1
|
Sohn AL, Bowman AP, Barnes MM, Kullman SW, Muddiman DC. Oversampling for Enhanced Spatial Resolution of Zebrafish by Top-Hat IR-MALDESI-MSI. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1959-1968. [PMID: 38985437 DOI: 10.1021/jasms.4c00219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Mass spectrometry imaging (MSI) has become a significant tool for measuring chemical species in biological tissues, where much of the impact of these platforms lies in their capability to report the spatial distribution of analytes for correlation to sample morphology. As a result, enhancement of spatial resolution has become a frontier of innovation in the field, and necessary developments are dependent on the ionization source. More particularly, laser-based imaging sources may require modifications to the optical train or alternative sampling techniques. These challenges are heightened for systems with infrared (IR) lasers, as their operating wavelength generates spot sizes that are inherently larger than their ultraviolet counterparts. Recently, the infrared matrix-assisted laser desorption electrospray ionization (IR-MALDESI) source has shown the utility of a diffractive optical element (DOE) to produce square ablation patterns, termed top-hat IR-MALDESI. If the DOE optic is combined with oversampling methods, smaller ablation volumes can be sampled to render higher spatial resolution imaging experiments. Further, this approach enables reproducible spot sizes and ablation volumes for better comparison between scans. Herein, we investigate the utility of oversampling with top-hat IR-MALDESI to enhance the spatial resolution of measured lipids localized within the head of sectioned zebrafish tissue. Four different spatial resolutions were evaluated for data quality (e.g., mass measurement accuracy, spectral accuracy) and quantity of annotations. Other experimental parameters to consider for high spatial resolution imaging are also discussed. Ultimately, 20 μm spatial resolution was achieved in this work and supports feasibility for use in future IR-MALDESI studies.
Collapse
Affiliation(s)
- Alexandria L Sohn
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | | | - Morgan M Barnes
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Seth W Kullman
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - David C Muddiman
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
2
|
Guan C, Kong L. Mass spectrometry imaging in pulmonary disorders. Clin Chim Acta 2024; 561:119835. [PMID: 38936534 DOI: 10.1016/j.cca.2024.119835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Mass Spectrometry Imaging (MSI) represents a novel and advancing technology that offers unparalleled in situ characterization of tissues. It provides comprehensive insights into the chemical structures, relative abundances, and spatial distributions of a vast array of both identified and unidentified endogenous and exogenous compounds, a capability not paralleled by existing analytical methodologies. Recent scholarly endeavors have increasingly explored the utility of MSI in the adjunct diagnosis and biomarker research of pulmonary disorders, including but not limited to lung cancer. Concurrently, MSI has proven instrumental in elucidating the spatiotemporal dynamics of various pharmacological agents. This review concisely delineates the fundamental principles underpinning MSI, its applications in pulmonary disease diagnosis, biomarker discovery, and drug distribution investigations. Additionally, it presents a forward-looking perspective on the prospective trajectories of MSI technological advancements.
Collapse
Affiliation(s)
- Chunliu Guan
- Key Laboratory of Environment Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Lu Kong
- Key Laboratory of Environment Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
3
|
Gardner W, Winkler DA, Bamford SE, Muir BW, Pigram PJ. Markedly Enhanced Analysis of Mass Spectrometry Images Using Weakly Supervised Machine Learning. SMALL METHODS 2024; 8:e2301230. [PMID: 38204217 DOI: 10.1002/smtd.202301230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/03/2023] [Indexed: 01/12/2024]
Abstract
Supervised and unsupervised machine learning algorithms are routinely applied to time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging data and, more broadly, to mass spectrometry imaging (MSI). These algorithms have accelerated large-scale, single-pixel analysis, classification, and regression. However, there is relatively little research on methods suited for so-called weakly supervised problems, where ground-truth class labels exist at the image level, but not at the individual pixel level. Unsupervised learning methods are usually applied to these problems. However, these methods cannot make use of available labels. Here a novel method specifically designed for weakly supervised MSI data is presented. A dual-stream multiple instance learning (MIL) approach is adapted from computational pathology that reveals the spatial-spectral characteristics distinguishing different classes of MSI images. The method uses an information entropy-regularized attention mechanism to identify characteristic class pixels that are then used to extract characteristic mass spectra. This work provides a proof-of-concept exemplification using printed ink samples imaged by ToF-SIMS. A second application-oriented study is also presented, focusing on the analysis of a mixed powder sample type. Results demonstrate the potential of the MIL method for broader application in MSI, with implications for understanding subtle spatial-spectral characteristics in various applications and contexts.
Collapse
Affiliation(s)
- Wil Gardner
- Centre for Materials and Surface Science and Department of Mathematical and Physical Sciences, La Trobe University, Bundoora, Victoria, 3086, Australia
| | - David A Winkler
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, 3086, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Advanced Materials and Healthcare Technologies, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Sarah E Bamford
- Centre for Materials and Surface Science and Department of Mathematical and Physical Sciences, La Trobe University, Bundoora, Victoria, 3086, Australia
| | | | - Paul J Pigram
- Centre for Materials and Surface Science and Department of Mathematical and Physical Sciences, La Trobe University, Bundoora, Victoria, 3086, Australia
| |
Collapse
|
4
|
Duncan KD, Pětrošová H, Lum JJ, Goodlett DR. Mass spectrometry imaging methods for visualizing tumor heterogeneity. Curr Opin Biotechnol 2024; 86:103068. [PMID: 38310648 PMCID: PMC11520788 DOI: 10.1016/j.copbio.2024.103068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 02/06/2024]
Abstract
Profiling spatial distributions of lipids, metabolites, and proteins in tumors can reveal unique cellular microenvironments and provide molecular evidence for cancer cell dysfunction and proliferation. Mass spectrometry imaging (MSI) is a label-free technique that can be used to map biomolecules in tumors in situ. Here, we discuss current progress in applying MSI to uncover molecular heterogeneity in tumors. First, the analytical strategies to profile small molecules and proteins are outlined, and current methods for multimodal imaging to maximize biological information are highlighted. Second, we present and summarize biological insights obtained by MSI of tumor tissue. Finally, we discuss important considerations for designing MSI experiments and several current analytical challenges.
Collapse
Affiliation(s)
- Kyle D Duncan
- Department of Chemistry, Vancouver Island University, Nanaimo, British Columbia, Canada; Department of Chemistry, University of Victoria, Victoria, British Columbia, Canada.
| | - Helena Pětrošová
- University of Victoria Genome British Columbia Proteomics Center, University of Victoria, Victoria, British Columbia, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada.
| | - Julian J Lum
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada; Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - David R Goodlett
- University of Victoria Genome British Columbia Proteomics Center, University of Victoria, Victoria, British Columbia, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
5
|
Ma X, Fernández FM. Advances in mass spectrometry imaging for spatial cancer metabolomics. MASS SPECTROMETRY REVIEWS 2024; 43:235-268. [PMID: 36065601 PMCID: PMC9986357 DOI: 10.1002/mas.21804] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 05/09/2023]
Abstract
Mass spectrometry (MS) has become a central technique in cancer research. The ability to analyze various types of biomolecules in complex biological matrices makes it well suited for understanding biochemical alterations associated with disease progression. Different biological samples, including serum, urine, saliva, and tissues have been successfully analyzed using mass spectrometry. In particular, spatial metabolomics using MS imaging (MSI) allows the direct visualization of metabolite distributions in tissues, thus enabling in-depth understanding of cancer-associated biochemical changes within specific structures. In recent years, MSI studies have been increasingly used to uncover metabolic reprogramming associated with cancer development, enabling the discovery of key biomarkers with potential for cancer diagnostics. In this review, we aim to cover the basic principles of MSI experiments for the nonspecialists, including fundamentals, the sample preparation process, the evolution of the mass spectrometry techniques used, and data analysis strategies. We also review MSI advances associated with cancer research in the last 5 years, including spatial lipidomics and glycomics, the adoption of three-dimensional and multimodal imaging MSI approaches, and the implementation of artificial intelligence/machine learning in MSI-based cancer studies. The adoption of MSI in clinical research and for single-cell metabolomics is also discussed. Spatially resolved studies on other small molecule metabolites such as amino acids, polyamines, and nucleotides/nucleosides will not be discussed in the context.
Collapse
Affiliation(s)
- Xin Ma
- School of Chemistry and Biochemistry and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Facundo M Fernández
- School of Chemistry and Biochemistry and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
6
|
Chen E, Ling AL, Reardon DA, Chiocca EA. Lessons learned from phase 3 trials of immunotherapy for glioblastoma: Time for longitudinal sampling? Neuro Oncol 2024; 26:211-225. [PMID: 37995317 PMCID: PMC10836778 DOI: 10.1093/neuonc/noad211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023] Open
Abstract
Glioblastoma (GBM)'s median overall survival is almost 21 months. Six phase 3 immunotherapy clinical trials have recently been published, yet 5/6 did not meet approval by regulatory bodies. For the sixth, approval is uncertain. Trial failures result from multiple factors, ranging from intrinsic tumor biology to clinical trial design. Understanding the clinical and basic science of these 6 trials is compelled by other immunotherapies reaching the point of advanced phase 3 clinical trial testing. We need to understand more of the science in human GBMs in early trials: the "window of opportunity" design may not be best to understand complex changes brought about by immunotherapeutic perturbations of the GBM microenvironment. The convergence of increased safety of image-guided biopsies with "multi-omics" of small cell numbers now permits longitudinal sampling of tumor and biofluids to dissect the complex temporal changes in the GBM microenvironment as a function of the immunotherapy.
Collapse
Affiliation(s)
- Ethan Chen
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Alexander L Ling
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Krestensen KK, Heeren RMA, Balluff B. State-of-the-art mass spectrometry imaging applications in biomedical research. Analyst 2023; 148:6161-6187. [PMID: 37947390 DOI: 10.1039/d3an01495a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Mass spectrometry imaging has advanced from a niche technique to a widely applied spatial biology tool operating at the forefront of numerous fields, most notably making a significant impact in biomedical pharmacological research. The growth of the field has gone hand in hand with an increase in publications and usage of the technique by new laboratories, and consequently this has led to a shift from general MSI reviews to topic-specific reviews. Given this development, we see the need to recapitulate the strengths of MSI by providing a more holistic overview of state-of-the-art MSI studies to provide the new generation of researchers with an up-to-date reference framework. Here we review scientific advances for the six largest biomedical fields of MSI application (oncology, pharmacology, neurology, cardiovascular diseases, endocrinology, and rheumatology). These publications thereby give examples for at least one of the following categories: they provide novel mechanistic insights, use an exceptionally large cohort size, establish a workflow that has the potential to become a high-impact methodology, or are highly cited in their field. We finally have a look into new emerging fields and trends in MSI (immunology, microbiology, infectious diseases, and aging), as applied MSI is continuously broadening as a result of technological breakthroughs.
Collapse
Affiliation(s)
- Kasper K Krestensen
- The Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Ron M A Heeren
- The Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Benjamin Balluff
- The Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
8
|
Yang E, Shen XE, West‐Foyle H, Hahm T, Siegler MA, Brown DR, Johnson CC, Kim JH, Roker LA, Tressler CM, Barman I, Kuo SC, Glunde K. FluoMALDI Microscopy: Matrix Co-Crystallization Simultaneously Enhances Fluorescence and MALDI Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304343. [PMID: 37908150 PMCID: PMC10724403 DOI: 10.1002/advs.202304343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/15/2023] [Indexed: 11/02/2023]
Abstract
Here, the authors report that co-crystallization of fluorophores with matrix-assisted laser desorption/ionization (MALDI) imaging matrices significantly enhances fluorophore brightness up to 79-fold, enabling the amplification of innate tissue autofluorescence. This discovery facilitates FluoMALDI, the imaging of the same biological sample by both fluorescence microscopy and MALDI imaging. The approach combines the high spatial resolution and specific labeling capabilities of fluorescence microscopy with the inherently multiplexed, versatile imaging capabilities of MALDI imaging. This new paradigm simplifies registration by avoiding physical changes between fluorescence and MALDI imaging, allowing to image the exact same cells in tissues with both modalities. Matrix-fluorophore co-crystallization also facilitates applications with insufficient fluorescence brightness. The authors demonstrate feasibility of FluoMALDI imaging with endogenous and exogenous fluorophores and autofluorescence-based FluoMALDI of brain and kidney tissue sections. FluoMALDI will advance structural-functional microscopic imaging in cell biology, biomedicine, and pathology.
Collapse
Affiliation(s)
- Ethan Yang
- Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Applied Imaging Mass Spectrometry CoreJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Xinyi Elaine Shen
- Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Applied Imaging Mass Spectrometry CoreJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Hoku West‐Foyle
- Microscope FacilityJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of Cell BiologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Tae‐Hun Hahm
- Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Applied Imaging Mass Spectrometry CoreJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | | | - Dalton R. Brown
- Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Applied Imaging Mass Spectrometry CoreJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Cole C. Johnson
- Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Applied Imaging Mass Spectrometry CoreJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Jeong Hee Kim
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - LaToya Ann Roker
- Microscope FacilityJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of Cell BiologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Caitlin M. Tressler
- Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Applied Imaging Mass Spectrometry CoreJohns Hopkins University School of MedicineBaltimoreMD21287USA
| | - Ishan Barman
- Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Sidney Kimmel Comprehensive Cancer CancerJohns Hopkins University School of MedicineBaltimoreMD21231USA
| | - Scot C. Kuo
- Microscope FacilityJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of Cell BiologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21218USA
| | - Kristine Glunde
- Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Applied Imaging Mass Spectrometry CoreJohns Hopkins University School of MedicineBaltimoreMD21287USA
- Sidney Kimmel Comprehensive Cancer CancerJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Department of Biological ChemistryJohns Hopkins University School of MedicineBaltimoreMD21205USA
| |
Collapse
|
9
|
Abstract
Imaging mass spectrometry is a well-established technology that can easily and succinctly communicate the spatial localization of molecules within samples. This review communicates the recent advances in the field, with a specific focus on matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) applied on tissues. The general sample preparation strategies for different analyte classes are explored, including special considerations for sample types (fresh frozen or formalin-fixed,) strategies for various analytes (lipids, metabolites, proteins, peptides, and glycans) and how multimodal imaging strategies can leverage the strengths of each approach is mentioned. This work explores appropriate experimental design approaches and standardization of processes needed for successful studies, as well as the various data analysis platforms available to analyze data and their strengths. The review concludes with applications of imaging mass spectrometry in various fields, with a focus on medical research, and some examples from plant biology and microbe metabolism are mentioned, to illustrate the breadth and depth of MALDI IMS.
Collapse
Affiliation(s)
- Jessica L Moore
- Department of Proteomics, Discovery Life Sciences, Huntsville, Alabama 35806, United States
| | - Georgia Charkoftaki
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, Connecticut 06520, United States
| |
Collapse
|
10
|
Chung HH, Huang P, Chen CL, Lee C, Hsu CC. Next-generation pathology practices with mass spectrometry imaging. MASS SPECTROMETRY REVIEWS 2023; 42:2446-2465. [PMID: 35815718 DOI: 10.1002/mas.21795] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 06/15/2023]
Abstract
Mass spectrometry imaging (MSI) is a powerful technique that reveals the spatial distribution of various molecules in biological samples, and it is widely used in pathology-related research. In this review, we summarize common MSI techniques, including matrix-assisted laser desorption/ionization and desorption electrospray ionization MSI, and their applications in pathological research, including disease diagnosis, microbiology, and drug discovery. We also describe the improvements of MSI, focusing on the accumulation of imaging data sets, expansion of chemical coverage, and identification of biological significant molecules, that have prompted the evolution of MSI to meet the requirements of pathology practices. Overall, this review details the applications and improvements of MSI techniques, demonstrating the potential of integrating MSI techniques into next-generation pathology practices.
Collapse
Affiliation(s)
- Hsin-Hsiang Chung
- Department of Chemistry, National Taiwan University, Taipei City, Taiwan
| | - Penghsuan Huang
- Department of Chemistry, National Taiwan University, Taipei City, Taiwan
| | - Chih-Lin Chen
- Department of Chemistry, National Taiwan University, Taipei City, Taiwan
| | - Chuping Lee
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei City, Taiwan
| |
Collapse
|
11
|
Wang Z, Zhu H, Xiong W. Advances in mass spectrometry-based multi-scale metabolomic methodologies and their applications in biological and clinical investigations. Sci Bull (Beijing) 2023; 68:2268-2284. [PMID: 37666722 DOI: 10.1016/j.scib.2023.08.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/25/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
Metabolomics is a nascent field of inquiry that emerged in the late 20th century. It encompasses the comprehensive profiling of metabolites across a spectrum of organisms, ranging from bacteria and cells to tissues. The rapid evolution of analytical methods and data analysis has greatly accelerated progress in this dynamic discipline over recent decades. Sophisticated techniques such as liquid chromatograph mass spectrometry (MS), gas chromatograph MS, capillary electrophoresis MS, and nuclear magnetic resonance serve as the cornerstone of metabolomic analysis. Building upon these methods, a plethora of modifications and combinations have emerged to propel the advancement of metabolomics. Despite this progress, scrutinizing metabolism at the single-cell or single-organelle level remains an arduous task over the decades. Some of the most thrilling advancements, such as single-cell and single-organelle metabolic profiling techniques, offer profound insights into the intricate mechanisms within cells and organelles. This allows for a comprehensive study of metabolic heterogeneity and its pivotal role in multiple biological processes. The progress made in MS imaging has enabled high-resolution in situ metabolic profiling of tissue sections and even individual cells. Spatial reconstruction techniques enable the direct representation of metabolic distribution and alteration in three-dimensional space. The application of novel metabolomic techniques has led to significant breakthroughs in biological and clinical studies, including the discovery of novel metabolic pathways, determination of cell fate in differentiation, anti-aging intervention through modulating metabolism, metabolomics-based clinicopathologic analysis, and surgical decision-making based on on-site intraoperative metabolic analysis. This review presents a comprehensive overview of both conventional and innovative metabolomic techniques, highlighting their applications in groundbreaking biological and clinical studies.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Hongying Zhu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; CAS Key Laboratory of Brain Function and Disease, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, Hefei 230026, China.
| | - Wei Xiong
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; CAS Key Laboratory of Brain Function and Disease, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, Hefei 230026, China.
| |
Collapse
|
12
|
King ME, Lin M, Spradlin M, Eberlin LS. Advances and Emerging Medical Applications of Direct Mass Spectrometry Technologies for Tissue Analysis. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2023; 16:1-25. [PMID: 36944233 DOI: 10.1146/annurev-anchem-061020-015544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Offering superb speed, chemical specificity, and analytical sensitivity, direct mass spectrometry (MS) technologies are highly amenable for the molecular analysis of complex tissues to aid in disease characterization and help identify new diagnostic, prognostic, and predictive markers. By enabling detection of clinically actionable molecular profiles from tissues and cells, direct MS technologies have the potential to guide treatment decisions and transform sample analysis within clinical workflows. In this review, we highlight recent health-related developments and applications of direct MS technologies that exhibit tangible potential to accelerate clinical research and disease diagnosis, including oncological and neurodegenerative diseases and microbial infections. We focus primarily on applications that employ direct MS technologies for tissue analysis, including MS imaging technologies to map spatial distributions of molecules in situ as well as handheld devices for rapid in vivo and ex vivo tissue analysis.
Collapse
Affiliation(s)
- Mary E King
- Department of Chemistry, The University of Texas at Austin, Austin, Texas, USA;
- Department of Surgery, Baylor College of Medicine, Houston, Texas, USA;
| | - Monica Lin
- Department of Chemistry, The University of Texas at Austin, Austin, Texas, USA;
| | - Meredith Spradlin
- Department of Chemistry, The University of Texas at Austin, Austin, Texas, USA;
| | - Livia S Eberlin
- Department of Surgery, Baylor College of Medicine, Houston, Texas, USA;
| |
Collapse
|
13
|
Benedetti E, Liu EM, Tang C, Kuo F, Buyukozkan M, Park T, Park J, Correa F, Hakimi AA, Intlekofer AM, Krumsiek J, Reznik E. A multimodal atlas of tumour metabolism reveals the architecture of gene-metabolite covariation. Nat Metab 2023; 5:1029-1044. [PMID: 37337120 PMCID: PMC10290959 DOI: 10.1038/s42255-023-00817-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 05/09/2023] [Indexed: 06/21/2023]
Abstract
Tumour metabolism is controlled by coordinated changes in metabolite abundance and gene expression, but simultaneous quantification of metabolites and transcripts in primary tissue is rare. To overcome this limitation and to study gene-metabolite covariation in cancer, we assemble the Cancer Atlas of Metabolic Profiles of metabolomic and transcriptomic data from 988 tumour and control specimens spanning 11 cancer types in published and newly generated datasets. Meta-analysis of the Cancer Atlas of Metabolic Profiles reveals two classes of gene-metabolite covariation that transcend cancer types. The first corresponds to gene-metabolite pairs engaged in direct enzyme-substrate interactions, identifying putative genes controlling metabolite pool sizes. A second class of gene-metabolite covariation represents a small number of hub metabolites, including quinolinate and nicotinamide adenine dinucleotide, which correlate to many genes specifically expressed in immune cell populations. These results provide evidence that gene-metabolite covariation in cellularly heterogeneous tissue arises, in part, from both mechanistic interactions between genes and metabolites, and from remodelling of the bulk metabolome in specific immune microenvironments.
Collapse
Affiliation(s)
- Elisa Benedetti
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Eric Minwei Liu
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cerise Tang
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fengshen Kuo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mustafa Buyukozkan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Tricia Park
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jinsung Park
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fabian Correa
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew M Intlekofer
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Ed Reznik
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
14
|
Tressler CM, Ayyappan V, Nakuchima S, Yang E, Sonkar K, Tan Z, Glunde K. A multimodal pipeline using NMR spectroscopy and MALDI-TOF mass spectrometry imaging from the same tissue sample. NMR IN BIOMEDICINE 2023; 36:e4770. [PMID: 35538020 PMCID: PMC9867920 DOI: 10.1002/nbm.4770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 06/14/2023]
Abstract
NMR spectroscopy and matrix assisted laser desorption ionization mass spectrometry imaging (MALDI MSI) are both commonly used to detect large numbers of metabolites and lipids in metabolomic and lipidomic studies. We have demonstrated a new workflow, highlighting the benefits of both techniques to obtain metabolomic and lipidomic data, which has realized for the first time the combination of these two complementary and powerful technologies. NMR spectroscopy is frequently used to obtain quantitative metabolite information from cells and tissues. Lipid detection is also possible with NMR spectroscopy, with changes being visible across entire classes of molecules. Meanwhile, MALDI MSI provides relative measures of metabolite and lipid concentrations, mapping spatial information of many specific metabolite and lipid molecules across cells or tissues. We have used these two complementary techniques in combination to obtain metabolomic and lipidomic measurements from triple-negative human breast cancer cells and tumor xenograft models. We have emphasized critical experimental procedures that ensured the success of achieving NMR spectroscopy and MALDI MSI in a combined workflow from the same sample. Our data show that several phospholipid metabolite species were differentially distributed in viable and necrotic regions of breast tumor xenografts. This study emphasizes the power of combined NMR spectroscopy-MALDI imaging to advance metabolomic and lipidomic studies.
Collapse
Affiliation(s)
- Caitlin M. Tressler
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vinay Ayyappan
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sofia Nakuchima
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ethan Yang
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kanchan Sonkar
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zheqiong Tan
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristine Glunde
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Straehla JP, Reardon DA, Wen PY, Agar NYR. The Blood-Brain Barrier: Implications for Experimental Cancer Therapeutics. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:265-289. [PMID: 38323268 PMCID: PMC10846865 DOI: 10.1146/annurev-cancerbio-061421-040433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
The blood-brain barrier is critically important for the treatment of both primary and metastatic cancers of the central nervous system (CNS). Clinical outcomes for patients with primary CNS tumors are poor and have not significantly improved in decades. As treatments for patients with extracranial solid tumors improve, the incidence of CNS metastases is on the rise due to suboptimal CNS exposure of otherwise systemically active agents. Despite state-of-the art surgical care and increasingly precise radiation therapy, clinical progress is limited by the ability to deliver an effective dose of a therapeutic agent to all cancerous cells. Given the tremendous heterogeneity of CNS cancers, both across cancer subtypes and within a single tumor, and the range of diverse therapies under investigation, a nuanced examination of CNS drug exposure is needed. With a shared goal, common vocabulary, and interdisciplinary collaboration, the field is poised for renewed progress in the treatment of CNS cancers.
Collapse
Affiliation(s)
- Joelle P Straehla
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Wang X, Zhang L, Xiang Y, Ye N, Liu K. Systematic study of tissue section thickness for MALDI MS profiling and imaging. Analyst 2023; 148:888-897. [PMID: 36661109 DOI: 10.1039/d2an01739c] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) has become a powerful method for studying the spatial distribution of molecules. Preparation of tissue sections is a critical step for obtaining high-quality imaging data. The thickness of the slice of tissue affects the feature quality of MALDI MSI. However, few studies involved in-depth and systematic examination of slice thickness. Herein, we investigate the effect of tissue slice thickness on MALDI MSI detection. We found that the thicker the slice, the worse the results obtained by MALDI MS, which we attributed to the charging effect. The optimal slice thickness of brain tissue obtained in this work is 2-6 μm. Comparisons of the effects of slice thickness on atmospheric pressure and vacuum MALDI assays indicated that the ion signals and imaging quality of vacuum MALDI were more seriously affected by the thickness, with atmospheric pressure (AP) MALDI having a greater tolerance for slice thickness than vacuum MALDI. The MALDI MSI of peptides after enzymatic digestion of tissue sections of different thicknesses was also studied, revealing that the most suitable tissue thickness for enzyme digestion is about 10 μm. Finally, we optimized the slice thicknesses of six tissues in mice to provide a reference for MALDI MSI studies. It is worth mentioning that in our study the values of slice thickness range from the nanometer level (400 nm) at the minimum to 150 μm at the maximum, values which were unprecedented. Detailed in-depth and systematic studies of slice thickness will promote the development of sample preparation technology of AP and vacuum MALDI MSI, which will provide important references for the selection of tissue section thickness.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Lu Zhang
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Yuhong Xiang
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Nengsheng Ye
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Kehui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
17
|
Yu J, Hermann M, Smith R, Tomm H, Metwally H, Kolwich J, Liu C, Le Blanc JCY, Covey TR, Ross AC, Oleschuk R. Hyperspectral Visualization-Based Mass Spectrometry Imaging by LMJ-SSP: A Novel Strategy for Rapid Natural Product Profiling in Bacteria. Anal Chem 2023; 95:2020-2028. [PMID: 36634199 DOI: 10.1021/acs.analchem.2c04550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Mass spectrometry imaging (MSI) has been widely used to discover natural products (NPs) from underexplored microbiological sources. However, the technique is limited by incompatibility with complicated/uneven surface topography and labor-intensive sample preparation, as well as lengthy compound profiling procedures. Here, liquid micro-junction surface sampling probe (LMJ-SSP)-based MSI is used for rapid profiling of natural products from Gram-negative marine bacteria Pseudoalteromonas on nutrient agar media without any sample preparation. A conductance-based autosampling platform with 1 mm spatial resolution and an innovative multivariant analysis-driven method was used to create one hyperspectral image for the sampling area. NP discovery requires general spatial correlation between m/z and colony location but not highly precise spatial resolution. The hyperspectral image was used to annotate different m/z by straightforward color differences without the need to directly interrogate the spectra. To demonstrate the utility of our approach, the rapid analysis of Pseudoalteromonas rubra DSM6842, Pseudoalteromonas tunicata DSM14096, Pseudoalteromonas piscicida JCM20779, and Pseudoalteromonas elyakovii ATCC700519 cultures was directly performed on Agar. Various natural products, including prodiginine and tambjamine analogues, were quickly identified from the hyperspectral image, and the dynamic extracellular environment was shown with compound heatmaps. Hyperspectral visualization-based MSI is an efficient and sensitive strategy for direct and rapid natural product profiling from different Pseudoalteromonas strains.
Collapse
Affiliation(s)
- Jian Yu
- Department of Chemistry, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Matthias Hermann
- Department of Chemistry, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Rachael Smith
- Department of Chemistry, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Hailey Tomm
- Department of Chemistry, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Haidy Metwally
- Department of Chemistry, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Jennifer Kolwich
- Department of Chemistry, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Chang Liu
- SCIEX, Concord, Ontario L4K 4 V8, Canada
| | | | | | - Avena C Ross
- Department of Chemistry, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Richard Oleschuk
- Department of Chemistry, Queen's University, Kingston, Ontario K7L 3N6, Canada
| |
Collapse
|
18
|
Hassan T, Firdous P, Nissar K, Ahmad MB, Imtiyaz Z. Role of proteomics in surgical oncology. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
19
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2019-2020. MASS SPECTROMETRY REVIEWS 2022:e21806. [PMID: 36468275 DOI: 10.1002/mas.21806] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
This review is the tenth update of the original article published in 1999 on the application of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2020. Also included are papers that describe methods appropriate to analysis by MALDI, such as sample preparation techniques, even though the ionization method is not MALDI. The review is basically divided into three sections: (1) general aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, fragmentation, quantification and the use of arrays. (2) Applications to various structural types such as oligo- and polysaccharides, glycoproteins, glycolipids, glycosides and biopharmaceuticals, and (3) other areas such as medicine, industrial processes and glycan synthesis where MALDI is extensively used. Much of the material relating to applications is presented in tabular form. The reported work shows increasing use of incorporation of new techniques such as ion mobility and the enormous impact that MALDI imaging is having. MALDI, although invented nearly 40 years ago is still an ideal technique for carbohydrate analysis and advancements in the technique and range of applications show little sign of diminishing.
Collapse
Affiliation(s)
- David J Harvey
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
- Department of Chemistry, University of Oxford, Oxford, Oxfordshire, United Kingdom
| |
Collapse
|
20
|
Hu H, Laskin J. Emerging Computational Methods in Mass Spectrometry Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203339. [PMID: 36253139 PMCID: PMC9731724 DOI: 10.1002/advs.202203339] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/17/2022] [Indexed: 05/10/2023]
Abstract
Mass spectrometry imaging (MSI) is a powerful analytical technique that generates maps of hundreds of molecules in biological samples with high sensitivity and molecular specificity. Advanced MSI platforms with capability of high-spatial resolution and high-throughput acquisition generate vast amount of data, which necessitates the development of computational tools for MSI data analysis. In addition, computation-driven MSI experiments have recently emerged as enabling technologies for further improving the MSI capabilities with little or no hardware modification. This review provides a critical summary of computational methods and resources developed for MSI data analysis and interpretation along with computational approaches for improving throughput and molecular coverage in MSI experiments. This review is focused on the recently developed artificial intelligence methods and provides an outlook for a future paradigm shift in MSI with transformative computational methods.
Collapse
Affiliation(s)
- Hang Hu
- Department of ChemistryPurdue University560 Oval DriveWest LafayetteIN47907USA
| | - Julia Laskin
- Department of ChemistryPurdue University560 Oval DriveWest LafayetteIN47907USA
| |
Collapse
|
21
|
Groessl M, Vogt B. Mass spectrometry imaging and its place in nephrology. Nephrol Dial Transplant 2022; 37:2363-2365. [PMID: 34940875 DOI: 10.1093/ndt/gfab359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Indexed: 12/31/2022] Open
Affiliation(s)
- Michael Groessl
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Azam KSF, Ryabchykov O, Bocklitz T. A Review on Data Fusion of Multidimensional Medical and Biomedical Data. Molecules 2022; 27:7448. [PMID: 36364272 PMCID: PMC9655963 DOI: 10.3390/molecules27217448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/19/2022] [Accepted: 10/21/2022] [Indexed: 08/05/2024] Open
Abstract
Data fusion aims to provide a more accurate description of a sample than any one source of data alone. At the same time, data fusion minimizes the uncertainty of the results by combining data from multiple sources. Both aim to improve the characterization of samples and might improve clinical diagnosis and prognosis. In this paper, we present an overview of the advances achieved over the last decades in data fusion approaches in the context of the medical and biomedical fields. We collected approaches for interpreting multiple sources of data in different combinations: image to image, image to biomarker, spectra to image, spectra to spectra, spectra to biomarker, and others. We found that the most prevalent combination is the image-to-image fusion and that most data fusion approaches were applied together with deep learning or machine learning methods.
Collapse
Affiliation(s)
- Kazi Sultana Farhana Azam
- Leibniz Institute of Photonic Technology, Member of Leibniz-Research Alliance “Health Technologies”, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - Oleg Ryabchykov
- Leibniz Institute of Photonic Technology, Member of Leibniz-Research Alliance “Health Technologies”, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - Thomas Bocklitz
- Leibniz Institute of Photonic Technology, Member of Leibniz-Research Alliance “Health Technologies”, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| |
Collapse
|
23
|
Körber A, Keelor JD, Claes BSR, Heeren RMA, Anthony IGM. Fast Mass Microscopy: Mass Spectrometry Imaging of a Gigapixel Image in 34 Minutes. Anal Chem 2022; 94:14652-14658. [PMID: 36223179 PMCID: PMC9607864 DOI: 10.1021/acs.analchem.2c02870] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/22/2022] [Indexed: 11/29/2022]
Abstract
Mass spectrometry imaging (MSI) maps the spatial distributions of chemicals on surfaces. MSI requires improvements in throughput and spatial resolution, and often one is compromised for the other. In microprobe-mode MSI, improvements in spatial resolution increase the imaging time quadratically, thus limiting the use of high spatial resolution MSI for large areas or sample cohorts and time-sensitive measurements. Here, we bypass this quadratic relationship by combining a Timepix3 detector with a continuously sampling secondary ion mass spectrometry mass microscope. By reconstructing the data into large-field mass images, this new method, fast mass microscopy, enables orders of magnitude higher throughput than conventional MSI albeit yet at lower mass resolution. We acquired submicron, gigapixel images of fingerprints and rat tissue at acquisition speeds of 600,000 and 15,500 pixels s-1, respectively. For the first image, a comparable microprobe-mode measurement would take more than 2 months, whereas our approach took 33.3 min.
Collapse
Affiliation(s)
- Aljoscha Körber
- The
Maastricht MultiModal Molecular Imaging Institute (M4i), Division
of Imaging Mass Spectrometry, Maastricht
University, Universiteitssingel 50, Maastricht 6229 ER, The Netherlands
| | - Joel D. Keelor
- Amsterdam
Scientific Instruments B.V. (ASI), Science Park 106, Amsterdam 1098 XG, The Netherlands
| | - Britt S. R. Claes
- The
Maastricht MultiModal Molecular Imaging Institute (M4i), Division
of Imaging Mass Spectrometry, Maastricht
University, Universiteitssingel 50, Maastricht 6229 ER, The Netherlands
| | - Ron M. A. Heeren
- The
Maastricht MultiModal Molecular Imaging Institute (M4i), Division
of Imaging Mass Spectrometry, Maastricht
University, Universiteitssingel 50, Maastricht 6229 ER, The Netherlands
| | - Ian G. M. Anthony
- The
Maastricht MultiModal Molecular Imaging Institute (M4i), Division
of Imaging Mass Spectrometry, Maastricht
University, Universiteitssingel 50, Maastricht 6229 ER, The Netherlands
| |
Collapse
|
24
|
Hu H, Helminiak D, Yang M, Unsihuay D, Hilger RT, Ye DH, Laskin J. High-Throughput Mass Spectrometry Imaging with Dynamic Sparse Sampling. ACS MEASUREMENT SCIENCE AU 2022; 2:466-474. [PMID: 36281292 PMCID: PMC9585637 DOI: 10.1021/acsmeasuresciau.2c00031] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 05/25/2023]
Abstract
Mass spectrometry imaging (MSI) enables label-free mapping of hundreds of molecules in biological samples with high sensitivity and unprecedented specificity. Conventional MSI experiments are relatively slow, limiting their utility for applications requiring rapid data acquisition, such as intraoperative tissue analysis or 3D imaging. Recent advances in MSI technology focus on improving the spatial resolution and molecular coverage, further increasing the acquisition time. Herein, a deep learning approach for dynamic sampling (DLADS) was employed to reduce the number of required measurements, thereby improving the throughput of MSI experiments in comparison with conventional methods. DLADS trains a deep learning model to dynamically predict molecularly informative tissue locations for active mass spectra sampling and reconstructs high-fidelity molecular images using only the sparsely sampled information. Experimental hardware and software integration of DLADS with nanospray desorption electrospray ionization (nano-DESI) MSI is reported for the first time, which demonstrates a 2.3-fold improvement in throughput for a linewise acquisition mode. Meanwhile, simulations indicate that a 5-10-fold throughput improvement may be achieved using the pointwise acquisition mode.
Collapse
Affiliation(s)
- Hang Hu
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - David Helminiak
- Electrical
and Computer Engineering, Marquette University, Milwaukee, Wisconsin 53233, United States
| | - Manxi Yang
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Daisy Unsihuay
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ryan T. Hilger
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Dong Hye Ye
- Electrical
and Computer Engineering, Marquette University, Milwaukee, Wisconsin 53233, United States
| | - Julia Laskin
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
25
|
Stopka SA, van der Reest J, Abdelmoula WM, Ruiz DF, Joshi S, Ringel AE, Haigis MC, Agar NYR. Spatially resolved characterization of tissue metabolic compartments in fasted and high-fat diet livers. PLoS One 2022; 17:e0261803. [PMID: 36067168 PMCID: PMC9447892 DOI: 10.1371/journal.pone.0261803] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 08/12/2022] [Indexed: 11/18/2022] Open
Abstract
Cells adapt their metabolism to physiological stimuli, and metabolic heterogeneity exists between cell types, within tissues, and subcellular compartments. The liver plays an essential role in maintaining whole-body metabolic homeostasis and is structurally defined by metabolic zones. These zones are well-understood on the transcriptomic level, but have not been comprehensively characterized on the metabolomic level. Mass spectrometry imaging (MSI) can be used to map hundreds of metabolites directly from a tissue section, offering an important advance to investigate metabolic heterogeneity in tissues compared to extraction-based metabolomics methods that analyze tissue metabolite profiles in bulk. We established a workflow for the preparation of tissue specimens for matrix-assisted laser desorption/ionization (MALDI) MSI that can be implemented to achieve broad coverage of central carbon, nucleotide, and lipid metabolism pathways. Herein, we used this approach to visualize the effect of nutrient stress and excess on liver metabolism. Our data revealed a highly organized metabolic tissue compartmentalization in livers, which becomes disrupted under high fat diet. Fasting caused changes in the abundance of several metabolites, including increased levels of fatty acids and TCA intermediates while fatty livers had higher levels of purine and pentose phosphate-related metabolites, which generate reducing equivalents to counteract oxidative stress. This spatially conserved approach allowed the visualization of liver metabolic compartmentalization at 30 μm pixel resolution and can be applied more broadly to yield new insights into metabolic heterogeneity in vivo.
Collapse
Affiliation(s)
- Sylwia A. Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United Statees of America
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United Statees of America
| | - Jiska van der Reest
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United Statees of America
- Department of Cell Biology, Blavatnik Institute, Ludwig Center, Harvard Medical School, Boston, MA, United Statees of America
| | - Walid M. Abdelmoula
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United Statees of America
| | - Daniela F. Ruiz
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United Statees of America
- Bouvé College of Health Sciences, Northeastern University, Boston, MA, United Statees of America
| | - Shakchhi Joshi
- Department of Cell Biology, Blavatnik Institute, Ludwig Center, Harvard Medical School, Boston, MA, United Statees of America
| | - Alison E. Ringel
- Department of Cell Biology, Blavatnik Institute, Ludwig Center, Harvard Medical School, Boston, MA, United Statees of America
| | - Marcia C. Haigis
- Department of Cell Biology, Blavatnik Institute, Ludwig Center, Harvard Medical School, Boston, MA, United Statees of America
- * E-mail: (MCH); (NYRA)
| | - Nathalie Y. R. Agar
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United Statees of America
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United Statees of America
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United Statees of America
- * E-mail: (MCH); (NYRA)
| |
Collapse
|
26
|
Coy S, Wang S, Stopka SA, Lin JR, Yapp C, Ritch CC, Salhi L, Baker GJ, Rashid R, Baquer G, Regan M, Khadka P, Cole KA, Hwang J, Wen PY, Bandopadhayay P, Santi M, De Raedt T, Ligon KL, Agar NYR, Sorger PK, Touat M, Santagata S. Single cell spatial analysis reveals the topology of immunomodulatory purinergic signaling in glioblastoma. Nat Commun 2022; 13:4814. [PMID: 35973991 PMCID: PMC9381513 DOI: 10.1038/s41467-022-32430-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/29/2022] [Indexed: 12/11/2022] Open
Abstract
How the glioma immune microenvironment fosters tumorigenesis remains incompletely defined. Here, we use single-cell RNA-sequencing and multiplexed tissue-imaging to characterize the composition, spatial organization, and clinical significance of extracellular purinergic signaling in glioma. We show that microglia are the predominant source of CD39, while tumor cells principally express CD73. In glioblastoma, CD73 is associated with EGFR amplification, astrocyte-like differentiation, and increased adenosine, and is linked to hypoxia. Glioblastomas enriched for CD73 exhibit inflammatory microenvironments, suggesting that purinergic signaling regulates immune adaptation. Spatially-resolved single-cell analyses demonstrate a strong spatial correlation between tumor-CD73 and microglial-CD39, with proximity associated with poor outcomes. Similar spatial organization is present in pediatric high-grade gliomas including H3K27M-mutant diffuse midline glioma. These data reveal that purinergic signaling in gliomas is shaped by genotype, lineage, and functional state, and that core enzymes expressed by tumor and myeloid cells are organized to promote adenosine-rich microenvironments potentially amenable to therapeutic targeting.
Collapse
Affiliation(s)
- Shannon Coy
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Shu Wang
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Harvard Graduate Program in Biophysics, Harvard University, Boston, MA, USA
| | - Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jia-Ren Lin
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Clarence Yapp
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Cecily C Ritch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
| | - Lisa Salhi
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle Epinière, and AP-HP Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - Gregory J Baker
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Rumana Rashid
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
- Pitt-CMU Medical Scientist Training Program, University of Pittsburgh-Carnegie Mellon, Pittsburgh, PA, USA
| | - Gerard Baquer
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Regan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Prasidda Khadka
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kristina A Cole
- Children's Hospital of Philadelphia, University of Pennsylvania, Pennsylvania, PA, USA
| | - Jaeho Hwang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrick Y Wen
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pratiti Bandopadhayay
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mariarita Santi
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Thomas De Raedt
- Children's Hospital of Philadelphia, University of Pennsylvania, Pennsylvania, PA, USA
| | - Keith L Ligon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Mehdi Touat
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle Epinière, and AP-HP Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France.
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA.
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Baijnath S, Kaya I, Nilsson A, Shariatgorji R, Andrén PE. Advances in spatial mass spectrometry enable in-depth neuropharmacodynamics. Trends Pharmacol Sci 2022; 43:740-753. [DOI: 10.1016/j.tips.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/06/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022]
|
28
|
Gardner W, Winkler DA, Cutts SM, Torney SA, Pietersz GA, Muir BW, Pigram PJ. Two-Dimensional and Three-Dimensional Time-of-Flight Secondary Ion Mass Spectrometry Image Feature Extraction Using a Spatially Aware Convolutional Autoencoder. Anal Chem 2022; 94:7804-7813. [PMID: 35616489 DOI: 10.1021/acs.analchem.1c05453] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Feature extraction algorithms are an important class of unsupervised methods used to reduce data dimensionality. They have been applied extensively for time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging─commonly, matrix factorization (MF) techniques such as principal component analysis have been used. A limitation of MF is the assumption of linearity, which is generally not accurate for ToF-SIMS data. Recently, nonlinear autoencoders have been shown to outperform MF techniques for ToF-SIMS image feature extraction. However, another limitation of most feature extraction methods (including autoencoders) that is particularly important for hyperspectral data is that they do not consider spatial information. To address this limitation, we describe the application of the convolutional autoencoder (CNNAE) to hyperspectral ToF-SIMS imaging data. The CNNAE is an artificial neural network developed specifically for hyperspectral data that uses convolutional layers for image encoding, thereby explicitly incorporating pixel neighborhood information. We compared the performance of the CNNAE with other common feature extraction algorithms for two biological ToF-SIMS imaging data sets. We investigated the extracted features and used the dimensionality-reduced data to train additional ML algorithms. By converting two-dimensional convolutional layers to three-dimensional (3D), we also showed how the CNNAE can be extended to 3D ToF-SIMS images. In general, the CNNAE produced features with significantly higher contrast and autocorrelation than other techniques. Furthermore, histologically recognizable features in the data were more accurately represented. The extension of the CNNAE to 3D data also provided an important proof of principle for the analysis of more complex 3D data sets.
Collapse
Affiliation(s)
- Wil Gardner
- Centre for Materials and Surface Science and Department of Chemistry and Physics, La Trobe University, Bundoora, Victoria 3086, Australia.,La Trobe Institute for Molecular Sciences, La Trobe University, Bundoora, Victoria 3086, Australia.,CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | - David A Winkler
- La Trobe Institute for Molecular Sciences, La Trobe University, Bundoora, Victoria 3086, Australia.,Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, U.K
| | - Suzanne M Cutts
- La Trobe Institute for Molecular Sciences, La Trobe University, Bundoora, Victoria 3086, Australia
| | - Steven A Torney
- La Trobe Institute for Molecular Sciences, La Trobe University, Bundoora, Victoria 3086, Australia
| | - Geoffrey A Pietersz
- Immune Therapies Laboratory, Burnet Institute, Melbourne, Victoria 3004, Australia.,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | | | - Paul J Pigram
- Centre for Materials and Surface Science and Department of Chemistry and Physics, La Trobe University, Bundoora, Victoria 3086, Australia
| |
Collapse
|
29
|
Boyaval F, Dalebout H, Van Zeijl R, Wang W, Fariña-Sarasqueta A, Lageveen-Kammeijer GSM, Boonstra JJ, McDonnell LA, Wuhrer M, Morreau H, Heijs B. High-Mannose N-Glycans as Malignant Progression Markers in Early-Stage Colorectal Cancer. Cancers (Basel) 2022; 14:1552. [PMID: 35326703 PMCID: PMC8945895 DOI: 10.3390/cancers14061552] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/09/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
The increase incidence of early colorectal cancer (T1 CRC) last years is mainly due to the introduction of population-based screening for CRC. T1 CRC staging based on histological criteria remains challenging and there is high variability among pathologists in the scoring of these criteria. It is crucial to unravel the biology behind the progression of adenoma into T1 CRC. Glycomic studies have reported extensively on alterations of the N-glycomic pattern in CRC; therefore, investigating these alterations may reveal new insights into the development of T1 CRC. We used matrix-assisted laser desorption ionization (MALDI) mass spectrometry imaging (MSI) to spatially profile the N-glycan species in a cohort of pT1 CRC using archival formalin-fixed and paraffin-embedded (FFPE) material. To generate structural information on the observed N-glycans, CE-ESI-MS/MS was used in conjunction with MALDI-MSI. Relative intensities and glycosylation traits were calculated based on a panel of 58 N-glycans. Our analysis showed pronounced differences between normal epithelium, dysplastic, and carcinoma regions. High-mannose-type N-glycans were higher in the dysplastic region than in carcinoma, which correlates to increased proliferation of the cells. We observed changes in the cancer invasive front, including higher expression of α2,3-linked sialic acids which followed the glycosylation pattern of the carcinoma region.
Collapse
Affiliation(s)
- Fanny Boyaval
- Department of Pathology, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands;
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Hans Dalebout
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - René Van Zeijl
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Wenjun Wang
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Arantza Fariña-Sarasqueta
- Department of Pathology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Guinevere S. M. Lageveen-Kammeijer
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Jurjen J. Boonstra
- Department of Gastroenterology and Hepatology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands;
| | - Liam A. McDonnell
- Fondazione Pisana per la Scienza ONLUS, Via Ferruccio Giovannini, 56017 San Giuliano Terme, Italy;
| | - Manfred Wuhrer
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands;
| | - Bram Heijs
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands; (H.D.); (R.V.Z.); (W.W.); (G.S.M.L.-K.); (M.W.)
| |
Collapse
|
30
|
DeLaney K, Phetsanthad A, Li L. ADVANCES IN HIGH-RESOLUTION MALDI MASS SPECTROMETRY FOR NEUROBIOLOGY. MASS SPECTROMETRY REVIEWS 2022; 41:194-214. [PMID: 33165982 PMCID: PMC8106695 DOI: 10.1002/mas.21661] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 09/13/2020] [Indexed: 05/08/2023]
Abstract
Research in the field of neurobiology and neurochemistry has seen a rapid expansion in the last several years due to advances in technologies and instrumentation, facilitating the detection of biomolecules critical to the complex signaling of neurons. Part of this growth has been due to the development and implementation of high-resolution Fourier transform (FT) mass spectrometry (MS), as is offered by FT ion cyclotron resonance (FTICR) and Orbitrap mass analyzers, which improves the accuracy of measurements and helps resolve the complex biological mixtures often analyzed in the nervous system. The coupling of matrix-assisted laser desorption/ionization (MALDI) with high-resolution MS has drastically expanded the information that can be obtained with these complex samples. This review discusses notable technical developments in MALDI-FTICR and MALDI-Orbitrap platforms and their applications toward molecules in the nervous system, including sequence elucidation and profiling with de novo sequencing, analysis of post-translational modifications, in situ analysis, key advances in sample preparation and handling, quantitation, and imaging. Notable novel applications are also discussed to highlight key developments critical to advancing our understanding of neurobiology and providing insight into the exciting future of this field. © 2020 John Wiley & Sons Ltd. Mass Spec Rev.
Collapse
Affiliation(s)
- Kellen DeLaney
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA
| | - Ashley Phetsanthad
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706, USA
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, USA
| |
Collapse
|
31
|
Applications of multivariate analysis and unsupervised machine learning to ToF-SIMS images of organic, bioorganic, and biological systems. Biointerphases 2022; 17:020802. [DOI: 10.1116/6.0001590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging offers a powerful, label-free method for exploring organic, bioorganic, and biological systems. The technique is capable of very high spatial resolution, while also producing an enormous amount of information about the chemical and molecular composition of a surface. However, this information is inherently complex, making interpretation and analysis of the vast amount of data produced by a single ToF-SIMS experiment a considerable challenge. Much research over the past few decades has focused on the application and development of multivariate analysis (MVA) and machine learning (ML) techniques that find meaningful patterns and relationships in these datasets. Here, we review the unsupervised algorithms—that is, algorithms that do not require ground truth labels—that have been applied to ToF-SIMS images, as well as other algorithms and approaches that have been used in the broader family of mass spectrometry imaging (MSI) techniques. We first give a nontechnical overview of several commonly used classes of unsupervised algorithms, such as matrix factorization, clustering, and nonlinear dimensionality reduction. We then review the application of unsupervised algorithms to various organic, bioorganic, and biological systems including cells and tissues, organic films, residues and coatings, and spatially structured systems such as polymer microarrays. We then cover several novel algorithms employed for other MSI techniques that have received little attention from ToF-SIMS imaging researchers. We conclude with a brief outline of potential future directions for the application of MVA and ML algorithms to ToF-SIMS images.
Collapse
|
32
|
Abdelmoula WM, Stopka SA, Randall EC, Regan M, Agar JN, Sarkaria JN, Wells WM, Kapur T, Agar NYR. massNet: integrated processing and classification of spatially resolved mass spectrometry data using deep learning for rapid tumor delineation. Bioinformatics 2022; 38:2015-2021. [PMID: 35040929 PMCID: PMC8963284 DOI: 10.1093/bioinformatics/btac032] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 01/21/2023] Open
Abstract
MOTIVATION Mass spectrometry imaging (MSI) provides rich biochemical information in a label-free manner and therefore holds promise to substantially impact current practice in disease diagnosis. However, the complex nature of MSI data poses computational challenges in its analysis. The complexity of the data arises from its large size, high-dimensionality and spectral nonlinearity. Preprocessing, including peak picking, has been used to reduce raw data complexity; however, peak picking is sensitive to parameter selection that, perhaps prematurely, shapes the downstream analysis for tissue classification and ensuing biological interpretation. RESULTS We propose a deep learning model, massNet, that provides the desired qualities of scalability, nonlinearity and speed in MSI data analysis. This deep learning model was used, without prior preprocessing and peak picking, to classify MSI data from a mouse brain harboring a patient-derived tumor. The massNet architecture established automatically learning of predictive features, and automated methods were incorporated to identify peaks with potential for tumor delineation. The model's performance was assessed using cross-validation, and the results demonstrate higher accuracy and a substantial gain in speed compared to the established classical machine learning method, support vector machine. AVAILABILITY AND IMPLEMENTATION https://github.com/wabdelmoula/massNet. The data underlying this article are available in the NIH Common Fund's National Metabolomics Data Repository (NMDR) Metabolomics Workbench under project id (PR001292) with http://dx.doi.org/10.21228/M8Q70T. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Walid M Abdelmoula
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA,Invicro LLC, Boston, MA 02210, USA
| | - Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA,Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth C Randall
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Regan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey N Agar
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02111, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55902, USA
| | - William M Wells
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA,Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02139, USA
| | - Tina Kapur
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA,Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA,Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA,To whom correspondence should be addressed.
| |
Collapse
|
33
|
Lopez BGC, Kohale IN, Du Z, Korsunsky I, Abdelmoula WM, Dai Y, Stopka SA, Gaglia G, Randall EC, Regan MS, Basu SS, Clark AR, Marin BM, Mladek AC, Burgenske DM, Agar JN, Supko JG, Grossman SA, Nabors LB, Raychaudhuri S, Ligon KL, Wen PY, Alexander B, Lee EQ, Santagata S, Sarkaria J, White FM, Agar NYR. Multimodal platform for assessing drug distribution and response in clinical trials. Neuro Oncol 2022; 24:64-77. [PMID: 34383057 PMCID: PMC8730776 DOI: 10.1093/neuonc/noab197] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Response to targeted therapy varies between patients for largely unknown reasons. Here, we developed and applied an integrative platform using mass spectrometry imaging (MSI), phosphoproteomics, and multiplexed tissue imaging for mapping drug distribution, target engagement, and adaptive response to gain insights into heterogeneous response to therapy. METHODS Patient-derived xenograft (PDX) lines of glioblastoma were treated with adavosertib, a Wee1 inhibitor, and tissue drug distribution was measured with MALDI-MSI. Phosphoproteomics was measured in the same tumors to identify biomarkers of drug target engagement and cellular adaptive response. Multiplexed tissue imaging was performed on sister sections to evaluate spatial co-localization of drug and cellular response. The integrated platform was then applied on clinical specimens from glioblastoma patients enrolled in the phase 1 clinical trial. RESULTS PDX tumors exposed to different doses of adavosertib revealed intra- and inter-tumoral heterogeneity of drug distribution and integration of the heterogeneous drug distribution with phosphoproteomics and multiplexed tissue imaging revealed new markers of molecular response to adavosertib. Analysis of paired clinical specimens from patients enrolled in the phase 1 clinical trial informed the translational potential of the identified biomarkers in studying patient's response to adavosertib. CONCLUSIONS The multimodal platform identified a signature of drug efficacy and patient-specific adaptive responses applicable to preclinical and clinical drug development. The information generated by the approach may inform mechanisms of success and failure in future early phase clinical trials, providing information for optimizing clinical trial design and guiding future application into clinical practice.
Collapse
Affiliation(s)
- Begoña G C Lopez
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ishwar N Kohale
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ziming Du
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ilya Korsunsky
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Walid M Abdelmoula
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yang Dai
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sylwia A Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Giorgio Gaglia
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth C Randall
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael S Regan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sankha S Basu
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amanda R Clark
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bianca-Maria Marin
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ann C Mladek
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Jeffrey N Agar
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, USA
| | - Jeffrey G Supko
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Stuart A Grossman
- Brain Cancer Program, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Louis B Nabors
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Keith L Ligon
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Brian Alexander
- Department of Radiation Oncology, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Sandro Santagata
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jann Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Forest M White
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Shedlock CJ, Stumpo KA. Data parsing in mass spectrometry imaging using R Studio and Cardinal: A tutorial. J Mass Spectrom Adv Clin Lab 2022; 23:58-70. [PMID: 35072143 PMCID: PMC8762469 DOI: 10.1016/j.jmsacl.2021.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023] Open
Abstract
Mass spectrometry imaging (MSI) has emerged as a rapidly expanding field in the MS community. The analysis of large spectral data is further complicated by the added spatial dimension of MSI. A plethora of resources exist for expert users to begin parsing MSI data in R, but there is a critical lack of guidance for absolute beginners. This tutorial is designed to serve as a one-stop guide to start using R with MSI data and describe the possibilities that data science can bring to MSI analysis.
Collapse
Key Words
- AuNP, gold nanoparticle
- Cardinal
- DESI, desorption electrospray ioniziation
- Data validation
- IACUC, Institutional Animal Care and Use Committee
- ITO, indium tin oxide
- MSI, mass spectrometry imaging
- Mass spectrometry imaging
- PCA, principal component analysis
- R Studio
- RAM, random access memory
- RMS, root mean squared
- SNR, signal to noise ratio
- SSC, spatial shrunken centroid
- SSD, solid state drive
- TIC, total ion current
Collapse
Affiliation(s)
- Cameron J. Shedlock
- Department of Chemistry, University of Scranton, Scranton, PA 18510, United States
| | - Katherine A. Stumpo
- Department of Chemistry, University of Scranton, Scranton, PA 18510, United States
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Bruker Scientific, Billerica, MA 01821, United States
| |
Collapse
|
35
|
Neumann JM, Freitag H, Hartmann JS, Niehaus K, Galanis M, Griesshammer M, Kellner U, Bednarz H. Subtyping non-small cell lung cancer by histology-guided spatial metabolomics. J Cancer Res Clin Oncol 2021; 148:351-360. [PMID: 34839410 PMCID: PMC8800912 DOI: 10.1007/s00432-021-03834-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/11/2021] [Indexed: 01/04/2023]
Abstract
Purpose Most cancer-related deaths worldwide are associated with lung cancer. Subtyping of non-small cell lung cancer (NSCLC) into adenocarcinoma (AC) and squamous cell carcinoma (SqCC) is of importance, as therapy regimes differ. However, conventional staining and immunohistochemistry have their limitations. Therefore, a spatial metabolomics approach was aimed to detect differences between subtypes and to discriminate tumor and stroma regions in tissues. Methods Fresh-frozen NSCLC tissues (n = 35) were analyzed by matrix-assisted laser desorption/ionization-mass spectrometry imaging (MALDI-MSI) of small molecules (< m/z 1000). Measured samples were subsequently stained and histopathologically examined. A differentiation of subtypes and a discrimination of tumor and stroma regions was performed by receiver operating characteristic analysis and machine learning algorithms. Results Histology-guided spatial metabolomics revealed differences between AC and SqCC and between NSCLC tumor and tumor microenvironment. A diagnostic ability of 0.95 was achieved for the discrimination of AC and SqCC. Metabolomic contrast to the tumor microenvironment was revealed with an area under the curve of 0.96 due to differences in phospholipid profile. Furthermore, the detection of NSCLC with rarely arising mutations of the isocitrate dehydrogenase (IDH) gene was demonstrated through 45 times enhanced oncometabolite levels. Conclusion MALDI-MSI of small molecules can contribute to NSCLC subtyping. Measurements can be performed intraoperatively on a single tissue section to support currently available approaches. Moreover, the technique can be beneficial in screening of IDH-mutants for the characterization of these seldom cases promoting the development of treatment strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03834-w.
Collapse
Affiliation(s)
- Judith Martha Neumann
- Faculty of Biology, Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Hinrich Freitag
- Institut für Pathologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Jasmin Saskia Hartmann
- Faculty of Biology, Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Karsten Niehaus
- Faculty of Biology, Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Michail Galanis
- Universitätsklinik für Allgemeinchirurgie, Viszeral-, Thorax- und Endokrine Chirurgie, Johannes Wesling Klinikum Minden, Minden, Germany.,Clinic for Thoracic Surgery and Thoracic Endoscopy, University Hospital Bielefeld Mitte, Bielefeld, Germany
| | - Martin Griesshammer
- Universitätsklinik für Hämatologie, Onkologie, Hämostaseologie und Palliativmedizin, Universitätszentrum Innere Medizin, Johannes Wesling Klinikum Minden, Minden, Germany
| | - Udo Kellner
- Institut für Pathologie, Medizinische Hochschule Hannover, Hannover, Germany. .,Institut für Pathologie, Johannes Wesling Klinikum, Minden, Germany.
| | - Hanna Bednarz
- Faculty of Biology, Proteome and Metabolome Research, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany. .,Medical School OWL, AG1: Sustainable Environmental Health Sciences, Bielefeld University, Bielefeld, Germany.
| |
Collapse
|
36
|
Peak learning of mass spectrometry imaging data using artificial neural networks. Nat Commun 2021; 12:5544. [PMID: 34545087 PMCID: PMC8452737 DOI: 10.1038/s41467-021-25744-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
Mass spectrometry imaging (MSI) is an emerging technology that holds potential for improving, biomarker discovery, metabolomics research, pharmaceutical applications and clinical diagnosis. Despite many solutions being developed, the large data size and high dimensional nature of MSI, especially 3D datasets, still pose computational and memory complexities that hinder accurate identification of biologically relevant molecular patterns. Moreover, the subjectivity in the selection of parameters for conventional pre-processing approaches can lead to bias. Therefore, we assess if a probabilistic generative model based on a fully connected variational autoencoder can be used for unsupervised analysis and peak learning of MSI data to uncover hidden structures. The resulting msiPL method learns and visualizes the underlying non-linear spectral manifold, revealing biologically relevant clusters of tissue anatomy in a mouse kidney and tumor heterogeneity in human prostatectomy tissue, colorectal carcinoma, and glioblastoma mouse model, with identification of underlying m/z peaks. The method is applied for the analysis of MSI datasets ranging from 3.3 to 78.9 GB, without prior pre-processing and peak picking, and acquired using different mass spectrometers at different centers.
Collapse
|
37
|
Zhang J, Sans M, Garza KY, Eberlin LS. MASS SPECTROMETRY TECHNOLOGIES TO ADVANCE CARE FOR CANCER PATIENTS IN CLINICAL AND INTRAOPERATIVE USE. MASS SPECTROMETRY REVIEWS 2021; 40:692-720. [PMID: 33094861 DOI: 10.1002/mas.21664] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 09/09/2020] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
Developments in mass spectrometry technologies have driven a widespread interest and expanded their use in cancer-related research and clinical applications. In this review, we highlight the developments in mass spectrometry methods and instrumentation applied to direct tissue analysis that have been tailored at enhancing performance in clinical research as well as facilitating translation and implementation of mass spectrometry in clinical settings, with a focus on cancer-related studies. Notable studies demonstrating the capabilities of direct mass spectrometry analysis in biomarker discovery, cancer diagnosis and prognosis, tissue analysis during oncologic surgeries, and other clinically relevant problems that have the potential to substantially advance cancer patient care are discussed. Key challenges that need to be addressed before routine clinical implementation including regulatory efforts are also discussed. Overall, the studies highlighted in this review demonstrate the transformative potential of mass spectrometry technologies to advance clinical research and care for cancer patients. © 2020 Wiley Periodicals, Inc. Mass Spec Rev.
Collapse
Affiliation(s)
- Jialing Zhang
- Department of Chemistry, University of Texas at Austin, Austin, TX
| | - Marta Sans
- Department of Chemistry, University of Texas at Austin, Austin, TX
| | - Kyana Y Garza
- Department of Chemistry, University of Texas at Austin, Austin, TX
| | - Livia S Eberlin
- Department of Chemistry, University of Texas at Austin, Austin, TX
| |
Collapse
|
38
|
Basu SS, Agar NYR. Bringing Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry Imaging to the Clinics. Clin Lab Med 2021; 41:309-324. [PMID: 34020766 DOI: 10.1016/j.cll.2021.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) is an emerging analytical technique that promises to change tissue-based diagnostics. This article provides a brief introduction to MALDI MSI as well as clinical diagnostic workflows and opportunities to apply this powerful approach. It describes various MALDI MSI applications, from more clinically mature applications such as cancer to emerging applications such as infectious diseases and drug distribution. In addition, it discusses the analytical considerations that need to be considered when bringing these approaches to different diagnostic problems and settings.
Collapse
Affiliation(s)
- Sankha S Basu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
39
|
Bensussan AV, Lin J, Guo C, Katz R, Krishnamurthy S, Cressman E, Eberlin LS. Distinguishing Non-Small Cell Lung Cancer Subtypes in Fine Needle Aspiration Biopsies by Desorption Electrospray Ionization Mass Spectrometry Imaging. Clin Chem 2021; 66:1424-1433. [PMID: 33141910 DOI: 10.1093/clinchem/hvaa207] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/17/2020] [Indexed: 11/13/2022]
Abstract
BACKGROUND Distinguishing adenocarcinoma and squamous cell carcinoma subtypes of non-small cell lung cancers is critical to patient care. Preoperative minimally-invasive biopsy techniques, such as fine needle aspiration (FNA), are increasingly used for lung cancer diagnosis and subtyping. Yet, histologic distinction of lung cancer subtypes in FNA material can be challenging. Here, we evaluated the usefulness of desorption electrospray ionization mass spectrometry imaging (DESI-MSI) to diagnose and differentiate lung cancer subtypes in tissues and FNA samples. METHODS DESI-MSI was used to analyze 22 normal, 26 adenocarcinoma, and 25 squamous cell carcinoma lung tissues. Mass spectra obtained from the tissue sections were used to generate and validate statistical classifiers for lung cancer diagnosis and subtyping. Classifiers were then tested on DESI-MSI data collected from 16 clinical FNA samples prospectively collected from 8 patients undergoing interventional radiology guided FNA. RESULTS Various metabolites and lipid species were detected in the mass spectra obtained from lung tissues. The classifiers generated from tissue sections yielded 100% accuracy, 100% sensitivity, and 100% specificity for lung cancer diagnosis, and 73.5% accuracy for lung cancer subtyping for the training set of tissues, per-patient. On the validation set of tissues, 100% accuracy for lung cancer diagnosis and 94.1% accuracy for lung cancer subtyping were achieved. When tested on the FNA samples, 100% diagnostic accuracy and 87.5% accuracy on subtyping were achieved per-slide. CONCLUSIONS DESI-MSI can be useful as an ancillary technique to conventional cytopathology for diagnosis and subtyping of non-small cell lung cancers.
Collapse
Affiliation(s)
- Alena V Bensussan
- Department of Chemistry, The University of Texas at Austin, Austin, TX
| | - John Lin
- Department of Chemistry, The University of Texas at Austin, Austin, TX
| | - Chunxiao Guo
- Department of Interventional Radiology, Division of Diagnostic Imaging, MD Anderson Cancer Center, Houston, TX
| | - Ruth Katz
- Department of Pathology, Division of Pathology and Laboratory Medicine, MD Anderson Cancer Center, Houston, TX
| | - Savitri Krishnamurthy
- Department of Pathology, Division of Pathology and Laboratory Medicine, MD Anderson Cancer Center, Houston, TX
| | - Erik Cressman
- Department of Interventional Radiology, Division of Diagnostic Imaging, MD Anderson Cancer Center, Houston, TX
| | - Livia S Eberlin
- Department of Chemistry, The University of Texas at Austin, Austin, TX
| |
Collapse
|
40
|
Vaysse PM, Grabsch HI, van den Hout MFCM, Bemelmans MHA, Heeren RMA, Olde Damink SWM, Porta Siegel T. Real-time lipid patterns to classify viable and necrotic liver tumors. J Transl Med 2021; 101:381-395. [PMID: 33483597 DOI: 10.1038/s41374-020-00526-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Real-time tissue classifiers based on molecular patterns are emerging tools for fast tumor diagnosis. Here, we used rapid evaporative ionization mass spectrometry (REIMS) and multivariate statistical analysis (principal component analysis-linear discriminant analysis) to classify tissues with subsequent comparison to gold standard histopathology. We explored whether REIMS lipid patterns can identify human liver tumors and improve the rapid characterization of their underlying metabolic features. REIMS-based classification of liver parenchyma (LP), hepatocellular carcinoma (HCC), and metastatic adenocarcinoma (MAC) reached an accuracy of 98.3%. Lipid patterns of LP were more similar to those of HCC than to those of MAC and allowed clear distinction between primary and metastatic liver tumors. HCC lipid patterns were more heterogeneous than those of MAC, which is consistent with the variation seen in the histopathological phenotype. A common ceramide pattern discriminated necrotic from viable tumor in MAC with 92.9% accuracy and in other human tumors. Targeted analysis of ceramide and related sphingolipid mass features in necrotic tissues may provide a new classification of tumor cell death based on metabolic shifts. Real-time lipid patterns may have a role in future clinical decision-making in cancer precision medicine.
Collapse
Affiliation(s)
- Pierre-Maxence Vaysse
- Maastricht MultiModal Molecular Imaging Institute (M4i), University of Maastricht, Maastricht, The Netherlands
- Department of Surgery, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Otorhinolaryngology, Head & Neck Surgery, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Heike I Grabsch
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, UK
| | - Mari F C M van den Hout
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Marc H A Bemelmans
- Department of Surgery, Maastricht University Medical Center+, Maastricht, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ron M A Heeren
- Maastricht MultiModal Molecular Imaging Institute (M4i), University of Maastricht, Maastricht, The Netherlands
| | - Steven W M Olde Damink
- Department of Surgery, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of General, Visceral and Transplantation Surgery, RWTH University Hospital Aachen, Aachen, Germany
- NUTRIM School of Nutrition and Translational Research in Metabolism Faculty of Health, University of Maastricht, Maastricht, The Netherlands
| | - Tiffany Porta Siegel
- Maastricht MultiModal Molecular Imaging Institute (M4i), University of Maastricht, Maastricht, The Netherlands.
| |
Collapse
|
41
|
Hu H, Yin R, Brown HM, Laskin J. Spatial Segmentation of Mass Spectrometry Imaging Data by Combining Multivariate Clustering and Univariate Thresholding. Anal Chem 2021; 93:3477-3485. [PMID: 33570915 PMCID: PMC7904669 DOI: 10.1021/acs.analchem.0c04798] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Spatial segmentation partitions mass spectrometry imaging (MSI) data into distinct regions, providing a concise visualization of the vast amount of data and identifying regions of interest (ROIs) for downstream statistical analysis. Unsupervised approaches are particularly attractive, as they may be used to discover the underlying subpopulations present in the high-dimensional MSI data without prior knowledge of the properties of the sample. Herein, we introduce an unsupervised spatial segmentation approach, which combines multivariate clustering and univariate thresholding to generate comprehensive spatial segmentation maps of the MSI data. This approach combines matrix factorization and manifold learning to enable high-quality image segmentation without an extensive hyperparameter search. In parallel, some ion images inadequately represented in the multivariate analysis were treated using univariate thresholding to generate complementary spatial segments. The final spatial segmentation map was assembled from segment candidates that were generated using both techniques. We demonstrate the performance and robustness of this approach for two MSI data sets of mouse uterine and kidney tissue sections that were acquired with different spatial resolutions. The resulting segmentation maps are easy to interpret and project onto the known anatomical regions of the tissue.
Collapse
Affiliation(s)
- Hang Hu
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ruichuan Yin
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Hilary M Brown
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Julia Laskin
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
42
|
Neumann EK, Djambazova KV, Caprioli RM, Spraggins JM. Multimodal Imaging Mass Spectrometry: Next Generation Molecular Mapping in Biology and Medicine. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:2401-2415. [PMID: 32886506 PMCID: PMC9278956 DOI: 10.1021/jasms.0c00232] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Imaging mass spectrometry has become a mature molecular mapping technology that is used for molecular discovery in many medical and biological systems. While powerful by itself, imaging mass spectrometry can be complemented by the addition of other orthogonal, chemically informative imaging technologies to maximize the information gained from a single experiment and enable deeper understanding of biological processes. Within this review, we describe MALDI, SIMS, and DESI imaging mass spectrometric technologies and how these have been integrated with other analytical modalities such as microscopy, transcriptomics, spectroscopy, and electrochemistry in a field termed multimodal imaging. We explore the future of this field and discuss forthcoming developments that will bring new insights to help unravel the molecular complexities of biological systems, from single cells to functional tissue structures and organs.
Collapse
Affiliation(s)
- Elizabeth K Neumann
- Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States
| | - Katerina V Djambazova
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States
| | - Richard M Caprioli
- Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States
- Department of Pharmacology, Vanderbilt University, 2220 Pierce Avenue, Nashville, Tennessee 37232, United States
- Department of Medicine, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States
| | - Jeffrey M Spraggins
- Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue S #9160, Nashville, Tennessee 37235, United States
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States
| |
Collapse
|
43
|
McLaughlin N, Bielinski TM, Tressler CM, Barton E, Glunde K, Stumpo KA. Pneumatically Sprayed Gold Nanoparticles for Mass Spectrometry Imaging of Neurotransmitters. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:2452-2461. [PMID: 32841002 DOI: 10.1021/jasms.0c00156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Using citrate-capped gold nanoparticles (AuNPs) for laser desorption ionization mass spectrometry (LDI-MS) is an approach that has demonstrated broad applicability to ionization of different classes of molecules. Here, we show a simple AuNP-based approach for the ionization of neurotransmitters. Specifically, the detection of acetylcholine, dopamine, epinephrine, glutamine, 4-aminobutyric acid, norepinephrine, octopamine, and serotonin was achieved at physiologically relevant concentrations in serum and homogenized tissue. Additionally, pneumatic spraying of AuNPs onto tissue sections facilitated mass spectrometry imaging (MSI) of rabbit brain tissue sections, zebrafish embryos, and neuroblastoma cells for several neurotransmitters simultaneously using this quick and simple sample preparation. AuNP LDI-MS achieved mapping of neurotransmitters in fine structures of zebrafish embryos and neuroblastoma cells at a lateral spatial resolution of 5 μm. The use of AuNPs to ionize small aminergic neurotransmitters in situ provides a fast, high-spatial resolution method for simultaneous detection of a class of molecules that typically evade comprehensive detection with traditional matrixes.
Collapse
Affiliation(s)
- Nolan McLaughlin
- Department of Chemistry, University of Scranton, Scranton, Pennsylvania 18510, United States
| | - Tyler M Bielinski
- Department of Chemistry, University of Scranton, Scranton, Pennsylvania 18510, United States
| | - Caitlin M Tressler
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Eric Barton
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Kristine Glunde
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Katherine A Stumpo
- Department of Chemistry, University of Scranton, Scranton, Pennsylvania 18510, United States
| |
Collapse
|
44
|
Pietkiewicz D, Horała A, Plewa S, Jasiński P, Nowak-Markwitz E, Kokot ZJ, Matysiak J. MALDI-MSI-A Step Forward in Overcoming the Diagnostic Challenges in Ovarian Tumors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E7564. [PMID: 33080944 PMCID: PMC7589662 DOI: 10.3390/ijerph17207564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023]
Abstract
This study presents the use of matrix-assisted laser desorption and ionization mass spectrometry imaging (MALDI-MSI) directly on the tissue of two ovarian tumors that often present a diagnostic challenge, a low-grade serous borderline ovarian tumor and ovarian fibrothecoma. Different spatial distribution of m/z values within the tissue samples was observed, and regiospecific peaks were identified. Among the 106 peaks in the borderline ovarian tumor five, regiospecific peaks (m/z: 2861.35; 2775.79; 3368.34; 3438.43; 4936.37) were selected using FlexImaging software. Subsequently, the distribution of those selected peaks was visualized on the fibrothecoma tissue section, which demonstrated the differences in the tissue homo-/heterogeneous structure of both tumors. The comparison with the histopathological staining of the ovarian borderline tumor tissue section, obtained during serial sectioning, showed a close correlation of the molecular map with the morphological and histopathological features of the tissue and allowed the identification of different tissue types within the sample. This study highlights the potential significance of MSI in enabling morphological characterization of ovarian tumors as well as correct diagnosis and further prognosis than thus far seen in the literature. Osteopontin, tropomyosin and orosomucoid are only a couple of the molecules investigated using MALDI-MSI in ovarian cancer research. This study, in line with the available literature, proves the potential of MALDI-MSI to overcome the current limitations of classic histopathological examination giving a more in-depth insight into the tissue structure and thus lead to the more accurate differential diagnosis of ovarian tumors, especially in the most challenging cases.
Collapse
Affiliation(s)
- Dagmara Pietkiewicz
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 6 Grunwaldzka Street, 60-780 Poznan, Poland; (D.P.); (S.P.)
| | - Agnieszka Horała
- Gynecologic Oncology Department, Poznan University of Medical Sciences, 33 Polna Street, 60-535 Poznan, Poland; (A.H.); (E.N.-M.)
| | - Szymon Plewa
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 6 Grunwaldzka Street, 60-780 Poznan, Poland; (D.P.); (S.P.)
| | - Piotr Jasiński
- Department of Pathology Gynecological and Obstetric Clinical Hospital, Poznan University of Medical Sciences, 33 Polna Street, 60-535 Poznan, Poland;
| | - Ewa Nowak-Markwitz
- Gynecologic Oncology Department, Poznan University of Medical Sciences, 33 Polna Street, 60-535 Poznan, Poland; (A.H.); (E.N.-M.)
| | - Zenon J. Kokot
- Faculty of Health Sciences, Calisia University, 13 Kaszubska Street, 62-800 Kalisz, Poland;
| | - Jan Matysiak
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, 6 Grunwaldzka Street, 60-780 Poznan, Poland; (D.P.); (S.P.)
| |
Collapse
|
45
|
Gardner W, Cutts SM, Phillips DR, Pigram PJ. Understanding mass spectrometry images: complexity to clarity with machine learning. Biopolymers 2020; 112:e23400. [PMID: 32937683 DOI: 10.1002/bip.23400] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 11/08/2022]
Abstract
The application of artificial intelligence and machine learning to hyperspectral mass spectrometry imaging (MSI) data has received considerable attention over recent years. Various methodologies have shown great promise in their ability to handle the complexity and size of MSI data sets. Advances in this area have been particularly appealing for MSI of biological samples, which typically produce highly complicated data with often subtle relationships between features. There are many different machine learning approaches that have been applied to MSI data over the past two decades. In this review, we focus on a subset of non-linear machine learning techniques that have mostly only been applied in the past 5 years. Specifically, we review the use of the self-organizing map (SOM), SOM with relational perspective mapping (SOM-RPM), t-distributed stochastic neighbor embedding (t-SNE) and uniform manifold approximation and projection (UMAP). While not their only functionality, we have grouped these techniques based on their ability to produce what we refer to as similarity maps. Similarity maps are color representations of hyperspectral data, in which spectral similarity between pixels-that is, their distance in high-dimensional space-is represented by relative color similarity. In discussing these techniques, we describe, briefly, their associated algorithms and functionalities, and also outline applications in MSI research with a strong focus on biological sample types. The aim of this review is therefore to introduce this relatively recent paradigm for visualizing and exploring hyperspectral MSI, while also providing a comparison between each technique discussed.
Collapse
Affiliation(s)
- Wil Gardner
- Centre for Materials and Surface Science and Department of Chemistry and Physics, La Trobe University, Melbourne, Victoria, Australia.,La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, Australia.,CSIRO Manufacturing, Clayton, Victoria, Australia
| | - Suzanne M Cutts
- La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Don R Phillips
- La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Paul J Pigram
- Centre for Materials and Surface Science and Department of Chemistry and Physics, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
46
|
Nomura F, Tsuchida S, Murata S, Satoh M, Matsushita K. Mass spectrometry-based microbiological testing for blood stream infection. Clin Proteomics 2020; 17:14. [PMID: 32435163 PMCID: PMC7222329 DOI: 10.1186/s12014-020-09278-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The most successful application of mass spectrometry (MS) in laboratory medicine is identification (ID) of microorganisms using matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) in blood stream infection. We describe MALDI-TOF MS-based bacterial ID with particular emphasis on the methods so far developed to directly identify microorganisms from positive blood culture bottles with MALDI-TOF MS including our own protocols. We touch upon the increasing roles of Liquid chromatography (LC) coupled with tandem mass spectrometry (MS/MS) as well. MAIN BODY Because blood culture bottles contain a variety of nonbacterial proteins that may interfere with analysis and interpretation, appropriate pretreatments are prerequisites for successful ID. Pretreatments include purification of bacterial pellets and short-term subcultures to form microcolonies prior to MALDI-TOF MS analysis. Three commercial protocols are currently available: the Sepsityper® kit (Bruker Daltonics), the Vitek MS blood culture kit (bioMerieux, Inc.), and the rapid BACpro® II kit (Nittobo Medical Co., Tokyo). Because these commercially available kits are costly and bacterial ID rates using these kits are not satisfactory, particularly for Gram-positive bacteria, various home-brew protocols have been developed: 1. Stepwise differential sedimentation of blood cells and microorganisms, 2. Combination of centrifugation and lysis procedures, 3. Lysis-vacuum filtration, and 4. Centrifugation and membrane filtration technique (CMFT). We prospectively evaluated the performance of this CMFT protocol compared with that of Sepsityper® using 170 monomicrobial positive blood cultures. Although preliminary, the performance of the CMFT was significantly better than that of Sepsityper®, particularly for Gram-positive isolates. MALDI-TOF MS-based testing of polymicrobial blood specimens, however, is still challenging. Also, its contribution to assessment of susceptibility and resistance to antibiotics is still limited. For this purpose, liquid chromatography (LC) coupled with tandem mass spectrometry (MS/MS) should be more useful because this approach can identify as many as several thousand peptide sequences. CONCLUSION MALDI-TOF MS is now an essential tool for rapid bacterial ID of pathogens that cause blood stream infection. For the purpose of assessment of susceptibility and resistance to antibiotics of the pathogens, the roles of liquid chromatography (LC) coupled with tandem mass spectrometry (MS/MS) will increase in the future.
Collapse
Affiliation(s)
- Fumio Nomura
- Division of Clinical Mass Spectrometry, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Sachio Tsuchida
- Division of Clinical Mass Spectrometry, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Syota Murata
- Division of Laboratory Medicine, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Mamoru Satoh
- Division of Clinical Mass Spectrometry, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Kazuyuki Matsushita
- Division of Laboratory Medicine, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| |
Collapse
|
47
|
Alexandrov T. Spatial Metabolomics and Imaging Mass Spectrometry in the Age of Artificial Intelligence. Annu Rev Biomed Data Sci 2020; 3:61-87. [PMID: 34056560 DOI: 10.1146/annurev-biodatasci-011420-031537] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Spatial metabolomics is an emerging field of omics research that has enabled localizing metabolites, lipids, and drugs in tissue sections, a feat considered impossible just two decades ago. Spatial metabolomics and its enabling technology-imaging mass spectrometry-generate big hyper-spectral imaging data that have motivated the development of tailored computational methods at the intersection of computational metabolomics and image analysis. Experimental and computational developments have recently opened doors to applications of spatial metabolomics in life sciences and biomedicine. At the same time, these advances have coincided with a rapid evolution in machine learning, deep learning, and artificial intelligence, which are transforming our everyday life and promise to revolutionize biology and healthcare. Here, we introduce spatial metabolomics through the eyes of a computational scientist, review the outstanding challenges, provide a look into the future, and discuss opportunities granted by the ongoing convergence of human and artificial intelligence.
Collapse
Affiliation(s)
- Theodore Alexandrov
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
48
|
Holzlechner M, Eugenin E, Prideaux B. Mass spectrometry imaging to detect lipid biomarkers and disease signatures in cancer. Cancer Rep (Hoboken) 2019; 2:e1229. [PMID: 32729258 PMCID: PMC7941519 DOI: 10.1002/cnr2.1229] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Current methods to identify, classify, and predict tumor behavior mostly rely on histology, immunohistochemistry, and molecular determinants. However, better predictive markers are required for tumor diagnosis and evaluation. Due, in part, to recent technological advancements, metabolomics and lipid biomarkers have become a promising area in cancer research. Therefore, there is a necessity for novel and complementary techniques to identify and visualize these molecular markers within tumors and surrounding tissue. RECENT FINDINGS Since its introduction, mass spectrometry imaging (MSI) has proven to be a powerful tool for mapping analytes in biological tissues. By adding the label-free specificity of mass spectrometry to the detailed spatial information of traditional histology, hundreds of lipids can be imaged simultaneously within a tumor. MSI provides highly detailed lipid maps for comparing intra-tumor, tumor margin, and healthy regions to identify biomarkers, patterns of disease, and potential therapeutic targets. In this manuscript, recent advancement in sample preparation and MSI technologies are discussed with special emphasis on cancer lipid research to identify tumor biomarkers. CONCLUSION MSI offers a unique approach for biomolecular characterization of tumor tissues and provides valuable complementary information to histology for lipid biomarker discovery and tumor classification in clinical and research cancer applications.
Collapse
Affiliation(s)
- Matthias Holzlechner
- Department of Neuroscience, Cell Biology, and AnatomyThe University of Texas Medical Branch at Galveston (UTMB)GalvestonTexas
| | - Eliseo Eugenin
- Department of Neuroscience, Cell Biology, and AnatomyThe University of Texas Medical Branch at Galveston (UTMB)GalvestonTexas
| | - Brendan Prideaux
- Department of Neuroscience, Cell Biology, and AnatomyThe University of Texas Medical Branch at Galveston (UTMB)GalvestonTexas
| |
Collapse
|