ionisation methods, instrumentation and techniques are continuously being improved and developed, novel mass spectrometry-based clinical applications will emerge for areas such as proteomics, metabolomics, haematology and anatomical pathology. This review will summarise the general principles of mass spectrometry and specifically highlight current and future clinical applications in anatomical pathology.
Collapse
Affiliation(s)
- Angela W S Fung
- Department of Pathology and Laboratory Medicine, St Paul's Hospital, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Vijithan Sugumar
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Annie He Ren
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Vathany Kulasingam
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada .,Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
56
|
L'Imperio V, Smith A, Pisani A, D'Armiento M, Scollo V, Casano S, Sinico RA, Nebuloni M, Tosoni A, Pieruzzi F, Magni F, Pagni F. MALDI imaging in Fabry nephropathy: a multicenter study. J Nephrol 2019; 33:299-306. [PMID: 31292888 DOI: 10.1007/s40620-019-00627-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/25/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND The current study evaluates the application of histology and in situ proteomics (MALDI-MSI) in Fabry nephropathy (FN), showing investigative and classification role for this coupled approach. METHODS A retrospective series of 14 formalin fixed paraffin embedded (FFPE) renal biopsies with diagnosis of FN and 1 biopsy from a patient bearing a galactosidase-α (GLA) genetic variant of unknown significance (GVUS, c.376A>G) have been classified for clinical characteristics. Groups were compared for histological differences (following the ISGFN scoring system). Moreover, renal biopsies from these cases have been analyzed with MALDI-MSI as previously described to find proteomic signatures among different mutations and phenotypes. RESULTS Comparison of clinical features revealed lower mean 24 h proteinuria in females (225 mg/24 h) than in males (1477.5 mg/24 h, p = 0.006). As for clinical characteristics, females significantly differed from males only for lower arterial sclerosis, with a mean value of 0.82 vs. 1.05 (p = 0.001). Proteomic analysis demonstrated specific signatures in different subgroups of FN patients. Moreover, MALDI correctly classified cases with undetermined mutation or GVUS. CONCLUSIONS The present study demonstrated the feasible application of MALDI-MSI in the analysis of FN FFPE renal biopsies, allowing the detection of putative signatures for phenotypic distinction and demonstrating genetic classification capabilities.
Collapse
Affiliation(s)
- Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Andrew Smith
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Monza, Italy
| | - Antonio Pisani
- Chair of Nephrology, University Federico II, Naples, Italy
| | - Maria D'Armiento
- Department of Biomorphological and Functional Sciences, Section of Anatomic Pathology, Federico II University, Naples, Italy
| | - Viviana Scollo
- Department of Medicine and Surgery, Nephrology Unit, University of Milano-Bicocca, Monza, Italy
| | - Stefano Casano
- Department of Medicine and Surgery, Pathology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Renato Alberto Sinico
- Department of Medicine and Surgery, Nephrology Unit, University of Milano-Bicocca, Monza, Italy
| | - Manuela Nebuloni
- Research Center for Renal Immunopathology, University of Milan, Milan, Italy.,Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, Milan, Italy
| | - Antonella Tosoni
- Research Center for Renal Immunopathology, University of Milan, Milan, Italy.,Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, Milan, Italy
| | - Federico Pieruzzi
- Department of Medicine and Surgery, Nephrology Unit, University of Milano-Bicocca, Monza, Italy
| | - Fulvio Magni
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Monza, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy. .,Research Center for Renal Immunopathology, University of Milan, Milan, Italy.
| |
Collapse
|
57
|
Wang J, Wang C, Han X. Tutorial on lipidomics. Anal Chim Acta 2019; 1061:28-41. [PMID: 30926037 PMCID: PMC7375172 DOI: 10.1016/j.aca.2019.01.043] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 12/20/2022]
Abstract
The mainstream of lipidomics involves mass spectrometry-based, systematic, and large-scale studies of the structure, composition, and quantity of lipids in biological systems such as organs, cells, and body fluids. As increasingly more researchers in broad fields are beginning to pay attention to and actively learn about the lipidomic technology, some introduction on the topic is needed to help the newcomers to better understand the field. This tutorial seeks to introduce the basic knowledge about lipidomics and to provide readers with some core ideas and the most important approaches for studying the field.
Collapse
Affiliation(s)
- Jianing Wang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Chunyan Wang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; Department of Medicine - Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
58
|
Chen K, Baluya D, Tosun M, Li F, Maletic-Savatic M. Imaging Mass Spectrometry: A New Tool to Assess Molecular Underpinnings of Neurodegeneration. Metabolites 2019; 9:E135. [PMID: 31295847 PMCID: PMC6681116 DOI: 10.3390/metabo9070135] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/19/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are prevalent and devastating. While extensive research has been done over the past decades, we are still far from comprehensively understanding what causes neurodegeneration and how we can prevent it or reverse it. Recently, systems biology approaches have led to a holistic examination of the interactions between genome, metabolome, and the environment, in order to shed new light on neurodegenerative pathogenesis. One of the new technologies that has emerged to facilitate such studies is imaging mass spectrometry (IMS). With its ability to map a wide range of small molecules with high spatial resolution, coupled with the ability to quantify them at once, without the need for a priori labeling, IMS has taken center stage in current research efforts in elucidating the role of the metabolome in driving neurodegeneration. IMS has already proven to be effective in investigating the lipidome and the proteome of various neurodegenerative diseases, such as Alzheimer's, Parkinson's, Huntington's, multiple sclerosis, and amyotrophic lateral sclerosis. Here, we review the IMS platform for capturing biological snapshots of the metabolic state to shed more light on the molecular mechanisms of the diseased brain.
Collapse
Affiliation(s)
- Kevin Chen
- Department of Biosciences, Rice University, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Dodge Baluya
- Chemical Imaging Research Core at MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Mehmet Tosun
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Feng Li
- Center for Drug Discovery and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mirjana Maletic-Savatic
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA.
- Department of Neuroscience and Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
59
|
Castellanos A, Ramirez CE, Michalkova V, Nouzova M, Noriega FG, Francisco FL. Three Dimensional Secondary Ion Mass Spectrometry Imaging (3D-SIMS) of Aedes aegypti ovarian follicles. JOURNAL OF ANALYTICAL ATOMIC SPECTROMETRY 2019; 34:874-883. [PMID: 31680712 PMCID: PMC6824543 DOI: 10.1039/c8ja00425k] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The mobilization of nutrient reserves into the ovaries of Aedes aegypti mosquitoes after sugar-feeding plays a vital role in the female's reproductive maturation. In the present work, three-dimensional secondary ion mass spectrometry imaging (3D-SIMS) was used to generate ultrahigh spatial resolution (~1 μm) chemical maps and study the composition and spatial distribution of lipids at the single ovarian follicle level (~100 μm in size). 3D-Mass Spectrometry Imaging (3D-MSI) allowed the identification of cellular types in the follicle (oocyte, nurse and follicular cells) using endogenous markers, and revealed that most of the triacyglycerides (TGs) were compartmentalized in the oocyte region. By comparing follicles from water-fed and sugar-fed females (n=2), 3D-MSI-Time of Flight-SIMS showed that TGs were more abundant in ovarian follicles of sugar-fed females; despite relative sample reproducibility per feeding condition, more biological replicates will better support the trends observed. While the current 3D-MSI-TOF-SIMS does not permit MS/MS analysis of the lipid species, complementary LC-MS/MS analysis of the ovarian follicles aided tentative lipid assignments of the SIMS data. The combination of these MS approaches is giving us a first glimpse of the distribution of functionally relevant ovarian lipid molecules at the cellular level. These new tools can be used to investigate the roles of different lipids on follicle fitness and overall mosquito reproductive output.
Collapse
Affiliation(s)
- Anthony Castellanos
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, United States
| | - Cesar E. Ramirez
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, United States
| | - Veronika Michalkova
- Department of Biological Sciences, Florida International University, Miami, Florida, 33199, United States
| | - Marcela Nouzova
- Department of Biological Sciences, Florida International University, Miami, Florida, 33199, United States
- Institute of Parasitology, Biology Centre CAS, Ceske, Budejovice, Czech Republic; and
| | - Fernando G. Noriega
- Department of Biological Sciences, Florida International University, Miami, Florida, 33199, United States
- Biomolecular Sciences Institute, Florida International University, Miami, Florida, 33199, United States
| | - Fernández-Lima Francisco
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, 33199, United States
- Biomolecular Sciences Institute, Florida International University, Miami, Florida, 33199, United States
- Corresponding author: Francisco A. Fernández-Lima, Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th St AHC4-233, Miami, FL 33199, USA;
| |
Collapse
|
60
|
Liao Y, Fu X, Zhou H, Rao W, Zeng L, Yang Z. Visualized analysis of within-tissue spatial distribution of specialized metabolites in tea (Camellia sinensis) using desorption electrospray ionization imaging mass spectrometry. Food Chem 2019; 292:204-210. [PMID: 31054666 DOI: 10.1016/j.foodchem.2019.04.055] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022]
Abstract
Although specialized metabolite distributions in different tea (Camellia sinensis) tissues has been studied extensively, little is known about their within-tissue distribution owing to the lack of nondestructive methodology. In this study, desorption electrospray ionization imaging mass spectrometry was used to investigate the within-tissue spatial distributions of specialized metabolites in tea. To overcome the negative effects of the large amount of wax on tea leaves, several sample preparation methods were compared, with a Teflon-imprint method established for tea leaves. Polyphenols are characteristic metabolites in tea leaves. Epicatechin gallate/catechin gallate, epigallocatechin gallate/gallocatechin gallate, and gallic acid were evenly distributed on both sides of the leaves, while epicatechin/catechin, epigallocatechin/gallocatechin, and assamicain A were distributed near the leaf vein. L-Theanine was mainly accumulated in tea roots. L-Theanine and valinol were distributed around the outer root cross-section. The results will advance our understanding of the precise localizations and in-vivo biosyntheses of specialized metabolites in tea.
Collapse
Affiliation(s)
- Yinyin Liao
- Guangdong Provincial Key Laboratory of Applied Botany & Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Xingke Road 723, Tianhe District, Guangzhou 510650, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiumin Fu
- Guangdong Provincial Key Laboratory of Applied Botany & Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Xingke Road 723, Tianhe District, Guangzhou 510650, China
| | - Haiyun Zhou
- Instrumental Analysis & Research Center, Sun Yat-Sen University, No. 135, Xingang Xi Road, Guangzhou 510275, China
| | - Wei Rao
- Waters Technologies (Shanghai) Ltd., No. 1000 Jinhai Road, Shanghai 201203, China
| | - Lanting Zeng
- Guangdong Provincial Key Laboratory of Applied Botany & Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Xingke Road 723, Tianhe District, Guangzhou 510650, China
| | - Ziyin Yang
- Guangdong Provincial Key Laboratory of Applied Botany & Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Xingke Road 723, Tianhe District, Guangzhou 510650, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China.
| |
Collapse
|
61
|
Dickmeis T, Feng Y, Mione MC, Ninov N, Santoro M, Spaink HP, Gut P. Nano-Sampling and Reporter Tools to Study Metabolic Regulation in Zebrafish. Front Cell Dev Biol 2019; 7:15. [PMID: 30873407 PMCID: PMC6401643 DOI: 10.3389/fcell.2019.00015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
In the past years, evidence has emerged that hallmarks of human metabolic disorders can be recapitulated in zebrafish using genetic, pharmacological or dietary interventions. An advantage of modeling metabolic diseases in zebrafish compared to other "lower organisms" is the presence of a vertebrate body plan providing the possibility to study the tissue-intrinsic processes preceding the loss of metabolic homeostasis. While the small size of zebrafish is advantageous in many aspects, it also has shortcomings such as the difficulty to obtain sufficient amounts for biochemical analyses in response to metabolic challenges. A workshop at the European Zebrafish Principal Investigator meeting in Trento, Italy, was dedicated to discuss the advantages and disadvantages of zebrafish to study metabolic disorders. This perspective article by the participants highlights strategies to achieve improved tissue-resolution for read-outs using "nano-sampling" approaches for metabolomics as well as live imaging of zebrafish expressing fluorescent reporter tools that inform on cellular or subcellular metabolic processes. We provide several examples, including the use of reporter tools to study the heterogeneity of pancreatic beta-cells within their tissue environment. While limitations exist, we believe that with the advent of new technologies and more labs developing methods that can be applied to minimal amounts of tissue or single cells, zebrafish will further increase their utility to study energy metabolism.
Collapse
Affiliation(s)
- Thomas Dickmeis
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Yi Feng
- Centre for Inflammation Research, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland
| | | | - Nikolay Ninov
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | | | - Herman P. Spaink
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Philipp Gut
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| |
Collapse
|
62
|
Tang W, Chen J, Zhou J, Ge J, Zhang Y, Li P, Li B. Quantitative MALDI Imaging of Spatial Distributions and Dynamic Changes of Tetrandrine in Multiple Organs of Rats. Am J Cancer Res 2019; 9:932-944. [PMID: 30867807 PMCID: PMC6401406 DOI: 10.7150/thno.30408] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/22/2018] [Indexed: 12/16/2022] Open
Abstract
Detailed spatio-temporal information on drug distribution in organs is of paramount importance to assess drug clinically-relevant properties and potential side-effects. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) as a label-free and sensitive imaging modality provides an additional means of accurately visualizing drug and its metabolites distributions in tissue sections. However, technical limitations, complex physiochemical environment of surface and low abundance of target drugs make quantitative MALDI imaging of drug and its metabolites quite challenging. Methods: In this study, an internal standard correction strategy was applied for quantitative MALDI imaging of tetrandrine in multiple organs of rats including lung, liver, kidney, spleen, and heart. The feasibility and reliability of the developed quantitative MSI method were validated by conventional liquid chromatography-tandem MS (LC-MS/MS) analysis, and the two methods showed a significant correlation. Results: The quantitative MALDI imaging method met the requirements of specificity, sensitivity and linearity. Tissue-specific spatio-temporal distribution patterns of tetrandrine in different organs were revealed after intravenous administration in the rat. Moreover, demethylated metabolite was detected in liver tissues. Conclusions: The current work illustrates that quantitative MALDI imaging provides an alternative means of accurately addressing the problem of drug and its metabolites distribution in tissues, complementary to traditional LC-MS/MS of tissue homogenates and whole-body autoradiography (WBA). Quantitative spatio-chemical information obtained here can improve our understanding of pharmacokinetics (PK), pharmacodynamics (PD), and potential transient toxicities of tetrandrine in organs, and possibly direct further optimization of drug properties to reduce drug-induced organ toxicity.
Collapse
|
63
|
Neagu AN. Proteome Imaging: From Classic to Modern Mass Spectrometry-Based Molecular Histology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1140:55-98. [PMID: 31347042 DOI: 10.1007/978-3-030-15950-4_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In order to overcome the limitations of classic imaging in Histology during the actually era of multiomics, the multi-color "molecular microscope" by its emerging "molecular pictures" offers quantitative and spatial information about thousands of molecular profiles without labeling of potential targets. Healthy and diseased human tissues, as well as those of diverse invertebrate and vertebrate animal models, including genetically engineered species and cultured cells, can be easily analyzed by histology-directed MALDI imaging mass spectrometry. The aims of this review are to discuss a range of proteomic information emerging from MALDI mass spectrometry imaging comparative to classic histology, histochemistry and immunohistochemistry, with applications in biology and medicine, concerning the detection and distribution of structural proteins and biological active molecules, such as antimicrobial peptides and proteins, allergens, neurotransmitters and hormones, enzymes, growth factors, toxins and others. The molecular imaging is very well suited for discovery and validation of candidate protein biomarkers in neuroproteomics, oncoproteomics, aging and age-related diseases, parasitoproteomics, forensic, and ecotoxicology. Additionally, in situ proteome imaging may help to elucidate the physiological and pathological mechanisms involved in developmental biology, reproductive research, amyloidogenesis, tumorigenesis, wound healing, neural network regeneration, matrix mineralization, apoptosis and oxidative stress, pain tolerance, cell cycle and transformation under oncogenic stress, tumor heterogeneity, behavior and aggressiveness, drugs bioaccumulation and biotransformation, organism's reaction against environmental penetrating xenobiotics, immune signaling, assessment of integrity and functionality of tissue barriers, behavioral biology, and molecular origins of diseases. MALDI MSI is certainly a valuable tool for personalized medicine and "Eco-Evo-Devo" integrative biology in the current context of global environmental challenges.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, "Alexandru Ioan Cuza" University of Iasi, Iasi, Romania.
| |
Collapse
|
64
|
Xu G, Li J. Recent advances in mass spectrometry imaging for multiomics application in neurology. J Comp Neurol 2018; 527:2158-2169. [DOI: 10.1002/cne.24571] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/14/2018] [Accepted: 10/24/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Guang Xu
- Hubei Education Cloud Service Engineering Technology Research CenterHubei University of Education Wuhan China
| | - Jianjun Li
- Human Health TherapeuticsNational Research Council Canada Ottawa Ontario
| |
Collapse
|
65
|
Smith A, L'Imperio V, Denti V, Mazza M, Ivanova M, Stella M, Piga I, Chinello C, Ajello E, Pieruzzi F, Pagni F, Magni F. High Spatial Resolution MALDI-MS Imaging in the Study of Membranous Nephropathy. Proteomics Clin Appl 2018; 13:e1800016. [PMID: 30548219 DOI: 10.1002/prca.201800016] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 11/30/2018] [Indexed: 12/15/2022]
Abstract
PURPOSE Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) technology has advanced rapidly during recent years with the development of instruments equipped with low-diameter lasers that are suitable for high spatial resolution imaging. This may provide significant advantages in certain fields of molecular pathology where more specific protein fingerprints of individual cell types are required, such as renal pathology. EXPERIMENTAL DESIGN Here MALDI-MSI analysis of a cohort of membranous nephropathy (MN) patients is performed among which patients either responded favorably (R; n = 6), or unfavorably (NR; n = 4), to immunosuppressive treatment (Ponticelli Regimen), employing a 10 µm laser spot diameter. RESULTS Specific tryptic peptide profiles of the different cellular regions within the glomerulus can be generated, similarly for the epithelial cells belonging to the proximal and distal tubules. Conversely, specific glomerular and sub-glomerular profiles cannot be obtained while using the pixel size performed in previous studies (50 µm). Furthermore, two proteins are highlighted, sonic hedgehog and α-smooth muscle actin, whose signal intensity and spatial localization within the sub-glomerular and tubulointerstitial compartments differ between treatment responders and non-responders. CONCLUSIONS AND CLINICAL RELEVANCE The present study exemplifies the advantage of using high spatial resolution MALDI-MSI for the study of MN and highlights that such findings have the potential to provide complementary support in the routine prognostic assessment of MN patients.
Collapse
Affiliation(s)
- Andrew Smith
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Vincenzo L'Imperio
- San Gerardo Hospital, Department of Medicine and Surgery, Pathology, University of Milano-Bicocca, Monza, 20900, Italy
| | - Vanna Denti
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Mariafrancesca Mazza
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Mariia Ivanova
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Martina Stella
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Isabella Piga
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Clizia Chinello
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Elena Ajello
- Department of Medicine and Surgery, Nephrology Unit, University of Milano-Bicocca, Monza, 20900, Italy
| | - Federico Pieruzzi
- Department of Medicine and Surgery, Nephrology Unit, University of Milano-Bicocca, Monza, 20900, Italy
| | - Fabio Pagni
- San Gerardo Hospital, Department of Medicine and Surgery, Pathology, University of Milano-Bicocca, Monza, 20900, Italy
| | - Fulvio Magni
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| |
Collapse
|
66
|
Corinti D, Crestoni ME, Fornarini S, Pieper M, Niehaus K, Giampà M. An integrated approach to study novel properties of a MALDI matrix (4-maleicanhydridoproton sponge) for MS imaging analyses. Anal Bioanal Chem 2018; 411:953-964. [DOI: 10.1007/s00216-018-1531-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/22/2018] [Accepted: 11/29/2018] [Indexed: 02/02/2023]
|
67
|
Leopold J, Popkova Y, Engel KM, Schiller J. Recent Developments of Useful MALDI Matrices for the Mass Spectrometric Characterization of Lipids. Biomolecules 2018; 8:biom8040173. [PMID: 30551655 PMCID: PMC6316665 DOI: 10.3390/biom8040173] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 12/24/2022] Open
Abstract
Matrix-assisted laser desorption/ionization (MALDI) is one of the most successful “soft” ionization methods in the field of mass spectrometry and enables the analysis of a broad range of molecules, including lipids. Although the details of the ionization process are still unknown, the importance of the matrix is commonly accepted. Both, the development of and the search for useful matrices was, and still is, an empirical process, since properties like vacuum stability, high absorption at the laser wavelength, etc. have to be fulfilled by a compound to become a useful matrix. This review provides a survey of successfully used MALDI matrices for the lipid analyses of complex biological samples. The advantages and drawbacks of the established organic matrix molecules (cinnamic or benzoic acid derivatives), liquid crystalline matrices, and mixtures of common matrices will be discussed. Furthermore, we will deal with nanocrystalline matrices, which are most suitable to analyze small molecules, such as free fatty acids. It will be shown that the analysis of mixtures and the quantitative analysis of small molecules can be easily performed if the matrix is carefully selected. Finally, some basic principles of how useful matrix compounds can be “designed” de novo will be introduced.
Collapse
Affiliation(s)
- Jenny Leopold
- Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstr. 16/18, Leipzig University, D-04107 Leipzig, Germany.
| | - Yulia Popkova
- Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstr. 16/18, Leipzig University, D-04107 Leipzig, Germany.
| | - Kathrin M Engel
- Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstr. 16/18, Leipzig University, D-04107 Leipzig, Germany.
| | - Jürgen Schiller
- Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstr. 16/18, Leipzig University, D-04107 Leipzig, Germany.
| |
Collapse
|
68
|
Vaysse PM, Heeren RMA, Porta T, Balluff B. Mass spectrometry imaging for clinical research - latest developments, applications, and current limitations. Analyst 2018. [PMID: 28642940 DOI: 10.1039/c7an00565b] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mass spectrometry is being used in many clinical research areas ranging from toxicology to personalized medicine. Of all the mass spectrometry techniques, mass spectrometry imaging (MSI), in particular, has continuously grown towards clinical acceptance. Significant technological and methodological improvements have contributed to enhance the performance of MSI recently, pushing the limits of throughput, spatial resolution, and sensitivity. This has stimulated the spread of MSI usage across various biomedical research areas such as oncology, neurological disorders, cardiology, and rheumatology, just to name a few. After highlighting the latest major developments and applications touching all aspects of translational research (i.e. from early pre-clinical to clinical research), we will discuss the present challenges in translational research performed with MSI: data management and analysis, molecular coverage and identification capabilities, and finally, reproducibility across multiple research centers, which is the largest remaining obstacle in moving MSI towards clinical routine.
Collapse
Affiliation(s)
- Pierre-Maxence Vaysse
- Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| | - Ron M A Heeren
- Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| | - Tiffany Porta
- Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| | - Benjamin Balluff
- Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|
69
|
Fernández R, Garate J, Martín-Saiz L, Galetich I, Fernández JA. Matrix Sublimation Device for MALDI Mass Spectrometry Imaging. Anal Chem 2018; 91:803-807. [DOI: 10.1021/acs.analchem.8b04765] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Roberto Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio
Sarriena S/N, Leioa 48940, Spain
| | - Jone Garate
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio
Sarriena S/N, Leioa 48940, Spain
| | - Lucia Martín-Saiz
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio
Sarriena S/N, Leioa 48940, Spain
| | - Igor Galetich
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio
Sarriena S/N, Leioa 48940, Spain
| | - José A. Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio
Sarriena S/N, Leioa 48940, Spain
| |
Collapse
|
70
|
Himmel LE, Hackett TA, Moore JL, Adams WR, Thomas G, Novitskaya T, Caprioli RM, Zijlstra A, Mahadevan-Jansen A, Boyd KL. Beyond the H&E: Advanced Technologies for in situ Tissue Biomarker Imaging. ILAR J 2018; 59:51-65. [PMID: 30462242 PMCID: PMC6645175 DOI: 10.1093/ilar/ily004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/27/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023] Open
Abstract
For decades, histopathology with routine hematoxylin and eosin staining has been and remains the gold standard for reaching a morphologic diagnosis in tissue samples from humans and veterinary species. However, within the past decade, there has been exponential growth in advanced techniques for in situ tissue biomarker imaging that bridge the divide between anatomic and molecular pathology. It is now possible to simultaneously observe localization and expression magnitude of multiple protein, nucleic acid, and molecular targets in tissue sections and apply machine learning to synthesize vast, image-derived datasets. As these technologies become more sophisticated and widely available, a team-science approach involving subspecialists with medical, engineering, and physics backgrounds is critical to upholding quality and validity in studies generating these data. The purpose of this manuscript is to detail the scientific premise, tools and training, quality control, and data collection and analysis considerations needed for the most prominent advanced imaging technologies currently applied in tissue sections: immunofluorescence, in situ hybridization, laser capture microdissection, matrix-assisted laser desorption ionization imaging mass spectrometry, and spectroscopic/optical methods. We conclude with a brief overview of future directions for ex vivo and in vivo imaging techniques.
Collapse
Affiliation(s)
- Lauren E Himmel
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Troy A Hackett
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Jessica L Moore
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Wilson R Adams
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Giju Thomas
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Tatiana Novitskaya
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Richard M Caprioli
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Andries Zijlstra
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Anita Mahadevan-Jansen
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Kelli L Boyd
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| |
Collapse
|
71
|
Solihat NN, Acter T, Kim D, Plante AF, Kim S. Analyzing Solid-Phase Natural Organic Matter Using Laser Desorption Ionization Ultrahigh Resolution Mass Spectrometry. Anal Chem 2018; 91:951-957. [DOI: 10.1021/acs.analchem.8b04032] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Nissa Nurfajrin Solihat
- Department of Chemistry, Kyungpook National University, Daegu 41566, Republic of Korea
- Research Center for Biomaterials, Indonesian Institute of Sciences (LIPI), Cibinong 16911, Indonesia
| | - Thamina Acter
- Department of Chemistry, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Donghwi Kim
- Department of Chemistry, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Alain F. Plante
- University of Pennsylvania, 240 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Sunghwan Kim
- Department of Chemistry, Kyungpook National University, Daegu 41566, Republic of Korea
- Green-Nano Materials Research Center, Daegu 41566, Republic of Korea
| |
Collapse
|
72
|
Michno W, Wehrli PM, Blennow K, Zetterberg H, Hanrieder J. Molecular imaging mass spectrometry for probing protein dynamics in neurodegenerative disease pathology. J Neurochem 2018; 151:488-506. [PMID: 30040875 DOI: 10.1111/jnc.14559] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/03/2018] [Accepted: 07/12/2018] [Indexed: 12/14/2022]
Abstract
Recent advances in the understanding of basic pathological mechanisms in various neurological diseases depend directly on the development of novel bioanalytical technologies that allow sensitive and specific chemical imaging at high resolution in cells and tissues. Mass spectrometry-based molecular imaging (IMS) has gained increasing popularity in biomedical research for mapping the spatial distribution of molecular species in situ. The technology allows for comprehensive, untargeted delineation of in situ distribution profiles of metabolites, lipids, peptides and proteins. A major advantage of IMS over conventional histochemical techniques is its superior molecular specificity. Imaging mass spectrometry has therefore great potential for probing molecular regulations in CNS-derived tissues and cells for understanding neurodegenerative disease mechanism. The goal of this review is to familiarize the reader with the experimental workflow, instrumental developments and methodological challenges as well as to give a concise overview of the major advances and recent developments and applications of IMS-based protein and peptide profiling with particular focus on neurodegenerative diseases. This article is part of the Special Issue "Proteomics".
Collapse
Affiliation(s)
- Wojciech Michno
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Patrick M Wehrli
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK.,Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
73
|
Legros N, Pohlentz G, Steil D, Müthing J. Shiga toxin-glycosphingolipid interaction: Status quo of research with focus on primary human brain and kidney endothelial cells. Int J Med Microbiol 2018; 308:1073-1084. [PMID: 30224239 DOI: 10.1016/j.ijmm.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
Shiga toxin (Stx)-mediated injury of the kidneys and the brain represent the major extraintestinal complications in humans upon infection by enterohemorrhagic Escherichia coli (EHEC). Damage of renal and cerebral endothelial cells is the key event in the pathogenesis of the life-threatening hemolytic uremic syndrome (HUS). Stxs are AB5 toxins and the B-pentamers of the two clinically important Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid globotriaosylceramide (Gb3Cer, Galα4Galβ4Glcβ1Cer) and to less extent to globotetraosylceramide (Gb4Cer, GalNAcβ3Galα4Galβ4Glcβ1), which are expected to reside in lipid rafts in the plasma membrane of the human endothelium. This review summarizes the current knowledge on the Stx glycosphingolipid receptors and their lipid membrane ensemble in primary human brain microvascular endothelial cells (pHBMECs) and primary human renal glomerular endothelial cells (pHRGECs). Increasing knowledge on the precise initial molecular mechanisms by which Stxs interact with cellular targets will help to develop specific therapeutics and/or preventive measures to combat EHEC-caused diseases.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | | | - Daniel Steil
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany; Interdisciplinary Center for Clinical Research (IZKF), University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
74
|
Greco V, Piras C, Pieroni L, Ronci M, Putignani L, Roncada P, Urbani A. Applications of MALDI-TOF mass spectrometry in clinical proteomics. Expert Rev Proteomics 2018; 15:683-696. [PMID: 30058389 DOI: 10.1080/14789450.2018.1505510] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION The development of precision medicine requires advanced technologies to address the multifactorial disease stratification and to support personalized treatments. Among omics techniques, proteomics based on Mass Spectrometry (MS) is becoming increasingly relevant in clinical practice allowing a phenotypic characterization of the dynamic functional status of the organism. From this perspective, Matrix Assisted Laser Desorption Ionization Time of Flight (MALDI-TOF) MS is a suitable platform for providing a high-throughput support to clinics. Areas covered: This review aims to provide an updated overview of MALDI-TOF MS applications in clinical proteomics. The most relevant features of this analysis have been discussed, highlighting both pre-analytical and analytical factors that are crucial in proteomics studies. Particular emphasis is placed on biofluids proteomics for biomarkers discovery and on recent progresses in clinical microbiology, drug monitoring, and minimal residual disease (MRD). Expert commentary: Despite some analytical limitations, the latest technological advances together with the easiness of use, the low time and low cost consuming and the high throughput are making MALDI-TOF MS instruments very attractive for the clinical practice. These features offer a significant potential for the routine of the clinical laboratory and ultimately for personalized medicine.
Collapse
Affiliation(s)
- Viviana Greco
- a Institute of Biochemistry and Clinical Biochemistry , Università Cattolica del Sacro Cuore , Rome , Italy.,b Department of Laboratory Diagnostic and Infectious Diseases , Fondazione Policlinico Universitario Agostino Gemelli-IRCCS , Rome , Italy
| | - Cristian Piras
- c Dipartimento di Medicina Veterinaria , Università degli studi di Milano , Milano , Italy
| | - Luisa Pieroni
- d Proteomics and Metabonomics Unit , IRCCS-Fondazione Santa Lucia , Rome , Italy
| | - Maurizio Ronci
- d Proteomics and Metabonomics Unit , IRCCS-Fondazione Santa Lucia , Rome , Italy.,e Department of Medical, Oral and Biotechnological Sciences , University "G. D'Annunzio" of Chieti-Pescara , Chieti , Italy
| | - Lorenza Putignani
- f Unit of Parasitology Bambino Gesù Children's Hospital , IRCCS , Rome , Italy.,g Unit of Human Microbiome , Bambino Gesù Children's Hospital, IRCCS , Rome , Italy
| | - Paola Roncada
- h Dipartimento di Scienze della Salute , Università degli studi "Magna Græcia" di Catanzaro , Catanzaro , Italy
| | - Andrea Urbani
- a Institute of Biochemistry and Clinical Biochemistry , Università Cattolica del Sacro Cuore , Rome , Italy.,b Department of Laboratory Diagnostic and Infectious Diseases , Fondazione Policlinico Universitario Agostino Gemelli-IRCCS , Rome , Italy
| |
Collapse
|
75
|
Kaushik AK, DeBerardinis RJ. Applications of metabolomics to study cancer metabolism. Biochim Biophys Acta Rev Cancer 2018; 1870:2-14. [PMID: 29702206 PMCID: PMC6193562 DOI: 10.1016/j.bbcan.2018.04.009] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/20/2018] [Indexed: 12/13/2022]
Abstract
Reprogrammed metabolism supports tumor growth and provides a potential source of therapeutic targets and disease biomarkers. Mass spectrometry-based metabolomics has emerged as a broadly informative technique for profiling metabolic features associated with specific oncogenotypes, disease progression, therapeutic liabilities and other clinically relevant aspects of tumor biology. In this review, we introduce the applications of metabolomics to study deregulated metabolism and metabolic vulnerabilities in cancer. We provide examples of studies that used metabolomics to discover novel metabolic regulatory mechanisms, including processes that link metabolic alterations with gene expression, protein function, and other aspects of systems biology. Finally, we discuss emerging applications of metabolomics for in vivo isotope tracing and metabolite imaging, both of which hold promise to advance our understanding of the role of metabolic reprogramming in cancer.
Collapse
Affiliation(s)
- Akash K Kaushik
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. Dallas, TX 75390-8502, United States
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. Dallas, TX 75390-8502, United States.
| |
Collapse
|
76
|
Santos T, Théron L, Chambon C, Viala D, Centeno D, Esbelin J, Hébraud M. MALDI mass spectrometry imaging and in situ microproteomics of Listeria monocytogenes biofilms. J Proteomics 2018; 187:152-160. [PMID: 30071319 DOI: 10.1016/j.jprot.2018.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/16/2018] [Accepted: 07/16/2018] [Indexed: 02/08/2023]
Abstract
MALDI-TOF Mass spectrometry Imaging (MSI) is a surface-sampling technology that can determine spatial information and relative abundance of analytes directly from biological samples. Human listeriosis cases are due to the ingestion of contaminated foods with the pathogenic bacteria Listeria monocytogenes. The reduction of water availability in food workshops by decreasing the air relative humidity (RH) is one strategy to improve the control of bacterial contamination. This study aims to develop and implement an MSI approach on L. monocytogenes biofilms and proof of concept using a dehumidified stress condition. MSI allowed examining the distribution of low molecular weight proteins within the biofilms subjected to a dehumidification environment, mimicking the one present in a food workshop (10 °C, 75% RH). Furthermore, a LC-MS/MS approach was made to link the dots between MSI and protein identification. Five identified proteins were assigned to registered MSI m/z, including two cold-shock proteins and a ligase involved in cell wall biogenesis. These data demonstrate how imaging can be used to dissect the proteome of an intact bacterial biofilm giving new insights into protein expression relating to a dehumidification stress adaptation. Data are available via ProteomeXchange with identifier PXD010444. BIOLOGICAL SIGNIFICANCE The ready-to-eat food processing industry has the daily challenge of controlling the contamination of surfaces and machines with spoilage and pathogenic microorganisms. In some cases, it is a lost cause due to these microorganisms' capacity to withstand the cleaning treatments, like desiccation procedures. Such a case is the ubiquitous Gram-positive Bacterium Listeria monocytogenes. Its surface proteins have particular importance for the interaction with its environment, being important factors contributing to adaptation to stress conditions. There are few reproducibly techniques to obtain the surface proteins of Gram-positive cells. Here, we developed a workflow that enables the use of MALDI imaging on Gram-positive bacterium biofilms to study the impact of dehumidification on sessile cells. It will be of the most interest to test this workflow with different environmental conditions and potentially apply it to other biofilm-forming bacteria.
Collapse
Affiliation(s)
- Tiago Santos
- Université Clermont Auvergne, INRA, UMR MEDiS, F-63122 Saint-Genès Champanelle, France
| | - Laëtitia Théron
- INRA, Plateforme d'Exploration du Métabolisme, composante protéomique (PFEMcp), F-63122 Saint-Genès Champanelle, France
| | - Christophe Chambon
- INRA, Plateforme d'Exploration du Métabolisme, composante protéomique (PFEMcp), F-63122 Saint-Genès Champanelle, France
| | - Didier Viala
- INRA, Plateforme d'Exploration du Métabolisme, composante protéomique (PFEMcp), F-63122 Saint-Genès Champanelle, France
| | - Delphine Centeno
- INRA, Plateforme d'Exploration du Métabolisme, composante protéomique (PFEMcp), F-63122 Saint-Genès Champanelle, France
| | - Julia Esbelin
- Université Clermont Auvergne, INRA, UMR MEDiS, F-63122 Saint-Genès Champanelle, France
| | - Michel Hébraud
- Université Clermont Auvergne, INRA, UMR MEDiS, F-63122 Saint-Genès Champanelle, France; INRA, Plateforme d'Exploration du Métabolisme, composante protéomique (PFEMcp), F-63122 Saint-Genès Champanelle, France.
| |
Collapse
|
77
|
Qin L, Zhang Y, Liu Y, He H, Han M, Li Y, Zeng M, Wang X. Recent advances in matrix-assisted laser desorption/ionisation mass spectrometry imaging (MALDI-MSI) for in situ analysis of endogenous molecules in plants. PHYTOCHEMICAL ANALYSIS : PCA 2018; 29:351-364. [PMID: 29667236 DOI: 10.1002/pca.2759] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 02/01/2018] [Accepted: 02/04/2018] [Indexed: 05/27/2023]
Abstract
INTRODUCTION Mass spectrometry imaging (MSI) as a label-free and powerful imaging technique enables in situ evaluation of a tissue metabolome and/or proteome, becoming increasingly popular in the detection of plant endogenous molecules. OBJECTIVE The characterisation of structure and spatial information of endogenous molecules in plants are both very important aspects to better understand the physiological mechanism of plant organism. METHODS Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) is a commonly-used tissue imaging technique, which requires matrix to assist in situ detection of a variety of molecules on the surface of a tissue section. In previous studies, MALDI-MSI was mostly used for the detection of molecules from animal tissue sections, compared to plant samples due to cell structural limitations, such as plant cuticles, epicuticular waxes, and cell walls. Despite the enormous progress that has been made in tissue imaging, there is still a challenge for MALDI-MSI suitable for the imaging of endogenous compounds in plants. RESULTS This review summarises the recent advances in MALDI-MSI, focusing on the application of in situ detection of endogenous molecules in different plant organs, i.e. root, stem, leaf, flower, fruit, and seed. CONCLUSION Further improvements on instrumentation sensitivity, matrix selection, image processing and sample preparation will expand the application of MALDI-MSI in plant research.
Collapse
Affiliation(s)
- Liang Qin
- Centre for Imaging & Systems Biology, Minzu University of China, Beijing, P. R. China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, P. R. China
| | - Yawen Zhang
- Centre for Imaging & Systems Biology, Minzu University of China, Beijing, P. R. China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, P. R. China
| | - Yaqin Liu
- Centre for Imaging & Systems Biology, Minzu University of China, Beijing, P. R. China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, P. R. China
| | - Huixin He
- Centre for Imaging & Systems Biology, Minzu University of China, Beijing, P. R. China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, P. R. China
| | - Manman Han
- Centre for Imaging & Systems Biology, Minzu University of China, Beijing, P. R. China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, P. R. China
| | - Yanyan Li
- The Hospital of Minzu University of China, Minzu University of China, Beijing, P. R. China
| | - Maomao Zeng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
- Collaborative Innovation Centre of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, P. R. China
| | - Xiaodong Wang
- Centre for Imaging & Systems Biology, Minzu University of China, Beijing, P. R. China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, P. R. China
| |
Collapse
|
78
|
Kriegsmann J, Casadonte R, Kriegsmann K, Longuespée R, Kriegsmann M. Mass spectrometry in pathology - Vision for a future workflow. Pathol Res Pract 2018; 214:1057-1063. [PMID: 29910062 DOI: 10.1016/j.prp.2018.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 04/23/2018] [Accepted: 05/11/2018] [Indexed: 02/09/2023]
Abstract
Mass spectrometric (MS) techniques are applied in various areas of medical diagnostics. For the detection of microbiological germs and genetic mutations, MS is a method used in routine. Since MS also allows the analysis of proteins and peptides, it seems an ideal candidate to supplement histopatholological diagnostics. Matrix-assisted laser desorption/ionization time-of-flight Imaging MS links molecular analysis of numerous analytes with morphological information about their spatial distribution in cells or tissues. Herein, we review principle MS techniques as well as potential applications in pathology and discuss our vision for a future workflow.
Collapse
Affiliation(s)
- Jörg Kriegsmann
- MVZ for Histology, Cytology and Molecular Diagnostics Trier, Trier, Germany; Proteopath GmbH, Trier, Germany
| | | | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Rémi Longuespée
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
79
|
Calvano CD, Monopoli A, Cataldi TRI, Palmisano F. MALDI matrices for low molecular weight compounds: an endless story? Anal Bioanal Chem 2018; 410:4015-4038. [DOI: 10.1007/s00216-018-1014-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/27/2018] [Accepted: 03/08/2018] [Indexed: 10/17/2022]
|
80
|
Schröter J, Fülöp A, Hopf C, Schiller J. The combination of 2,5-dihydroxybenzoic acid and 2,5-dihydroxyacetophenone matrices for unequivocal assignment of phosphatidylethanolamine species in complex mixtures. Anal Bioanal Chem 2018; 410:2437-2447. [PMID: 29445834 DOI: 10.1007/s00216-018-0926-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/19/2018] [Accepted: 01/29/2018] [Indexed: 12/13/2022]
Abstract
Unequivocal assignment of phospholipid peaks in complex mixtures is difficult if only the m/z values but no tandem mass spectrometry (MS/MS) data are available. This is usually the case for matrix-assisted laser/desorption ionization time-of-flight (MALDI-TOF) MS imaging experiments and the analysis has normally to be performed without prior separation. Another problem might be the often matrix-induced loss of one methyl group in phosphatidylcholine (PC) species, which makes them detectable as negative ions becoming isomers of some phosphatidylethanolamines (PEs). Selected lipid mixtures of known compositions were investigated by negative ion MALDI-TOF MS and various imaging experiments. In addition to common matrices such as 2,5-dihydroxybenzoic acid (DHB) and 9-aminoacridine (9-AA), different binary matrices, including 2,5-dihydroxyacetophenone (2,5-DHAP) as matrix additive to DHB, were tested to probe their performance in both ionization modes. Beside artificial PC and PE mixtures of known compositions, egg yolk and liver extracts as well as cryosections from liver and pancreas tissue were selected as biologically relevant systems. The majority of the binary MALDI matrices used here leads to the loss of a methyl group from PC in the negative ion mode, which makes the clear identification of PE species ambiguous. However, this problem does not apply if a mixture of DHB and 2,5-DHAP is used. Therefore, the application of DHB/2,5-DHAP as matrix is a simple method to unequivocally identify PEs even in complex mixtures and tissue sections as negative ions and without the necessity to separate the individual lipid classes prior to MS detection. Graphical abstract Many common MALDI matrices (such as 9-AA) induce the loss of a methyl group from PC rendering the PC detectable as negative ion. These ions (m/z 744.6 in the upper trace) represent isomers of typical PE species. It will be shown that this problem can be avoided if mixtures between DHB and 2,5-DHAP are applied. At these conditions, POPC is exclusively detectable as a matrix adduct with DHB (at m/z 912.6, lower trace) and does not interfere with PE. This approach can also be used in MALDI MS imaging.
Collapse
Affiliation(s)
- Jenny Schröter
- Faculty of Medicine, Institute for Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107, Leipzig, Germany.
| | - Annabelle Fülöp
- Center for Biomedical Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Strasse 10, 68163, Mannheim, Germany
| | - Carsten Hopf
- Center for Biomedical Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Strasse 10, 68163, Mannheim, Germany
| | - Jürgen Schiller
- Faculty of Medicine, Institute for Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107, Leipzig, Germany
| |
Collapse
|
81
|
Dong J, Ning W, Mans DJ, Mans JD. A binary matrix for the rapid detection and characterization of small-molecule cardiovascular drugs by MALDI-MS and MS/MS. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2018; 10:572-578. [PMID: 30319716 PMCID: PMC6178826 DOI: 10.1039/c7ay02583a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
A mixture of α-cyano-4-hydroxycinnamic acid and 1,5-diaminonaphthalene was discovered as a novel binary matrix for the qualitative analysis of 14 small-molecule (~250-550 Da) cardiovascular drugs by matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) and MS/MS in either positive or negative ion mode.
Collapse
Affiliation(s)
- Jinlan Dong
- Division of Pharmaceutical Analysis, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, MO, 63110, USA
| | - Wenjing Ning
- Division of Pharmaceutical Analysis, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, MO, 63110, USA
| | - Daniel J Mans
- Division of Pharmaceutical Analysis, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, MO, 63110, USA
| | - Jamie D Mans
- Division of Pharmaceutical Analysis, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, MO, 63110, USA
| |
Collapse
|
82
|
Szigyarto CAK, Spitali P. Biomarkers of Duchenne muscular dystrophy: current findings. Degener Neurol Neuromuscul Dis 2018; 8:1-13. [PMID: 30050384 PMCID: PMC6053903 DOI: 10.2147/dnnd.s121099] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Numerous biomarkers have been unveiled in the rapidly evolving biomarker discovery field, with an aim to improve the clinical management of disorders. In rare diseases, such as Duchenne muscular dystrophy, this endeavor has created a wealth of knowledge that, if effectively exploited, will benefit affected individuals, with respect to health care, therapy, improved quality of life and increased life expectancy. The most promising findings and molecular biomarkers are inspected in this review, with an aim to provide an overview of currently known biomarkers and the technological developments used. Biomarkers as cells, genetic variations, miRNAs, proteins, lipids and/or metabolites indicative of disease severity, progression and treatment response have the potential to improve development and approval of therapies, clinical management of DMD and patients’ life quality. We highlight the complexity of translating research results to clinical use, emphasizing the need for biomarkers, fit for purpose and describe the challenges associated with qualifying biomarkers for clinical applications.
Collapse
Affiliation(s)
- Cristina Al-Khalili Szigyarto
- Division of Proteomics, School of Biotechnology, AlbaNova University Center, KTH-Royal Institute of Technology, Stockholm, Sweden, .,Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden,
| | - Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands,
| |
Collapse
|
83
|
Keller C, Maeda J, Jayaraman D, Chakraborty S, Sussman MR, Harris JM, Ané JM, Li L. Comparison of Vacuum MALDI and AP-MALDI Platforms for the Mass Spectrometry Imaging of Metabolites Involved in Salt Stress in Medicago truncatula. FRONTIERS IN PLANT SCIENCE 2018; 9:1238. [PMID: 30210517 PMCID: PMC6121006 DOI: 10.3389/fpls.2018.01238] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/06/2018] [Indexed: 05/23/2023]
Abstract
Matrix-assisted laser desorption/ionization-mass spectrometry imaging (MALDI-MSI) is routinely used to determine the spatial distributions of various biomolecules in tissues. Recently, there has been an increased interest in creating higher resolution images using sources with more focused beams. One such source, an atmospheric pressure (AP) MALDI source from MassTech, has a laser capable of reaching spatial resolutions of 10 μm. Here, the AP-MALDI source coupled with a Q Exactive HF Orbitrap platform is compared to the commercial MALDI LTQ Orbitrap XL system using Medicago truncatula root nodules. AP-MALDI parameters, such as the S-lens value, capillary temperature, and spray voltage, were optimized on the Q Exactive-HF platform for optimal detection of plant metabolites. The performance of the two systems was evaluated for sensitivity, spatial resolution, and overall ability to detect plant metabolites. The commercial MALDI LTQ Orbitrap XL was superior regarding the number of compounds detected, as at least two times more m/z were detected compared to the AP-MALDI system. However, although the AP-MALDI source requires a spatial resolution higher than 10 μm to get the best signal, the spatial resolution at 30 μm is still superior compared to the 75 μm spatial resolution achieved on the MALDI platform. The AP-MALDI system was also used to investigate the metabolites present in M. truncatula roots and root nodules under high salt and low salt conditions. A discriminative analysis with SCiLS software revealed m/z ions specific to the control and salt conditions. This analysis revealed 44 m/z ions present at relatively higher abundances in the control samples, and 77 m/z enriched in the salt samples. Liquid chromatography-tandem MS was performed to determine the putative molecular identities of some of the mass ions enriched in each sample, including, asparagine, adenosine, and nicotianamine in the control samples, and arginine and soyasaponin I in the salt treated samples.
Collapse
Affiliation(s)
- Caitlin Keller
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI, United States
| | - Junko Maeda
- Department of Agronomy, University of Wisconsin–Madison, Madison, WI, United States
| | | | - Sanhita Chakraborty
- Department of Plant Biology, University of Vermont, Burlington, VT, United States
| | - Michael R. Sussman
- Department of Biochemistry, University of Wisconsin–Madison, Madison, WI, United States
| | - Jeanne M. Harris
- Department of Plant Biology, University of Vermont, Burlington, VT, United States
| | - Jean-Michel Ané
- Department of Agronomy, University of Wisconsin–Madison, Madison, WI, United States
- Department of Bacteriology, University of Wisconsin–Madison, Madison, WI, United States
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI, United States
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI, United States
- *Correspondence: Lingjun Li, ;
| |
Collapse
|
84
|
Drake RR, West CA, Mehta AS, Angel PM. MALDI Mass Spectrometry Imaging of N-Linked Glycans in Tissues. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1104:59-76. [PMID: 30484244 DOI: 10.1007/978-981-13-2158-0_4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) has been used for two decades to profile the glycan constituents of biological samples. An adaptation of the method to tissues, MALDI mass spectrometry imaging (MALDI-MSI), allows high-throughput spatial profiling of hundreds to thousands of molecules within a single thin tissue section. The ability to profile N-glycans within tissues using MALDI-MSI is a recently developed method that allows identification and localization of 40 or more N-glycans. The key component is to apply a molecular coating of peptide-N-glycosidase to tissues, an enzyme that releases N-glycans from their protein carrier. In this chapter, the methods and approaches to robustly and reproducibly generate two-dimensional N-glycan tissue maps by MALDI-MSI workflows are summarized. Current strengths and limitations of the approach are discussed, as well as potential future applications of the method.
Collapse
Affiliation(s)
- Richard R Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA.
| | - Connor A West
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Anand S Mehta
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Peggi M Angel
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
85
|
Matrix-Assisted Laser Desorption/Ionisation Mass Spectrometry Imaging in the Study of Gastric Cancer: A Mini Review. Int J Mol Sci 2017; 18:ijms18122588. [PMID: 29194417 PMCID: PMC5751191 DOI: 10.3390/ijms18122588] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/25/2017] [Accepted: 11/28/2017] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related deaths worldwide and the disease outcome commonly depends upon the tumour stage at the time of diagnosis. However, this cancer can often be asymptomatic during the early stages and remain undetected until the later stages of tumour development, having a significant impact on patient prognosis. However, our comprehension of the mechanisms underlying the development of gastric malignancies is still lacking. For these reasons, the search for new diagnostic and prognostic markers for gastric cancer is an ongoing pursuit. Modern mass spectrometry imaging (MSI) techniques, in particular matrix-assisted laser desorption/ionisation (MALDI), have emerged as a plausible tool in clinical pathology as a whole. More specifically, MALDI-MSI is being increasingly employed in the study of gastric cancer and has already elucidated some important disease checkpoints that may help us to better understand the molecular mechanisms underpinning this aggressive cancer. Here we report the state of the art of MALDI-MSI approaches, ranging from sample preparation to statistical analysis, and provide a complete review of the key findings that have been reported in the literature thus far.
Collapse
|
86
|
Xu LX, Wang TT, Geng YY, Wang WY, Li Y, Duan XK, Xu B, Liu CC, Liu WH. The direct analysis of drug distribution of rotigotine-loaded microspheres from tissue sections by LESA coupled with tandem mass spectrometry. Anal Bioanal Chem 2017; 409:5217-5223. [DOI: 10.1007/s00216-017-0440-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/17/2017] [Accepted: 05/30/2017] [Indexed: 10/19/2022]
|
87
|
Hansen RL, Lee YJ. High-Spatial Resolution Mass Spectrometry Imaging: Toward Single Cell Metabolomics in Plant Tissues. CHEM REC 2017; 18:65-77. [PMID: 28685965 DOI: 10.1002/tcr.201700027] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Indexed: 12/27/2022]
Abstract
Mass spectrometry imaging (MSI) is a powerful tool that has advanced our understanding of complex biological processes by enabling unprecedented details of metabolic biology to be uncovered. Through the use of high-spatial resolution MSI, metabolite localizations can be obtained with high precision. Here we describe our recent progress to enhance the spatial resolution of matrix-assisted laser desorption/ionization (MALDI) MSI from ∼50 μm with the commercial configuration to ∼5 μm. Additionally, we describe our efforts to develop a 'multiplex MSI' data acquisition method to allow more chemical information to be obtained on a single tissue in a single instrument run, and the development of new matrices to improve the ionization efficiency for a variety of small molecule metabolites. In combination, these contributions, along with the efforts of others, will bring MSI experiments closer to achieving metabolomic scale.
Collapse
Affiliation(s)
- Rebecca L Hansen
- Department of Chemistry, Iowa State University, 35 A Roy J Carver Co-Lab, 1111 WOI Road Ames, IA 50011, United States of America
| | - Young Jin Lee
- Department of Chemistry, Iowa State University, 35 A Roy J Carver Co-Lab, 1111 WOI Road Ames, IA 50011, United States of America
| |
Collapse
|
88
|
MALDI (matrix assisted laser desorption ionization) Imaging Mass Spectrometry (IMS) of skin: Aspects of sample preparation. Talanta 2017; 174:325-335. [PMID: 28738588 DOI: 10.1016/j.talanta.2017.06.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/15/2017] [Accepted: 06/02/2017] [Indexed: 12/15/2022]
Abstract
MALDI (matrix assisted laser desorption ionization) Imaging Mass Spectrometry (IMS) allows molecular analysis of biological materials making possible the identification and localization of molecules in tissues, and has been applied to address many questions on skin pathophysiology, as well as on studies about drug absorption and metabolism. Sample preparation for MALDI IMS is the most important part of the workflow, comprising specimen collection and preservation, tissue embedding, cryosectioning, washing, and matrix application. These steps must be carefully optimized for specific analytes of interest (lipids, proteins, drugs, etc.), representing a challenge for skin analysis. In this review, critical parameters for MALDI IMS sample preparation of skin samples will be described. In addition, specific applications of MALDI IMS of skin samples will be presented including wound healing, neoplasia, and infection.
Collapse
|
89
|
Quanico J, Franck J, Wisztorski M, Salzet M, Fournier I. Integrated mass spectrometry imaging and omics workflows on the same tissue section using grid-aided, parafilm-assisted microdissection. Biochim Biophys Acta Gen Subj 2017; 1861:1702-1714. [PMID: 28300637 DOI: 10.1016/j.bbagen.2017.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 03/08/2017] [Accepted: 03/10/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND In spite of the number of applications describing the use of MALDI MSI, one of its major drawbacks is the limited capability of identifying multiple compound classes directly on the same tissue section. METHODS We demonstrate the use of grid-aided, parafilm-assisted microdissection to perform MALDI MS imaging and shotgun proteomics and metabolomics in a combined workflow and using only a single tissue section. The grid is generated by microspotting acid dye 25 using a piezoelectric microspotter, and this grid was used as a guide to locate regions of interest and as an aid during manual microdissection. Subjecting the dissected pieces to the modified Folch method allows to separate the metabolites from proteins. The proteins can then be subjected to digestion under controlled conditions to improve protein identification yields. RESULTS The proof of concept experiment on rat brain generated 162 and 140 metabolite assignments from three ROIs (cerebellum, hippocampus and midbrain/hypothalamus) in positive and negative modes, respectively, and 890, 1303 and 1059 unique proteins. Integrated metabolite and protein overrepresentation analysis identified pathways associated with the biological functions of each ROI, most of which were not identified when looking at the protein and metabolite lists individually. CONCLUSIONS This combined MALDI MS imaging and multi-omics approach further extends the amount of information that can be generated from single tissue sections. GENERAL SIGNIFICANCE To the best of our knowledge, this is the first report combining both imaging and multi-omics analyses in the same workflow and on the same tissue section.
Collapse
Affiliation(s)
- Jusal Quanico
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Julien Franck
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Maxence Wisztorski
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Michel Salzet
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Isabelle Fournier
- Université de Lille 1, INSERM, U1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France.
| |
Collapse
|
90
|
Pharmacokinetic study based on a matrix-assisted laser desorption/ionization quadrupole ion trap time-of-flight imaging mass microscope combined with a novel relative exposure approach: A case of octreotide in mouse target tissues. Anal Chim Acta 2017; 952:71-80. [DOI: 10.1016/j.aca.2016.11.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/03/2016] [Accepted: 11/24/2016] [Indexed: 12/17/2022]
|
91
|
Kumaran S, Abdelhamid HN, Wu HF. Quantification analysis of protein and mycelium contents upon inhibition of melanin for Aspergillus niger: a study of matrix assisted laser desorption/ionization mass spectrometry (MALDI-MS). RSC Adv 2017. [DOI: 10.1039/c7ra03741d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mass spectrometry (MS) provides a simple discrimination method for microorganisms.
Collapse
Affiliation(s)
- Sekar Kumaran
- Department of Chemistry and Center for Nanoscience and Nanotechnology
- National Sun Yat-Sen University
- Kaohsiung
- Taiwan
- School of Pharmacy
| | - Hani Nasser Abdelhamid
- Department of Chemistry and Center for Nanoscience and Nanotechnology
- National Sun Yat-Sen University
- Kaohsiung
- Taiwan
- School of Pharmacy
| | - Hui-Fen Wu
- Department of Chemistry and Center for Nanoscience and Nanotechnology
- National Sun Yat-Sen University
- Kaohsiung
- Taiwan
- School of Pharmacy
| |
Collapse
|
92
|
Lipidomics-Reshaping the Analysis and Perception of Type 2 Diabetes. Int J Mol Sci 2016; 17:ijms17111841. [PMID: 27827927 PMCID: PMC5133841 DOI: 10.3390/ijms17111841] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 10/28/2016] [Accepted: 10/31/2016] [Indexed: 12/11/2022] Open
Abstract
As a consequence of a sedentary lifestyle as well as changed nutritional behavior, today's societies are challenged by the rapid propagation of metabolic disorders. A common feature of diseases, such as obesity and type 2 diabetes (T2D), is the dysregulation of lipid metabolism. Our understanding of the mechanisms underlying these diseases is hampered by the complexity of lipid metabolic pathways on a cellular level. Furthermore, overall lipid homeostasis in higher eukaryotic organisms needs to be maintained by a highly regulated interplay between tissues, such as adipose tissue, liver and muscle. Unraveling pathological mechanisms underlying metabolic disorders therefore requires a diversified approach, integrating basic cellular research with clinical research, ultimately relying on the analytical power of mass spectrometry-based techniques. Here, we discuss recent progress in the development of lipidomics approaches to resolve the pathological mechanisms of metabolic diseases and to identify suitable biomarkers for clinical application. Due to its growing impact worldwide, we focus on T2D to highlight the key role of lipidomics in our current understanding of this disease, discuss remaining questions and suggest future strategies to address them.
Collapse
|