1
|
Dong M, Yang W, Hao J, Jia X, Yang O, Lo MKF, Cao B, Hu S, Lin Y. Cross-Scale Multimodal Imaging for Organic Matter in Extraterrestrial Samples. Anal Chem 2025; 97:8258-8267. [PMID: 40102193 PMCID: PMC12019776 DOI: 10.1021/acs.analchem.4c05804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/07/2025] [Accepted: 03/13/2025] [Indexed: 03/20/2025]
Abstract
The analysis of extraterrestrial organic matter in samples returned by space missions provides a unique opportunity to study prebiotic chemistry. A comprehensive understanding of the occurrence and composition of organic matter is fundamental to unraveling its origin and evolutionary history. However, the scarcity and complexity of these materials pose considerable analytical challenges. Here, we developed a cross-scale multimodal imaging workflow that integrated mass spectrometry imaging (MSI) and vibrational spectroscopy imaging, including desorption electrospray ionization coupled quadrupole-time-of-flight mass spectrometry (DESI-Q-TOF/MS), time-of-flight secondary ion mass spectrometry (TOF-SIMS), nanoscale secondary ion mass spectrometry (NanoSIMS), focal plane array-Fourier transform infrared spectroscopy (FPA-FTIR), and optical photothermal infrared spectroscopy (O-PTIR). This workflow was applied to the Murchison meteorite, with the objective of establishing spatial associations between mineral phases, molecular composition, functional groups, and isotopic composition on a scale from the millimeter to the submicron. The spatial resolution of DESI has been improved from 100 to 200 to 20 μm, enabling spatial correlation with other imaging techniques. For the first time, the enrichment of organic matter─including CHN, CHO, and CHNO compounds and polycyclic aromatic hydrocarbons (PAHs)─in fine-grained rims (FGRs) surrounding silicate chondrules has been observed. Furthermore, the cross-scale multimodal imaging also reveals differences in organic matter composition between Ca-carbonate and phyllosilicates, as well as spatial heterogeneity within the latter. This workflow provides a new paradigm for studying the complex occurrence and composition of organic matter in various research fields, enhancing our understanding of prebiotic materials in the solar system.
Collapse
Affiliation(s)
- Mingtan Dong
- Key
Laboratory of Earth and Planetary Physics, Institute of Geology and
Geophysics, Chinese Academy of Sciences, Beijing 100029, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Yang
- Key
Laboratory of Earth and Planetary Physics, Institute of Geology and
Geophysics, Chinese Academy of Sciences, Beijing 100029, China
| | - Jialong Hao
- Key
Laboratory of Earth and Planetary Physics, Institute of Geology and
Geophysics, Chinese Academy of Sciences, Beijing 100029, China
| | | | - Ou Yang
- ULVAC-PHI
Instrument Co. Ltd., Nanjing 211102, China
| | - Michael K. F. Lo
- Photothermal
Spectroscopy Corporation, 325 Chapala Street, Santa
Barbara, California 93101, United States
| | - Bobo Cao
- Department
of Chemistry, Tsinghua University, Beijing 100084, China
| | - Sen Hu
- Key
Laboratory of Earth and Planetary Physics, Institute of Geology and
Geophysics, Chinese Academy of Sciences, Beijing 100029, China
| | - Yangting Lin
- Key
Laboratory of Earth and Planetary Physics, Institute of Geology and
Geophysics, Chinese Academy of Sciences, Beijing 100029, China
| |
Collapse
|
2
|
Fields L, Miles HN, Adrian AE, Patrenets E, Ricke WA, Li L. MSIght: A Modular Platform for Improved Confidence in Global, Untargeted Mass Spectrometry Imaging Annotation. J Proteome Res 2025. [PMID: 40197022 DOI: 10.1021/acs.jproteome.4c01140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Mass spectrometry imaging (MSI) has gained popularity in clinical analyses due to its high sensitivity, specificity, and throughput. However, global profiling experiments are often still restricted to LC-MS/MS analyses that lack spatial localization due to low-throughput methods for on-tissue peptide identification and confirmation. Additionally, the integration of parallel LC-MS/MS peptide confirmation, as well as histological stains for accurate mapping of identifications, presents a large bottleneck for data analysis, limiting throughput for untargeted profiling experiments. Here, we present a novel platform, termed MSIght, which automates the integration of these multiple modalities into an accessible and modular platform. Histological stains of tissue sections are coregistered to their respective MSI data sets to improve spatial localization and resolution of identified peptides. MS/MS peptide identifications via untargeted LC-MS/MS are used to confirm putative MSI identifications, thus generating MS images with greater confidence in a high-throughput, global manner. This platform has the potential to enable large-scale clinical cohorts to utilize MSI in the future for global proteomic profiling that uncovers novel biomarkers in a spatially resolved manner, thus widely expanding the utility of MSI in clinical discovery.
Collapse
Affiliation(s)
- Lauren Fields
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Hannah N Miles
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
- Department of Urology, George M. O'Brien Center of Research Excellence, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Alexis E Adrian
- Department of Urology, George M. O'Brien Center of Research Excellence, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
- School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Elliot Patrenets
- Department of Urology, George M. O'Brien Center of Research Excellence, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
- Department of Integrative Biology, University of Wisconsin-Madison, 250 N Mills St, Madison, Wisconsin 53706, United States
| | - William A Ricke
- Department of Urology, George M. O'Brien Center of Research Excellence, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
- School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
3
|
Gorman BL, Shafer CC, Ragi N, Sharma K, Neumann EK, Anderton CR. Imaging and spatially resolved mass spectrometry applications in nephrology. Nat Rev Nephrol 2025:10.1038/s41581-025-00946-1. [PMID: 40148534 DOI: 10.1038/s41581-025-00946-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
The application of spatially resolved mass spectrometry (MS) and MS imaging approaches for studying biomolecular processes in the kidney is rapidly growing. These powerful methods, which enable label-free and multiplexed detection of many molecular classes across omics domains (including metabolites, drugs, proteins and protein post-translational modifications), are beginning to reveal new molecular insights related to kidney health and disease. The complexity of the kidney often necessitates multiple scales of analysis for interrogating biofluids, whole organs, functional tissue units, single cells and subcellular compartments. Various MS methods can generate omics data across these spatial domains and facilitate both basic science and pathological assessment of the kidney. Optimal processes related to sample preparation and handling for different MS applications are rapidly evolving. Emerging technology and methods, improvement of spatial resolution, broader molecular characterization, multimodal and multiomics approaches and the use of machine learning and artificial intelligence approaches promise to make these applications even more valuable in the field of nephology. Overall, spatially resolved MS and MS imaging methods have the potential to fill much of the omics gap in systems biology analysis of the kidney and provide functional outputs that cannot be obtained using genomics and transcriptomic methods.
Collapse
Affiliation(s)
- Brittney L Gorman
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Catelynn C Shafer
- Department of Chemistry, University of California, Davis, Davis, CA, 95695, USA
| | - Nagarjunachary Ragi
- Center for Precision Medicine, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Kumar Sharma
- Center for Precision Medicine, The University of Texas Health San Antonio, San Antonio, TX, USA
- Division of Nephrology, Department of Medicine, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Elizabeth K Neumann
- Department of Chemistry, University of California, Davis, Davis, CA, 95695, USA
| | - Christopher R Anderton
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
4
|
Tafintseva V, Nippolainen E, Virtanen V, Solheim JH, Zimmermann B, Saarakkala S, Kröger H, Kohler A, Töyräs J, Afara IO, Shaikh R. Machine Learning Approaches for the Fusion of Near-Infrared, Mid-Infrared, and Raman Data to Identify Cartilage Degradation in Human Osteochondral Plugs. APPLIED SPECTROSCOPY 2025; 79:385-395. [PMID: 39512222 DOI: 10.1177/00037028241285583] [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: 11/15/2024]
Abstract
Vibrational spectroscopy methods such as mid-infrared (MIR), near-infrared (NIR), and Raman spectroscopies have been shown to have great potential for in vivo biomedical applications, such as arthroscopic evaluation of joint injuries and degeneration. Considering that these techniques provide complementary chemical information, in this study, we hypothesized that combining the MIR, NIR, and Raman data from human osteochondral samples can improve the detection of cartilage degradation. This study evaluated 272 osteochondral samples from 18 human knee joins, comprising both healthy and damaged tissue according to the reference Osteoarthritis Research Society International grading system. We established the one-block and multi-block classification models using partial least squares discriminant analysis (PLSDA), random forest, and support vector machine (SVM) algorithms. Feature modeling by principal component analysis was tested for the SVM (PCA-SVM) models. The best one-block models were built using MIR and Raman data, discriminating healthy cartilage from damaged with an accuracy of 77.5% for MIR and 77.8% for Raman using the PCA-SVM algorithm, whereas the NIR data did not perform as well achieving only 68.5% accuracy for the best model using PCA-SVM. The multi-block approach allowed an improvement with an accuracy of 81.4% for the best model by PCA-SVM. Fusing three blocks using MIR, NIR, and Raman by multi-block PLSDA significantly improved the performance of the single-block models to 79.1% correct classification. The significance was proven by statistical testing using analysis of variance. Thus, the study suggests the potential and the complementary value of the fusion of different spectroscopic techniques and provides valuable data analysis tools for the diagnostics of cartilage health.
Collapse
Affiliation(s)
- Valeria Tafintseva
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Ervin Nippolainen
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Department of Orthopedics, Traumatology and Hand Surgery, Kuopio University Hospital, Kuopio, Finland
| | - Vesa Virtanen
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Johanne Heitmann Solheim
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
- The Norwegian Metrology Service (Justervesenet), Kjeller, Norway
| | - Boris Zimmermann
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Simo Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Heikki Kröger
- Department of Orthopedics, Traumatology and Hand Surgery, Kuopio University Hospital, Kuopio, Finland
- Kuopio Musculoskeletal Research Unit, University of Eastern Finland, Kuopio, Finland
| | - Achim Kohler
- Faculty of Science and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Juha Töyräs
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Science Service Center, Kuopio University Hospital, Kuopio, Finland
- School of Electrical Engineering and Computer Science, The University of Queensland, Brisbane, Australia
| | - Isaac O Afara
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Department of Orthopedics, Traumatology and Hand Surgery, Kuopio University Hospital, Kuopio, Finland
| | - Rubina Shaikh
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- Department of Orthopedics, Traumatology and Hand Surgery, Kuopio University Hospital, Kuopio, Finland
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Lopes T, Cavaco R, Capela D, Dias F, Teixeira J, Monteiro CS, Lima A, Guimarães D, Jorge PAS, Silva NA. Improving LIBS-based mineral identification with Raman imaging and spectral knowledge distillation. Talanta 2025; 283:127110. [PMID: 39520923 DOI: 10.1016/j.talanta.2024.127110] [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: 08/02/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Combining data from different sensing modalities has been a promising research topic for building better and more reliable data-driven models. In particular, it is known that multimodal spectral imaging can improve the analytical capabilities of standalone spectroscopy techniques through fusion, hyphenation, or knowledge distillation techniques. In this manuscript, we focus on the latter, exploring how one can increase the performance of a Laser-induced Breakdown Spectroscopy system for mineral classification problems using additional spectral imaging techniques. Specifically, focusing on a scenario where Raman spectroscopy delivers accurate mineral classification performance, we show how to deploy a knowledge distillation pipeline where Raman spectroscopy may act as an autonomous supervisor for LIBS. For a case study concerning a challenging Li-bearing mineral identification of spodumene and petalite, our results demonstrate the advantages of this method in improving the performance of a single-technique system. LIBS trained with labels obtained by Raman presents an enhanced classification performance. Furthermore, leveraging the interpretability of the model deployed, the workflow opens opportunities for the deployment of assisted feature discovery pipelines, which may impact future academic and industrial applications.
Collapse
Affiliation(s)
- Tomás Lopes
- Center for Applied Photonics, INESC TEC, Rua do Campo Alegre 687, Porto, 4169-007, Portugal; Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre 687, Porto, 4169-007, Portugal.
| | - Rafael Cavaco
- Center for Applied Photonics, INESC TEC, Rua do Campo Alegre 687, Porto, 4169-007, Portugal; Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre 687, Porto, 4169-007, Portugal
| | - Diana Capela
- Center for Applied Photonics, INESC TEC, Rua do Campo Alegre 687, Porto, 4169-007, Portugal; Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre 687, Porto, 4169-007, Portugal
| | - Filipa Dias
- Departamento de Geociências, Ambiente e Ordenamento do Território, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre 687, Porto, 4169-007, Portugal
| | - Joana Teixeira
- Center for Applied Photonics, INESC TEC, Rua do Campo Alegre 687, Porto, 4169-007, Portugal; Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre 687, Porto, 4169-007, Portugal
| | - Catarina S Monteiro
- Center for Applied Photonics, INESC TEC, Rua do Campo Alegre 687, Porto, 4169-007, Portugal
| | - Alexandre Lima
- Departamento de Geociências, Ambiente e Ordenamento do Território, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre 687, Porto, 4169-007, Portugal
| | - Diana Guimarães
- Center for Applied Photonics, INESC TEC, Rua do Campo Alegre 687, Porto, 4169-007, Portugal
| | - Pedro A S Jorge
- Center for Applied Photonics, INESC TEC, Rua do Campo Alegre 687, Porto, 4169-007, Portugal; Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre 687, Porto, 4169-007, Portugal
| | - Nuno A Silva
- Center for Applied Photonics, INESC TEC, Rua do Campo Alegre 687, Porto, 4169-007, Portugal; Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre 687, Porto, 4169-007, Portugal
| |
Collapse
|
6
|
Maimó-Barceló A, Pérez-Romero K, Rodríguez RM, Huergo C, Calvo I, Fernández JA, Barceló-Coblijn G. To image or not to image: Use of imaging mass spectrometry in biomedical lipidomics. Prog Lipid Res 2025; 97:101319. [PMID: 39765282 DOI: 10.1016/j.plipres.2025.101319] [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: 04/21/2024] [Revised: 11/19/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Lipid imaging mass spectrometry (LIMS) allows for establishing the bidimensional distribution of lipid species within a tissue section. One of the main advantages is the generation of spatial information on lipid species distribution at a spatial (lateral) resolution bordering on single-cell resolution with no need to isolate cells. Thus, LIMS images demonstrate, with a level of detail never described before, that lipid profiles are highly sensitive to cell type and pathophysiological state. The wealth and relevance of the information conveyed by LIMS makes up for the lack of a separation stage before sample injection into the mass analyzer, which can somehow be circumvented by other means. Hence, the possibility of describing the lipidome at the cellular level while preserving the microenvironment offers an incomparable opportunity to investigate physiological and pathological contexts. However, to fully grasp the biological implications of the lipid profiles, it is essential to contextualize LIMS data within the broader multiscale 'omic' landscape, entailing genomics, epigenomics, and proteomics, each offering a unique window into the regulatory layers of the cell. In this line, the number of techniques that can be combined with LIMS to delve into the molecular mechanisms underlying differential lipid profiles is continuously increasing. Herein, we aim to describe the key features of LIMS analyses, from sample preparation to data interpretation, as well as the current methodologies to enrich and complete the final outcome. While the field is rapidly advancing, we consider there is solid evidence to foresee the incorporation of LIMS into clinical environments.
Collapse
Affiliation(s)
- Albert Maimó-Barceló
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa) - Health Research Institute of the Balearic Islands, Ctra. Valldemossa 79, Section G, Floor -1, E-07120 Palma, Balearic Islands, Spain; Research Unit, University Hospital Son Espases, Ctra Valldemossa 79, E-07120 Palma, Balearic Islands, Spain
| | - Karim Pérez-Romero
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa) - Health Research Institute of the Balearic Islands, Ctra. Valldemossa 79, Section G, Floor -1, E-07120 Palma, Balearic Islands, Spain; Research Unit, University Hospital Son Espases, Ctra Valldemossa 79, E-07120 Palma, Balearic Islands, Spain
| | - Ramón M Rodríguez
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa) - Health Research Institute of the Balearic Islands, Ctra. Valldemossa 79, Section G, Floor -1, E-07120 Palma, Balearic Islands, Spain; Research Unit, University Hospital Son Espases, Ctra Valldemossa 79, E-07120 Palma, Balearic Islands, Spain
| | - Cristina Huergo
- Department of Physical Chemistry, Fac. of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Ibai Calvo
- Department of Physical Chemistry, Fac. of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - José A Fernández
- Department of Physical Chemistry, Fac. of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain.
| | - Gwendolyn Barceló-Coblijn
- Lipids in Human Pathology, Institut d'Investigació Sanitària Illes Balears (IdISBa) - Health Research Institute of the Balearic Islands, Ctra. Valldemossa 79, Section G, Floor -1, E-07120 Palma, Balearic Islands, Spain; Research Unit, University Hospital Son Espases, Ctra Valldemossa 79, E-07120 Palma, Balearic Islands, Spain.
| |
Collapse
|
7
|
van der Vloet L, Hilaire PBS, Bouillod C, Isin EM, Heeren RMA, Vandenbosch M. How can MSI enhance our understanding of ASO distribution? Drug Discov Today 2025; 30:104275. [PMID: 39701373 DOI: 10.1016/j.drudis.2024.104275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 12/05/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
In the dynamic field of drug discovery and development, a comprehensive understanding of drug absorption, distribution, metabolism, excretion, and toxicity is crucial. Mass spectrometry imaging (MSI) has become a key analytical tool in the pharmaceutical industry, allowing evaluation of drug biodistribution and molecular profiles. Antisense oligonucleotides (ASOs) are emerging drug candidates for treating neurologic diseases. This review explores the potential of MSI in investigating ASOs' spatial distribution within neurological disease models. Here, we focus on multimodal molecular imaging to gain insights into ASO distribution, simultaneously with a better understanding of the molecular pathways affected by ASOs. An improved understanding of therapeutic ASOs in tissue will potentially improve neurologic therapies, emphasizing their importance in patient care.
Collapse
Affiliation(s)
- Laura van der Vloet
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | | | - Christophe Bouillod
- Institut de Recherche et Développement Servier Paris-Saclay, Rue Francis Perrin, 91190 Gif-sur-Yvette, France
| | - Emre M Isin
- Institut de Recherche et Développement Servier Paris-Saclay, Rue Francis Perrin, 91190 Gif-sur-Yvette, France
| | - Ron M A Heeren
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.
| | - Michiel Vandenbosch
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
8
|
Seydoux C, Ezzedine JA, Larbi GS, Ravanel S, Maréchal E, Barnes JP, Jouneau PH. Subcellular ToF-SIMS Imaging of the Snow Alga Sanguina nivaloides by Combining High Mass and High Lateral Resolution Acquisitions. Anal Chem 2024; 96:19917-19925. [PMID: 39642022 DOI: 10.1021/acs.analchem.4c03826] [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: 12/08/2024]
Abstract
Time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging has demonstrated great potential for metabolic imaging; however, achieving sufficiently high lateral and mass resolution to reach the organelle scale remains challenging. To address this, we have developed an approach that combines imaging acquisitions close to the highest lateral resolution (<150 nm) and mass resolution (9,000) reachable by ToF-SIMS. The data were then merged and processed using multivariate analysis (MVA), providing the identification and annotation of 85% of the main contributors to the multivariate analysis components at high lateral resolution. Insights into the electron microscopy sample preparation were provided, especially as we revealed that at least three different osmium-containing complexes can be found depending on the specific chemical environment of organelles. In cells of the snow alga Sanguina nivaloides, living in a natural environment limited in nutrients such as phosphorus (P), we mapped elements and molecules within their subcellular context, allowing for the molecular fingerprinting of organelles at a resolution of ∼150 nm, as confirmed by correlative electron microscopy. It was thus possible to highlight that S. nivaloides likely absorbed selectively some inorganic P forms provided by P-rich dust deposited on the snow surface. S. nivaloides cells could maintain phosphorylations in the stroma of the chloroplast, consistently with the preservation of photosynthetic activity. The presented method can thus overcome the current limitations of ToF-SIMS for subcellular imaging and contributes to the understanding of key questions, such as P homeostasis and other cell physiological processes.
Collapse
Affiliation(s)
- Claire Seydoux
- CEA, IRIG-Laboratoire Modélisation et Exploration des Matériaux, Université Grenoble Alpes, Grenoble 38000, France
| | - Jade A Ezzedine
- CEA, CNRS, INRAE, IRIG-Laboratoire de Physiologie Cellulaire et Végétale, Université Grenoble Alpes, Grenoble 38000, France
| | - Grégory Si Larbi
- CEA, CNRS, INRAE, IRIG-Laboratoire de Physiologie Cellulaire et Végétale, Université Grenoble Alpes, Grenoble 38000, France
| | - Stéphane Ravanel
- CEA, CNRS, INRAE, IRIG-Laboratoire de Physiologie Cellulaire et Végétale, Université Grenoble Alpes, Grenoble 38000, France
| | - Eric Maréchal
- CEA, CNRS, INRAE, IRIG-Laboratoire de Physiologie Cellulaire et Végétale, Université Grenoble Alpes, Grenoble 38000, France
| | | | - Pierre-Henri Jouneau
- CEA, IRIG-Laboratoire Modélisation et Exploration des Matériaux, Université Grenoble Alpes, Grenoble 38000, France
| |
Collapse
|
9
|
Shamraeva M, Visvikis T, Zoidis S, Anthony IGM, Van Nuffel S. The Application of a Random Forest Classifier to ToF-SIMS Imaging Data. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:2801-2814. [PMID: 39455427 PMCID: PMC11622239 DOI: 10.1021/jasms.4c00324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging is a potent analytical tool that provides spatially resolved chemical information on surfaces at the microscale. However, the hyperspectral nature of ToF-SIMS datasets can be challenging to analyze and interpret. Both supervised and unsupervised machine learning (ML) approaches are increasingly useful to help analyze ToF-SIMS data. Random Forest (RF) has emerged as a robust and powerful algorithm for processing mass spectrometry data. This machine learning approach offers several advantages, including accommodating nonlinear relationships, robustness to outliers in the data, managing the high-dimensional feature space, and mitigating the risk of overfitting. The application of RF to ToF-SIMS imaging facilitates the classification of complex chemical compositions and the identification of features contributing to these classifications. This tutorial aims to assist nonexperts in either machine learning or ToF-SIMS to apply Random Forest to complex ToF-SIMS datasets.
Collapse
Affiliation(s)
- Mariya
A. Shamraeva
- Maastricht
MultiModal Molecular Imaging Institute (M4i), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Theodoros Visvikis
- Faculty
of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan 1, Maastricht 6229EN, The Netherlands
| | - Stefanos Zoidis
- Faculty
of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan 1, Maastricht 6229EN, The Netherlands
| | - Ian G. M. Anthony
- Maastricht
MultiModal Molecular Imaging Institute (M4i), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Sebastiaan Van Nuffel
- Maastricht
MultiModal Molecular Imaging Institute (M4i), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Faculty
of Science and Engineering, Maastricht University, Paul-Henri Spaaklaan 1, Maastricht 6229EN, The Netherlands
| |
Collapse
|
10
|
Dannhorn A, Kazanc E, Flint L, Guo F, Carter A, Hall AR, Jones SA, Poulogiannis G, Barry ST, Sansom OJ, Bunch J, Takats Z, Goodwin RJA. Morphological and molecular preservation through universal preparation of fresh-frozen tissue samples for multimodal imaging workflows. Nat Protoc 2024; 19:2685-2711. [PMID: 38806741 DOI: 10.1038/s41596-024-00987-z] [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: 07/21/2023] [Accepted: 02/14/2024] [Indexed: 05/30/2024]
Abstract
The landscape of tissue-based imaging modalities is constantly and rapidly evolving. While formalin-fixed, paraffin-embedded material is still useful for histological imaging, the fixation process irreversibly changes the molecular composition of the sample. Therefore, many imaging approaches require fresh-frozen material to get meaningful results. This is particularly true for molecular imaging techniques such as mass spectrometry imaging, which are widely used to probe the spatial arrangement of the tissue metabolome. As high-quality fresh-frozen tissues are limited in their availability, any sample preparation workflow they are subjected to needs to ensure morphological and molecular preservation of the tissues and be compatible with as many of the established and emerging imaging techniques as possible to obtain the maximum possible insights from the tissues. Here we describe a universal sample preparation workflow, from the initial step of freezing the tissues to the cold embedding in a new hydroxypropyl methylcellulose/polyvinylpyrrolidone-enriched hydrogel and the generation of thin tissue sections for analysis. Moreover, we highlight the optimized storage conditions that limit molecular and morphological degradation of the sections. The protocol is compatible with human and plant tissues and can be easily adapted for the preparation of alternative sample formats (e.g., three-dimensional cell cultures). The integrated workflow is universally compatible with histological tissue analysis, mass spectrometry imaging and imaging mass cytometry, as well as spatial proteomic, genomic and transcriptomic tissue analysis. The protocol can be completed within 4 h and requires minimal prior experience in the preparation of tissue samples for multimodal imaging experiments.
Collapse
Affiliation(s)
- Andreas Dannhorn
- Imaging and Data analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Department of Digestion, Metabolism and Reproduction, Sir Alexander Fleming Building, Imperial College London, London, UK
| | - Emine Kazanc
- Department of Digestion, Metabolism and Reproduction, Sir Alexander Fleming Building, Imperial College London, London, UK
| | - Lucy Flint
- Imaging and Data analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Fei Guo
- Imaging and Data analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Safety Innovations, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Alfie Carter
- Imaging and Data analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Safety Innovations, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Andrew R Hall
- Safety Innovations, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Stewart A Jones
- Imaging and Data analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | | | - Simon T Barry
- Bioscience, Discovery, Oncology R&D, AstraZeneca, Cambridge, UK
| | | | - Josephine Bunch
- National Centre of Excellence in Mass Spectrometry Imaging (NiCE-MSI), National Physical Laboratory, Teddington, UK
| | - Zoltan Takats
- Department of Digestion, Metabolism and Reproduction, Sir Alexander Fleming Building, Imperial College London, London, UK
| | - Richard J A Goodwin
- Imaging and Data analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
11
|
Zhan L, Huang Y, Wang G. Multi-modal mass spectrometry imaging of a single tissue section. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5074. [PMID: 39017393 DOI: 10.1002/jms.5074] [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: 02/08/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024]
Abstract
Mass spectrometry imaging (MSI) was developed to visualize spatial chemical information within tissues, thereby facilitating spatial multi-omic analysis. However, due to the limited spatial information provided by individual modal MSI, correlating various chemical data within tissues remains a significant challenge. In recent years, multimodal MSI has garnered considerable attention due to its ability to visualize the spatial distributions of multiple biomolecules within tissues. Among the strategies employed in this field, multimodal imaging on a single tissue section circumvents multiple issues introduced by integration of images of consecutive tissue sections. In this minireview, we provide an overview of multimodal MSI on a single tissue section, with a particular focus on the use of Matrix-Assisted Laser Desorption/Ionization-MSI for spatial multi-omic investigations that offer a comprehensive and in-depth elucidation of the biological state and activities, aiming to inspire the development of new approaches in this field.
Collapse
Affiliation(s)
- Lingpeng Zhan
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yanyi Huang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Guanbo Wang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| |
Collapse
|
12
|
Rahman MF, Islam A, Islam MM, Mamun MA, Xu L, Sakamoto T, Sato T, Takahashi Y, Kahyo T, Aoyagi S, Kaibuchi K, Setou M. Mass Spectrometry Imaging Combined with Sparse Autoencoder Method Reveals Altered Phosphorylcholine Distribution in Imipramine Treated Wild-Type Mice Brains. Int J Mol Sci 2024; 25:7969. [PMID: 39063212 PMCID: PMC11276679 DOI: 10.3390/ijms25147969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Mass spectrometry imaging (MSI) is essential for visualizing drug distribution, metabolites, and significant biomolecules in pharmacokinetic studies. This study mainly focuses on imipramine, a tricyclic antidepressant that affects endogenous metabolite concentrations. The aim was to use atmospheric pressure matrix-assisted laser desorption/ionization (AP-MALDI)-MSI combined with different dimensionality reduction methods to examine the distribution and impact of imipramine on endogenous metabolites in the brains of treated wild-type mice. Brain sections from both control and imipramine-treated mice underwent AP-MALDI-MSI. Dimensionality reduction methods, including principal component analysis, multivariate curve resolution, and sparse autoencoder (SAE), were employed to extract valuable information from the MSI data. Only the SAE method identified phosphorylcholine (ChoP) as a potential marker distinguishing between the control and treated mice brains. Additionally, a significant decrease in ChoP accumulation was observed in the cerebellum, hypothalamus, thalamus, midbrain, caudate putamen, and striatum ventral regions of the treated mice brains. The application of dimensionality reduction methods, particularly the SAE method, to the AP-MALDI-MSI data is a novel approach for peak selection in AP-MALDI-MSI data analysis. This study revealed a significant decrease in ChoP in imipramine-treated mice brains.
Collapse
Affiliation(s)
- Md Foyzur Rahman
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Ariful Islam
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Md. Monirul Islam
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Md. Al Mamun
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- Preppers Co., Ltd., 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Lili Xu
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Takumi Sakamoto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- Preppers Co., Ltd., 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Tomohito Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Yutaka Takahashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- Preppers Co., Ltd., 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- Quantum Imaging Laboratory, Division of Research and Development in Photonics Technology/International Mass Imaging and Spatial Omics Center, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Satoka Aoyagi
- Faculty of Science and Technology, Seikei University, 3-3-1 Kichijoji-kitamachi, Musashino-shi 180-8633, Tokyo, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- International Mass Imaging and Spatial Omics Center, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| |
Collapse
|
13
|
Lopes T, Capela D, Guimarães D, Ferreira MFS, Jorge PAS, Silva NA. From sensor fusion to knowledge distillation in collaborative LIBS and hyperspectral imaging for mineral identification. Sci Rep 2024; 14:9123. [PMID: 38643168 PMCID: PMC11032373 DOI: 10.1038/s41598-024-59553-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/12/2024] [Indexed: 04/22/2024] Open
Abstract
Multimodal spectral imaging offers a unique approach to the enhancement of the analytical capabilities of standalone spectroscopy techniques by combining information gathered from distinct sources. In this manuscript, we explore such opportunities by focusing on two well-known spectral imaging techniques, namely laser-induced breakdown spectroscopy, and hyperspectral imaging, and explore the opportunities of collaborative sensing for a case study involving mineral identification. In specific, the work builds upon two distinct approaches: a traditional sensor fusion, where we strive to increase the information gathered by including information from the two modalities; and a knowledge distillation approach, where the Laser Induced Breakdown spectroscopy is used as an autonomous supervisor for hyperspectral imaging. Our results show the potential of both approaches in enhancing the performance over a single modality sensing system, highlighting, in particular, the advantages of the knowledge distillation framework in maximizing the potential benefits of using multiple techniques to build more interpretable models and paving for industrial applications.
Collapse
Affiliation(s)
- Tomás Lopes
- INESC TEC, Center for Applied Photonics, 4169-007, Porto, Portugal
- Departamento de Física, Faculdade de Ciências da Universidade do Porto, 4169-007, Porto, Portugal
| | - Diana Capela
- INESC TEC, Center for Applied Photonics, 4169-007, Porto, Portugal
- Departamento de Física, Faculdade de Ciências da Universidade do Porto, 4169-007, Porto, Portugal
| | - Diana Guimarães
- INESC TEC, Center for Applied Photonics, 4169-007, Porto, Portugal
| | - Miguel F S Ferreira
- INESC TEC, Center for Applied Photonics, 4169-007, Porto, Portugal
- Departamento de Física, Faculdade de Ciências da Universidade do Porto, 4169-007, Porto, Portugal
| | - Pedro A S Jorge
- INESC TEC, Center for Applied Photonics, 4169-007, Porto, Portugal
- Departamento de Física, Faculdade de Ciências da Universidade do Porto, 4169-007, Porto, Portugal
| | - Nuno A Silva
- INESC TEC, Center for Applied Photonics, 4169-007, Porto, Portugal.
| |
Collapse
|
14
|
Klein D, Rivera ES, Caprioli RM, Spraggins JM. Imaging Mass Spectrometry of Isotopically Resolved Intact Proteins on a Trapped Ion-Mobility Quadrupole Time-of-Flight Mass Spectrometer. Anal Chem 2024; 96:5065-5070. [PMID: 38517028 PMCID: PMC10993197 DOI: 10.1021/acs.analchem.3c05252] [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: 11/20/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
In this work, we demonstrate rapid, high spatial, and high spectral resolution imaging of intact proteins by matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) on a hybrid quadrupole-reflectron time-of-flight (qTOF) mass spectrometer equipped with trapped ion mobility spectrometry (TIMS). Historically, untargeted MALDI IMS of proteins has been performed on TOF mass spectrometers. While advances in TOF instrumentation have enabled rapid, high spatial resolution IMS of intact proteins, TOF mass spectrometers generate relatively low-resolution mass spectra with limited mass accuracy. Conversely, the implementation of MALDI sources on high-resolving power Fourier transform (FT) mass spectrometers has allowed IMS experiments to be conducted with high spectral resolution with the caveat of increasingly long data acquisition times. As illustrated here, qTOF mass spectrometers enable protein imaging with the combined advantages of TOF and FT mass spectrometers. Protein isotope distributions were resolved for both a protein standard mixture and proteins detected from a whole-body mouse pup tissue section. Rapid (∼10 pixels/s) 10 μm lateral spatial resolution IMS was performed on a rat brain tissue section while maintaining isotopic spectral resolution. Lastly, proof-of-concept MALDI-TIMS data was acquired from a protein mixture to demonstrate the ability to differentiate charge states by ion mobility. These experiments highlight the advantages of qTOF and timsTOF platforms for resolving and interpreting complex protein spectra generated from tissue by IMS.
Collapse
Affiliation(s)
- Dustin
R. Klein
- Mass
Spectrometry Research Center, Vanderbilt
University, Nashville, Tennessee 37235, United States
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Emilio S. Rivera
- Mass
Spectrometry Research Center, Vanderbilt
University, Nashville, Tennessee 37235, United States
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Richard M. Caprioli
- Mass
Spectrometry Research Center, Vanderbilt
University, Nashville, Tennessee 37235, United States
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department
of Medicine, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jeffrey M. Spraggins
- Mass
Spectrometry Research Center, Vanderbilt
University, Nashville, Tennessee 37235, United States
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department
of Cell and Developmental Biology, Vanderbilt
University, Nashville, Tennessee 37235, United States
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37235, United States
| |
Collapse
|
15
|
Croslow SW, Trinklein TJ, Sweedler JV. Advances in multimodal mass spectrometry for single-cell analysis and imaging enhancement. FEBS Lett 2024; 598:591-601. [PMID: 38243373 PMCID: PMC10963143 DOI: 10.1002/1873-3468.14798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 01/21/2024]
Abstract
Multimodal mass spectrometry (MMS) incorporates an imaging modality with probe-based mass spectrometry (MS) to enable precise, targeted data acquisition and provide additional biological and chemical data not available by MS alone. Two categories of MMS are covered; in the first, an imaging modality guides the MS probe to target individual cells and to reduce acquisition time by automatically defining regions of interest. In the second category, imaging and MS data are coupled in the data analysis pipeline to increase the effective spatial resolution using a higher resolution imaging method, correct for tissue deformation, and incorporate fine morphological features in an MS imaging dataset. Recent methodological and computational developments are covered along with their application to single-cell and imaging analyses.
Collapse
Affiliation(s)
- Seth W. Croslow
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Timothy J. Trinklein
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jonathan V. Sweedler
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
16
|
Gorman BL, Torti SV, Torti FM, Anderton CR. Mass spectrometry imaging of metals in tissues and cells: Methods and biological applications. Biochim Biophys Acta Gen Subj 2024; 1868:130329. [PMID: 36791830 PMCID: PMC10423302 DOI: 10.1016/j.bbagen.2023.130329] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Metals are pervasive throughout biological processes, where they play essential structural and catalytic roles. Metals can also exhibit deleterious effects on human health. Powerful analytical techniques, such as mass spectrometry imaging (MSI), are required to map metals due to their low concentrations within biological tissue. SCOPE OF REVIEW This Mini Review focuses on key MSI technology that can image metal distributions in situ, describing considerations for each technique (e.g., resolution, sensitivity, etc.). We highlight recent work using MSI for mapping trace metals in tissues, detecting metal-based drugs, and simultaneously imaging metals and biomolecules. MAJOR CONCLUSIONS MSI has enabled significant advances in locating bioactive metals at high spatial resolution and correlating their distributions with that of biomolecules. The use of metal-based immunochemistry has enabled simultaneous high-throughput protein and biomolecule imaging. GENERAL SIGNIFICANCE The techniques and examples described herein can be applied to many biological questions concerning the important biological roles of metals, metal toxicity, and localization of metal-based drugs.
Collapse
Affiliation(s)
- Brittney L Gorman
- Environmental Molecular Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, United States of America
| | - Suzy V Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Frank M Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Christopher R Anderton
- Environmental Molecular Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, United States of America.
| |
Collapse
|
17
|
Levasseur M, Nicol E, Elie N, Houël E, Eparvier V, Touboul D. Spatialized Metabolomic Annotation Combining MALDI Imaging and Molecular Networks. Anal Chem 2024; 96:18-22. [PMID: 38134413 DOI: 10.1021/acs.analchem.3c03482] [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: 12/24/2023]
Abstract
MALDI mass spectrometry imaging has gained major interest in the field of chemical imaging. This technique makes it possible to locate tens to hundreds of ionic signals on the sample surface without any a priori. One of the current challenges is still the limited ability to annotate signals in order to convert m/z values into probable chemical structures. At the same time, data obtained by LC-MS/MS have benefited from the development of numerous chemoinformatics tools, in particular molecular networks, for their efficient annotation. For the first time, we present here the combination of MALDI-FT-ICR imaging with molecular networks from MALDI-MS/MS data directly acquired on plant tissue sections. Annotation improvements are demonstrated, paving the way for new annotation pipelines for MALDI imaging.
Collapse
Affiliation(s)
- Marceau Levasseur
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198 Gif-sur-Yvette, France
| | - Edith Nicol
- Laboratoire de Chimie Moléculaire (LCM), CNRS, École Polytechnique, Institut Polytechnique de Paris, 91120 Palaiseau, France
| | - Nicolas Elie
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198 Gif-sur-Yvette, France
| | - Emeline Houël
- Sorbonne Université, CNRS, Laboratoire de Biodiversité et Biotechnologies Microbiennes, LBBM, Observatoire Océanologique, 66 650 Banyuls-sur-Mer, France
| | - Véronique Eparvier
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198 Gif-sur-Yvette, France
| | - David Touboul
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198 Gif-sur-Yvette, France
- Laboratoire de Chimie Moléculaire (LCM), CNRS, École Polytechnique, Institut Polytechnique de Paris, 91120 Palaiseau, France
| |
Collapse
|
18
|
Barut I, Fletcher JS. Cell and tissue imaging by secondary ion mass spectrometry. Biointerphases 2023; 18:061202. [PMID: 38108477 DOI: 10.1116/6.0003140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023] Open
Abstract
This Tutorial focuses on the use of secondary ion mass spectrometry for the analysis of cellular and tissue samples. The Tutorial aims to cover the considerations in sample preparation analytical set up and some specific aspects of data interpretation associated with such analysis.
Collapse
Affiliation(s)
- Inci Barut
- Department of Pharmacy, Basic Pharmaceutical Sciences, Gazi University, Ankara 06330, Turkey
| | - John S Fletcher
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg 413 90, Sweden
| |
Collapse
|
19
|
Liu M, Salinas G, Yu J, Cornet A, Li H, Kuhn A, Sojic N. Endogenous and exogenous wireless multimodal light-emitting chemical devices. Chem Sci 2023; 14:10664-10670. [PMID: 37829015 PMCID: PMC10566513 DOI: 10.1039/d3sc03678b] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/04/2023] [Indexed: 10/14/2023] Open
Abstract
Multimodal imaging is a powerful and versatile approach that integrates and correlates multiple optical modalities within a single device. This concept has gained considerable attention due to its potential applications ranging from sensing to medicine. Herein, we develop several wireless multimodal light-emitting chemical systems by coupling two light sources based on different physical principles: electrochemiluminescence (ECL) occurring at the electrode interface and a light-emitting diode (LED) switched on by an electrochemically triggered electron flow. Endogenous (thermodynamically spontaneous redox process) and exogenous (requiring an external power source) bipolar electrochemistry acts as a driving force to trigger both light emissions at different wavelengths. The results presented here interconnect optical imaging and electrochemical reactions, providing a novel and so far unexplored alternative to design autonomous hybrid systems with multimodal and multicolor optical readouts for complex bio-chemical systems.
Collapse
Affiliation(s)
- Miaoxia Liu
- Univ. Bordeaux, Bordeaux INP, ISM, UMR 5255 CNRS, Site ENSMAC 33607 Pessac France
| | - Gerardo Salinas
- Univ. Bordeaux, Bordeaux INP, ISM, UMR 5255 CNRS, Site ENSMAC 33607 Pessac France
| | - Jing Yu
- Univ. Bordeaux, Bordeaux INP, ISM, UMR 5255 CNRS, Site ENSMAC 33607 Pessac France
| | - Antoine Cornet
- Univ. Bordeaux, Bordeaux INP, ISM, UMR 5255 CNRS, Site ENSMAC 33607 Pessac France
| | - Haidong Li
- College of Chemistry and Chemical Engineering, Yangzhou University 225002 Yangzhou China
| | - Alexander Kuhn
- Univ. Bordeaux, Bordeaux INP, ISM, UMR 5255 CNRS, Site ENSMAC 33607 Pessac France
| | - Neso Sojic
- Univ. Bordeaux, Bordeaux INP, ISM, UMR 5255 CNRS, Site ENSMAC 33607 Pessac France
| |
Collapse
|
20
|
Moore JL, Patterson NH, Norris JL, Caprioli RM. Prospective on Imaging Mass Spectrometry in Clinical Diagnostics. Mol Cell Proteomics 2023; 22:100576. [PMID: 37209813 PMCID: PMC10545939 DOI: 10.1016/j.mcpro.2023.100576] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/22/2023] Open
Abstract
Imaging mass spectrometry (IMS) is a molecular technology utilized for spatially driven research, providing molecular maps from tissue sections. This article reviews matrix-assisted laser desorption ionization (MALDI) IMS and its progress as a primary tool in the clinical laboratory. MALDI mass spectrometry has been used to classify bacteria and perform other bulk analyses for plate-based assays for many years. However, the clinical application of spatial data within a tissue biopsy for diagnoses and prognoses is still an emerging opportunity in molecular diagnostics. This work considers spatially driven mass spectrometry approaches for clinical diagnostics and addresses aspects of new imaging-based assays that include analyte selection, quality control/assurance metrics, data reproducibility, data classification, and data scoring. It is necessary to implement these tasks for the rigorous translation of IMS to the clinical laboratory; however, this requires detailed standardized protocols for introducing IMS into the clinical laboratory to deliver reliable and reproducible results that inform and guide patient care.
Collapse
Affiliation(s)
| | - Nathan Heath Patterson
- Frontier Diagnostics, Nashville, Tennessee, USA; Vanderbilt University Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Jeremy L Norris
- Frontier Diagnostics, Nashville, Tennessee, USA; Vanderbilt University Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Richard M Caprioli
- Frontier Diagnostics, Nashville, Tennessee, USA; Vanderbilt University Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA; Departments of Biochemistry, Pharmacology, Chemistry, and Medicine, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
21
|
Rittel MF, Schmidt S, Weis CA, Birgin E, van Marwick B, Rädle M, Diehl SJ, Rahbari NN, Marx A, Hopf C. Spatial Omics Imaging of Fresh-Frozen Tissue and Routine FFPE Histopathology of a Single Cancer Needle Core Biopsy: A Freezing Device and Multimodal Workflow. Cancers (Basel) 2023; 15:2676. [PMID: 37345020 DOI: 10.3390/cancers15102676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/16/2023] [Accepted: 05/06/2023] [Indexed: 06/23/2023] Open
Abstract
The complex molecular alterations that underlie cancer pathophysiology are studied in depth with omics methods using bulk tissue extracts. For spatially resolved tissue diagnostics using needle biopsy cores, however, histopathological analysis using stained FFPE tissue and the immunohistochemistry (IHC) of a few marker proteins is currently the main clinical focus. Today, spatial omics imaging using MSI or IRI is an emerging diagnostic technology for the identification and classification of various cancer types. However, to conserve tissue-specific metabolomic states, fast, reliable, and precise methods for the preparation of fresh-frozen (FF) tissue sections are crucial. Such methods are often incompatible with clinical practice, since spatial metabolomics and the routine histopathology of needle biopsies currently require two biopsies for FF and FFPE sampling, respectively. Therefore, we developed a device and corresponding laboratory and computational workflows for the multimodal spatial omics analysis of fresh-frozen, longitudinally sectioned needle biopsies to accompany standard FFPE histopathology of the same biopsy core. As a proof-of-concept, we analyzed surgical human liver cancer specimens using IRI and MSI with precise co-registration and, following FFPE processing, by sequential clinical pathology analysis of the same biopsy core. This workflow allowed for a spatial comparison between different spectral profiles and alterations in tissue histology, as well as a direct comparison for histological diagnosis without the need for an extra biopsy.
Collapse
Affiliation(s)
- Miriam F Rittel
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
- Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Stefan Schmidt
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
| | - Cleo-Aron Weis
- Institute of Pathology, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Emrullah Birgin
- Department of Surgery, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Björn van Marwick
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
| | - Matthias Rädle
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
- Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Steffen J Diehl
- Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Clinic of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Nuh N Rahbari
- Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Department of Surgery, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Alexander Marx
- Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
- Institute of Pathology, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Carsten Hopf
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, Paul-Wittsack-Str. 10, 68163 Mannheim, Germany
- Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| |
Collapse
|
22
|
Krijnen K, Keelor JD, Böhm S, Ellis SR, Köster C, Höhndorf J, Heeren RMA, Anthony IGM. A Multimodal SIMS/MALDI Mass Spectrometry Imaging Source with Secondary Electron Imaging Capabilities for Use with timsTOF Instruments. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:720-727. [PMID: 36891615 PMCID: PMC10080675 DOI: 10.1021/jasms.2c00381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 06/18/2023]
Abstract
Mass spectrometry imaging (MSI) is a surface analysis technique that produces chemical images and is commonly used for biological and biomedical research. Multimodal imaging combines multiple imaging modes in order to get a more comprehensive view of a sample. Multimodal MSI images are often acquired using multiple MSI instruments, which leads to issues regarding image registration and increases the chance of sample damage or degradation during sample transfer. These problems can be solved by using a single instrument that can image in multiple modes. In order to improve the efficiency of multimodal imaging and investigate complementary modes of MSI, we have modified a prototype Bruker timsTOF fleX by adding secondary ion mass spectrometry (SIMS) and secondary electron (SE) imaging capabilities while preserving the ability to perform matrix-assisted laser desorption/ionization (MALDI). We show multimodal images collected on this instrument that required only trivial registration and were acquired without sample transfer between imaging trials. Furthermore, we characterize the performance of SIMS, SE, and MALDI imaging and compare the performance of the modified instrument to a commercial timsTOF fleX.
Collapse
Affiliation(s)
- Kasper Krijnen
- Maastricht
MultiModal Molecular Imaging (M4i) Institute, Division of Imaging
Mass Spectrometry, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Joel D. Keelor
- Maastricht
MultiModal Molecular Imaging (M4i) Institute, Division of Imaging
Mass Spectrometry, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Sebastian Böhm
- Bruker
Daltonics GmbH & Co KG, Fahrenheitstraße 4, 28359 Bremen, Germany
| | - Shane R. Ellis
- Maastricht
MultiModal Molecular Imaging (M4i) Institute, Division of Imaging
Mass Spectrometry, Maastricht University, 6229 ER Maastricht, The Netherlands
- Molecular
Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Claus Köster
- Bruker
Daltonics GmbH & Co KG, Fahrenheitstraße 4, 28359 Bremen, Germany
| | - Jens Höhndorf
- Bruker
Daltonics GmbH & Co KG, Fahrenheitstraße 4, 28359 Bremen, Germany
| | - Ron M. A. Heeren
- Maastricht
MultiModal Molecular Imaging (M4i) Institute, Division of Imaging
Mass Spectrometry, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ian G. M. Anthony
- Maastricht
MultiModal Molecular Imaging (M4i) Institute, Division of Imaging
Mass Spectrometry, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
23
|
Lin BJ, Kuo TC, Chung HH, Huang YC, Wang MY, Hsu CC, Yao PY, Tseng YJ. MSIr: Automatic Registration Service for Mass Spectrometry Imaging and Histology. Anal Chem 2023; 95:3317-3324. [PMID: 36724516 PMCID: PMC9933042 DOI: 10.1021/acs.analchem.2c04360] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Mass spectrometry imaging (MSI) is a powerful tool that can be used to simultaneously investigate the spatial distribution of different molecules in samples. However, it is difficult to comprehensively analyze complex biological systems with only a single analytical technique due to different analytical properties and application limitations. Therefore, many analytical methods have been combined to extend data interpretation, evaluate data credibility, and facilitate data mining to explore important temporal and spatial relationships in biological systems. Image registration is an initial and critical step for multimodal imaging data fusion. However, the image registration of multimodal images is not a simple task. The property difference between each data modality may include spatial resolution, image characteristics, or both. The image registrations between MSI and different imaging techniques are often achieved indirectly through histology. Many methods exist for image registration between MSI data and histological images. However, most of them are manual or semiautomatic and have their prerequisites. Here, we built MSI Registrar (MSIr), a web service for automatic registration between MSI and histology. It can help to reduce subjectivity and processing time efficiently. MSIr provides an interface for manually selecting region of interests from histological images; the user selects regions of interest to extract the corresponding spectrum indices in MSI data. In the performance evaluation, MSIr can quickly map MSI data to histological images and help pinpoint molecular components at specific locations in tissues. Most registrations were adequate and were without excessive shifts. MSIr is freely available at https://msir.cmdm.tw and https://github.com/CMDM-Lab/MSIr.
Collapse
Affiliation(s)
- Bo-Jhang Lin
- Graduate
Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan
| | - Tien-Chueh Kuo
- The
Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Hsin-Hsiang Chung
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Ying-Chen Huang
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Ming-Yang Wang
- Department
of Surgery, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Cheng-Chih Hsu
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Po-Yang Yao
- Graduate
Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan
| | - Yufeng Jane Tseng
- Graduate
Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan,The
Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei 10617, Taiwan,Department
of Computer Science and Information Engineering, National Taiwan University, Taipei 10617, Taiwan,School of
Pharmacy, College of Medicine, National
Taiwan University, Taipei 10002, Taiwan,. Phone: +886.2.3366.4888#529. Fax: +886.2.23628167
| |
Collapse
|
24
|
Xi Y, Sohn AL, Joignant AN, Cologna SM, Prentice BM, Muddiman DC. SMART: A data reporting standard for mass spectrometry imaging. JOURNAL OF MASS SPECTROMETRY : JMS 2023; 58:e4904. [PMID: 36740651 PMCID: PMC10078510 DOI: 10.1002/jms.4904] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
Mass spectrometry imaging (MSI) is an important analytical technique that simultaneously reports the spatial location and abundance of detected ions in biological, chemical, clinical, and pharmaceutical studies. As MSI grows in popularity, it has become evident that data reporting varies among different research groups and between techniques. The lack of consistency in data reporting inherently creates additional challenges in comparing intra- and inter-laboratory MSI data. In this tutorial, we propose a unified data reporting system, SMART, based on the common features shared between techniques. While there are limitations to any reporting system, SMART was decided upon after significant discussion to more easily understand and benchmark MSI data. SMART is not intended to be comprehensive but rather capture essential baseline information for a given MSI study; this could be within a study (e.g., effect of spot size on the measured ion signals) or between two studies (e.g., different MSI platform technologies applied to the same tissue type). This tutorial does not attempt to address the confidence with which annotations are made nor does it deny the importance of other parameters that are not included in the current SMART format. Ultimately, the goal of this tutorial is to discuss the necessity of establishing a uniform reporting system to communicate MSI data in publications and presentations in a simple format to readily interpret the parameters and baseline outcomes of the data.
Collapse
Affiliation(s)
- Ying Xi
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina, USA
- Molecular Education, Technology and Research Innovation Center, North Carolina State University, Raleigh, North Carolina, USA
| | - Alexandria L Sohn
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina, USA
| | - Alena N Joignant
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina, USA
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Boone M Prentice
- Department of Chemistry, University of Florida, Gainesville, Florida, USA
| | - David C Muddiman
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina, USA
- Molecular Education, Technology and Research Innovation Center, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
25
|
Golpelichi F, Parastar H. Quantitative Mass Spectrometry Imaging Using Multivariate Curve Resolution and Deep Learning: A Case Study. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:236-244. [PMID: 36594891 DOI: 10.1021/jasms.2c00268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In the present contribution, a novel approach based on multivariate curve resolution and deep learning (DL) is proposed for quantitative mass spectrometry imaging (MSI) as a potent technique for identifying different compounds and creating their distribution maps in biological tissues without need for sample preparation. As a case study, chlordecone as a carcinogenic pesticide was quantitatively determined in mouse liver using matrix-assisted laser desorption ionization-MSI (MALDI-MSI). For this purpose, data from seven standard spots containing 0 to 20 picomoles of chlordecone and four unknown tissues from the mouse livers infected with chlordecone for 1, 5, and 10 days were analyzed using a convolutional neural network (CNN). To solve the lack of sufficient data for CNN model training, each pixel was considered as a sample, the designed CNN models were trained by pixels in training sets, and their corresponding amounts of chlordecone were obtained by multivariate curve resolution-alternating least-squares (MCR-ALS). The trained models were then externally evaluated using calibration pixels in test sets for 1, 5, and 10 days of exposure, respectively. Prediction R2 for all three data sets ranged from 0.93 to 0.96, which was superior to support vector machine (SVM) and partial least-squares (PLS). The trained CNN models were finally used to predict the amount of chlordecone in mouse liver tissues, and their results were compared with MALDI-MSI and GC-MS methods, which were comparable. Inspection of the results confirmed the validity of the proposed method.
Collapse
Affiliation(s)
- Fatemeh Golpelichi
- Department of Chemistry, Sharif University of Technology, P.O. Box 11155-9516, 1458889694Tehran, Iran
| | - Hadi Parastar
- Department of Chemistry, Sharif University of Technology, P.O. Box 11155-9516, 1458889694Tehran, Iran
| |
Collapse
|
26
|
Guo A, Chen Z, Li F, Luo Q. Delineating regions of interest for mass spectrometry imaging by multimodally corroborated spatial segmentation. Gigascience 2022; 12:giad021. [PMID: 37039115 PMCID: PMC10087011 DOI: 10.1093/gigascience/giad021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/17/2023] [Accepted: 03/13/2023] [Indexed: 04/12/2023] Open
Abstract
Mass spectrometry imaging (MSI), which localizes molecules in a tag-free, spatially resolved manner, is a powerful tool for the understanding of underlying biochemical mechanisms of biological phenomena. When analyzing MSI data, it is essential to delineate regions of interest (ROIs) that correspond to tissue areas of different anatomical or pathological labels. Spatial segmentation, obtained by clustering MSI pixels according to their mass spectral similarities, is a popular approach to automate ROI definition. However, how to select the number of clusters (#Clusters), which determines the granularity of segmentation, remains to be resolved, and an inappropriate #Clusters may lead to ROIs not biologically real. Here we report a multimodal fusion strategy to enable an objective and trustworthy selection of #Clusters by utilizing additional information from corresponding histology images. A deep learning-based algorithm is proposed to extract "histomorphological feature spectra" across an entire hematoxylin and eosin image. Clustering is then similarly performed to produce histology segmentation. Since ROIs originating from instrumental noise or artifacts would not be reproduced cross-modally, the consistency between histology and MSI segmentation becomes an effective measure of the biological validity of the results. So, #Clusters that maximize the consistency is deemed as most probable. We validated our strategy on mouse kidney and renal tumor specimens by producing multimodally corroborated ROIs that agreed excellently with ground truths. Downstream analysis based on the said ROIs revealed lipid molecules highly specific to tissue anatomy or pathology. Our work will greatly facilitate MSI-mediated spatial lipidomics, metabolomics, and proteomics research by providing intelligent software to automatically and reliably generate ROIs.
Collapse
Affiliation(s)
- Ang Guo
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhiyu Chen
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qian Luo
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
27
|
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: 24] [Impact Index Per Article: 8.0] [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
|
28
|
Hou JJ, Zhang ZJ, Wu WY, He QQ, Zhang TQ, Liu YW, Wang ZJ, Gao L, Long HL, Lei M, Wu WY, Guo DA. Mass spectrometry imaging: new eyes on natural products for drug research and development. Acta Pharmacol Sin 2022; 43:3096-3111. [PMID: 36229602 PMCID: PMC9712638 DOI: 10.1038/s41401-022-00990-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/25/2022] [Indexed: 11/09/2022]
Abstract
Natural products (NPs) and their structural analogs represent a major source of novel drug development for disease prevention and treatment. The development of new drugs from NPs includes two crucial aspects. One is the discovery of NPs from medicinal plants/microorganisms, and the other is the evaluation of the NPs in vivo at various physiological and pathological states. The heterogeneous spatial distribution of NPs in medicinal plants/microorganisms or in vivo can provide valuable information for drug development. However, few molecular imaging technologies can detect thousands of compounds simultaneously on a label-free basis. Over the last two decades, mass spectrometry imaging (MSI) methods have progressively improved and diversified, thereby allowing for the development of various applications of NPs in plants/microorganisms and in vivo NP research. Because MSI allows for the spatial mapping of the production and distribution of numerous molecules in situ without labeling, it provides a visualization tool for NP research. Therefore, we have focused this mini-review on summarizing the applications of MSI technology in discovering NPs from medicinal plants and evaluating NPs in preclinical studies from the perspective of new drug research and development (R&D). Additionally, we briefly reviewed the factors that should be carefully considered to obtain the desired MSI results. Finally, the future development of MSI in new drug R&D is proposed.
Collapse
Affiliation(s)
- Jin-Jun Hou
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zi-Jia Zhang
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wen-Yong Wu
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Qing-Qing He
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Teng-Qian Zhang
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ya-Wen Liu
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhao-Jun Wang
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Gao
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hua-Li Long
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Min Lei
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wan-Ying Wu
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - De-An Guo
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
29
|
Godeffroy L, Lemineur JF, Shkirskiy V, Miranda Vieira M, Noël JM, Kanoufi F. Bridging the Gap between Single Nanoparticle Imaging and Global Electrochemical Response by Correlative Microscopy Assisted By Machine Vision. SMALL METHODS 2022; 6:e2200659. [PMID: 35789075 DOI: 10.1002/smtd.202200659] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/17/2022] [Indexed: 06/15/2023]
Abstract
The nanostructuration of an electrochemical interface dictates its micro- and macroscopic behavior. It is generally highly complex and often evolves under operating conditions. Electrochemistry at these nanostructurations can be imaged both operando and/or ex situ at the single nanoobject or nanoparticle (NP) level by diverse optical, electron, and local probe microscopy techniques. However, they only probe a tiny random fraction of interfaces that are by essence highly heterogeneous. Given the above background, correlative multimicroscopy strategy coupled to electrochemistry in a droplet cell provides a unique solution to gain mechanistic insights in electrocatalysis. To do so, a general machine-vision methodology is depicted enabling the automated local identification of various physical and chemical descriptors of NPs (size, composition, activity) obtained from multiple complementary operando and ex situ microscopy imaging of the electrode. These multifarious microscopically probed descriptors for each and all individual NPs are used to reconstruct the global electrochemical response. Herein the methodology unveils the competing processes involved in the electrocatalysis of hydrogen evolution reaction at nickel based NPs, showing that Ni metal activity is comparable to that of platinum.
Collapse
Affiliation(s)
| | | | | | | | - Jean-Marc Noël
- Université Paris Cité, ITODYS, CNRS, 75013, Paris, France
| | | |
Collapse
|
30
|
Nwabufo CK, Aigbogun OP. The Role of Mass Spectrometry Imaging in Pharmacokinetic Studies. Xenobiotica 2022; 52:811-827. [PMID: 36048000 DOI: 10.1080/00498254.2022.2119900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Although liquid chromatography-tandem mass spectrometry is the gold standard analytical platform for the quantification of drugs, metabolites, and biomarkers in biological samples, it cannot localize them in target tissues.The localization and quantification of drugs and/or their associated metabolites in target tissues is a more direct measure of bioavailability, biodistribution, efficacy, and regional toxicity compared to the traditional substitute studies using plasma.Therefore, combining high spatial resolution imaging functionality with the superior selectivity and sensitivity of mass spectrometry into one analytical technique will be a valuable tool for targeted localization and quantification of drugs, metabolites, and biomarkers.Mass spectrometry imaging (MSI) is a tagless analytical technique that allows for the direct localization and quantification of drugs, metabolites, and biomarkers in biological tissues, and has been used extensively in pharmaceutical research.The overall goal of this current review is to provide a detailed description of the working principle of MSI and its application in pharmacokinetic studies encompassing absorption, distribution, metabolism, excretion, and toxicity processes, followed by a discussion of the strategies for addressing the challenges associated with the functional utility of MSI in pharmacokinetic studies that support drug development.
Collapse
Affiliation(s)
- Chukwunonso K Nwabufo
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Omozojie P Aigbogun
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada.,Department of Chemistry, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
31
|
Baquer G, Sementé L, Mahamdi T, Correig X, Ràfols P, García-Altares M. What are we imaging? Software tools and experimental strategies for annotation and identification of small molecules in mass spectrometry imaging. MASS SPECTROMETRY REVIEWS 2022:e21794. [PMID: 35822576 DOI: 10.1002/mas.21794] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Mass spectrometry imaging (MSI) has become a widespread analytical technique to perform nonlabeled spatial molecular identification. The Achilles' heel of MSI is the annotation and identification of molecular species due to intrinsic limitations of the technique (lack of chromatographic separation and the difficulty to apply tandem MS). Successful strategies to perform annotation and identification combine extra analytical steps, like using orthogonal analytical techniques to identify compounds; with algorithms that integrate the spectral and spatial information. In this review, we discuss different experimental strategies and bioinformatics tools to annotate and identify compounds in MSI experiments. We target strategies and tools for small molecule applications, such as lipidomics and metabolomics. First, we explain how sample preparation and the acquisition process influences annotation and identification, from sample preservation to the use of orthogonal techniques. Then, we review twelve software tools for annotation and identification in MSI. Finally, we offer perspectives on two current needs of the MSI community: the adaptation of guidelines for communicating confidence levels in identifications; and the creation of a standard format to store and exchange annotations and identifications in MSI.
Collapse
Affiliation(s)
- Gerard Baquer
- Department of Electronic Engineering, University Rovira I Virgili, Tarragona, Spain
| | - Lluc Sementé
- Department of Electronic Engineering, University Rovira I Virgili, Tarragona, Spain
| | - Toufik Mahamdi
- Department of Electronic Engineering, University Rovira I Virgili, Tarragona, Spain
| | - Xavier Correig
- Department of Electronic Engineering, University Rovira I Virgili, Tarragona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Institut D'Investigacio Sanitaria Pere Virgili, Tarragona, Spain
| | - Pere Ràfols
- Department of Electronic Engineering, University Rovira I Virgili, Tarragona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Institut D'Investigacio Sanitaria Pere Virgili, Tarragona, Spain
| | - María García-Altares
- Department of Electronic Engineering, University Rovira I Virgili, Tarragona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| |
Collapse
|
32
|
Spruill ML, Maletic-Savatic M, Martin H, Li F, Liu X. Spatial analysis of drug absorption, distribution, metabolism, and toxicology using mass spectrometry imaging. Biochem Pharmacol 2022; 201:115080. [PMID: 35561842 PMCID: PMC9744413 DOI: 10.1016/j.bcp.2022.115080] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
Abstract
Mass spectrometry imaging (MSI) is emerging as a powerful analytical tool for detection, quantification, and simultaneous spatial molecular imaging of endogenous and exogenous molecules via in situ mass spectrometry analysis of thin tissue sections without the requirement of chemical labeling. The MSI generates chemically specific and spatially resolved ion distribution information for administered drugs and metabolites, which allows numerous applications for studies involving various stages of drug absorption, distribution, metabolism, excretion, and toxicity (ADMET). MSI-based pharmacokinetic imaging analysis provides a histological context and cellular environment regarding dynamic drug distribution and metabolism processes, and facilitates the understanding of the spatial pharmacokinetics and pharmacodynamic properties of drugs. Herein, we discuss the MSI's current technological developments that offer qualitative, quantitative, and spatial location information of small molecule drugs, antibody, and oligonucleotides macromolecule drugs, and their metabolites in preclinical and clinical tissue specimens. We highlight the macro and micro drug-distribution in the whole-body, brain, lung, liver, kidney, stomach, intestine tissue sections, organoids, and the latest applications of MSI in pharmaceutical ADMET studies.
Collapse
Affiliation(s)
- Michelle L Spruill
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Mirjana Maletic-Savatic
- Department of Pediatrics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | | | - Feng Li
- Center for Drug Discovery and Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA.
| |
Collapse
|
33
|
Dong Y, Aharoni A. Image to insight: exploring natural products through mass spectrometry imaging. Nat Prod Rep 2022; 39:1510-1530. [PMID: 35735199 DOI: 10.1039/d2np00011c] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Covering: 2017 to 2022Mass spectrometry imaging (MSI) has become a mature molecular imaging technique that is well-matched for natural product (NP) discovery. Here we present a brief overview of MSI, followed by a thorough discussion of different MSI applications in NP research. This review will mainly focus on the recent progress of MSI in plants and microorganisms as they are the main producers of NPs. Specifically, the opportunity and potential of combining MSI with other imaging modalities and stable isotope labeling are discussed. Throughout, we focus on both the strengths and weaknesses of MSI, with an eye on future improvements that are necessary for the progression of MSI toward routine NP studies. Finally, we discuss new areas of research, future perspectives, and the overall direction that the field may take in the years to come.
Collapse
Affiliation(s)
- Yonghui Dong
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Asaph Aharoni
- Department of Plant Sciences, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
34
|
Kuerschner L, Thiele C. Tracing Lipid Metabolism by Alkyne Lipids and Mass Spectrometry: The State of the Art. Front Mol Biosci 2022; 9:880559. [PMID: 35669564 PMCID: PMC9163959 DOI: 10.3389/fmolb.2022.880559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/19/2022] [Indexed: 01/22/2023] Open
Abstract
Lipid tracing studies are a key method to gain a better understanding of the complex metabolic network lipids are involved in. In recent years, alkyne lipid tracers and mass spectrometry have been developed as powerful tools for such studies. This study aims to review the present standing of the underlying technique, highlight major findings the strategy allowed for, summarize its advantages, and discuss some limitations. In addition, an outlook on future developments is given.
Collapse
|
35
|
Tuck M, Grélard F, Blanc L, Desbenoit N. MALDI-MSI Towards Multimodal Imaging: Challenges and Perspectives. Front Chem 2022; 10:904688. [PMID: 35615316 PMCID: PMC9124797 DOI: 10.3389/fchem.2022.904688] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/14/2022] [Indexed: 01/22/2023] Open
Abstract
Multimodal imaging is a powerful strategy for combining information from multiple images. It involves several fields in the acquisition, processing and interpretation of images. As multimodal imaging is a vast subject area with various combinations of imaging techniques, it has been extensively reviewed. Here we focus on Matrix-assisted Laser Desorption Ionization Mass Spectrometry Imaging (MALDI-MSI) coupling other imaging modalities in multimodal approaches. While MALDI-MS images convey a substantial amount of chemical information, they are not readily informative about the morphological nature of the tissue. By providing a supplementary modality, MALDI-MS images can be more informative and better reflect the nature of the tissue. In this mini review, we emphasize the analytical and computational strategies to address multimodal MALDI-MSI.
Collapse
|
36
|
Ajith A, Milnes PJ, Johnson GN, Lockyer NP. Mass Spectrometry Imaging for Spatial Chemical Profiling of Vegetative Parts of Plants. PLANTS 2022; 11:plants11091234. [PMID: 35567235 PMCID: PMC9102225 DOI: 10.3390/plants11091234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022]
Abstract
The detection of chemical species and understanding their respective localisations in tissues have important implications in plant science. The conventional methods for imaging spatial localisation of chemical species are often restricted by the number of species that can be identified and is mostly done in a targeted manner. Mass spectrometry imaging combines the ability of traditional mass spectrometry to detect numerous chemical species in a sample with their spatial localisation information by analysing the specimen in a 2D manner. This article details the popular mass spectrometry imaging methodologies which are widely pursued along with their respective sample preparation and the data analysis methods that are commonly used. We also review the advancements through the years in the usage of the technique for the spatial profiling of endogenous metabolites, detection of xenobiotic agrochemicals and disease detection in plants. As an actively pursued area of research, we also address the hurdles in the analysis of plant tissues, the future scopes and an integrated approach to analyse samples combining different mass spectrometry imaging methods to obtain the most information from a sample of interest.
Collapse
Affiliation(s)
- Akhila Ajith
- Department of Chemistry, Photon Science Institute, University of Manchester, Manchester M13 9PL, UK;
| | - Phillip J. Milnes
- Syngenta, Jeolott’s Hill International Research Centre, Bracknell RG42 6EY, UK;
| | - Giles N. Johnson
- Department of Earth and Environmental Sciences, University of Manchester, Manchester M13 9PY, UK;
| | - Nicholas P. Lockyer
- Department of Chemistry, Photon Science Institute, University of Manchester, Manchester M13 9PL, UK;
- Correspondence:
| |
Collapse
|
37
|
Technical steps towards enhanced localization of proteins in cultural heritage samples by immunofluorescence microscopy and micro-reflectance imaging spectroscopy. Microchem J 2022. [DOI: 10.1016/j.microc.2022.107243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
38
|
Simultaneous Determination of Lamotrigine, Oxcarbazepine, Lacosamide, and Topiramate in Rat Plasma by Ultra-Performance Liquid Chromatography-Tandem Mass Spectrometry. Int J Anal Chem 2022; 2022:1838645. [PMID: 35321047 PMCID: PMC8938153 DOI: 10.1155/2022/1838645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/24/2022] [Indexed: 12/21/2022] Open
Abstract
This study established an ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method to study the pharmacokinetics of four antiepileptic drugs, lamotrigine, oxcarbazepine, lacosamide, and topiramate, in rats after oral administration. The gradient elution was performed on a UPLC HSS T3 (2.1 mm × 100 mm, 1.8 μm) column with acetonitrile-0.1% formic acid as the mobile phase at a flow rate of 0.4 mL/min. Protein precipitation by acetonitrile was adopted for plasma sample pretreatment. Electrospray- (ESI-) positive/negative ion switching and multiple reaction monitoring (MRM) modes were adopted for ion quantitative determination of antiepileptic drugs. UPLC-MS/MS detection and Drug and Statistics (DAS) software fitting were performed to blood samples collected from rats after oral administration of lamotrigine, oxcarbazepine, lacosamide, and topiramate (5 mg/kg). All drugs examined showed linearity within 5–5000 ng/ml (R2 > 0.9987), the intraday accuracy was within 92%–108%, and the interday accuracy was within 93%–109%. The relative standard deviations (RSD) of intraday and interday were less than 15%. The matrix effect was within 91%–105%, and the recovery was better than 88%. The established UPLC-MS/MS method was successfully applied to the pharmacokinetic study of lamotrigine, oxcarbazepine, lacosamide, and topiramate in rats.
Collapse
|
39
|
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
|
40
|
Ritschar S, Schirmer E, Hufnagl B, Löder MGJ, Römpp A, Laforsch C. Classification of target tissues of Eisenia fetida using sequential multimodal chemical analysis and machine learning. Histochem Cell Biol 2022; 157:127-137. [PMID: 34750664 PMCID: PMC8847259 DOI: 10.1007/s00418-021-02037-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2021] [Indexed: 12/22/2022]
Abstract
Acquiring comprehensive knowledge about the uptake of pollutants, impact on tissue integrity and the effects at the molecular level in organisms is of increasing interest due to the environmental exposure to numerous contaminants. The analysis of tissues can be performed by histological examination, which is still time-consuming and restricted to target-specific staining methods. The histological approaches can be complemented with chemical imaging analysis. Chemical imaging of tissue sections is typically performed using a single imaging approach. However, for toxicological testing of environmental pollutants, a multimodal approach combined with improved data acquisition and evaluation is desirable, since it may allow for more rapid tissue characterization and give further information on ecotoxicological effects at the tissue level. Therefore, using the soil model organism Eisenia fetida as a model, we developed a sequential workflow combining Fourier transform infrared spectroscopy (FTIR) and matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) for chemical analysis of the same tissue sections. Data analysis of the FTIR spectra via random decision forest (RDF) classification enabled the rapid identification of target tissues (e.g., digestive tissue), which are relevant from an ecotoxicological point of view. MALDI imaging analysis provided specific lipid species which are sensitive to metabolic changes and environmental stressors. Taken together, our approach provides a fast and reproducible workflow for label-free histochemical tissue analyses in E. fetida, which can be applied to other model organisms as well.
Collapse
Affiliation(s)
- Sven Ritschar
- Department of Animal Ecology i and BayCEER, University of Bayreuth, Bayreuth, Germany
| | - Elisabeth Schirmer
- Department of Bioanalytical Sciences and Food Analysis, University of Bayreuth, Bayreuth, Germany
| | - Benedikt Hufnagl
- Institute of Chemical Technologies and Analytics, Vienna, TU, Austria
- Purency GmbH, Walfischgasse 8/34, T1010, Vienna, Austria
| | - Martin G J Löder
- Department of Animal Ecology i and BayCEER, University of Bayreuth, Bayreuth, Germany
| | - Andreas Römpp
- Department of Bioanalytical Sciences and Food Analysis, University of Bayreuth, Bayreuth, Germany.
| | - Christian Laforsch
- Department of Animal Ecology i and BayCEER, University of Bayreuth, Bayreuth, Germany.
| |
Collapse
|
41
|
Engel KM, Prabutzki P, Leopold J, Nimptsch A, Lemmnitzer K, Vos DRN, Hopf C, Schiller J. A new update of MALDI-TOF mass spectrometry in lipid research. Prog Lipid Res 2022; 86:101145. [PMID: 34995672 DOI: 10.1016/j.plipres.2021.101145] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 01/06/2023]
Abstract
Matrix-assisted laser desorption and ionization (MALDI) mass spectrometry (MS) is an indispensable tool in modern lipid research since it is fast, sensitive, tolerates sample impurities and provides spectra without major analyte fragmentation. We will discuss some methodological aspects, the related ion-forming processes and the MALDI MS characteristics of the different lipid classes (with the focus on glycerophospholipids) and the progress, which was achieved during the last ten years. Particular attention will be given to quantitative aspects of MALDI MS since this is widely considered as the most serious drawback of the method. Although the detailed role of the matrix is not yet completely understood, it will be explicitly shown that the careful choice of the matrix is crucial (besides the careful evaluation of the positive and negative ion mass spectra) in order to be able to detect all lipid classes of interest. Two developments will be highlighted: spatially resolved Imaging MS is nowadays well established and the distribution of lipids in tissues merits increasing interest because lipids are readily detectable and represent ubiquitous compounds. It will also be shown that a combination of MALDI MS with thin-layer chromatography (TLC) enables a fast spatially resolved screening of an entire TLC plate which makes the method competitive with LC/MS.
Collapse
Affiliation(s)
- Kathrin M Engel
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - Patricia Prabutzki
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - Jenny Leopold
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - Ariane Nimptsch
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - Katharina Lemmnitzer
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany
| | - D R Naomi Vos
- Center for Biomedical Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Strasse 10, D-68163 Mannheim, Germany
| | - Carsten Hopf
- Center for Biomedical Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack-Strasse 10, D-68163 Mannheim, Germany
| | - Jürgen Schiller
- Leipzig University, Faculty of Medicine, Institute for Medical Physics and Biophysics, Härtelstraße 16-18, D-04107, Germany.
| |
Collapse
|
42
|
Van Nuffel S, Brunelle A. TOF-SIMS Imaging of Biological Tissue Sections and Structural Determination Using Tandem MS. Methods Mol Biol 2022; 2437:77-86. [PMID: 34902141 DOI: 10.1007/978-1-0716-2030-4_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over the past couple of years, imaging mass spectrometry (IMS) has arisen as a powerful tool to answer research questions in the biomedical field. Imaging mass spectrometry allows for label-free chemical imaging by providing full molecular information. The IMS technique best positioned for cell and tissue analysis is time-of-flight secondary ion mass spectrometry (ToF-SIMS) because it has the best spatial resolution of all the molecular IMS techniques and can detect many biochemical species and especially lipids with high sensitivity. Because one must rely on the mass and isotopic pattern of an ion in combination with positive correlations with lower mass fragments to help identify its structure, one major problem during ToF-SIMS experiments is the ambiguity when assigning a molecule to a certain mass peak. The solution are instruments with tandem MS capabilities as was already the case for many MALDI-ToF instruments more than a decade ago. It has been a few years since instruments with this capability were introduced and a number of interesting publications have been produced highlighting the advantages in biological SIMS work. Here, we present a protocol describing how tandem MS can be used to elucidate the structure of unknown or ambiguous mass peaks in biological tissue samples observed during ToF-SIMS imaging based on our experiences.
Collapse
Affiliation(s)
- Sebastiaan Van Nuffel
- Faculty of Science and Engineering, Maastricht University, Maastricht, The Netherlands
- Maastricht MultiModal Molecular Imaging Institute (M4i), Maastricht University, Maastricht, The Netherlands
| | - Alain Brunelle
- Sorbonne Université, CNRS, Laboratoire d'Archéologie Moléculaire et Structurale, LAMS, Paris, France.
| |
Collapse
|
43
|
Blanc L, Ferraro GB, Tuck M, Prideaux B, Dartois V, Jain RK, Desbenoit N. Kendrick Mass Defect Variation to Decipher Isotopic Labeling in Brain Metastases Studied by Mass Spectrometry Imaging. Anal Chem 2021; 93:16314-16319. [PMID: 34860501 PMCID: PMC9841243 DOI: 10.1021/acs.analchem.1c03916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Besides many other applications, isotopic labeling is commonly used to decipher the metabolism of living biological systems. By giving a stable isotopically labeled compound as a substrate, the biological system will use this labeled nutrient as it would with a regular substrate and incorporate stable heavy atoms into new metabolites. Utilizing mass spectrometry, by comparing heavy atom enriched isotopic profiles and naturally occurring ones, it is possible to identify these metabolites and deduce valuable information about metabolism and biochemical pathways. The coupling of this approach with mass spectrometry imaging (MSI) allows one then to obtain 2D maps of metabolisms used by living specimens. As metabolic networks are convoluted, a global overview of the isotopically labeled data set to detect unexpected metabolites is crucial. Unfortunately, due to the complexity of MSI spectra, such untargeted processing approaches are difficult to decipher. In this technical note, we demonstrate the potential of a variation around the Kendrick analysis concept to detect the incorporation of stable heavy atoms into metabolites. The Kendrick analysis uses as a base unit the difference between the mass of the most abundant isotope and the mass of the corresponding stable isotopic tracer (namely, 12C and 13C). The resulting Kendrick plot offers an alternative method to process the MSI data set with a new perspective allowing for the rapid detection of the 13C-enriched metabolites and separating unrelated compounds. This processing method of MS data could therefore be a useful tool to decipher isotopic labeling and study metabolic networks, especially as it does not require advanced computational capabilities.
Collapse
Affiliation(s)
- Landry Blanc
- Univ. Bordeaux, CNRS, CBMN, UMR 5248, F-33600 Pessac, France
| | - Gino B. Ferraro
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Michael Tuck
- Univ. Bordeaux, CNRS, CBMN, UMR 5248, F-33600 Pessac, France
| | - Brendan Prideaux
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Department of Medical Sciences, Hackensack Meridian Health, Nutley, New Jersey 07601, United States
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | | |
Collapse
|
44
|
Puthongkham P, Wirojsaengthong S, Suea-Ngam A. Machine learning and chemometrics for electrochemical sensors: moving forward to the future of analytical chemistry. Analyst 2021; 146:6351-6364. [PMID: 34585185 DOI: 10.1039/d1an01148k] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Electrochemical sensors and biosensors have been successfully used in a wide range of applications, but systematic optimization and nonlinear relationships have been compromised for electrode fabrication and data analysis. Machine learning and experimental designs are chemometric tools that have been proved to be useful in method development and data analysis. This minireview summarizes recent applications of machine learning and experimental designs in electroanalytical chemistry. First, experimental designs, e.g., full factorial, central composite, and Box-Behnken are discussed as systematic approaches to optimize electrode fabrication to consider the effects from individual variables and their interactions. Then, the principles of machine learning algorithms, including linear and logistic regressions, neural network, and support vector machine, are introduced. These machine learning models have been implemented to extract complex relationships between chemical structures and their electrochemical properties and to analyze complicated electrochemical data to improve calibration and analyte classification, such as in electronic tongues. Lastly, the future of machine learning and experimental designs in electrochemical sensors is outlined. These chemometric strategies will accelerate the development and enhance the performance of electrochemical devices for point-of-care diagnostics and commercialization.
Collapse
Affiliation(s)
- Pumidech Puthongkham
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand. .,Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Chulalongkorn University, Bangkok 10330, Thailand.,Center of Excellence in Responsive Wearable Materials, Chulalongkorn University, Bangkok 10330, Thailand
| | - Supacha Wirojsaengthong
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Akkapol Suea-Ngam
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
45
|
Surowka AD, Czyzycki M, Ziomber-Lisiak A, Migliori A, Szczerbowska-Boruchowska M. On 2D-FTIR-XRF microscopy - A step forward correlative tissue studies by infrared and hard X-ray radiation. Ultramicroscopy 2021; 232:113408. [PMID: 34706307 DOI: 10.1016/j.ultramic.2021.113408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/14/2021] [Accepted: 10/03/2021] [Indexed: 11/28/2022]
Abstract
Correlative Fourier Transform Infra-Red (FTIR) and hard X-Ray Fluorescence (XRF) microscopy studies of thin biological samples have recently evolved as complementary methods for biochemical fingerprinting of animal/human tissues. These are seen particularly useful for tracking the mechanisms of neurological diseases, i.e., in Alzheimer/Parkinson disease, in the brain where mishandling of trace metals (Fe, Cu, Zn) seems to be often associated with ongoing damage to molecular components via, among others, oxidative/reductive stress neurotoxicity. Despite substantial progress in state-of-the-art detection and data analysis methods, combined FTIR-XRF experiments have never benefited from correlation and co-localization analysis of molecular moieties and chemical elements, respectively. We here propose for the first time a completely novel data analysis pipeline, utilizing the idea of 2D correlation spectrometry for brain tissue analysis. In this paper, we utilized combined benchtop FTIR - synchrotron XRF mapping experiments on thin brain samples mounted on polypropylene membranes. By implementing our recently developed Multiple Linear Regression Multi-Reference (MLR-MR) algorithm, along with advanced image processing, artifact-free 2D FTIR-XRF spectra could be obtained by mitigating the impact of spectral artifacts, such as Etalon fringes and mild scattering Mie-like signatures, in the FTIR data. We demonstrated that the method is a powerful tool for co-localizing and correlating molecular arrangements and chemical elements (and vice versa) using visually attractive 2D correlograms. Moreover, the methods' applicability for fostering the identification of distinct (biological) materials, involving chemical elements and molecular arrangements, is also shown. Taken together, the 2D FTIR-XRF method opens up for new measures for in-situ investigating hidden complex biochemical correlations, and yet unraveled mechanisms in a biological sample. This step seems crucial for developing new strategies for facilitating the research on the interaction of metals/nonmetals with organic components. This is particularly important for enhancing our understanding of the diseases associated with metal/nonmetal mishandling.
Collapse
Affiliation(s)
- Artur D Surowka
- Faculty of Physics and Applied Computer Science, AGH University of Science and Technology, al. A. Mickiewicza 30, Krakow 30-059, Poland.
| | - Mateusz Czyzycki
- Faculty of Physics and Applied Computer Science, AGH University of Science and Technology, al. A. Mickiewicza 30, Krakow 30-059, Poland; Laboratory for Applications of Synchrotron Radiation, Karlsruhe Institute of Technology, Kaiser Str. 12, Karlsruhe 76131, Germany; Nuclear Science and Instrumentation Laboratory, International Atomic Energy Agency (IAEA) Laboratories, Seibersdorf, Austria
| | - Agata Ziomber-Lisiak
- Department of Pathophysiology, Jagiellonian University, Medical College, Czysta 18, Krakow 31-121, Poland
| | - Alessandro Migliori
- Nuclear Science and Instrumentation Laboratory, International Atomic Energy Agency (IAEA) Laboratories, Seibersdorf, Austria
| | | |
Collapse
|
46
|
Rapid brain structure and tumour margin detection on whole frozen tissue sections by fast multiphotometric mid-infrared scanning. Sci Rep 2021; 11:11307. [PMID: 34050224 PMCID: PMC8163866 DOI: 10.1038/s41598-021-90777-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/17/2021] [Indexed: 01/31/2023] Open
Abstract
Frozen section analysis is a frequently used method for examination of tissue samples, especially for tumour detection. In the majority of cases, the aim is to identify characteristic tissue morphologies or tumour margins. Depending on the type of tissue, a high number of misdiagnoses are associated with this process. In this work, a fast spectroscopic measurement device and workflow was developed that significantly improves the speed of whole frozen tissue section analyses and provides sufficient information to visualize tissue structures and tumour margins, dependent on their lipid and protein molecular vibrations. That optical and non-destructive method is based on selected wavenumbers in the mid-infrared (MIR) range. We present a measuring system that substantially outperforms a commercially available Fourier Transform Infrared (FT-IR) Imaging system, since it enables acquisition of reduced spectral information at a scan field of 1 cm2 in 3 s, with a spatial resolution of 20 µm. This allows fast visualization of segmented structure areas with little computational effort. For the first time, this multiphotometric MIR system is applied to biomedical tissue sections. We are referencing our novel MIR scanner on cryopreserved murine sagittal and coronal brain sections, especially focusing on the hippocampus, and show its usability for rapid identification of primary hepatocellular carcinoma (HCC) in mouse liver.
Collapse
|
47
|
Design of a Multimodal Imaging System and Its First Application to Distinguish Grey and White Matter of Brain Tissue. A Proof-of-Concept-Study. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11114777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multimodal imaging gains increasing popularity for biomedical applications. This article presents the design of a novel multimodal imaging system. The centerpiece is a light microscope operating in the incident and transmitted light mode. Additionally, Raman spectroscopy and VIS/NIR reflectance spectroscopy are adapted. The proof-of-concept is realized to distinguish between grey matter (GM) and white matter (WM) of normal mouse brain tissue. Besides Raman and VIS/NIR spectroscopy, the following optical microscopy techniques are applied in the incident light mode: brightfield, darkfield, and polarization microscopy. To complement the study, brightfield images of a hematoxylin and eosin (H&E) stained cryosection in the transmitted light mode are recorded using the same imaging system. Data acquisition based on polarization microscopy and Raman spectroscopy gives the best results regarding the tissue differentiation of the unstained section. In addition to the discrimination of GM and WM, both modalities are suited to highlight differences in the density of myelinated axons. For Raman spectroscopy, this is achieved by calculating the sum of two intensity peak ratios (I2857 + I2888)/I2930 in the high-wavenumber region. For an optimum combination of the modalities, it is recommended to apply the molecule-specific but time-consuming Raman spectroscopy to smaller regions of interest, which have previously been identified by the microscopic modes.
Collapse
|