1
|
Tang W, Li Z, Zou Y, Liao J, Li B. A multimodal pipeline for image correction and registration of mass spectrometry imaging with microscopy. Anal Chim Acta 2023; 1283:341969. [PMID: 37977791 DOI: 10.1016/j.aca.2023.341969] [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: 05/28/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023]
Abstract
The integration of matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) and histology plays a pivotal role in advancing our understanding of complex heterogeneous tissues, which provides a comprehensive description of biological tissue with both wide molecule coverage and high lateral resolution. Herein, we proposed a novel strategy for the correction and registration of MALDI MSI data with hematoxylin & eosin (H&E) staining images. To overcome the challenges of discrepancies in spatial resolution towards the unification of the two imaging modalities, a deep learning-based interpolation algorithm for MALDI MSI data was constructed, which enables spatial coherence and the following orientation matching between images. Coupled with the affine transformation (AT) and the subsequent moving least squares algorithm, the two types of images from one rat brain tissue section were aligned automatically with high accuracy. Moreover, we demonstrated the practicality of the developed pipeline by projecting it to a rat cerebral ischemia-reperfusion injury model, which would help decipher the link between molecular metabolism and pathological interpretation towards microregion. This new approach offers the chance for other types of bioimaging to boost the field of multimodal image fusion.
Collapse
Affiliation(s)
- Weiwei Tang
- State Key Laboratory of Natural Medicines and School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhen Li
- School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuchen Zou
- State Key Laboratory of Natural Medicines and School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jun Liao
- School of Science, China Pharmaceutical University, Nanjing, 211198, China.
| | - Bin Li
- State Key Laboratory of Natural Medicines and School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
2
|
Akbari B, Huber BR, Sherman JH. Unlocking the Hidden Depths: Multi-Modal Integration of Imaging Mass Spectrometry-Based and Molecular Imaging Techniques. Crit Rev Anal Chem 2023; 55:109-138. [PMID: 37847593 DOI: 10.1080/10408347.2023.2266838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Multimodal imaging (MMI) has emerged as a powerful tool in clinical research, combining different imaging modes to acquire comprehensive information and enabling scientists and surgeons to study tissue identification, localization, metabolic activity, and molecular discovery, thus aiding in disease progression analysis. While multimodal instruments are gaining popularity, challenges such as non-standardized characteristics, custom software, inadequate commercial support, and integration issues with other instruments need to be addressed. The field of multimodal imaging or multiplexed imaging allows for simultaneous signal reproduction from multiple imaging strategies. Intraoperatively, MMI can be integrated into frameless stereotactic surgery. Recent developments in medical imaging modalities such as magnetic resonance imaging (MRI), and Positron Emission Topography (PET) have brought new perspectives to multimodal imaging, enabling early cancer detection, molecular tracking, and real-time progression monitoring. Despite the evidence supporting the role of MMI in surgical decision-making, there is a need for comprehensive studies to validate and perform integration at the intersection of multiple imaging technologies. They were integrating mass spectrometry-based technologies (e.g., imaging mass spectrometry (IMS), imaging mass cytometry (IMC), and Ion mobility mass spectrometry ((IM-IM) with medical imaging modalities, offering promising avenues for molecular discovery and clinical applications. This review emphasizes the potential of multi-omics approaches in tissue mapping using MMI integrated into desorption electrospray ionization (DESI) and matrix-assisted laser desorption ionization (MALDI), allowing for sequential analyses of the same section. By addressing existing knowledge gaps, this review encourages future research endeavors toward multi-omics approaches, providing a roadmap for future research and enhancing the value of MMI in molecular pathology for diagnosis.
Collapse
Affiliation(s)
- Behnaz Akbari
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| | - Bertrand Russell Huber
- Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
- US Department of Veteran Affairs, VA Boston Healthcare System, Boston, Massachusetts USA
- US Department of Veterans Affairs, National Center for PTSD, Boston, Massachusetts USA
| | - Janet Hope Sherman
- Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Sadeghi M, Ramos-Prats A, Neto P, Castaldi F, Crowley D, Matulewicz P, Paradiso E, Freysinger W, Ferraguti F, Goebel G. Localization and Registration of 2D Histological Mouse Brain Images in 3D Atlas Space. Neuroinformatics 2023; 21:615-630. [PMID: 37357231 PMCID: PMC10406728 DOI: 10.1007/s12021-023-09632-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 06/27/2023]
Abstract
To accurately explore the anatomical organization of neural circuits in the brain, it is crucial to map the experimental brain data onto a standardized system of coordinates. Studying 2D histological mouse brain slices remains the standard procedure in many laboratories. Mapping these 2D brain slices is challenging; due to deformations, artifacts, and tilted angles introduced during the standard preparation and slicing process. In addition, analysis of experimental mouse brain slices can be highly dependent on the level of expertise of the human operator. Here we propose a computational tool for Accurate Mouse Brain Image Analysis (AMBIA), to map 2D mouse brain slices on the 3D brain model with minimal human intervention. AMBIA has a modular design that comprises a localization module and a registration module. The localization module is a deep learning-based pipeline that localizes a single 2D slice in the 3D Allen Brain Atlas and generates a corresponding atlas plane. The registration module is built upon the Ardent python package that performs deformable 2D registration between the brain slice to its corresponding atlas. By comparing AMBIA's performance in localization and registration to human ratings, we demonstrate that it performs at a human expert level. AMBIA provides an intuitive and highly efficient way for accurate registration of experimental 2D mouse brain images to 3D digital mouse brain atlas. Our tool provides a graphical user interface and it is designed to be used by researchers with minimal programming knowledge.
Collapse
Affiliation(s)
- Maryam Sadeghi
- Department of Medical Statistics and Informatics, Medical University of Innsbruck, Innsbruck, Austria.
| | - Arnau Ramos-Prats
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Pedro Neto
- Faculty of Engineering, University of Porto, Porto, Portugal
| | - Federico Castaldi
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Devin Crowley
- Biomedical Engineering, Johns Hopkins University, Baltimore, United States
| | - Pawel Matulewicz
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Enrica Paradiso
- KNAW, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | | | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Goebel
- Department of Medical Statistics and Informatics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
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: 22] [Impact Index Per Article: 7.3] [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
|
5
|
Grady FS, Graff SA, Aldridge GM, Geerling JC. BoutonNet: an automatic method to detect anterogradely labeled presynaptic boutons in brain tissue sections. Brain Struct Funct 2022; 227:1921-1932. [PMID: 35648216 PMCID: PMC10597056 DOI: 10.1007/s00429-022-02504-y] [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: 02/06/2022] [Accepted: 04/22/2022] [Indexed: 11/29/2022]
Abstract
Neurons emit axons, which form synapses, the fundamental unit of the nervous system. Neuroscientists use genetic anterograde tracing methods to label the synaptic output of specific neuronal subpopulations, but the resulting data sets are too large for manual analysis, and current automated methods have significant limitations in cost and quality. In this paper, we describe a pipeline optimized to identify anterogradely labeled presynaptic boutons in brain tissue sections. Our histologic pipeline labels boutons with high sensitivity and low background. To automatically detect labeled boutons in slide-scanned tissue sections, we developed BoutonNet. This detector uses a two-step approach: an intensity-based method proposes possible boutons, which are checked by a neural network-based confirmation step. BoutonNet was compared to expert annotation on a separate validation data set and achieved a result within human inter-rater variance. This open-source technique will allow quantitative analysis of the fundamental unit of the brain on a whole-brain scale.
Collapse
Affiliation(s)
- Fillan S Grady
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, PBDB 1320, 169 Newton Rd, Iowa City, IA, 52246, USA
| | - Shantelle A Graff
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, PBDB 1320, 169 Newton Rd, Iowa City, IA, 52246, USA
| | - Georgina M Aldridge
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, PBDB 1320, 169 Newton Rd, Iowa City, IA, 52246, USA
| | - Joel C Geerling
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, PBDB 1320, 169 Newton Rd, Iowa City, IA, 52246, USA.
| |
Collapse
|
6
|
Noun M, Akoumeh R, Abbas I. Cell and Tissue Imaging by TOF-SIMS and MALDI-TOF: An Overview for Biological and Pharmaceutical Analysis. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2022; 28:1-26. [PMID: 34809729 DOI: 10.1017/s1431927621013593] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The potential of mass spectrometry imaging (MSI) has been demonstrated in cell and tissue research since 1970. MSI can reveal the spatial distribution of a wide range of atomic and molecular ions detected from biological sample surfaces, it is a powerful and valuable technique used to monitor and detect diverse chemical and biological compounds, such as drugs, lipids, proteins, and DNA. MSI techniques, notably matrix-assisted laser desorption/ionization time of flight (MALDI-TOF) and time of flight secondary ion mass spectrometry (TOF-SIMS), witnessed a dramatic upsurge in studying and investigating biological samples especially, cells and tissue sections. This advancement is attributed to the submicron lateral resolution, the high sensitivity, the good precision, and the accurate chemical specificity, which make these techniques suitable for decoding and understanding complex mechanisms of certain diseases, as well as monitoring the spatial distribution of specific elements, and compounds. While the application of both techniques for the analysis of cells and tissues is thoroughly discussed, a briefing of MALDI-TOF and TOF-SIMS basis and the adequate sampling before analysis are briefly covered. The importance of MALDI-TOF and TOF-SIMS as diagnostic tools and robust analytical techniques in the medicinal, pharmaceutical, and toxicology fields is highlighted through representative published studies.
Collapse
Affiliation(s)
- Manale Noun
- Lebanese Atomic Energy Commission - NCSR, Beirut, Lebanon
| | - Rayane Akoumeh
- Lebanese Atomic Energy Commission - NCSR, Beirut, Lebanon
| | - Imane Abbas
- Lebanese Atomic Energy Commission - NCSR, Beirut, Lebanon
| |
Collapse
|
7
|
Balluff B, Heeren RM, Race AM. An overview of image registration for aligning mass spectrometry imaging with clinically relevant imaging modalities. J Mass Spectrom Adv Clin Lab 2022; 23:26-38. [PMID: 35156074 PMCID: PMC8821033 DOI: 10.1016/j.jmsacl.2021.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 01/25/2023] Open
Abstract
Mass spectrometry imaging (MSI) is a powerful molecular imaging technique. Integration with other imaging modalities is essential in clinical MSI. Image integration is performed by image registration techniques. Technical potential of image registration in MSI has not been fully exploited. Roadmap proposed to improve registration accuracy.
Mass spectrometry imaging (MSI) is used in many aspects of clinical research, including pharmacokinetics, toxicology, personalised medicine, and surgical decision-making. Maximising its potential requires the spatial integration of MSI images with imaging data from existing clinical imaging modalities, such as histology and MRI. To ensure that the information is properly integrated, all contributing images must be accurately aligned. This process is called image registration and is the focus of this review. In light of the ever-increasing spatial resolution of MSI instrumentation and a diversification of multi-modal MSI studies (e.g., spatial omics, 3D-MSI), the accuracy, versatility, and precision of image registration must increase accordingly. We review the application of image registration to align MSI data with different clinically relevant ex vivo and in vivo imaging techniques. Based on this, we identify steps in the current image registration processes where there is potential for improvement. Finally, we propose a roadmap for community efforts to address these challenges in order to increase registration quality and help MSI to fully exploit its multi-modal potential.
Collapse
|
8
|
Tian S, Hou Z, Zuo X, Xiong W, Huang G. Automatic Registration of the Mass Spectrometry Imaging Data of Sagittal Brain Slices to the Reference Atlas. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:1789-1797. [PMID: 34096712 DOI: 10.1021/jasms.1c00137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The registration of the mass spectrometry imaging (MSI) data with mouse brain tissue slices from the atlases could perform automatic anatomical interpretation, and the comparison of MSI data in particular brain regions from different mice could be accelerated. However, the current registration of MSI data with mouse brain tissue slices is mainly focused on the coronal. Although the sagittal plane is able to provide more information about brain regions on a single histological slice than the coronal, it is difficult to directly register the complete sagittal brain slices of a mouse as a result of the more significant individualized differences and more positional shifts of brain regions. Herein, by adding the auxiliary line on the two brain regions of central canal (CC) and cerebral peduncle (CP), the registration accuracy of the MSI data with sagittal brain slices has been improved (∼2-5-folds for different brain regions). Moreover, the histological sections with different degrees deformation and different dyeing effects have been used to verify that this pipeline has a certain universality. Our method facilitates the rapid comparison of sagittal plane MSI data from different animals and accelerates the application in the discovery of disease markers.
Collapse
Affiliation(s)
- Shuangshuang Tian
- Department of Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei Anhui 230026, P. R. China
| | - Zhuanghao Hou
- Department of Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei Anhui 230026, P. R. China
| | - Xin Zuo
- School of Life Sciences, Neurodegenerative Disorder Research Center, University of Science and Technology of China, Hefei Anhui 230026, P. R. China
| | - Wei Xiong
- School of Life Sciences, Neurodegenerative Disorder Research Center, University of Science and Technology of China, Hefei Anhui 230026, P. R. China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guangming Huang
- Department of Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei Anhui 230026, P. R. China
| |
Collapse
|
9
|
Vos DRN, Ellis SR, Balluff B, Heeren RMA. Experimental and Data Analysis Considerations for Three-Dimensional Mass Spectrometry Imaging in Biomedical Research. Mol Imaging Biol 2021; 23:149-159. [PMID: 33025328 PMCID: PMC7910367 DOI: 10.1007/s11307-020-01541-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 09/10/2020] [Indexed: 10/26/2022]
Abstract
Mass spectrometry imaging (MSI) enables the visualization of molecular distributions on complex surfaces. It has been extensively used in the field of biomedical research to investigate healthy and diseased tissues. Most of the MSI studies are conducted in a 2D fashion where only a single slice of the full sample volume is investigated. However, biological processes occur within a tissue volume and would ideally be investigated as a whole to gain a more comprehensive understanding of the spatial and molecular complexity of biological samples such as tissues and cells. Mass spectrometry imaging has therefore been expanded to the 3D realm whereby molecular distributions within a 3D sample can be visualized. The benefit of investigating volumetric data has led to a quick rise in the application of single-sample 3D-MSI investigations. Several experimental and data analysis aspects need to be considered to perform successful 3D-MSI studies. In this review, we discuss these aspects as well as ongoing developments that enable 3D-MSI to be routinely applied to multi-sample studies.
Collapse
Affiliation(s)
- D R N Vos
- The Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - S R Ellis
- The Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| | - B Balluff
- The Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - R M A Heeren
- The Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|
10
|
Zhang W, Claesen M, Moerman T, Groseclose MR, Waelkens E, De Moor B, Verbeeck N. Spatially aware clustering of ion images in mass spectrometry imaging data using deep learning. Anal Bioanal Chem 2021; 413:2803-2819. [PMID: 33646352 PMCID: PMC8007517 DOI: 10.1007/s00216-021-03179-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/25/2020] [Accepted: 01/15/2021] [Indexed: 11/12/2022]
Abstract
Computational analysis is crucial to capitalize on the wealth of spatio-molecular information generated by mass spectrometry imaging (MSI) experiments. Currently, the spatial information available in MSI data is often under-utilized, due to the challenges of in-depth spatial pattern extraction. The advent of deep learning has greatly facilitated such complex spatial analysis. In this work, we use a pre-trained neural network to extract high-level features from ion images in MSI data, and test whether this improves downstream data analysis. The resulting neural network interpretation of ion images, coined neural ion images, is used to cluster ion images based on spatial expressions. We evaluate the impact of neural ion images on two ion image clustering pipelines, namely DBSCAN clustering, combined with UMAP-based dimensionality reduction, and k-means clustering. In both pipelines, we compare regular and neural ion images from two different MSI datasets. All tested pipelines could extract underlying spatial patterns, but the neural network-based pipelines provided better assignment of ion images, with more fine-grained clusters, and greater consistency in the spatial structures assigned to individual clusters. Additionally, we introduce the relative isotope ratio metric to quantitatively evaluate clustering quality. The resulting scores show that isotopical m/z values are more often clustered together in the neural network-based pipeline, indicating improved clustering outcomes. The usefulness of neural ion images extends beyond clustering towards a generic framework to incorporate spatial information into any MSI-focused machine learning pipeline, both supervised and unsupervised.
Collapse
Affiliation(s)
- Wanqiu Zhang
- KU Leuven, Department of Electrical Engineering (ESAT), STADIUS Center for Dynamical Systems, Signal Processing and Data Analytics, Kasteelpark Arenberg 10, 3001, Leuven, Belgium.
- Aspect Analytics NV, C-mine 12, 3600, Genk, Belgium.
| | - Marc Claesen
- Aspect Analytics NV, C-mine 12, 3600, Genk, Belgium
| | | | | | - Etienne Waelkens
- KU Leuven, Department of Cellular and Molecular Medicine, Campus Gasthuisberg O&N1, Herestraat 49, Box 901, 3000, Leuven, Belgium
| | - Bart De Moor
- KU Leuven, Department of Electrical Engineering (ESAT), STADIUS Center for Dynamical Systems, Signal Processing and Data Analytics, Kasteelpark Arenberg 10, 3001, Leuven, Belgium
| | | |
Collapse
|
11
|
Race AM, Sutton D, Hamm G, Maglennon G, Morton JP, Strittmatter N, Campbell A, Sansom OJ, Wang Y, Barry ST, Takáts Z, Goodwin RJA, Bunch J. Deep Learning-Based Annotation Transfer between Molecular Imaging Modalities: An Automated Workflow for Multimodal Data Integration. Anal Chem 2021; 93:3061-3071. [PMID: 33534548 DOI: 10.1021/acs.analchem.0c02726] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
An ever-increasing array of imaging technologies are being used in the study of complex biological samples, each of which provides complementary, occasionally overlapping information at different length scales and spatial resolutions. It is important to understand the information provided by one technique in the context of the other to achieve a more holistic overview of such complex samples. One way to achieve this is to use annotations from one modality to investigate additional modalities. For microscopy-based techniques, these annotations could be manually generated using digital pathology software or automatically generated by machine learning (including deep learning) methods. Here, we present a generic method for using annotations from one microscopy modality to extract information from complementary modalities. We also present a fast, general, multimodal registration workflow [evaluated on multiple mass spectrometry imaging (MSI) modalities, matrix-assisted laser desorption/ionization, desorption electrospray ionization, and rapid evaporative ionization mass spectrometry] for automatic alignment of complex data sets, demonstrating an order of magnitude speed-up compared to previously published work. To demonstrate the power of the annotation transfer and multimodal registration workflows, we combine MSI, histological staining (such as hematoxylin and eosin), and deep learning (automatic annotation of histology images) to investigate a pancreatic cancer mouse model. Neoplastic pancreatic tissue regions, which were histologically indistinguishable from one another, were observed to be metabolically different. We demonstrate the use of the proposed methods to better understand tumor heterogeneity and the tumor microenvironment by transferring machine learning results freely between the two modalities.
Collapse
Affiliation(s)
- Alan M Race
- Imaging and AI, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Daniel Sutton
- Imaging and AI, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Gregory Hamm
- Imaging and AI, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Gareth Maglennon
- Oncology Safety, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Jennifer P Morton
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, U.K
| | - Nicole Strittmatter
- Imaging and AI, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Andrew Campbell
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, U.K
| | - Yinhai Wang
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Zoltan Takáts
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, U.K
| | - Richard J A Goodwin
- Imaging and AI, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Josephine Bunch
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, U.K
- National Centre of Excellence in Mass Spectrometry Imaging (NiCE-MSI), National Physical Laboratory, Teddington TW11 0LW, U.K
| |
Collapse
|
12
|
Ščupáková K, Dewez F, Walch AK, Heeren RMA, Balluff B. Morphometric Cell Classification for Single-Cell MALDI-Mass Spectrometry Imaging. Angew Chem Int Ed Engl 2020; 59:17447-17450. [PMID: 32668069 PMCID: PMC7540554 DOI: 10.1002/anie.202007315] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/04/2020] [Indexed: 12/14/2022]
Abstract
The large-scale and label-free molecular characterization of single cells in their natural tissue habitat remains a major challenge in molecular biology. We present a method that integrates morphometric image analysis to delineate and classify individual cells with their single-cell-specific molecular profiles. This approach provides a new means to study spatial biological processes such as cancer field effects and the relationship between morphometric and molecular features.
Collapse
Affiliation(s)
- Klára Ščupáková
- Maastricht MultiModal Molecular Imaging Institute (M4I)University of MaastrichtUniversiteitssingel 506200 MDMaastrichtThe Netherlands
| | - Frédéric Dewez
- Maastricht MultiModal Molecular Imaging Institute (M4I)University of MaastrichtUniversiteitssingel 506200 MDMaastrichtThe Netherlands
- Mass Spectrometry Laboratory (MSLab)University of LiègeBelgium
| | - Axel K. Walch
- Research Unit Analytical PathologyHelmholtz Zentrum MünchenOberschleißheimGermany
| | - Ron M. A. Heeren
- Maastricht MultiModal Molecular Imaging Institute (M4I)University of MaastrichtUniversiteitssingel 506200 MDMaastrichtThe Netherlands
| | - Benjamin Balluff
- Maastricht MultiModal Molecular Imaging Institute (M4I)University of MaastrichtUniversiteitssingel 506200 MDMaastrichtThe Netherlands
| |
Collapse
|
13
|
Ščupáková K, Dewez F, Walch AK, Heeren RMA, Balluff B. Morphometric Cell Classification for Single‐Cell MALDI‐Mass Spectrometry Imaging. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202007315] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Klára Ščupáková
- Maastricht MultiModal Molecular Imaging Institute (M4I) University of Maastricht Universiteitssingel 50 6200 MD Maastricht The Netherlands
| | - Frédéric Dewez
- Maastricht MultiModal Molecular Imaging Institute (M4I) University of Maastricht Universiteitssingel 50 6200 MD Maastricht The Netherlands
- Mass Spectrometry Laboratory (MSLab) University of Liège Belgium
| | - Axel K. Walch
- Research Unit Analytical Pathology Helmholtz Zentrum München Oberschleißheim Germany
| | - Ron M. A. Heeren
- Maastricht MultiModal Molecular Imaging Institute (M4I) University of Maastricht Universiteitssingel 50 6200 MD Maastricht The Netherlands
| | - Benjamin Balluff
- Maastricht MultiModal Molecular Imaging Institute (M4I) University of Maastricht Universiteitssingel 50 6200 MD Maastricht The Netherlands
| |
Collapse
|
14
|
Ščupáková K, Balluff B, Tressler C, Adelaja T, Heeren RM, Glunde K, Ertaylan G. Cellular resolution in clinical MALDI mass spectrometry imaging: the latest advancements and current challenges. Clin Chem Lab Med 2020; 58:914-929. [PMID: 31665113 PMCID: PMC9867918 DOI: 10.1515/cclm-2019-0858] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023]
Abstract
Mass spectrometry (MS) is the workhorse of metabolomics, proteomics and lipidomics. Mass spectrometry imaging (MSI), its extension to spatially resolved analysis of tissues, is a powerful tool for visualizing molecular information within the histological context of tissue. This review summarizes recent developments in MSI and highlights current challenges that remain to achieve molecular imaging at the cellular level of clinical specimens. We focus on matrix-assisted laser desorption/ionization (MALDI)-MSI. We discuss the current status of each of the analysis steps and remaining challenges to reach the desired level of cellular imaging. Currently, analyte delocalization and degradation, matrix crystal size, laser focus restrictions and detector sensitivity are factors that are limiting spatial resolution. New sample preparation devices and laser optic systems are being developed to push the boundaries of these limitations. Furthermore, we review the processing of cellular MSI data and images, and the systematic integration of these data in the light of available algorithms and databases. We discuss roadblocks in the data analysis pipeline and show how technology from other fields can be used to overcome these. Finally, we conclude with curative and community efforts that are needed to enable contextualization of the information obtained.
Collapse
Affiliation(s)
- Klára Ščupáková
- Maastricht MultiModal Molecular Imaging Institute (M4I), University of Maastricht, Maastricht, The Netherlands
| | - Benjamin Balluff
- Maastricht MultiModal Molecular Imaging Institute (M4I), University of Maastricht, Maastricht, The Netherlands
| | - Caitlin Tressler
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tobi Adelaja
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ron M.A. Heeren
- Corresponding author: Ron M.A. Heeren, Maastricht MultiModal Molecular Imaging Institute (M4I), University of Maastricht, Maastricht, The Netherlands,
| | - Kristine Glunde
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; and The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gökhan Ertaylan
- Unit Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
| |
Collapse
|
15
|
Verbeeck N, Caprioli RM, Van de Plas R. Unsupervised machine learning for exploratory data analysis in imaging mass spectrometry. MASS SPECTROMETRY REVIEWS 2020; 39:245-291. [PMID: 31602691 PMCID: PMC7187435 DOI: 10.1002/mas.21602] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/27/2018] [Indexed: 05/20/2023]
Abstract
Imaging mass spectrometry (IMS) is a rapidly advancing molecular imaging modality that can map the spatial distribution of molecules with high chemical specificity. IMS does not require prior tagging of molecular targets and is able to measure a large number of ions concurrently in a single experiment. While this makes it particularly suited for exploratory analysis, the large amount and high-dimensional nature of data generated by IMS techniques make automated computational analysis indispensable. Research into computational methods for IMS data has touched upon different aspects, including spectral preprocessing, data formats, dimensionality reduction, spatial registration, sample classification, differential analysis between IMS experiments, and data-driven fusion methods to extract patterns corroborated by both IMS and other imaging modalities. In this work, we review unsupervised machine learning methods for exploratory analysis of IMS data, with particular focus on (a) factorization, (b) clustering, and (c) manifold learning. To provide a view across the various IMS modalities, we have attempted to include examples from a range of approaches including matrix assisted laser desorption/ionization, desorption electrospray ionization, and secondary ion mass spectrometry-based IMS. This review aims to be an entry point for both (i) analytical chemists and mass spectrometry experts who want to explore computational techniques; and (ii) computer scientists and data mining specialists who want to enter the IMS field. © 2019 The Authors. Mass Spectrometry Reviews published by Wiley Periodicals, Inc. Mass SpecRev 00:1-47, 2019.
Collapse
Affiliation(s)
- Nico Verbeeck
- Delft Center for Systems and ControlDelft University of Technology ‐ TU DelftDelftThe Netherlands
- Aspect Analytics NVGenkBelgium
- STADIUS Center for Dynamical Systems, Signal Processing, and Data Analytics, Department of Electrical Engineering (ESAT)KU LeuvenLeuvenBelgium
| | - Richard M. Caprioli
- Mass Spectrometry Research CenterVanderbilt UniversityNashvilleTN
- Department of BiochemistryVanderbilt UniversityNashvilleTN
- Department of ChemistryVanderbilt UniversityNashvilleTN
- Department of PharmacologyVanderbilt UniversityNashvilleTN
- Department of MedicineVanderbilt UniversityNashvilleTN
| | - Raf Van de Plas
- Delft Center for Systems and ControlDelft University of Technology ‐ TU DelftDelftThe Netherlands
- Mass Spectrometry Research CenterVanderbilt UniversityNashvilleTN
- Department of BiochemistryVanderbilt UniversityNashvilleTN
| |
Collapse
|
16
|
|
17
|
Liang H, Dabrowska N, Kapur J, Weller DS. Structure-Based Intensity Propagation for 3-D Brain Reconstruction With Multilayer Section Microscopy. IEEE TRANSACTIONS ON MEDICAL IMAGING 2019; 38:1106-1115. [PMID: 30371359 PMCID: PMC6488466 DOI: 10.1109/tmi.2018.2878488] [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] [Indexed: 06/08/2023]
Abstract
Microscopy is widely used for brain research because of its high resolution and ability to stain for many different biomarkers. Since whole brains are usually sectioned for tissue staining and imaging, reconstruction of 3D brain volumes from these sections is important for visualization and analysis. Recently developed tissue clearing techniques and advanced confocal microscopy enable multilayer sections to be imaged without compromising the resolution. However, noticeable structure inconsistence occurs if surface layers are used to align these sections. In this paper, a structure-based intensity propagation method is designed for the robust representation of multilayer sections. The 3D structures in reconstructed brains are more consistent using the proposed methods. Experiments are conducted on 367 multilayer sections from 20 mouse brains. The average reconstruction quality measured by the structure consistence index increases by 45% with the tissue flattening method and 29% further with the structure-based intensity propagation.
Collapse
|
18
|
Abdelmoula WM, Regan MS, Lopez BGC, Randall EC, Lawler S, Mladek AC, Nowicki MO, Marin BM, Agar JN, Swanson KR, Kapur T, Sarkaria JN, Wells W, Agar NYR. Automatic 3D Nonlinear Registration of Mass Spectrometry Imaging and Magnetic Resonance Imaging Data. Anal Chem 2019; 91:6206-6216. [PMID: 30932478 DOI: 10.1021/acs.analchem.9b00854] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Multimodal integration between mass spectrometry imaging (MSI) and radiology-established modalities such as magnetic resonance imaging (MRI) would allow the investigations of key questions in complex biological systems such as the central nervous system. Such integration would provide complementary multiscale data to bridge the gap between molecular and anatomical phenotypes, potentially revealing new insights into molecular mechanisms underlying anatomical pathologies presented on MRI. Automatic coregistration between 3D MSI/MRI is a computationally challenging process due to dimensional complexity, MSI data sparsity, lack of direct spatial-correspondences, and nonlinear tissue deformation. Here, we present a new computational approach based on stochastic neighbor embedding to nonlinearly align 3D MSI to MRI data, identify and reconstruct biologically relevant molecular patterns in 3D, and fuse the MSI datacube to the MRI space. We demonstrate our method using multimodal high-spectral resolution matrix-assisted laser desorption ionization (MALDI) 9.4 T MSI and 7 T in vivo MRI data, acquired from a patient-derived, xenograft mouse brain model of glioblastoma following administration of the EGFR inhibitor drug of Erlotinib. Results show the distribution of some identified molecular ions of the EGFR inhibitor erlotinib, a phosphatidylcholine lipid, and cholesterol, which were reconstructed in 3D and mapped to the MRI space. The registration quality was evaluated on two normal mouse brains using the Dice coefficient for the regions of brainstem, hippocampus, and cortex. The method is generic and can therefore be applied to hyperspectral images from different mass spectrometers and integrated with other established in vivo imaging modalities such as computed tomography (CT) and positron emission tomography (PET).
Collapse
Affiliation(s)
- Walid M Abdelmoula
- Department of Neurosurgery, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Michael S Regan
- Department of Neurosurgery, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Begona G C Lopez
- Department of Neurosurgery, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Elizabeth C Randall
- Department of Radiology, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Sean Lawler
- Department of Neurosurgery, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Ann C Mladek
- Department of Radiation Oncology , Mayo Clinic , 200 First Street SW , Rochester , Minnesota 55902 , United States
| | - Michal O Nowicki
- Department of Neurosurgery, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Bianca M Marin
- Department of Radiation Oncology , Mayo Clinic , 200 First Street SW , Rochester , Minnesota 55902 , United States
| | - Jeffrey N Agar
- Department of Chemistry and Chemical Biology , Northeastern University , 412 TF (140 The Fenway) , Boston , Massachusetts 02111 , United States
| | - Kristin R Swanson
- Mathematical NeuroOncology Lab, Department of Neurosurgery , Mayo Clinic , 5777 East Mayo Boulevard , Phoenix , Arizona 85054 , United States
| | - Tina Kapur
- Department of Radiology, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Jann N Sarkaria
- Department of Radiation Oncology , Mayo Clinic , 200 First Street SW , Rochester , Minnesota 55902 , United States
| | - William Wells
- Department of Radiology, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States.,Computer Science and Artificial Intelligence Laboratory , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States.,Department of Radiology, Brigham and Women's Hospital , Harvard Medical School , Boston , Massachusetts 02115 , United States.,Department of Cancer Biology, Dana-Farber Cancer Institute , Harvard Medical School , Boston , Massachusetts 02115 , United States
| |
Collapse
|
19
|
Liao Y, Fu X, Zhou H, Rao W, Zeng L, Yang Z. Visualized analysis of within-tissue spatial distribution of specialized metabolites in tea (Camellia sinensis) using desorption electrospray ionization imaging mass spectrometry. Food Chem 2019; 292:204-210. [PMID: 31054666 DOI: 10.1016/j.foodchem.2019.04.055] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022]
Abstract
Although specialized metabolite distributions in different tea (Camellia sinensis) tissues has been studied extensively, little is known about their within-tissue distribution owing to the lack of nondestructive methodology. In this study, desorption electrospray ionization imaging mass spectrometry was used to investigate the within-tissue spatial distributions of specialized metabolites in tea. To overcome the negative effects of the large amount of wax on tea leaves, several sample preparation methods were compared, with a Teflon-imprint method established for tea leaves. Polyphenols are characteristic metabolites in tea leaves. Epicatechin gallate/catechin gallate, epigallocatechin gallate/gallocatechin gallate, and gallic acid were evenly distributed on both sides of the leaves, while epicatechin/catechin, epigallocatechin/gallocatechin, and assamicain A were distributed near the leaf vein. L-Theanine was mainly accumulated in tea roots. L-Theanine and valinol were distributed around the outer root cross-section. The results will advance our understanding of the precise localizations and in-vivo biosyntheses of specialized metabolites in tea.
Collapse
Affiliation(s)
- Yinyin Liao
- Guangdong Provincial Key Laboratory of Applied Botany & Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Xingke Road 723, Tianhe District, Guangzhou 510650, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiumin Fu
- Guangdong Provincial Key Laboratory of Applied Botany & Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Xingke Road 723, Tianhe District, Guangzhou 510650, China
| | - Haiyun Zhou
- Instrumental Analysis & Research Center, Sun Yat-Sen University, No. 135, Xingang Xi Road, Guangzhou 510275, China
| | - Wei Rao
- Waters Technologies (Shanghai) Ltd., No. 1000 Jinhai Road, Shanghai 201203, China
| | - Lanting Zeng
- Guangdong Provincial Key Laboratory of Applied Botany & Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Xingke Road 723, Tianhe District, Guangzhou 510650, China
| | - Ziyin Yang
- Guangdong Provincial Key Laboratory of Applied Botany & Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Xingke Road 723, Tianhe District, Guangzhou 510650, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China.
| |
Collapse
|
20
|
Mass Spectrometry Imaging and Integration with Other Imaging Modalities for Greater Molecular Understanding of Biological Tissues. Mol Imaging Biol 2019; 20:888-901. [PMID: 30167993 PMCID: PMC6244545 DOI: 10.1007/s11307-018-1267-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Over the last two decades, mass spectrometry imaging (MSI) has been increasingly employed to investigate the spatial distribution of a wide variety of molecules in complex biological samples. MSI has demonstrated its potential in numerous applications from drug discovery, disease state evaluation through proteomic and/or metabolomic studies. Significant technological and methodological advancements have addressed natural limitations of the techniques, i.e., increased spatial resolution, increased detection sensitivity especially for large molecules, higher throughput analysis and data management. One of the next major evolutions of MSI is linked to the introduction of imaging mass cytometry (IMC). IMC is a multiplexed method for tissue phenotyping, imaging signalling pathway or cell marker assessment, at sub-cellular resolution (1 μm). It uses MSI to simultaneously detect and quantify up to 30 different antibodies within a tissue section. The combination of MSI with other molecular imaging techniques can also provide highly relevant complementary information to explore new scientific fields. Traditionally, classical histology (especially haematoxylin and eosin–stained sections) is overlaid with molecular profiles obtained by MSI. Thus, MSI-based molecular histology provides a snapshot of a tissue microenvironment and enables the correlation of drugs, metabolites, lipids, peptides or proteins with histological/pathological features or tissue substructures. Recently, many examples combining MSI with other imaging modalities such as fluorescence, confocal Raman spectroscopy and MRI have emerged. For instance, brain pathophysiology has been studied using both MRI and MSI, establishing correlations between in and ex vivo molecular imaging techniques. Endogenous metabolite and small peptide modulation were evaluated depending on disease state. Here, we review advanced ‘hot topics’ in MSI development and explore the combination of MSI with established molecular imaging techniques to improve our understanding of biological and pathophysiological processes.
Collapse
|
21
|
Han J, Permentier H, Bischoff R, Groothuis G, Casini A, Horvatovich P. Imaging of protein distribution in tissues using mass spectrometry: An interdisciplinary challenge. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2018.12.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
22
|
Xiong J, Ren J, Luo L, Horowitz M. Mapping Histological Slice Sequences to the Allen Mouse Brain Atlas Without 3D Reconstruction. Front Neuroinform 2018; 12:93. [PMID: 30618698 PMCID: PMC6297281 DOI: 10.3389/fninf.2018.00093] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/22/2018] [Indexed: 11/13/2022] Open
Abstract
Histological brain slices are widely used in neuroscience to study the anatomical organization of neural circuits. Systematic and accurate comparisons of anatomical data from multiple brains, especially from different studies, can benefit tremendously from registering histological slices onto a common reference atlas. Most existing methods rely on an initial reconstruction of the volume before registering it to a reference atlas. Because these slices are prone to distortions during the sectioning process and often sectioned with non-standard angles, reconstruction is challenging and often inaccurate. Here we describe a framework that maps each slice to its corresponding plane in the Allen Mouse Brain Atlas (2015) to build a plane-wise mapping and then perform 2D nonrigid registration to build a pixel-wise mapping. We use the L2 norm of the histogram of oriented gradients difference of two patches as the similarity metric for both steps and a Markov random field formulation that incorporates tissue coherency to compute the nonrigid registration. To fix significantly distorted regions that are misshaped or much smaller than the control grids, we train a context-aggregation network to segment and warp them to their corresponding regions with thin plate spline. We have shown that our method generates results comparable to an expert neuroscientist and is significantly better than reconstruction-first approaches. Code and sample dataset are available at sites.google.com/view/brain-mapping.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Electrical Engineering, Stanford University, Stanford, CA, United States
| | - Jing Ren
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
| | - Mark Horowitz
- Department of Electrical Engineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
23
|
Patterson NH, Tuck M, Van de Plas R, Caprioli RM. Advanced Registration and Analysis of MALDI Imaging Mass Spectrometry Measurements through Autofluorescence Microscopy. Anal Chem 2018; 90:12395-12403. [PMID: 30272960 DOI: 10.1021/acs.analchem.8b02884] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The correlation of imaging mass spectrometry (IMS) with histopathology can help relate novel molecular findings obtained through IMS to the well-characterized and validated histopathology knowledge base. The quality of correlation between these two modalities is limited by the quality of the spatial mapping that is obtained by registration of the two image types. In this work, we develop novel workflows for MALDI IMS-to-microscopy data registration and analysis using nondestructive IMS-compatible wide field autofluorescence (AF) microscopy combined with computational image registration. First, a substantially automated procedure for high-accuracy registration between IMS and microscopy data of the same section is described that explicitly links the MALDI laser ablation pattern imaged by microscopy to its corresponding IMS pixel. Subsequent examination of the registered data allows for high-confidence colocalization of image features between the two modalities, down to single-cell scales within tissue. Building on this IMS-microscopy spatial mapping, we furthermore demonstrate the automated spatial correlation between IMS measurements from serial sections. This AF-registration-driven inter-section analysis, using a combination of nonlinear AF-to-AF and IMS-to-AF image registrations, can be applied to tissue sections that are prepared and imaged with different sample preparations (e.g., lipids vs proteins) and/or that are measured using different spatial resolutions. Importantly, all registrations, whether within a single section or across serial sections, are entirely independent of the IMS intensity signal content and thus unbiased by it.
Collapse
Affiliation(s)
| | | | - Raf Van de Plas
- Delft Center for Systems and Control (DCSC) , Delft University of Technology , 2628 CD Delft , The Netherlands
| | | |
Collapse
|
24
|
Integrated Brain Atlas for Unbiased Mapping of Nervous System Effects Following Liraglutide Treatment. Sci Rep 2018; 8:10310. [PMID: 29985439 PMCID: PMC6037685 DOI: 10.1038/s41598-018-28496-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023] Open
Abstract
Light Sheet Fluorescence Microscopy (LSFM) of whole organs, in particular the brain, offers a plethora of biological data imaged in 3D. This technique is however often hindered by cumbersome non-automated analysis methods. Here we describe an approach to fully automate the analysis by integrating with data from the Allen Institute of Brain Science (AIBS), to provide precise assessment of the distribution and action of peptide-based pharmaceuticals in the brain. To illustrate this approach, we examined the acute central nervous system effects of the glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide. Peripherally administered liraglutide accessed the hypothalamus and brainstem, and led to activation in several brain regions of which most were intersected by projections from neurons in the lateral parabrachial nucleus. Collectively, we provide a rapid and unbiased analytical framework for LSFM data which enables quantification and exploration based on data from AIBS to support basic and translational discovery.
Collapse
|
25
|
Rabe JH, A Sammour D, Schulz S, Munteanu B, Ott M, Ochs K, Hohenberger P, Marx A, Platten M, Opitz CA, Ory DS, Hopf C. Fourier Transform Infrared Microscopy Enables Guidance of Automated Mass Spectrometry Imaging to Predefined Tissue Morphologies. Sci Rep 2018; 8:313. [PMID: 29321555 PMCID: PMC5762902 DOI: 10.1038/s41598-017-18477-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/12/2017] [Indexed: 12/27/2022] Open
Abstract
Multimodal imaging combines complementary platforms for spatially resolved tissue analysis that are poised for application in life science and personalized medicine. Unlike established clinical in vivo multimodality imaging, automated workflows for in-depth multimodal molecular ex vivo tissue analysis that combine the speed and ease of spectroscopic imaging with molecular details provided by mass spectrometry imaging (MSI) are lagging behind. Here, we present an integrated approach that utilizes non-destructive Fourier transform infrared (FTIR) microscopy and matrix assisted laser desorption/ionization (MALDI) MSI for analysing single-slide tissue specimen. We show that FTIR microscopy can automatically guide high-resolution MSI data acquisition and interpretation without requiring prior histopathological tissue annotation, thus circumventing potential human-annotation-bias while achieving >90% reductions of data load and acquisition time. We apply FTIR imaging as an upstream modality to improve accuracy of tissue-morphology detection and to retrieve diagnostic molecular signatures in an automated, unbiased and spatially aware manner. We show the general applicability of multimodal FTIR-guided MALDI-MSI by demonstrating precise tumor localization in mouse brain bearing glioma xenografts and in human primary gastrointestinal stromal tumors. Finally, the presented multimodal tissue analysis method allows for morphology-sensitive lipid signature retrieval from brains of mice suffering from lipidosis caused by Niemann-Pick type C disease.
Collapse
Affiliation(s)
- Jan-Hinrich Rabe
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), 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
| | - Denis A Sammour
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), 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
| | - Sandra Schulz
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), 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
| | - Bogdan Munteanu
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), 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
| | - Martina Ott
- German Cancer Consortium (DKTK) CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katharina Ochs
- German Cancer Consortium (DKTK) CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and National Center of Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Hohenberger
- University Medical Center Mannheim of Heidelberg University, Mannheim, Germany
| | - Alexander Marx
- University Medical Center Mannheim of Heidelberg University, Mannheim, Germany
| | - Michael Platten
- German Cancer Consortium (DKTK) CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Medical Center Mannheim of Heidelberg University, Mannheim, Germany
| | - Christiane A Opitz
- Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and National Center of Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center and Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, 63110, USA
| | - Carsten Hopf
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), 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.
| |
Collapse
|
26
|
Rae Buchberger A, DeLaney K, Johnson J, Li L. Mass Spectrometry Imaging: A Review of Emerging Advancements and Future Insights. Anal Chem 2018; 90:240-265. [PMID: 29155564 PMCID: PMC5959842 DOI: 10.1021/acs.analchem.7b04733] [Citation(s) in RCA: 601] [Impact Index Per Article: 85.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Amanda Rae Buchberger
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Kellen DeLaney
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Jillian Johnson
- School of Pharmacy, University of Wisconsin-Madison, 777 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
| |
Collapse
|
27
|
Bulk M, Abdelmoula WM, Nabuurs RJA, van der Graaf LM, Mulders CWH, Mulder AA, Jost CR, Koster AJ, van Buchem MA, Natté R, Dijkstra J, van der Weerd L. Postmortem MRI and histology demonstrate differential iron accumulation and cortical myelin organization in early- and late-onset Alzheimer's disease. Neurobiol Aging 2017; 62:231-242. [PMID: 29195086 DOI: 10.1016/j.neurobiolaging.2017.10.017] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 10/18/2017] [Accepted: 10/18/2017] [Indexed: 11/15/2022]
Abstract
Previous MRI studies reported cortical iron accumulation in early-onset (EOAD) compared to late-onset (LOAD) Alzheimer disease patients. However, the pattern and origin of iron accumulation is poorly understood. This study investigated the histopathological correlates of MRI contrast in both EOAD and LOAD. T2*-weighted MRI was performed on postmortem frontal cortex of controls, EOAD, and LOAD. Images were ordinally scored using predefined criteria followed by histology. Nonlinear histology-MRI registration was used to calculate pixel-wise spatial correlations based on the signal intensity. EOAD and LOAD were distinguishable based on 7T MRI from controls and from each other. Histology-MRI correlation analysis of the pixel intensities showed that the MRI contrast is best explained by increased iron accumulation and changes in cortical myelin, whereas amyloid and tau showed less spatial correspondence with T2*-weighted MRI. Neuropathologically, subtypes of Alzheimer's disease showed different patterns of iron accumulation and cortical myelin changes independent of amyloid and tau that may be detected by high-field susceptibility-based MRI.
Collapse
Affiliation(s)
- Marjolein Bulk
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Percuros BV, Leiden, the Netherlands.
| | - Walid M Abdelmoula
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rob J A Nabuurs
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Linda M van der Graaf
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Coen W H Mulders
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Aat A Mulder
- Department of Molecular Cell Biology, Electron Microscopy Section, Leiden University Medical Center, Leiden, the Netherlands
| | - Carolina R Jost
- Department of Molecular Cell Biology, Electron Microscopy Section, Leiden University Medical Center, Leiden, the Netherlands
| | - Abraham J Koster
- Department of Molecular Cell Biology, Electron Microscopy Section, Leiden University Medical Center, Leiden, the Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Remco Natté
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jouke Dijkstra
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
28
|
Dexter A, Race AM, Steven RT, Barnes JR, Hulme H, Goodwin RJA, Styles IB, Bunch J. Two-Phase and Graph-Based Clustering Methods for Accurate and Efficient Segmentation of Large Mass Spectrometry Images. Anal Chem 2017; 89:11293-11300. [DOI: 10.1021/acs.analchem.7b01758] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Alex Dexter
- PSIBS
Doctoral Training Centre, University of Birmingham Edgbaston, Birmingham B15 2TT, United Kingdom
- National Physical
Laboratory, Teddington, Middlesex TW11 0LW, United Kingdom
| | - Alan M. Race
- National Physical
Laboratory, Teddington, Middlesex TW11 0LW, United Kingdom
| | - Rory T. Steven
- National Physical
Laboratory, Teddington, Middlesex TW11 0LW, United Kingdom
| | - Jennifer R. Barnes
- AstraZeneca, Drug Safety and Metabolism, Cambridge CB4 0WG, United Kingdom
| | - Heather Hulme
- AstraZeneca, Drug Safety and Metabolism, Cambridge CB4 0WG, United Kingdom
- University
of Glasgow, University Avenue, Glasgow, G12 8QQ, United Kingdom
| | | | - Iain B. Styles
- School
of Computer Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Josephine Bunch
- National Physical
Laboratory, Teddington, Middlesex TW11 0LW, United Kingdom
- School
of
Pharmacy, University of Nottingham, Nottingham, Nottinghamshire NG7 2RD, United Kingdom
| |
Collapse
|
29
|
Abstract
Magnetic resonance imaging, positron emission tomography, and optical imaging have emerged as key tools to understand brain function and neurological disorders in preclinical mouse models. They offer the unique advantage of monitoring individual structural and functional changes over time. What remained unsolved until recently was to generate whole-brain microscopy data which can be correlated to the 3D in vivo neuroimaging data. Conventional histological sections are inappropriate especially for neuronal tracing or the unbiased screening for molecular targets through the whole brain. As part of the European Society for Molecular Imaging (ESMI) meeting 2016 in Utrecht, the Netherlands, we addressed this issue in the Molecular Neuroimaging study group meeting. Presentations covered new brain clearing methods, light sheet microscopes for large samples, and automatic registration of microscopy to in vivo imaging data. In this article, we summarize the discussion; give an overview of the novel techniques; and discuss the practical needs, benefits, and limitations.
Collapse
|
30
|
Hattab G, Schlüter JP, Becker A, Nattkemper TW. ViCAR: An Adaptive and Landmark-Free Registration of Time Lapse Image Data from Microfluidics Experiments. Front Genet 2017; 8:69. [PMID: 28620411 PMCID: PMC5449445 DOI: 10.3389/fgene.2017.00069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/12/2017] [Indexed: 11/25/2022] Open
Abstract
In order to understand gene function in bacterial life cycles, time lapse bioimaging is applied in combination with different marker protocols in so called microfluidics chambers (i.e., a multi-well plate). In one experiment, a series of T images is recorded for one visual field, with a pixel resolution of 60 nm/px. Any (semi-)automatic analysis of the data is hampered by a strong image noise, low contrast and, last but not least, considerable irregular shifts during the acquisition. Image registration corrects such shifts enabling next steps of the analysis (e.g., feature extraction or tracking). Image alignment faces two obstacles in this microscopic context: (a) highly dynamic structural changes in the sample (i.e., colony growth) and (b) an individual data set-specific sample environment which makes the application of landmarks-based alignments almost impossible. We present a computational image registration solution, we refer to as ViCAR: (Vi)sual (C)ues based (A)daptive (R)egistration, for such microfluidics experiments, consisting of (1) the detection of particular polygons (outlined and segmented ones, referred to as visual cues), (2) the adaptive retrieval of three coordinates throughout different sets of frames, and finally (3) an image registration based on the relation of these points correcting both rotation and translation. We tested ViCAR with different data sets and have found that it provides an effective spatial alignment thereby paving the way to extract temporal features pertinent to each resulting bacterial colony. By using ViCAR, we achieved an image registration with 99.9% of image closeness, based on the average rmsd of 4.10−2 pixels, and superior results compared to a state of the art algorithm.
Collapse
Affiliation(s)
- Georges Hattab
- International Research Training Group 1906, Computational Methods for the Analysis of the Diversity and Dynamics of Genomes, Faculty of Technology, Bielefeld UniversityBielefeld, Germany.,Biodata Mining Group, Faculty of Technology, Center for Biotechnology, Bielefeld UniversityBielefeld, Germany
| | - Jan-Philip Schlüter
- SYNMIKRO, LOEWE-Center for Synthetic Microbiology, Philipps University of MarburgMarburg, Germany
| | - Anke Becker
- SYNMIKRO, LOEWE-Center for Synthetic Microbiology, Philipps University of MarburgMarburg, Germany
| | - Tim W Nattkemper
- Biodata Mining Group, Faculty of Technology, Center for Biotechnology, Bielefeld UniversityBielefeld, Germany
| |
Collapse
|
31
|
Mahfouz A, Huisman SMH, Lelieveldt BPF, Reinders MJT. Brain transcriptome atlases: a computational perspective. Brain Struct Funct 2017; 222:1557-1580. [PMID: 27909802 PMCID: PMC5406417 DOI: 10.1007/s00429-016-1338-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/15/2016] [Indexed: 01/31/2023]
Abstract
The immense complexity of the mammalian brain is largely reflected in the underlying molecular signatures of its billions of cells. Brain transcriptome atlases provide valuable insights into gene expression patterns across different brain areas throughout the course of development. Such atlases allow researchers to probe the molecular mechanisms which define neuronal identities, neuroanatomy, and patterns of connectivity. Despite the immense effort put into generating such atlases, to answer fundamental questions in neuroscience, an even greater effort is needed to develop methods to probe the resulting high-dimensional multivariate data. We provide a comprehensive overview of the various computational methods used to analyze brain transcriptome atlases.
Collapse
Affiliation(s)
- Ahmed Mahfouz
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
- Delft Bioinformatics Laboratory, Delft University of Technology, Delft, The Netherlands.
| | - Sjoerd M H Huisman
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Delft Bioinformatics Laboratory, Delft University of Technology, Delft, The Netherlands
| | - Boudewijn P F Lelieveldt
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Delft Bioinformatics Laboratory, Delft University of Technology, Delft, The Netherlands
| | - Marcel J T Reinders
- Delft Bioinformatics Laboratory, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
32
|
Verbeeck N, Spraggins JM, Murphy MJM, Wang HD, Deutch AY, Caprioli RM, Van de Plas R. Connecting imaging mass spectrometry and magnetic resonance imaging-based anatomical atlases for automated anatomical interpretation and differential analysis. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:967-977. [PMID: 28254588 DOI: 10.1016/j.bbapap.2017.02.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 02/13/2017] [Indexed: 12/16/2022]
Abstract
Imaging mass spectrometry (IMS) is a molecular imaging technology that can measure thousands of biomolecules concurrently without prior tagging, making it particularly suitable for exploratory research. However, the data size and dimensionality often makes thorough extraction of relevant information impractical. To help guide and accelerate IMS data analysis, we recently developed a framework that integrates IMS measurements with anatomical atlases, opening up opportunities for anatomy-driven exploration of IMS data. One example is the automated anatomical interpretation of ion images, where empirically measured ion distributions are automatically decomposed into their underlying anatomical structures. While offering significant potential, IMS-atlas integration has thus far been restricted to the Allen Mouse Brain Atlas (AMBA) and mouse brain samples. Here, we expand the applicability of this framework by extending towards new animal species and a new set of anatomical atlases retrieved from the Scalable Brain Atlas (SBA). Furthermore, as many SBA atlases are based on magnetic resonance imaging (MRI) data, a new registration pipeline was developed that enables direct non-rigid IMS-to-MRI registration. These developments are demonstrated on protein-focused FTICR IMS measurements from coronal brain sections of a Parkinson's disease (PD) rat model. The measurements are integrated with an MRI-based rat brain atlas from the SBA. The new rat-focused IMS-atlas integration is used to perform automated anatomical interpretation and to find differential ions between healthy and diseased tissue. IMS-atlas integration can serve as an important accelerator in IMS data exploration, and with these new developments it can now be applied to a wider variety of animal species and modalities. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.
Collapse
Affiliation(s)
- Nico Verbeeck
- Delft Center for Systems and Control (DCSC), Delft University of Technology, Mekelweg 2, 2628 CD Delft, the Netherlands.
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center (MSRC), Vanderbilt University, 465 21st Ave. South, 9160 Medical Research Building III, Nashville, TN 37240, USA; Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, TN 37205, USA; Department of Chemistry, Vanderbilt University, 7330 Stevenson Center Station B 351822, Nashville, TN 37235, USA.
| | - Monika J M Murphy
- Program in Neuroscience, Vanderbilt University, U-1205 Medical Research Building III, 465 21st Ave. South, Nashville, TN 37232, USA.
| | - Hui-Dong Wang
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr. Nashville, TN 37232, USA.
| | - Ariel Y Deutch
- Program in Neuroscience, Vanderbilt University, U-1205 Medical Research Building III, 465 21st Ave. South, Nashville, TN 37232, USA; Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1211 Medical Center Dr. Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, 460B Preston Research Building, Nashville, TN 37232, USA.
| | - Richard M Caprioli
- Mass Spectrometry Research Center (MSRC), Vanderbilt University, 465 21st Ave. South, 9160 Medical Research Building III, Nashville, TN 37240, USA; Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, TN 37205, USA; Department of Chemistry, Vanderbilt University, 7330 Stevenson Center Station B 351822, Nashville, TN 37235, USA; Department of Medicine, Vanderbilt University Medical Center, 1161 21st Ave. South, D-3100 Medical Center North, Nashville, TN 37232, USA.
| | - Raf Van de Plas
- Delft Center for Systems and Control (DCSC), Delft University of Technology, Mekelweg 2, 2628 CD Delft, the Netherlands; Mass Spectrometry Research Center (MSRC), Vanderbilt University, 465 21st Ave. South, 9160 Medical Research Building III, Nashville, TN 37240, USA; Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, TN 37205, USA.
| |
Collapse
|
33
|
Ucal Y, Durer ZA, Atak H, Kadioglu E, Sahin B, Coskun A, Baykal AT, Ozpinar A. Clinical applications of MALDI imaging technologies in cancer and neurodegenerative diseases. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:795-816. [PMID: 28087424 DOI: 10.1016/j.bbapap.2017.01.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 12/08/2016] [Accepted: 01/06/2017] [Indexed: 12/25/2022]
Abstract
Matrix-assisted laser desorption/ionization (MALDI) time-of-flight (TOF) imaging mass spectrometry (IMS) enables localization of analytes of interest along with histology. More specifically, MALDI-IMS identifies the distributions of proteins, peptides, small molecules, lipids, and drugs and their metabolites in tissues, with high spatial resolution. This unique capacity to directly analyze tissue samples without the need for lengthy sample preparation reduces technical variability and renders MALDI-IMS ideal for the identification of potential diagnostic and prognostic biomarkers and disease gradation. MALDI-IMS has evolved rapidly over the last decade and has been successfully used in both medical and basic research by scientists worldwide. In this review, we explore the clinical applications of MALDI-IMS, focusing on the major cancer types and neurodegenerative diseases. In particular, we re-emphasize the diagnostic potential of IMS and the challenges that must be confronted when conducting MALDI-IMS in clinical settings. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.
Collapse
Affiliation(s)
- Yasemin Ucal
- Acibadem University, Department of Medical Biochemistry, School of Medicine, Istanbul, Turkey
| | - Zeynep Aslıhan Durer
- Acibadem University, Department of Medical Biochemistry, School of Medicine, Istanbul, Turkey
| | - Hakan Atak
- Acibadem University, Department of Medical Biochemistry, School of Medicine, Istanbul, Turkey
| | - Elif Kadioglu
- Acibadem University, Department of Medical Biochemistry, School of Medicine, Istanbul, Turkey
| | - Betul Sahin
- Acibadem University, Department of Medical Biochemistry, School of Medicine, Istanbul, Turkey
| | - Abdurrahman Coskun
- Acibadem University, Department of Medical Biochemistry, School of Medicine, Istanbul, Turkey
| | - Ahmet Tarık Baykal
- Acibadem University, Department of Medical Biochemistry, School of Medicine, Istanbul, Turkey
| | - Aysel Ozpinar
- Acibadem University, Department of Medical Biochemistry, School of Medicine, Istanbul, Turkey.
| |
Collapse
|
34
|
Abstract
Over the last decade mass spectrometry imaging (MSI) has been integrated in to many areas of drug discovery and development. It can have significant impact in oncology drug discovery as it allows efficacy and safety of compounds to be assessed against the backdrop of the complex tumour microenvironment. We will discuss the roles of MSI in investigating compound and metabolite biodistribution and defining pharmacokinetic -pharmacodynamic relationships, analysis that is applicable to all drug discovery projects. We will then look more specifically at how MSI can be used to understand tumour metabolism and other applications specific to oncology research. This will all be described alongside the challenges of applying MSI to industry research with increased use of metrology for MSI.
Collapse
|
35
|
Dufresne M, Guneysu D, Patterson NH, Marcinkiewicz MM, Regina A, Demeule M, Chaurand P. Multimodal detection of GM2 and GM3 lipid species in the brain of mucopolysaccharidosis type II mouse by serial imaging mass spectrometry and immunohistochemistry. Anal Bioanal Chem 2016; 409:1425-1433. [DOI: 10.1007/s00216-016-0076-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/24/2016] [Accepted: 10/31/2016] [Indexed: 11/24/2022]
|
36
|
Dexter A, Race AM, Styles IB, Bunch J. Testing for Multivariate Normality in Mass Spectrometry Imaging Data: A Robust Statistical Approach for Clustering Evaluation and the Generation of Synthetic Mass Spectrometry Imaging Data Sets. Anal Chem 2016; 88:10893-10899. [DOI: 10.1021/acs.analchem.6b02139] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Alex Dexter
- National Physical Laboratory, Teddington, Middlesex TW11 0LW, United Kingdom
| | - Alan M. Race
- National Physical Laboratory, Teddington, Middlesex TW11 0LW, United Kingdom
| | | | - Josephine Bunch
- National Physical Laboratory, Teddington, Middlesex TW11 0LW, United Kingdom
- School
of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire NG7 2RD, United Kingdom
| |
Collapse
|
37
|
Chen SP, Tolner EA, Eikermann-Haerter K. Animal models of monogenic migraine. Cephalalgia 2016; 36:704-21. [PMID: 27154999 DOI: 10.1177/0333102416645933] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 04/01/2016] [Indexed: 01/18/2023]
Abstract
Migraine is a highly prevalent and disabling neurological disorder with a strong genetic component. Rare monogenic forms of migraine, or syndromes in which migraine frequently occurs, help scientists to unravel pathogenetic mechanisms of migraine and its comorbidities. Transgenic mouse models for rare monogenic mutations causing familial hemiplegic migraine (FHM), cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), and familial advanced sleep-phase syndrome (FASPS), have been created. Here, we review the current state of research using these mutant mice. We also discuss how currently available experimental approaches, including epigenetic studies, biomolecular analysis and optogenetic technologies, can be used for characterization of migraine genes to further unravel the functional and molecular pathways involved in migraine.
Collapse
Affiliation(s)
- Shih-Pin Chen
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taiwan Faculty of Medicine, National Yang-Ming University School of Medicine, Taiwan Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Else A Tolner
- Departments of Human Genetics and Neurology, Leiden University Medical Centre, the Netherlands
| | - Katharina Eikermann-Haerter
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| |
Collapse
|
38
|
Schubert N, Axer M, Schober M, Huynh AM, Huysegoms M, Palomero-Gallagher N, Bjaalie JG, Leergaard TB, Kirlangic ME, Amunts K, Zilles K. 3D Reconstructed Cyto-, Muscarinic M2 Receptor, and Fiber Architecture of the Rat Brain Registered to the Waxholm Space Atlas. Front Neuroanat 2016; 10:51. [PMID: 27199682 PMCID: PMC4853377 DOI: 10.3389/fnana.2016.00051] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 04/18/2016] [Indexed: 12/25/2022] Open
Abstract
High-resolution multiscale and multimodal 3D models of the brain are essential tools to understand its complex structural and functional organization. Neuroimaging techniques addressing different aspects of brain organization should be integrated in a reference space to enable topographically correct alignment and subsequent analysis of the various datasets and their modalities. The Waxholm Space (http://software.incf.org/software/waxholm-space) is a publicly available 3D coordinate-based standard reference space for the mapping and registration of neuroanatomical data in rodent brains. This paper provides a newly developed pipeline combining imaging and reconstruction steps with a novel registration strategy to integrate new neuroimaging modalities into the Waxholm Space atlas. As a proof of principle, we incorporated large scale high-resolution cyto-, muscarinic M2 receptor, and fiber architectonic images of rat brains into the 3D digital MRI based atlas of the Sprague Dawley rat in Waxholm Space. We describe the whole workflow, from image acquisition to reconstruction and registration of these three modalities into the Waxholm Space rat atlas. The registration of the brain sections into the atlas is performed by using both linear and non-linear transformations. The validity of the procedure is qualitatively demonstrated by visual inspection, and a quantitative evaluation is performed by measurement of the concordance between representative atlas-delineated regions and the same regions based on receptor or fiber architectonic data. This novel approach enables for the first time the generation of 3D reconstructed volumes of nerve fibers and fiber tracts, or of muscarinic M2 receptor density distributions, in an entire rat brain. Additionally, our pipeline facilitates the inclusion of further neuroimaging datasets, e.g., 3D reconstructed volumes of histochemical stainings or of the regional distributions of multiple other receptor types, into the Waxholm Space. Thereby, a multiscale and multimodal rat brain model was created in the Waxholm Space atlas of the rat brain. Since the registration of these multimodal high-resolution datasets into the same coordinate system is an indispensable requisite for multi-parameter analyses, this approach enables combined studies on receptor and cell distributions as well as fiber densities in the same anatomical structures at microscopic scales for the first time.
Collapse
Affiliation(s)
- Nicole Schubert
- Institute of Neuroscience and Medicine 1, Research Centre JülichJülich, Germany
| | - Markus Axer
- Institute of Neuroscience and Medicine 1, Research Centre JülichJülich, Germany
| | - Martin Schober
- Institute of Neuroscience and Medicine 1, Research Centre JülichJülich, Germany
| | - Anh-Minh Huynh
- Institute of Neuroscience and Medicine 1, Research Centre JülichJülich, Germany
| | - Marcel Huysegoms
- Institute of Neuroscience and Medicine 1, Research Centre JülichJülich, Germany
| | | | - Jan G. Bjaalie
- Institute of Basic Medical Sciences, University of OsloOslo, Norway
| | | | - Mehmet E. Kirlangic
- Institute of Neuroscience and Medicine 1, Research Centre JülichJülich, Germany
| | - Katrin Amunts
- Institute of Neuroscience and Medicine 1, Research Centre JülichJülich, Germany
- C. and O. Vogt Institute for Brain Research, Heinrich-Heine University DüsseldorfDüsseldorf, Germany
| | - Karl Zilles
- Institute of Neuroscience and Medicine 1, Research Centre JülichJülich, Germany
- Translational Brain Medicine, Jülich-Aachen Research AllianceAachen, Germany
- Department of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen UniversityAachen, Germany
| |
Collapse
|
39
|
Gorzolka K, Kölling J, Nattkemper TW, Niehaus K. Spatio-Temporal Metabolite Profiling of the Barley Germination Process by MALDI MS Imaging. PLoS One 2016; 11:e0150208. [PMID: 26938880 PMCID: PMC4777520 DOI: 10.1371/journal.pone.0150208] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/10/2016] [Indexed: 12/11/2022] Open
Abstract
MALDI mass spectrometry imaging was performed to localize metabolites during the first seven days of the barley germination. Up to 100 mass signals were detected of which 85 signals were identified as 48 different metabolites with highly tissue-specific localizations. Oligosaccharides were observed in the endosperm and in parts of the developed embryo. Lipids in the endosperm co-localized in dependency on their fatty acid compositions with changes in the distributions of diacyl phosphatidylcholines during germination. 26 potentially antifungal hordatines were detected in the embryo with tissue-specific localizations of their glycosylated, hydroxylated, and O-methylated derivates. In order to reveal spatio-temporal patterns in local metabolite compositions, multiple MSI data sets from a time series were analyzed in one batch. This requires a new preprocessing strategy to achieve comparability between data sets as well as a new strategy for unsupervised clustering. The resulting spatial segmentation for each time point sample is visualized in an interactive cluster map and enables simultaneous interactive exploration of all time points. Using this new analysis approach and visualization tool germination-dependent developments of metabolite patterns with single MS position accuracy were discovered. This is the first study that presents metabolite profiling of a cereals' germination process over time by MALDI MSI with the identification of a large number of peaks of agronomically and industrially important compounds such as oligosaccharides, lipids and antifungal agents. Their detailed localization as well as the MS cluster analyses for on-tissue metabolite profile mapping revealed important information for the understanding of the germination process, which is of high scientific interest.
Collapse
Affiliation(s)
- Karin Gorzolka
- Proteome and Metabolome Research, Faculty of Biology, Center for Biotechnology (CeBiTec), Bielefeld, Germany
| | - Jan Kölling
- Biodata Mining, Faculty of Technology, Center for Biotechnology (CeBiTec), Bielefeld, Germany.,International Research Training Group "Computational Methods for the Analysis of the Diversity and Dynamics of Genomes", Bielefeld University, Bielefeld, Germany
| | - Tim W Nattkemper
- Biodata Mining, Faculty of Technology, Center for Biotechnology (CeBiTec), Bielefeld, Germany
| | - Karsten Niehaus
- Proteome and Metabolome Research, Faculty of Biology, Center for Biotechnology (CeBiTec), Bielefeld, Germany
| |
Collapse
|
40
|
Fernández R, Garate J, Lage S, Terés S, Higuera M, Bestard-Escalas J, Martin ML, López DH, Guardiola-Serrano F, Escribá PV, Barceló-Coblijn G, Fernández JA. Optimized Protocol To Analyze Changes in the Lipidome of Xenografts after Treatment with 2-Hydroxyoleic Acid. Anal Chem 2016; 88:1022-9. [PMID: 26607740 PMCID: PMC5017204 DOI: 10.1021/acs.analchem.5b03978] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Xenografts are a popular model for the study of the action of new antitumor drugs. However, xenografts are highly heterogeneous structures, and therefore it is sometimes difficult to evaluate the effects of the compounds on tumor metabolism. In this context, imaging mass spectrometry (IMS) may yield the required information, due to its inherent characteristics of sensitivity and spatial resolution. To the best of our knowledge, there is still no clear analysis protocol to properly evaluate the changes between samples due to the treatment. Here we present a protocol for the evaluation of the effect of 2-hydroxyoleic acid (2-OHOA), an antitumor compound, on xenografts lipidome based on IMS. Direct treated/control comparison did not show conclusive results. As we will demonstrate, a more sophisticated protocol was required to evaluate these changes including the following: (1) identification of different areas in the xenograft, (2) classification of these areas (necrotic/viable) to compare similar types of tissues, (3) suppression of the effect of the variation of adduct formation between samples, and (4) normalization of the variables using the standard deviation to eliminate the excessive impact of the stronger peaks in the statistical analysis. In this way, the 36 lipid species that experienced the largest changes between treated and control were identified. Furthermore, incorporation of 2-hydroxyoleic acid to a sphinganine base was also confirmed by MS/MS. Comparison of the changes observed here with previous results obtained with different techniques demonstrates the validity of the protocol.
Collapse
Affiliation(s)
- Roberto Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Jone Garate
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Sergio Lage
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| | - Silvia Terés
- Unité de recherche Inserm 0916, Institut Européen de Chimie et Biologie (IECB)-INSERM, 2 rue Robert Escarpit, 33607 Pessac, France
| | - Mónica Higuera
- Laboratory of Molecular and Cell Biomedicine, Department of Biology, University of the Balearic Islands, E-07122 Palma, Balearic Islands, Spain
| | - Joan Bestard-Escalas
- Research Unit, Hospital Universitari Son Espases, Institut d’Investigació Sanitária de Palma (IdISPa), Carretera Valldemossa 79, E-07010 Palma, Balearic Islands, Spain
| | - M. Laura Martin
- Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, 415 East 68th Street, New York, New York 10065, United States
| | - Daniel H. López
- Research Unit, Hospital Universitari Son Espases, Institut d’Investigació Sanitária de Palma (IdISPa), Carretera Valldemossa 79, E-07010 Palma, Balearic Islands, Spain
| | - Francisca Guardiola-Serrano
- Laboratory of Molecular and Cell Biomedicine, Department of Biology, University of the Balearic Islands, E-07122 Palma, Balearic Islands, Spain
| | - Pablo V. Escribá
- Laboratory of Molecular and Cell Biomedicine, Department of Biology, University of the Balearic Islands, E-07122 Palma, Balearic Islands, Spain
| | - Gwendolyn Barceló-Coblijn
- Research Unit, Hospital Universitari Son Espases, Institut d’Investigació Sanitária de Palma (IdISPa), Carretera Valldemossa 79, E-07010 Palma, Balearic Islands, Spain
| | - José A. Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain
| |
Collapse
|
41
|
Bodzon-Kulakowska A, Suder P. Imaging mass spectrometry: Instrumentation, applications, and combination with other visualization techniques. MASS SPECTROMETRY REVIEWS 2016; 35:147-69. [PMID: 25962625 DOI: 10.1002/mas.21468] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 01/23/2015] [Indexed: 05/18/2023]
Abstract
Imaging Mass Spectrometry (IMS) is strengthening its position as a valuable analytical tool. It has unique ability to identify structures and to unravel molecular changes that occur in the precisely defined part of the sample. These unique features open new possibilities in the field of various aspects of biological research. In this review we briefly discuss the main imaging mass spectrometry techniques, as well as the nature of biological samples and molecules, which might be analyzed by such methodology. Moreover, a novel approach, where different analytical techniques might be combined with the results of IMS study, is emphasized and discussed. With such a fast development of IMS and related methods, we can foresee the promising future of this technique.
Collapse
Affiliation(s)
- Anna Bodzon-Kulakowska
- Department of Biochemistry and Neurobiology, Faculty of Materials Sciences and Ceramics, AGH University of Science and Technology, 30-059 Krakow, Poland
| | - Piotr Suder
- Department of Biochemistry and Neurobiology, Faculty of Materials Sciences and Ceramics, AGH University of Science and Technology, 30-059 Krakow, Poland
- Academic Centre for Materials and Nanotechnology (ACMiN), AGH University of Science and Technology, 30-059 Krakow, Poland
| |
Collapse
|
42
|
Heijs B, Abdelmoula WM, Lou S, Briaire-de Bruijn IH, Dijkstra J, Bovée JVMG, McDonnell LA. Histology-Guided High-Resolution Matrix-Assisted Laser Desorption Ionization Mass Spectrometry Imaging. Anal Chem 2015; 87:11978-83. [DOI: 10.1021/acs.analchem.5b03610] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Bram Heijs
- Center
for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333ZC Leiden, The Netherlands
| | - Walid M. Abdelmoula
- Division
of Image Processing, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, The Netherlands
| | - Sha Lou
- Center
for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333ZC Leiden, The Netherlands
| | - Inge H. Briaire-de Bruijn
- Department
of Pathology, Leiden University Medical Center, Albinusdreef
2, 2333ZA Leiden, The Netherlands
| | - Jouke Dijkstra
- Division
of Image Processing, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, The Netherlands
| | - Judith V. M. G. Bovée
- Department
of Pathology, Leiden University Medical Center, Albinusdreef
2, 2333ZA Leiden, The Netherlands
| | - Liam A. McDonnell
- Center
for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333ZC Leiden, The Netherlands
- Department
of Pathology, Leiden University Medical Center, Albinusdreef
2, 2333ZA Leiden, The Netherlands
- Fondazione Pisana per la Scienza ONLUS, 56125 Pisa, Italy
| |
Collapse
|
43
|
Carreira RJ, Shyti R, Balluff B, Abdelmoula WM, van Heiningen SH, van Zeijl RJ, Dijkstra J, Ferrari MD, Tolner EA, McDonnell LA, van den Maagdenberg AMJM. Large-scale mass spectrometry imaging investigation of consequences of cortical spreading depression in a transgenic mouse model of migraine. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:853-61. [PMID: 25877011 PMCID: PMC4422864 DOI: 10.1007/s13361-015-1136-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/10/2015] [Accepted: 03/10/2015] [Indexed: 05/04/2023]
Abstract
Cortical spreading depression (CSD) is the electrophysiological correlate of migraine aura. Transgenic mice carrying the R192Q missense mutation in the Cacna1a gene, which in patients causes familial hemiplegic migraine type 1 (FHM1), exhibit increased propensity to CSD. Herein, mass spectrometry imaging (MSI) was applied for the first time to an animal cohort of transgenic and wild type mice to study the biomolecular changes following CSD in the brain. Ninety-six coronal brain sections from 32 mice were analyzed by MALDI-MSI. All MSI datasets were registered to the Allen Brain Atlas reference atlas of the mouse brain so that the molecular signatures of distinct brain regions could be compared. A number of metabolites and peptides showed substantial changes in the brain associated with CSD. Among those, different mass spectral features showed significant (t-test, P < 0.05) changes in the cortex, 146 and 377 Da, and in the thalamus, 1820 and 1834 Da, of the CSD-affected hemisphere of FHM1 R192Q mice. Our findings reveal CSD- and genotype-specific molecular changes in the brain of FHM1 transgenic mice that may further our understanding about the role of CSD in migraine pathophysiology. The results also demonstrate the utility of aligning MSI datasets to a common reference atlas for large-scale MSI investigations.
Collapse
Affiliation(s)
- Ricardo J. Carreira
- />Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Reinald Shyti
- />Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Benjamin Balluff
- />Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Walid M. Abdelmoula
- />Division of Image Processing, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Rene J. van Zeijl
- />Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Jouke Dijkstra
- />Division of Image Processing, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michel D. Ferrari
- />Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Else A. Tolner
- />Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Liam A. McDonnell
- />Center for Proteomics and Metabolomics, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
- />Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | - Arn M. J. M. van den Maagdenberg
- />Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- />Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
44
|
Škrášková K, Khmelinskii A, Abdelmoula WM, De Munter S, Baes M, McDonnell L, Dijkstra J, Heeren RMA. Precise Anatomic Localization of Accumulated Lipids in Mfp2 Deficient Murine Brains Through Automated Registration of SIMS Images to the Allen Brain Atlas. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:948-57. [PMID: 25916600 PMCID: PMC4422856 DOI: 10.1007/s13361-015-1146-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/19/2015] [Accepted: 03/19/2015] [Indexed: 05/04/2023]
Abstract
Mass spectrometry imaging (MSI) is a powerful tool for the molecular characterization of specific tissue regions. Histochemical staining provides anatomic information complementary to MSI data. The combination of both modalities has been proven to be beneficial. However, direct comparison of histology based and mass spectrometry-based molecular images can become problematic because of potential tissue damages or changes caused by different sample preparation. Curated atlases such as the Allen Brain Atlas (ABA) offer a collection of highly detailed and standardized anatomic information. Direct comparison of MSI brain data to the ABA allows for conclusions to be drawn on precise anatomic localization of the molecular signal. Here we applied secondary ion mass spectrometry imaging at high spatial resolution to study brains of knock-out mouse models with impaired peroxisomal β-oxidation. Murine models were lacking D-multifunctional protein (MFP2), which is involved in degradation of very long chain fatty acids. SIMS imaging revealed deposits of fatty acids within distinct brain regions. Manual comparison of the MSI data with the histologic stains did not allow for an unequivocal anatomic identification of the fatty acids rich regions. We further employed an automated pipeline for co-registration of the SIMS data to the ABA. The registration enabled precise anatomic annotation of the brain structures with the revealed lipid deposits. The precise anatomic localization allowed for a deeper insight into the pathology of Mfp2 deficient mouse models.
Collapse
Affiliation(s)
- Karolina Škrášková
- />FOM-Institute AMOLF, Amsterdam, The Netherlands
- />TI-COAST, Amsterdam, The Netherlands
| | - Artem Khmelinskii
- />FOM-Institute AMOLF, Amsterdam, The Netherlands
- />Percuros B.V., Enschede, The Netherlands
- />Division of Image Processing, Department of Radiology, LUMC, Leiden, The Netherlands
| | - Walid M. Abdelmoula
- />Division of Image Processing, Department of Radiology, LUMC, Leiden, The Netherlands
| | | | - Myriam Baes
- />Laboratory of Cellular Metabolism, KU Leuven, Leuven, Belgium
| | - Liam McDonnell
- />Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
- />Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | - Jouke Dijkstra
- />Division of Image Processing, Department of Radiology, LUMC, Leiden, The Netherlands
| | - Ron M. A. Heeren
- />FOM-Institute AMOLF, Amsterdam, The Netherlands
- />TI-COAST, Amsterdam, The Netherlands
- />M4I, The Maastricht MultiModal Molecular Imaging Institute, University of Maastricht, Maastricht, The Netherlands
| |
Collapse
|
45
|
Addie RD, Balluff B, Bovée JVMG, Morreau H, McDonnell LA. Current State and Future Challenges of Mass Spectrometry Imaging for Clinical Research. Anal Chem 2015; 87:6426-33. [DOI: 10.1021/acs.analchem.5b00416] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Ruben D. Addie
- Center for Proteomics
and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Benjamin Balluff
- Center for Proteomics
and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Liam A. McDonnell
- Center for Proteomics
and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
- Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| |
Collapse
|
46
|
Rawlins CM, Salisbury JP, Feldman DR, Isim S, Agar NYR, Luther E, Agar JN. Imaging and Mapping of Tissue Constituents at the Single-Cell Level Using MALDI MSI and Quantitative Laser Scanning Cytometry. Methods Mol Biol 2015; 1346:133-49. [PMID: 26542720 DOI: 10.1007/978-1-4939-2987-0_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
For nearly a century, histopathology involved the laborious morphological analyses of tissues stained with broad-spectrum dyes (i.e., eosin to label proteins). With the advent of antibody-labeling, immunostaining (fluorescein and rhodamine for fluorescent labeling) and immunohistochemistry (DAB and hematoxylin), it became possible to identify specific immunological targets in cells and tissue preparations. Technical advances, including the development of monoclonal antibody technology, led to an ever-increasing palate of dyes, both fluorescent and chromatic. This provides an incredibly rich menu of molecular entities that can be visualized and quantified in cells-giving rise to the new discipline of Molecular Pathology. We describe the evolution of two analytical techniques, cytometry and mass spectrometry, which complement histopathological visual analysis by providing automated, cellular-resolution constituent maps. For the first time, laser scanning cytometry (LSC) and matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) are combined for the analysis of tissue sections. The utility of the marriage of these techniques is demonstrated by analyzing mouse brains with neuron-specific, genetically encoded, fluorescent proteins. We present a workflow that: (1) can be used with or without expensive matrix deposition methods, (2) uses LSC images to reveal the diverse landscape of neural tissue as well as the matrix, and (3) uses a tissue fixation method compatible with a DNA stain. The proposed workflow can be adapted for a variety of sample preparation and matrix deposition methods.
Collapse
Affiliation(s)
- Catherine M Rawlins
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, USA.
| | - Joseph P Salisbury
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, USA.
| | - Daniel R Feldman
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Sinan Isim
- Life Sciences Department, Brandeis University, Waltham, MA, USA.
| | - Nathalie Y R Agar
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ed Luther
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA.
| | - Jeffery N Agar
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, USA. .,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Verbeeck N, Yang J, De Moor B, Caprioli R, Waelkens E, Van de Plas R. Automated anatomical interpretation of ion distributions in tissue: linking imaging mass spectrometry to curated atlases. Anal Chem 2014; 86:8974-82. [PMID: 25153352 PMCID: PMC4165455 DOI: 10.1021/ac502838t] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 07/30/2014] [Indexed: 12/17/2022]
Abstract
Imaging mass spectrometry (IMS) has become a prime tool for studying the distribution of biomolecules in tissue. Although IMS data sets can become very large, computational methods have made it practically feasible to search these experiments for relevant findings. However, these methods lack access to an important source of information that many human interpretations rely upon: anatomical insight. In this work, we address this need by (1) integrating a curated anatomical data source with an empirically acquired IMS data source, establishing an algorithm-accessible link between them and (2) demonstrating the potential of such an IMS-anatomical atlas link by applying it toward automated anatomical interpretation of ion distributions in tissue. The concept is demonstrated in mouse brain tissue, using the Allen Mouse Brain Atlas as the curated anatomical data source that is linked to MALDI-based IMS experiments. We first develop a method to spatially map the anatomical atlas to the IMS data sets using nonrigid registration techniques. Once a mapping is established, a second computational method, called correlation-based querying, gives an elementary demonstration of the link by delivering basic insight into relationships between ion images and anatomical structures. Finally, a third algorithm moves further beyond both registration and correlation by providing automated anatomical interpretation of ion images. This task is approached as an optimization problem that deconstructs ion distributions as combinations of known anatomical structures. We demonstrate that establishing a link between an IMS experiment and an anatomical atlas enables automated anatomical annotation, which can serve as an important accelerator both for human and machine-guided exploration of IMS experiments.
Collapse
Affiliation(s)
- Nico Verbeeck
- Department
of Electrical Engineering (ESAT), STADIUS Center for
Dynamical Systems, Signal Processing, and Data Analytics, KU Leuven, Kasteelpark
Arenberg 10, Box 2446, 3001 Leuven, Belgium
- iMinds
Medical IT, Kasteelpark
Arenberg 10, Box 2446, 3001 Leuven, Belgium
| | - Junhai Yang
- Mass
Spectrometry Research Center and Departments of Biochemistry, Chemistry,
Pharmacology, and Medicine, Vanderbilt University, 465 21st Avenue South, MRB III Suite
9160, Nashville, Tennessee 37232, United States
| | - Bart De Moor
- Department
of Electrical Engineering (ESAT), STADIUS Center for
Dynamical Systems, Signal Processing, and Data Analytics, KU Leuven, Kasteelpark
Arenberg 10, Box 2446, 3001 Leuven, Belgium
- iMinds
Medical IT, Kasteelpark
Arenberg 10, Box 2446, 3001 Leuven, Belgium
| | - Richard
M. Caprioli
- Mass
Spectrometry Research Center and Departments of Biochemistry, Chemistry,
Pharmacology, and Medicine, Vanderbilt University, 465 21st Avenue South, MRB III Suite
9160, Nashville, Tennessee 37232, United States
| | - Etienne Waelkens
- Sybioma, KU Leuven, Campus Gasthuisberg O&N 2, Herestraat 49, Box
802, 3000 Leuven, Belgium
- Department
of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O&N 1, Herestraat
49, Box 901, 3000 Leuven, Belgium
| | - Raf Van de Plas
- Mass
Spectrometry Research Center and Departments of Biochemistry, Chemistry,
Pharmacology, and Medicine, Vanderbilt University, 465 21st Avenue South, MRB III Suite
9160, Nashville, Tennessee 37232, United States
| |
Collapse
|
48
|
Abdelmoula WM, Škrášková K, Balluff B, Carreira RJ, Tolner EA, Lelieveldt BPF, van der Maaten L, Morreau H, van den Maagdenberg AMJM, Heeren RMA, McDonnell LA, Dijkstra J. Automatic generic registration of mass spectrometry imaging data to histology using nonlinear stochastic embedding. Anal Chem 2014; 86:9204-11. [PMID: 25133861 DOI: 10.1021/ac502170f] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The combination of mass spectrometry imaging and histology has proven a powerful approach for obtaining molecular signatures from specific cells/tissues of interest, whether to identify biomolecular changes associated with specific histopathological entities or to determine the amount of a drug in specific organs/compartments. Currently there is no software that is able to explicitly register mass spectrometry imaging data spanning different ionization techniques or mass analyzers. Accordingly, the full capabilities of mass spectrometry imaging are at present underexploited. Here we present a fully automated generic approach for registering mass spectrometry imaging data to histology and demonstrate its capabilities for multiple mass analyzers, multiple ionization sources, and multiple tissue types.
Collapse
Affiliation(s)
- Walid M Abdelmoula
- Division of Image Processing, Department of Radiology, ‡Center for Proteomics and Metabolomics, §Department of Human Genetics, ∥Department of Neurology, and ⊥Department of Pathology, Leiden University Medical Center , 2333 ZA Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ahlf DR, Masyuko RN, Hummon AB, Bohn PW. Correlated mass spectrometry imaging and confocal Raman microscopy for studies of three-dimensional cell culture sections. Analyst 2014; 139:4578-85. [DOI: 10.1039/c4an00826j] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|