1
|
Good CJ, Butrico CE, Colley ME, Emmerson LN, Gibson-Corley KN, Cassat JE, Spraggins JM, Caprioli RM. Uncovering lipid dynamics in Staphylococcus aureus osteomyelitis using multimodal imaging mass spectrometry. Cell Chem Biol 2024; 31:1852-1868.e5. [PMID: 39389064 PMCID: PMC11977171 DOI: 10.1016/j.chembiol.2024.09.005] [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: 01/19/2024] [Revised: 06/20/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
Osteomyelitis occurs when Staphylococcus aureus invades the bone microenvironment, resulting in a bone marrow abscess with a spatially defined architecture of cells and biomolecules. Imaging mass spectrometry and microscopy are tools that can be employed to interrogate the lipidome of S. aureus-infected murine femurs and reveal metabolic and signaling consequences of infection. Here, nearly 250 lipids were spatially mapped to healthy and infection-associated morphological features throughout the femur, establishing composition profiles for tissue types. Ether lipids and arachidonoyl lipids were altered between cells and tissue structures in abscesses, suggesting their roles in abscess formation and inflammatory signaling. Sterols, triglycerides, bis(monoacylglycero)phosphates, and gangliosides possessed ring-like distributions throughout the abscess, suggesting a hypothesized dysregulation of lipid metabolism in a population of cells that cannot be discerned with traditional microscopy. These data provide insight into the signaling function and metabolism of cells in the fibrotic border of abscesses, likely characteristic of lipid-laden macrophages.
Collapse
Affiliation(s)
- Christopher J Good
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Casey E Butrico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Madeline E Colley
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Lauren N Emmerson
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37235, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA.
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
2
|
Good CJ, Butrico CE, Colley ME, Gibson-Corley KN, Cassat JE, Spraggins JM, Caprioli RM. In situ lipidomics of Staphylococcus aureus osteomyelitis using imaging mass spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569690. [PMID: 38077019 PMCID: PMC10705574 DOI: 10.1101/2023.12.01.569690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Osteomyelitis occurs when Staphylococcus aureus invades the bone microenvironment, resulting in a bone marrow abscess with a spatially defined architecture of cells and biomolecules. Imaging mass spectrometry and microscopy are invaluable tools that can be employed to interrogate the lipidome of S. aureus-infected murine femurs to reveal metabolic and signaling consequences of infection. Here, nearly 250 lipids were spatially mapped to healthy and infection-associated morphological features throughout the femur, establishing composition profiles for tissue types. Ether lipids and arachidonoyl lipids were significantly altered between cells and tissue structures in abscesses, suggesting their roles in abscess formation and inflammatory signaling. Sterols, triglycerides, bis(monoacylglycero)phosphates, and gangliosides possessed ring-like distributions throughout the abscess, indicating dysregulated lipid metabolism in a subpopulation of leukocytes that cannot be discerned with traditional microscopy. These data provide chemical insight into the signaling function and metabolism of cells in the fibrotic border of abscesses, likely characteristic of lipid-laden macrophages.
Collapse
Affiliation(s)
- Christopher J. Good
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Casey E. Butrico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Madeline E. Colley
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Katherine N. Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E. Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey M. Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Richard M. Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
3
|
Azam KSF, Ryabchykov O, Bocklitz T. A Review on Data Fusion of Multidimensional Medical and Biomedical Data. Molecules 2022; 27:7448. [PMID: 36364272 PMCID: PMC9655963 DOI: 10.3390/molecules27217448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/19/2022] [Accepted: 10/21/2022] [Indexed: 08/05/2024] Open
Abstract
Data fusion aims to provide a more accurate description of a sample than any one source of data alone. At the same time, data fusion minimizes the uncertainty of the results by combining data from multiple sources. Both aim to improve the characterization of samples and might improve clinical diagnosis and prognosis. In this paper, we present an overview of the advances achieved over the last decades in data fusion approaches in the context of the medical and biomedical fields. We collected approaches for interpreting multiple sources of data in different combinations: image to image, image to biomarker, spectra to image, spectra to spectra, spectra to biomarker, and others. We found that the most prevalent combination is the image-to-image fusion and that most data fusion approaches were applied together with deep learning or machine learning methods.
Collapse
Affiliation(s)
- Kazi Sultana Farhana Azam
- Leibniz Institute of Photonic Technology, Member of Leibniz-Research Alliance “Health Technologies”, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - Oleg Ryabchykov
- Leibniz Institute of Photonic Technology, Member of Leibniz-Research Alliance “Health Technologies”, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - Thomas Bocklitz
- Leibniz Institute of Photonic Technology, Member of Leibniz-Research Alliance “Health Technologies”, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| |
Collapse
|
4
|
Subasinghe SAAS, Pautler RG, Samee MAH, Yustein JT, Allen MJ. Dual-Mode Tumor Imaging Using Probes That Are Responsive to Hypoxia-Induced Pathological Conditions. BIOSENSORS 2022; 12:478. [PMID: 35884281 PMCID: PMC9313010 DOI: 10.3390/bios12070478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 05/02/2023]
Abstract
Hypoxia in solid tumors is associated with poor prognosis, increased aggressiveness, and strong resistance to therapeutics, making accurate monitoring of hypoxia important. Several imaging modalities have been used to study hypoxia, but each modality has inherent limitations. The use of a second modality can compensate for the limitations and validate the results of any single imaging modality. In this review, we describe dual-mode imaging systems for the detection of hypoxia that have been reported since the start of the 21st century. First, we provide a brief overview of the hallmarks of hypoxia used for imaging and the imaging modalities used to detect hypoxia, including optical imaging, ultrasound imaging, photoacoustic imaging, single-photon emission tomography, X-ray computed tomography, positron emission tomography, Cerenkov radiation energy transfer imaging, magnetic resonance imaging, electron paramagnetic resonance imaging, magnetic particle imaging, and surface-enhanced Raman spectroscopy, and mass spectrometric imaging. These overviews are followed by examples of hypoxia-relevant imaging using a mixture of probes for complementary single-mode imaging techniques. Then, we describe dual-mode molecular switches that are responsive in multiple imaging modalities to at least one hypoxia-induced pathological change. Finally, we offer future perspectives toward dual-mode imaging of hypoxia and hypoxia-induced pathophysiological changes in tumor microenvironments.
Collapse
Affiliation(s)
| | - Robia G. Pautler
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; (R.G.P.); (M.A.H.S.)
| | - Md. Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; (R.G.P.); (M.A.H.S.)
| | - Jason T. Yustein
- Integrative Molecular and Biomedical Sciences and the Department of Pediatrics in the Texas Children’s Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Matthew J. Allen
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA;
| |
Collapse
|
5
|
Tuck M, Grélard F, Blanc L, Desbenoit N. MALDI-MSI Towards Multimodal Imaging: Challenges and Perspectives. Front Chem 2022; 10:904688. [PMID: 35615316 PMCID: PMC9124797 DOI: 10.3389/fchem.2022.904688] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/14/2022] [Indexed: 01/22/2023] Open
Abstract
Multimodal imaging is a powerful strategy for combining information from multiple images. It involves several fields in the acquisition, processing and interpretation of images. As multimodal imaging is a vast subject area with various combinations of imaging techniques, it has been extensively reviewed. Here we focus on Matrix-assisted Laser Desorption Ionization Mass Spectrometry Imaging (MALDI-MSI) coupling other imaging modalities in multimodal approaches. While MALDI-MS images convey a substantial amount of chemical information, they are not readily informative about the morphological nature of the tissue. By providing a supplementary modality, MALDI-MS images can be more informative and better reflect the nature of the tissue. In this mini review, we emphasize the analytical and computational strategies to address multimodal MALDI-MSI.
Collapse
|
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
|
Analysis of the Manganese and MntR Regulon in Corynebacterium diphtheriae. J Bacteriol 2021; 203:e0027421. [PMID: 34370555 DOI: 10.1128/jb.00274-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Corynebacterium diphtheriae is the causative agent of a severe respiratory disease in humans. The bacterial systems required for infection are poorly understood, but the acquisition of metals such as manganese (Mn) is likely critical for host colonization. MntR is a Mn-dependent transcriptional regulator in C. diphtheriae that represses the expression of the mntABCD genes, which encode a putative ABC metal transporter. However, other targets of Mn and MntR regulation in C. diphtheriae have not been identified. In this study, we use comparisons between the gene expression profiles of wild-type C. diphtheriae strain 1737 grown without or with Mn supplementation and comparisons of gene expression between wild-type and an mntR deletion mutant to characterize the C. diphtheriae Mn and MntR regulon. MntR was observed to both repress and induce various target genes in a Mn-dependent manner. Genes induced by MntR include the Mn-superoxide dismutase, sodA, and the putative ABC transporter locus, iutABCD. DNA binding studies showed that MntR interacts with the promoter regions for several genes identified in the expression study, and a 17-bp consensus MntR DNA binding site was identified. We found that an mntR mutant displayed increased sensitivity to Mn and cadmium that could be alleviated by the additional deletion of the mntABCD transport locus, providing evidence that the MntABCD transporter functions as a Mn uptake system in C. diphtheriae. The findings in this study further our understanding of metal uptake systems and global metal regulatory networks in this important human pathogen. Importance Mechanisms for metal scavenging are critical to the survival and success of bacterial pathogens, including Corynebacterium diphtheriae. Metal import systems in pathogenic bacteria have been studied as possible vaccine components due to high conservation, critical functionality, and surface localization. In this study, we expand our understanding of the genes controlled by the global manganese regulator, MntR. We determined a role for the MntABCD transporter in manganese import using evidence from manganese and cadmium toxicity assays. Understanding the nutritional requirements of C. diphtheriae and the tools used to acquire essential metals will aid in the development of future vaccines.
Collapse
|
8
|
Skaar EP. Imaging Infection Across Scales of Size: From Whole Animals to Single Molecules. Annu Rev Microbiol 2021; 75:407-426. [PMID: 34343016 DOI: 10.1146/annurev-micro-041521-121457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Infectious diseases are a leading cause of global morbidity and mortality, and the threat of infectious diseases to human health is steadily increasing as new diseases emerge, existing diseases reemerge, and antimicrobial resistance expands. The application of imaging technology to the study of infection biology has the potential to uncover new factors that are critical to the outcome of host-pathogen interactions and to lead to innovations in diagnosis and treatment of infectious diseases. This article reviews current and future opportunities for the application of imaging to the study of infectious diseases, with a particular focus on the power of imaging objects across a broad range of sizes to expand the utility of these approaches. Expected final online publication date for the Annual Review of Microbiology, Volume 75 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Eric P Skaar
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA;
| |
Collapse
|
9
|
Application and Perspectives of MALDI-TOF Mass Spectrometry in Clinical Microbiology Laboratories. Microorganisms 2021; 9:microorganisms9071539. [PMID: 34361974 PMCID: PMC8307939 DOI: 10.3390/microorganisms9071539] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/06/2021] [Accepted: 07/18/2021] [Indexed: 12/11/2022] Open
Abstract
Early diagnosis of severe infections requires of a rapid and reliable diagnosis to initiate appropriate treatment, while avoiding unnecessary antimicrobial use and reducing associated morbidities and healthcare costs. It is a fact that conventional methods usually require more than 24–48 h to culture and profile bacterial species. Mass spectrometry (MS) is an analytical technique that has emerged as a powerful tool in clinical microbiology for identifying peptides and proteins, which makes it a promising tool for microbial identification. Matrix assisted laser desorption ionization–time of flight MS (MALDI–TOF MS) offers a cost- and time-effective alternative to conventional methods, such as bacterial culture and even 16S rRNA gene sequencing, for identifying viruses, bacteria and fungi and detecting virulence factors and mechanisms of resistance. This review provides an overview of the potential applications and perspectives of MS in clinical microbiology laboratories and proposes its use as a first-line method for microbial identification and diagnosis.
Collapse
|
10
|
Geier B, Oetjen J, Ruthensteiner B, Polikarpov M, Gruber-Vodicka HR, Liebeke M. Connecting structure and function from organisms to molecules in small-animal symbioses through chemo-histo-tomography. Proc Natl Acad Sci U S A 2021; 118:e2023773118. [PMID: 34183413 PMCID: PMC8300811 DOI: 10.1073/pnas.2023773118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Our understanding of metabolic interactions between small symbiotic animals and bacteria or parasitic eukaryotes that reside within their bodies is extremely limited. This gap in knowledge originates from a methodological challenge, namely to connect histological changes in host tissues induced by beneficial and parasitic (micro)organisms to the underlying metabolites. We addressed this challenge and developed chemo-histo-tomography (CHEMHIST), a culture-independent approach to connect anatomic structure and metabolic function in millimeter-sized symbiotic animals. CHEMHIST combines chemical imaging of metabolites based on mass spectrometry imaging (MSI) and microanatomy-based micro-computed X-ray tomography (micro-CT) on the same animal. Both high-resolution MSI and micro-CT allowed us to correlate the distribution of metabolites to the same animal's three-dimensional (3D) histology down to submicrometer resolutions. Our protocol is compatible with tissue-specific DNA sequencing and fluorescence in situ hybridization for the taxonomic identification and localization of the associated micro(organisms). Building CHEMHIST upon in situ imaging, we sampled an earthworm from its natural habitat and created an interactive 3D model of its physical and chemical interactions with bacteria and parasitic nematodes in its tissues. Combining MSI and micro-CT, we present a methodological groundwork for connecting metabolic and anatomic phenotypes of small symbiotic animals that often represent keystone species for ecosystem functioning.
Collapse
Affiliation(s)
- Benedikt Geier
- Max Planck Institute for Marine Microbiology, 28359 Bremen, Germany;
| | - Janina Oetjen
- MALDI Imaging Lab, University of Bremen, 28334 Bremen, Germany
| | | | - Maxim Polikarpov
- European Molecular Biology Laboratory, Hamburg Unit c/o Deutsches Elektronen-Synchrotron, 22607 Hamburg, Germany
| | | | - Manuel Liebeke
- Max Planck Institute for Marine Microbiology, 28359 Bremen, Germany;
| |
Collapse
|
11
|
Veis DJ, Cassat JE. Infectious Osteomyelitis: Marrying Bone Biology and Microbiology to Shed New Light on a Persistent Clinical Challenge. J Bone Miner Res 2021; 36:636-643. [PMID: 33740314 DOI: 10.1002/jbmr.4279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/01/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022]
Abstract
Infections of bone occur in a variety of clinical settings, ranging from spontaneous isolated infections arising from presumed hematogenous spread to those associated with skin and soft tissue wounds or medical implants. The majority are caused by the ubiquitous bacterium Staphyloccocus (S.) aureus, which can exist as a commensal organism on human skin as well as an invasive pathogen, but a multitude of other microbes are also capable of establishing bone infections. While studies of clinical isolates and small animal models have advanced our understanding of the role of various pathogen and host factors in infectious osteomyelitis (iOM), many questions remain unaddressed. Thus, there are many opportunities to elucidate host-pathogen interactions that may be leveraged toward treatment or prevention of this troublesome problem. Herein, we combine perspectives from bone biology and microbiology and suggest that interdisciplinary approaches will bring new insights to the field. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Deborah J Veis
- Division of Bone and Mineral Diseases, Departments of Medicine and Pathology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.,Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Shriners Hospitals for Children, St. Louis, MO, USA
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville,, TN, USA.,Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Institute for Infection, Immunology and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
12
|
Hagras M, Abutaleb NS, Elhosseiny NM, Abdelghany TM, Omara M, Elsebaei MM, Alhashimi M, Norvil AB, Gutay MI, Gowher H, Attia AS, Seleem MN, Mayhoub AS. Development of Biphenylthiazoles Exhibiting Improved Pharmacokinetics and Potent Activity Against Intracellular Staphylococcus aureus. ACS Infect Dis 2020; 6:2887-2900. [PMID: 32897045 DOI: 10.1021/acsinfecdis.0c00137] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exploring the structure-activity relationship (SAR) at the cationic part of arylthiazole antibiotics revealed hydrazine as an active moiety. The main objective of the study is to overcome the inherited toxicity associated with the free hydrazine. A series of hydrocarbon bridges was inserted in between the groups, to separate the two amino groups. Hence, the aminomethylpiperidine-containing analog 16 was identified as a new promising antibacterial agent with efficient antibacterial and pharmacokinetic profiles. Briefly, compound 16 outperformed vancomycin in terms of the antibacterial spectrum against vancomycin-resistant staphylococcal and enterococcal strains with minimum inhibitory concentrations (MICs) ranging from 2 to 4 μg/mL, which is a faster bactericidal mode of action, completely eradicating the high staphylococcal burden within 6-8 h, and it has a unique ability to completely clear intracellular staphylococci. In addition, the initial pharmacokinetic assessment confirmed the high metabolic stability of compound 16 (biological half-life >4 h); it had a good extravascular distribution and maintained a plasma concentration higher than the average MIC value for over 12 h. Moreover, compound 16 significantly reduced MRSA burden in an in vivo MRSA skin infection mouse experiment. These attributes collectively suggest that compound 16 is a good therapeutic candidate for invasive staphylococcal and enterococcal infections. From a mechanistic point of view, compound 16 inhibited undecaprenyl diphosphate phosphatase (UppP) with an IC50 value of 29 μM.
Collapse
Affiliation(s)
- Mohamed Hagras
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Nader S. Abutaleb
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
| | - Noha M. Elhosseiny
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Tamer M. Abdelghany
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Mariam Omara
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Mohamed M. Elsebaei
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
| | - Marwa Alhashimi
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
| | - Allison B Norvil
- Department of Biochemistry, College of Agriculture, Purdue University, West Lafayette, Indiana 47907, United States
| | - Mark I Gutay
- Department of Biochemistry, College of Agriculture, Purdue University, West Lafayette, Indiana 47907, United States
| | - Humaira Gowher
- Department of Biochemistry, College of Agriculture, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ahmed S. Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Mohamed N. Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, 725 Harrison Street, West Lafayette, Indiana 47907, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061, United States
| | - Abdelrahman S. Mayhoub
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University, 1-Elmokhayem Eldaem Street, Cairo 11884, Egypt
- University of Science and Technology, Nanoscience Program, Zewail City of Science and Technology, Ahmed Zewail Street, October Gardens, sixth of October, Giza 12578, Egypt
| |
Collapse
|
13
|
Blutke A, Sun N, Xu Z, Buck A, Harrison L, Schriever SC, Pfluger PT, Wiles D, Kunzke T, Huber K, Schlegel J, Aichler M, Feuchtinger A, Matiasek K, Hauck SM, Walch A. Light sheet fluorescence microscopy guided MALDI-imaging mass spectrometry of cleared tissue samples. Sci Rep 2020; 10:14461. [PMID: 32879402 PMCID: PMC7468256 DOI: 10.1038/s41598-020-71465-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 08/10/2020] [Indexed: 02/08/2023] Open
Abstract
Light sheet fluorescence microscopy (LSFM) of optically cleared biological samples represents a powerful tool to analyze the 3-dimensional morphology of tissues and organs. Multimodal combinations of LSFM with additional analyses of the identical sample help to limit the consumption of restricted specimen and reduce inter-sample variation. Here, we demonstrate the proof-of-concept that LSFM of cleared brain tissue samples can be combined with Matrix Assisted Laser Desorption/Ionization-Mass Spectrometry Imaging (MALDI-MSI) for detection and quantification of proteins. Samples of freshly dissected murine brain and of archived formalin-fixed paraffin-embedded (FFPE) human brain tissue were cleared (3DISCO). Tissue regions of interest were defined by LSFM and excised, (re)-embedded in paraffin, and sectioned. Mouse sections were coated with sinapinic acid matrix. Human brain sections were pre-digested with trypsin and coated with α-cyano-4-hydroxycinnamic acid matrix. Subsequently, sections were subjected to MALDI-time-of-flight (TOF)-MSI in mass ranges between 0.8 to 4 kDa (human tissue sections), or 2.5–25 kDa (mouse tissue sections) with a lateral resolution of 50 µm. Protein- and peptide-identities corresponding to acquired MALDI-MSI spectra were confirmed by parallel liquid chromatography tandem mass spectrometry (LC–MS/MS) analysis. The spatial abundance- and intensity-patterns of established marker proteins detected by MALDI-MSI were also confirmed by immunohistochemistry.
Collapse
Affiliation(s)
- Andreas Blutke
- Research Unit Analytical Pathology, Helmholtz Zentrum München, 8576, Neuherberg, Germany
| | - Na Sun
- Research Unit Analytical Pathology, Helmholtz Zentrum München, 8576, Neuherberg, Germany
| | - Zhihao Xu
- Research Unit Analytical Pathology, Helmholtz Zentrum München, 8576, Neuherberg, Germany
| | - Achim Buck
- Research Unit Analytical Pathology, Helmholtz Zentrum München, 8576, Neuherberg, Germany
| | - Luke Harrison
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, 85764, Neuherberg, Germany.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.,Division of Metabolic Diseases, Technische Universität München, 80333, Munich, Germany
| | - Sonja C Schriever
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, 85764, Neuherberg, Germany.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Paul T Pfluger
- Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, 85764, Neuherberg, Germany.,Institute for Diabetes and Obesity, Helmholtz Zentrum München, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | | | - Thomas Kunzke
- Research Unit Analytical Pathology, Helmholtz Zentrum München, 8576, Neuherberg, Germany
| | - Katharina Huber
- Research Unit Analytical Pathology, Helmholtz Zentrum München, 8576, Neuherberg, Germany
| | - Jürgen Schlegel
- Institute for Pathology, Department of Neuropathology, Technische Universität München, 80333, Munich, Germany
| | - Michaela Aichler
- Research Unit Analytical Pathology, Helmholtz Zentrum München, 8576, Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München, 8576, Neuherberg, Germany.
| | - Kaspar Matiasek
- Institute for Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, 80539, Munich, Germany
| | - Stefanie M Hauck
- Research Unit for Protein Science, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München, 8576, Neuherberg, Germany
| |
Collapse
|
14
|
Fincher JA, Korte AR, Yadavilli S, Morris NJ, Vertes A. Multimodal imaging of biological tissues using combined MALDI and NAPA-LDI mass spectrometry for enhanced molecular coverage. Analyst 2020; 145:6910-6918. [DOI: 10.1039/d0an00836b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sequential imaging of a tissue section by MALDI and NAPA-LDI mass spectrometry provides enhanced molecular coverage.
Collapse
Affiliation(s)
- Jarod A. Fincher
- Department of Chemistry
- The George Washington University
- Washington
- USA
| | - Andrew R. Korte
- Department of Chemistry
- The George Washington University
- Washington
- USA
| | - Sridevi Yadavilli
- Research Center for Genetic Medicine
- Children's National Medical Center
- Washington
- USA
| | | | - Akos Vertes
- Department of Chemistry
- The George Washington University
- Washington
- USA
| |
Collapse
|
15
|
Sikora KN, Hardie JM, Castellanos-García LJ, Liu Y, Reinhardt BM, Farkas ME, Rotello VM, Vachet RW. Dual Mass Spectrometric Tissue Imaging of Nanocarrier Distributions and Their Biochemical Effects. Anal Chem 2019; 92:2011-2018. [PMID: 31825199 DOI: 10.1021/acs.analchem.9b04398] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nanomaterial-based drug delivery vehicles are able to deliver therapeutics in a controlled, targeted manner. Currently, however, there are limited analytical methods that can detect both nanomaterial distributions and their biochemical effects concurrently. In this study, we demonstrate that matrix assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) and laser ablation inductively coupled plasma mass spectrometry imaging (LA-ICP-MSI) can be used together to obtain nanomaterial distributions and biochemical consequences. These studies employ nanoparticle-stabilized capsules (NPSCs) loaded with siRNA as a testbed. MALDI-MSI experiments on spleen tissues from intravenously injected mice indicate that NPSCs loaded with anti-TNF-α siRNA cause changes to the lipid composition in white pulp regions of the spleen, as anticipated, based on pathways known to be affected by TNF-α, whereas NPSCs loaded with scrambled siRNA do not cause the predicted changes. Interestingly, LA-ICP-MSI experiments reveal that the NPSCs primarily localize in the red pulp, suggesting that the observed changes in lipid composition are due to diffusive rather than localized effects on TNF-α production. Such information is only accessible by combining data from the two modalities, which we accomplish by using the heme signals from MALDI-MSI and iron signals from LA-ICP-MSI to overlay the images. Several unexpected changes in lipid composition also occur in regions where the NPSCs are found, suggesting that the NPSCs themselves can influence tissue biochemistry as well.
Collapse
Affiliation(s)
- Kristen N Sikora
- Department of Chemistry , University of Massachusetts , Amherst , Massachusetts 01003 , United States
| | - Joseph M Hardie
- Department of Chemistry , University of Massachusetts , Amherst , Massachusetts 01003 , United States
| | | | - Yuanchang Liu
- Department of Chemistry , University of Massachusetts , Amherst , Massachusetts 01003 , United States
| | - Biidaaban M Reinhardt
- Department of Chemistry , University of Massachusetts , Amherst , Massachusetts 01003 , United States
| | - Michelle E Farkas
- Department of Chemistry , University of Massachusetts , Amherst , Massachusetts 01003 , United States
| | - Vincent M Rotello
- Department of Chemistry , University of Massachusetts , Amherst , Massachusetts 01003 , United States
| | - Richard W Vachet
- Department of Chemistry , University of Massachusetts , Amherst , Massachusetts 01003 , United States
| |
Collapse
|
16
|
Cassat JE, Moore JL, Wilson KJ, Stark Z, Prentice BM, Van de Plas R, Perry WJ, Zhang Y, Virostko J, Colvin DC, Rose KL, Judd AM, Reyzer ML, Spraggins JM, Grunenwald CM, Gore JC, Caprioli RM, Skaar EP. Integrated molecular imaging reveals tissue heterogeneity driving host-pathogen interactions. Sci Transl Med 2019. [PMID: 29540616 DOI: 10.1126/scitranslmed.aan6361] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diseases are characterized by distinct changes in tissue molecular distribution. Molecular analysis of intact tissues traditionally requires preexisting knowledge of, and reagents for, the targets of interest. Conversely, label-free discovery of disease-associated tissue analytes requires destructive processing for downstream identification platforms. Tissue-based analyses therefore sacrifice discovery to gain spatial distribution of known targets or sacrifice tissue architecture for discovery of unknown targets. To overcome these obstacles, we developed a multimodality imaging platform for discovery-based molecular histology. We apply this platform to a model of disseminated infection triggered by the pathogen Staphylococcus aureus, leading to the discovery of infection-associated alterations in the distribution and abundance of proteins and elements in tissue in mice. These data provide an unbiased, three-dimensional analysis of how disease affects the molecular architecture of complex tissues, enable culture-free diagnosis of infection through imaging-based detection of bacterial and host analytes, and reveal molecular heterogeneity at the host-pathogen interface.
Collapse
Affiliation(s)
- James E Cassat
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jessica L Moore
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Kevin J Wilson
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN 37232, USA
| | - Zach Stark
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN 37232, USA
| | - Boone M Prentice
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Raf Van de Plas
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA.,Delft Center for Systems and Control, Delft University of Technology, Delft, Netherlands
| | - William J Perry
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Yaofang Zhang
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - John Virostko
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN 37232, USA
| | - Daniel C Colvin
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN 37232, USA
| | - Kristie L Rose
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Audra M Judd
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Michelle L Reyzer
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Caroline M Grunenwald
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John C Gore
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN 37232, USA.,Departments of Radiology and Radiologic Sciences, Biomedical Engineering, Molecular Physiology and Biophysics, and Physics and Astronomy, Vanderbilt University, Nashville, TN 37232, USA
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA.,Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA. .,U.S. Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37232, USA
| |
Collapse
|
17
|
Ryan DJ, Patterson NH, Putnam NE, Wilde AD, Weiss A, Perry WJ, Cassat JE, Skaar EP, Caprioli RM, Spraggins JM. MicroLESA: Integrating Autofluorescence Microscopy, In Situ Micro-Digestions, and Liquid Extraction Surface Analysis for High Spatial Resolution Targeted Proteomic Studies. Anal Chem 2019; 91:7578-7585. [PMID: 31149808 PMCID: PMC6652190 DOI: 10.1021/acs.analchem.8b05889] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The ability to target discrete features within tissue using liquid surface extractions enables the identification of proteins while maintaining the spatial integrity of the sample. Here, we present a liquid extraction surface analysis (LESA) workflow, termed microLESA, that allows proteomic profiling from discrete tissue features of ∼110 μm in diameter by integrating nondestructive autofluorescence microscopy and spatially targeted liquid droplet micro-digestion. Autofluorescence microscopy provides the visualization of tissue foci without the need for chemical stains or the use of serial tissue sections. Tryptic peptides are generated from tissue foci by applying small volume droplets (∼250 pL) of enzyme onto the surface prior to LESA. The microLESA workflow reduced the diameter of the sampled area almost 5-fold compared to previous LESA approaches. Experimental parameters, such as tissue thickness, trypsin concentration, and enzyme incubation duration, were tested to maximize proteomics analysis. The microLESA workflow was applied to the study of fluorescently labeled Staphylococcus aureus infected murine kidney to identify unique proteins related to host defense and bacterial pathogenesis. Proteins related to nutritional immunity and host immune response were identified by performing microLESA at the infectious foci and surrounding abscess. These identifications were then used to annotate specific proteins observed in infected kidney tissue by MALDI FT-ICR IMS through accurate mass matching.
Collapse
Affiliation(s)
- Daniel J. Ryan
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue South #9160, Nashville, Tennessee 37235, United States
| | - Nathan Heath Patterson
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue South #9160, Nashville, Tennessee 37235, United States
- Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
| | - Nicole E. Putnam
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Aimee D. Wilde
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Andy Weiss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - William J. Perry
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue South #9160, Nashville, Tennessee 37235, United States
| | - James E. Cassat
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Eric P. Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- United States (U.S.) Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United States
| | - Richard M. Caprioli
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue South #9160, Nashville, Tennessee 37235, United States
- Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
- Department of Pharmacology, Vanderbilt University, 442 Robinson Research Building, 2220 Pierce Avenue, Nashville, Tennessee 37232, United States
- Department of Medicine, Vanderbilt University, 465 21st Ave South #9160, Nashville, Tennessee 37235, United States
| | - Jeffrey M. Spraggins
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, Tennessee 37235, United States
- Mass Spectrometry Research Center, Vanderbilt University, 465 21st Avenue South #9160, Nashville, Tennessee 37235, United States
- Department of Biochemistry, Vanderbilt University, 607 Light Hall, Nashville, Tennessee 37205, United States
| |
Collapse
|
18
|
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
|
19
|
|
20
|
Jaegger CF, Negrão F, Assis DM, Belaz KRA, Angolini CFF, Fernandes AMAP, Santos VG, Pimentel A, Abánades DR, Giorgio S, Eberlin MN, Rocha DFO. MALDI MS imaging investigation of the host response to visceral leishmaniasis. MOLECULAR BIOSYSTEMS 2018; 13:1946-1953. [PMID: 28758666 DOI: 10.1039/c7mb00306d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mass spectrometry imaging (MSI) of animal tissues has become an important tool for in situ molecular analyses and biomarker studies in several clinical areas, but there are few applications in parasitological studies. Leishmaniasis is a neglected tropical disease, and experimental mouse models have been essential to evaluate pathological and immunological processes and to develop diagnostic methods. Herein we have employed MALDI MSI to examine peptides and low molecular weight proteins (2 to 20 kDa) differentially expressed in the liver during visceral leishmaniasis in mice models. We analyzed liver sections of Balb/c mice infected with Leishmania infantum using the SCiLS Lab software for statistical analysis, which facilitated data interpretation and thus highlighted several key proteins and/or peptides. We proposed a decision tree classification for visceral leishmaniasis with distinct phases of the disease, which are named here as healthy, acute infection and chronic infection. Among others, the ion of m/z 4963 was the most important to identify acute infection and was tentatively identified as Thymosin β4. This peptide was previously established as a recovery factor in the human liver and might participate in the response of mice to Leishmania infection. This preliminary investigation shows the potential of MALDI MSI to complement classical compound selective imaging techniques and to explore new features not yet recognized by these approaches.
Collapse
Affiliation(s)
- C F Jaegger
- ThoMSon Mass Spectrometry Laboratory, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Host-based lipid inflammation drives pathogenesis in Francisella infection. Proc Natl Acad Sci U S A 2017; 114:12596-12601. [PMID: 29109289 DOI: 10.1073/pnas.1712887114] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mass spectrometry imaging (MSI) was used to elucidate host lipids involved in the inflammatory signaling pathway generated at the host-pathogen interface during a septic bacterial infection. Using Francisella novicida as a model organism, a bacterial lipid virulence factor (endotoxin) was imaged and identified along with host phospholipids involved in the splenic response in murine tissues. Here, we demonstrate detection and distribution of endotoxin in a lethal murine F. novicida infection model, in addition to determining the temporally and spatially resolved innate lipid inflammatory response in both 2D and 3D renderings using MSI. Further, we show that the cyclooxygenase-2-dependent lipid inflammatory pathway is responsible for lethality in F. novicida infection due to overproduction of proinflammatory effectors including prostaglandin E2. The results of this study emphasize that spatial determination of the host lipid components of the immune response is crucial to identifying novel strategies to effectively address highly pathogenic and lethal infections stemming from bacterial, fungal, and viral origins.
Collapse
|
22
|
Prentice BM, Caprioli RM, Vuiblet V. Label-free molecular imaging of the kidney. Kidney Int 2017; 92:580-598. [PMID: 28750926 PMCID: PMC6193761 DOI: 10.1016/j.kint.2017.03.052] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/25/2022]
Abstract
In this review, we will highlight technologies that enable scientists to study the molecular characteristics of tissues and/or cells without the need for antibodies or other labeling techniques. Specifically, we will focus on matrix-assisted laser desorption/ionization imaging mass spectrometry, infrared spectroscopy, and Raman spectroscopy.
Collapse
Affiliation(s)
- Boone M Prentice
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Richard M Caprioli
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA; Departments of Pharmacology and Medicine, Vanderbilt University, Nashville, Tennessee, USA; Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA.
| | - Vincent Vuiblet
- Biophotonic Laboratory, UMR CNRS 7369 URCA, Reims, France; Nephropathology, Department of Biopathology Laboratory, CHU de Reims, Reims, France; Nephrology and Renal Transplantation department, CHU de Reims, Reims, France.
| |
Collapse
|
23
|
Mass spectrometry imaging identifies palmitoylcarnitine as an immunological mediator during Salmonella Typhimurium infection. Sci Rep 2017; 7:2786. [PMID: 28584281 PMCID: PMC5459799 DOI: 10.1038/s41598-017-03100-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
Salmonella Typhimurium causes a self-limiting gastroenteritis that may lead to systemic disease. Bacteria invade the small intestine, crossing the intestinal epithelium from where they are transported to the mesenteric lymph nodes (MLNs) within migrating immune cells. MLNs are an important site at which the innate and adaptive immune responses converge but their architecture and function is severely disrupted during S. Typhimurium infection. To further understand host-pathogen interactions at this site, we used mass spectrometry imaging (MSI) to analyse MLN tissue from a murine model of S. Typhimurium infection. A molecule, identified as palmitoylcarnitine (PalC), was of particular interest due to its high abundance at loci of S. Typhimurium infection and MLN disruption. High levels of PalC localised to sites within the MLNs where B and T cells were absent and where the perimeter of CD169+ sub capsular sinus macrophages was disrupted. MLN cells cultured ex vivo and treated with PalC had reduced CD4+CD25+ T cells and an increased number of B220+CD19+ B cells. The reduction in CD4+CD25+ T cells was likely due to apoptosis driven by increased caspase-3/7 activity. These data indicate that PalC significantly alters the host response in the MLNs, acting as a decisive factor in infection outcome.
Collapse
|
24
|
The Future in Disease Models for Mass Spectrometry Imaging, Ethical Issues, and the Way Forward. Methods Mol Biol 2017. [PMID: 28523509 DOI: 10.1007/978-1-4939-7051-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Mass Spectrometry Imaging (MSI) has evolved into a valuable tool for research into and the diagnosis of disease pathology. The ability to perform multiplex analysis of a wide range of molecules (e.g., proteins, lipids, and metabolites) simultaneously per tissue section while retaining the histological structure of the sample allows molecular information and tissue morphology to be correlated, thus increasing our understanding of a particular disease. Further development of MSI is required to improve suitability to the alternative models available, so that the combined approach can successfully provide the information required in disease characterization and prevention. MSI has been shown to be capable of providing spatiomolecular information in tumor spheroids, living skin equivalents, and ex vivo human tissues. Due to a considerable interest and scientific effort there are many more designed alternative disease models available which would benefit from the information MSI could provide.
Collapse
|
25
|
Ho YN, Shu LJ, Yang YL. Imaging mass spectrometry for metabolites: technical progress, multimodal imaging, and biological interactions. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2017; 9. [PMID: 28488813 DOI: 10.1002/wsbm.1387] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/24/2017] [Accepted: 02/28/2017] [Indexed: 12/19/2022]
Abstract
Imaging mass spectrometry (IMS) allows the study of the spatial distribution of small molecules in biological samples. IMS is able to identify and quantify chemicals in situ from whole tissue sections to single cells. Both vacuum mass spectrometry (MS) and ambient MS systems have advanced considerably over the last decade; however, some limitations are still hard to surmount. Sample pretreatment, matrix or solvent choices, and instrument improvement are the key factors that determine the successful application of IMS to different samples and analytes. IMS with innovative MS analyzers, powerful MS spectrum databases, and analysis tools can efficiently dereplicate, identify, and quantify natural products. Moreover, multimodal imaging systems and multiple MS-based systems provide additional structural, chemical, and morphological information and are applied as complementary tools to explore new fields. IMS has been applied to reveal interactions between living organisms at molecular level. Recently, IMS has helped solve many previously unidentifiable relations between bacteria, fungi, plants, animals, and insects. Other significant interactions on the chemical level can also be resolved using expanding IMS techniques. WIREs Syst Biol Med 2017, 9:e1387. doi: 10.1002/wsbm.1387 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Ying-Ning Ho
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Lin-Jie Shu
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Liang Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
26
|
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
|
27
|
Profiling and identification of new proteins involved in brain ischemia using MALDI-imaging-mass-spectrometry. J Proteomics 2017; 152:243-253. [DOI: 10.1016/j.jprot.2016.11.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 10/24/2016] [Accepted: 11/19/2016] [Indexed: 12/21/2022]
|
28
|
FDR-controlled metabolite annotation for high-resolution imaging mass spectrometry. Nat Methods 2016; 14:57-60. [PMID: 27842059 DOI: 10.1038/nmeth.4072] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 10/13/2016] [Indexed: 02/08/2023]
Abstract
High-mass-resolution imaging mass spectrometry promises to localize hundreds of metabolites in tissues, cell cultures, and agar plates with cellular resolution, but it is hampered by the lack of bioinformatics tools for automated metabolite identification. We report pySM, a framework for false discovery rate (FDR)-controlled metabolite annotation at the level of the molecular sum formula, for high-mass-resolution imaging mass spectrometry (https://github.com/alexandrovteam/pySM). We introduce a metabolite-signal match score and a target-decoy FDR estimate for spatial metabolomics.
Collapse
|
29
|
Lough G, Kyriazakis I, Bergmann S, Lengeling A, Doeschl-Wilson AB. Health trajectories reveal the dynamic contributions of host genetic resistance and tolerance to infection outcome. Proc Biol Sci 2016; 282:rspb.2015.2151. [PMID: 26582028 PMCID: PMC4685823 DOI: 10.1098/rspb.2015.2151] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Resistance and tolerance are two alternative strategies hosts can adopt to survive infections. Both strategies may be genetically controlled. To date, the relative contribution of resistance and tolerance to infection outcome is poorly understood. Here, we use a bioluminescent Listeria monocytogenes (Lm) infection challenge model to study the genetic determination and dynamic contributions of host resistance and tolerance to listeriosis in four genetically diverse mouse strains. Using conventional statistical analyses, we detect significant genetic variation in both resistance and tolerance, but cannot capture the time-dependent relative importance of either host strategy. We overcome these limitations through the development of novel statistical tools to analyse individual infection trajectories portraying simultaneous changes in infection severity and health. Based on these tools, early expression of resistance followed by expression of tolerance emerge as important hallmarks for surviving Lm infections. Our trajectory analysis further reveals that survivors and non-survivors follow distinct infection paths (which are also genetically determined) and provides new survival thresholds as objective endpoints in infection experiments. Future studies may use trajectories as novel traits for mapping and identifying genes that control infection dynamics and outcome. A Matlab script for user-friendly trajectory analysis is provided.
Collapse
Affiliation(s)
- Graham Lough
- Genetics and Genomics Division, The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, UK
| | - Ilias Kyriazakis
- School of Agriculture, Food and Rural Development, Newcastle University, Newcastle upon Tyne, UK
| | - Silke Bergmann
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Lengeling
- Infection and Immunity Division, The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, UK
| | - Andrea B Doeschl-Wilson
- Genetics and Genomics Division, The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
30
|
Canela N, Rodríguez MÁ, Baiges I, Nadal P, Arola L. Foodomics imaging by mass spectrometry and magnetic resonance. Electrophoresis 2016; 37:1748-67. [DOI: 10.1002/elps.201500494] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 12/22/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Núria Canela
- Group of Research on Omic Methodologies (GROM); Universitat Rovira i Virgili; Reus Spain
- Centre for Omic Sciences (COS); Universitat Rovira i Virgili; Reus Spain
| | - Miguel Ángel Rodríguez
- Group of Research on Omic Methodologies (GROM); Universitat Rovira i Virgili; Reus Spain
- Centre for Omic Sciences (COS); Universitat Rovira i Virgili; Reus Spain
| | - Isabel Baiges
- Group of Research on Omic Methodologies (GROM); Universitat Rovira i Virgili; Reus Spain
- Centre for Omic Sciences (COS); Universitat Rovira i Virgili; Reus Spain
| | - Pedro Nadal
- Group of Research on Omic Methodologies (GROM); Universitat Rovira i Virgili; Reus Spain
- Centre for Omic Sciences (COS); Universitat Rovira i Virgili; Reus Spain
- Centre Tecnològic de Nutriciò i Salut (CTNS); Reus Spain
| | - Lluís Arola
- Centre for Omic Sciences (COS); Universitat Rovira i Virgili; Reus Spain
- Centre Tecnològic de Nutriciò i Salut (CTNS); Reus Spain
| |
Collapse
|
31
|
Longuespée R, Casadonte R, Kriegsmann M, Pottier C, Picard de Muller G, Delvenne P, Kriegsmann J, De Pauw E. MALDI mass spectrometry imaging: A cutting-edge tool for fundamental and clinical histopathology. Proteomics Clin Appl 2016; 10:701-19. [PMID: 27188927 DOI: 10.1002/prca.201500140] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 04/07/2016] [Accepted: 05/13/2016] [Indexed: 01/16/2023]
Abstract
Histopathological diagnoses have been done in the last century based on hematoxylin and eosin staining. These methods were complemented by histochemistry, electron microscopy, immunohistochemistry (IHC), and molecular techniques. Mass spectrometry (MS) methods allow the thorough examination of various biocompounds in extracts and tissue sections. Today, mass spectrometry imaging (MSI), and especially matrix-assisted laser desorption ionization (MALDI) imaging links classical histology and molecular analyses. Direct mapping is a major advantage of the combination of molecular profiling and imaging. MSI can be considered as a cutting edge approach for molecular detection of proteins, peptides, carbohydrates, lipids, and small molecules in tissues. This review covers the detection of various biomolecules in histopathological sections by MSI. Proteomic methods will be introduced into clinical histopathology within the next few years.
Collapse
Affiliation(s)
- Rémi Longuespée
- Proteopath GmbH, Trier, Germany.,Mass Spectrometry Laboratory, GIGA-Research, Department of Chemistry, University of Liège, Liège, Belgium
| | | | - Mark Kriegsmann
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Charles Pottier
- Laboratory of Experimental Pathology, GIGA-Cancer, Department of Pathology, University of Liège, Liège, Belgium
| | | | - Philippe Delvenne
- Laboratory of Experimental Pathology, GIGA-Cancer, Department of Pathology, University of Liège, Liège, Belgium
| | - Jörg Kriegsmann
- Proteopath GmbH, Trier, Germany.,MVZ for Histology, Cytology and Molecular Diagnostics Trier, Trier, Germany
| | - Edwin De Pauw
- Mass Spectrometry Laboratory, GIGA-Research, Department of Chemistry, University of Liège, Liège, Belgium
| |
Collapse
|
32
|
Patterson NH, Doonan RJ, Daskalopoulou SS, Dufresne M, Lenglet S, Montecucco F, Thomas A, Chaurand P. Three-dimensional imaging MS of lipids in atherosclerotic plaques: Open-source methods for reconstruction and analysis. Proteomics 2016; 16:1642-51. [PMID: 26958804 DOI: 10.1002/pmic.201500490] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/01/2016] [Accepted: 03/03/2016] [Indexed: 01/01/2023]
Affiliation(s)
| | - Robert J. Doonan
- Department of Medicine, Faculty of Medicine; McGill University; Montreal Quebec Canada
| | | | - Martin Dufresne
- Department of Chemistry; University of Montreal; Montreal Quebec Canada
| | - Sébastien Lenglet
- Unit of Toxicology; University Centre of Legal Medicine; Geneva-Lausanne Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine; University of Genoa; Genoa Italy
- Division of Cardiology, Foundation for Medical Researches, Faculty of Medicine; University of Geneva; Geneva Switzerland
| | - Aurélien Thomas
- Unit of Toxicology; University Centre of Legal Medicine; Geneva-Lausanne Switzerland
- Faculty of Biology and Medicine; Lausanne University Hospital; University of Lausanne; Lausanne Switzerland
| | - Pierre Chaurand
- Department of Chemistry; University of Montreal; Montreal Quebec Canada
| |
Collapse
|
33
|
Olive AJ, Sassetti CM. Metabolic crosstalk between host and pathogen: sensing, adapting and competing. Nat Rev Microbiol 2016; 14:221-34. [PMID: 26949049 DOI: 10.1038/nrmicro.2016.12] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Our understanding of bacterial pathogenesis is dominated by the cell biology of the host-pathogen interaction. However, the majority of metabolites that are used in prokaryotic and eukaryotic physiology and signalling are chemically similar or identical. Therefore, the metabolic crosstalk between pathogens and host cells may be as important as the interactions between bacterial effector proteins and their host targets. In this Review we focus on host-pathogen interactions at the metabolic level: chemical signalling events that enable pathogens to sense anatomical location and the local physiology of the host; microbial metabolic pathways that are dedicated to circumvent host immune mechanisms; and a few metabolites as central points of competition between the host and bacterial pathogens.
Collapse
Affiliation(s)
- Andrew J Olive
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Christopher M Sassetti
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| |
Collapse
|
34
|
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: 127] [Impact Index Per Article: 14.1] [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
|
35
|
Spraggins JM, Rizzo DG, Moore JL, Rose KL, Hammer ND, Skaar EP, Caprioli RM. MALDI FTICR IMS of Intact Proteins: Using Mass Accuracy to Link Protein Images with Proteomics Data. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:974-85. [PMID: 25904064 PMCID: PMC4442642 DOI: 10.1007/s13361-015-1147-5] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 03/20/2015] [Accepted: 03/20/2015] [Indexed: 05/11/2023]
Abstract
MALDI imaging mass spectrometry is a highly sensitive and selective tool used to visualize biomolecules in tissue. However, identification of detected proteins remains a difficult task. Indirect identification strategies have been limited by insufficient mass accuracy to confidently link ion images to proteomics data. Here, we demonstrate the capabilities of MALDI FTICR MS for imaging intact proteins. MALDI FTICR IMS provides an unprecedented combination of mass resolving power (~75,000 at m/z 5000) and accuracy (<5ppm) for proteins up to ~12kDa, enabling identification based on correlation with LC-MS/MS proteomics data. Analysis of rat brain tissue was performed as a proof-of-concept highlighting the capabilities of this approach by imaging and identifying a number of proteins including N-terminally acetylated thymosin β(4) (m/z 4,963.502, 0.6ppm) and ATP synthase subunit ε (m/z 5,636.074, -2.3ppm). MALDI FTICR IMS was also used to differentiate a series of oxidation products of S100A8 (m/z 10,164.03, -2.1ppm), a subunit of the heterodimer calprotectin, in kidney tissue from mice infected with Staphylococcus aureus. S100A8 - M37O/C42O(3) (m/z 10228.00, -2.6ppm) was found to co-localize with bacterial microcolonies at the center of infectious foci. The ability of MALDI FTICR IMS to distinguish S100A8 modifications is critical to understanding calprotectin's roll in nutritional immunity.
Collapse
Affiliation(s)
- Jeffrey M Spraggins
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37205, USA,
| | | | | | | | | | | | | |
Collapse
|
36
|
Palmer AD, Alexandrov T. Serial 3D imaging mass spectrometry at its tipping point. Anal Chem 2015; 87:4055-62. [PMID: 25817912 DOI: 10.1021/ac504604g] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Since biology is by and large a 3-dimensional phenomenon, it is hardly surprising that 3D imaging has had a significant impact on many challenges in the life sciences. Imaging mass spectrometry (MS) is a spatially resolved label-free analytical technique that recently maturated into a powerful tool for in situ localization of hundreds of molecular species. Serial 3D imaging MS reconstructs 3D molecular images from serial sections imaged with mass spectrometry. As such, it provides a novel 3D imaging modality inheriting the advantages of imaging MS. Serial 3D imaging MS has been steadily developing over the past decade, and many of the technical challenges have been met. Essential tools and protocols were developed, in particular to improve the reproducibility of sample preparation, speed up data acquisition, and enable computationally intensive analysis of the big data generated. As a result, experimental data is starting to emerge that takes advantage of the extra spatial dimension that 3D imaging MS offers. Most studies still focus on method development rather than on exploring specific biological problems. The future success of 3D imaging MS requires it to find its own niche alongside existing 3D imaging modalities through finding applications that benefit from 3D imaging and at the same time utilize the unique chemical sensitivity of imaging mass spectrometry. This perspective critically reviews the challenges encountered during the development of serial-sectioning 3D imaging MS and discusses the steps needed to tip it from being an academic curiosity into a tool of choice for answering biological and medical questions.
Collapse
Affiliation(s)
- Andrew D Palmer
- †European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany.,‡Center for Industrial Mathematics, University of Bremen, 28359 Bremen, Germany
| | - Theodore Alexandrov
- †European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany.,‡Center for Industrial Mathematics, University of Bremen, 28359 Bremen, Germany.,§SCiLS GmbH, 28359 Bremen, Germany.,∥Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, San Diego, California 92161, United States
| |
Collapse
|
37
|
Ly A, Schöne C, Becker M, Rattke J, Meding S, Aichler M, Suckau D, Walch A, Hauck SM, Ueffing M. High-resolution MALDI mass spectrometric imaging of lipids in the mammalian retina. Histochem Cell Biol 2014; 143:453-62. [DOI: 10.1007/s00418-014-1303-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2014] [Indexed: 12/12/2022]
|
38
|
Sun N, Ly A, Meding S, Witting M, Hauck SM, Ueffing M, Schmitt-Kopplin P, Aichler M, Walch A. High-resolution metabolite imaging of light and dark treated retina using MALDI-FTICR mass spectrometry. Proteomics 2014; 14:913-23. [PMID: 24459044 DOI: 10.1002/pmic.201300407] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/06/2013] [Accepted: 12/20/2013] [Indexed: 11/06/2022]
Abstract
MS imaging (MSI) is a valuable tool for diagnostics and systems biology studies, being a highly sensitive, label-free technique capable of providing comprehensive spatial distribution of different classes of biomolecules. The application of MSI to the study of endogenous compounds has received considerable attention because metabolites are the result of the interactions of a biosystem with its environment. MSI can therefore enhance understanding of disease mechanisms and elucidate mechanisms for biological variation. We present the in situ comparative metabolomics imaging data for analyses of light- and dark-treated retina using MALDI-FTICR. A wide variety of tissue metabolites were imaged at a high spatial resolution. These include nucleotides, central carbon metabolism pathway intermediates, 2-oxocarboxylic acid metabolism, oxidative phosphorylation, glycerophospholipid metabolism, and cysteine and methionine metabolites. The high lateral resolution enabled the differentiation of retinal layers, allowing determination of the spatial distributions of different endogenous compounds. A number of metabolites demonstrated differences between light and dark conditions. These findings add to the understanding of metabolic activity in the retina.
Collapse
Affiliation(s)
- Na Sun
- Research Unit Analytical Pathology, Institute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Charwat V, Joksch M, Sticker D, Purtscher M, Rothbauer M, Ertl P. Monitoring cellular stress responses using integrated high-frequency impedance spectroscopy and time-resolved ELISA. Analyst 2014; 139:5271-82. [PMID: 25137192 DOI: 10.1039/c4an00824c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We have developed a lab-on-a-chip system for continuous and non-invasive monitoring of microfluidic cell cultures using integrated high-frequency contactless impedance spectroscopy. Electrically insulated microfabricated interdigitated electrode structures were embedded into four individually addressable microchambers to reliably and reproducibly detect cell-substrate interactions, cell viability and metabolic activity. While silicon nitride passivated sensor substrates provided a homogeneous cell culture surface that minimized cell orientation along interdigitated electrode structures, the application of high-frequency AC fields reduced the impact of the 300 nm thick passivation layer on sensor sensitivity. The additional implementation of multivariate data analysis methods such as partial least square (PLS) for high-frequency impedance spectra provided unambiguous information on intracellular pathway activation, up and down-regulation of protein synthesis as well as global cellular stress responses. A comparative cell analysis using connective tissue fibroblasts showed that high-frequency contactless impedance spectroscopy and time-resolved quantification of IL-6 secretion using ELISA provided similar results following stimulation with circulating pro-inflammatory cytokines IL-1β and TNFα. The combination of microfluidics with contactless impedance sensing and time-resolved quantification of stress factor release will provide biologist with a new tool to (a) establish a variety of uniform cell culture surfaces that feature complex biochemistries, micro- and nanopatterns; and (b) to simultaneously characterize cell responses under physiologically relevant conditions using a complementary non-invasive cell analysis method.
Collapse
Affiliation(s)
- Verena Charwat
- BioSensor Technologies, AIT Austrian Institute of Technology GmbH, Muthgasse 11, 1190 Vienna, Austria.
| | | | | | | | | | | |
Collapse
|
40
|
Moore JL, Caprioli RM, Skaar EP. Advanced mass spectrometry technologies for the study of microbial pathogenesis. Curr Opin Microbiol 2014; 19:45-51. [PMID: 24997399 PMCID: PMC4125470 DOI: 10.1016/j.mib.2014.05.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 05/12/2014] [Accepted: 05/28/2014] [Indexed: 02/08/2023]
Abstract
Matrix-assisted laser desorption/ionization mass spectrometry (MALDI MS) has been successfully applied to the field of microbial pathogenesis with promising results, principally in diagnostic microbiology to rapidly identify bacteria based on the molecular profiles of small cell populations. Direct profiling of molecules from serum and tissue samples by MALDI MS provides a means to study the pathogen-host interaction and to discover potential markers of infection. Systematic molecular profiling across tissue sections represents a new imaging modality, enabling regiospecific molecular measurements to be made in situ, in both two-dimensional and three-dimensional analyses. Herein, we briefly summarize work that employs MALDI MS to study the pathogenesis of microbial infection.
Collapse
Affiliation(s)
- Jessica L Moore
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, United States
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, United States.
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States.
| |
Collapse
|
41
|
Seeley EH, Wilson KJ, Yankeelov TE, Johnson RW, Gore JC, Caprioli RM, Matrisian LM, Sterling JA. Co-registration of multi-modality imaging allows for comprehensive analysis of tumor-induced bone disease. Bone 2014; 61:208-16. [PMID: 24487126 PMCID: PMC4005328 DOI: 10.1016/j.bone.2014.01.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 01/22/2014] [Accepted: 01/24/2014] [Indexed: 01/08/2023]
Abstract
Bone metastases are a clinically significant problem that arises in approximately 70% of metastatic breast cancer patients. Once established in the bone, tumor cells induce changes in the bone microenvironment that lead to bone destruction, pain, and significant morbidity. While much is known about the later stages of bone disease, less is known about the earlier stages or the changes in protein expression in the tumor micro-environment. Due to promising results of combining magnetic resonance imaging (MRI) and Matrix-Assisted Laser Desorption/Ionization Imaging Mass Spectrometry (MALDI IMS) ion images in the brain, we developed methods for applying these modalities to models of tumor-induced bone disease in order to better understand the changes in protein expression that occur within the tumor-bone microenvironment. Specifically, we integrated 3-dimensional-volume reconstructions of spatially resolved MALDI IMS with high-resolution anatomical and diffusion weighted MRI data and histology in an intratibial model of breast tumor-induced bone disease. This approach enables us to analyze proteomic profiles from MALDI IMS data with corresponding in vivo imaging and ex vivo histology data. To the best of our knowledge, this is the first time that these three modalities have been rigorously registered in the bone. The MALDI mass-to-charge ratio peaks indicate differential expression of calcyclin, ubiquitin, and other proteins within the tumor cells, while peaks corresponding to hemoglobin A and calgranulin A provided molecular information that aided in the identification of areas rich in red and white blood cells, respectively. This multi-modality approach will allow us to comprehensively understand the bone-tumor microenvironment and thus may allow us to better develop and test approaches for inhibiting bone metastases.
Collapse
Affiliation(s)
- Erin H Seeley
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
| | - Kevin J Wilson
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Thomas E Yankeelov
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Physics and Astronomy, Vanderbilt University, Nashville, TN, USA
| | - Rachelle W Johnson
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Bone Biology, Vanderbilt University, Nashville, TN, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Physics and Astronomy, Vanderbilt University, Nashville, TN, USA
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, USA; Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Lynn M Matrisian
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Julie A Sterling
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Bone Biology, Vanderbilt University, Nashville, TN, USA; Department of Medicine, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
42
|
Gessel MM, Norris JL, Caprioli RM. MALDI imaging mass spectrometry: spatial molecular analysis to enable a new age of discovery. J Proteomics 2014; 107:71-82. [PMID: 24686089 DOI: 10.1016/j.jprot.2014.03.021] [Citation(s) in RCA: 190] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 03/20/2014] [Indexed: 12/26/2022]
Abstract
Matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) combines the sensitivity and selectivity of mass spectrometry with spatial analysis to provide a new dimension for histological analyses to provide unbiased visualization of the arrangement of biomolecules in tissue. As such, MALDI IMS has the capability to become a powerful new molecular technology for the biological and clinical sciences. In this review, we briefly describe several applications of MALDI IMS covering a range of molecular weights, from drugs to proteins. Current limitations and challenges are discussed along with recent developments to address these issues. This article is part of a Special Issue entitled: 20years of Proteomics in memory of Viatliano Pallini. Guest Editors: Luca Bini, Juan J. Calvete, Natacha Turck, Denis Hochstrasser and Jean-Charles Sanchez.
Collapse
Affiliation(s)
- Megan M Gessel
- National Research Resource for Imaging Mass Spectrometry, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, 9160 Medical Research Building III, 465 21st Avenue South, Nashville, TN 37232-8575, United States; Department of Biochemistry, Vanderbilt University School of Medicine, 9160 Medical Research Building III, 465 21st Avenue South, Nashville, TN 37232-8575, United States
| | - Jeremy L Norris
- National Research Resource for Imaging Mass Spectrometry, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, 9160 Medical Research Building III, 465 21st Avenue South, Nashville, TN 37232-8575, United States; Department of Biochemistry, Vanderbilt University School of Medicine, 9160 Medical Research Building III, 465 21st Avenue South, Nashville, TN 37232-8575, United States
| | - Richard M Caprioli
- National Research Resource for Imaging Mass Spectrometry, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, 9160 Medical Research Building III, 465 21st Avenue South, Nashville, TN 37232-8575, United States; Department of Biochemistry, Vanderbilt University School of Medicine, 9160 Medical Research Building III, 465 21st Avenue South, Nashville, TN 37232-8575, United States.
| |
Collapse
|
43
|
Cillero-Pastor B, Heeren RMA. Matrix-Assisted Laser Desorption Ionization Mass Spectrometry Imaging for Peptide and Protein Analyses: A Critical Review of On-Tissue Digestion. J Proteome Res 2013; 13:325-35. [DOI: 10.1021/pr400743a] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Berta Cillero-Pastor
- FOM Institute AMOLF, Biomolecular Imaging Mass Spectrometry (BIMS), AMOLF Science Park 104, 1098 XG Amsterdam, The Netherlands
| | - Ron M. A. Heeren
- FOM Institute AMOLF, Biomolecular Imaging Mass Spectrometry (BIMS), AMOLF Science Park 104, 1098 XG Amsterdam, The Netherlands
| |
Collapse
|
44
|
Oetjen J, Aichler M, Trede D, Strehlow J, Berger J, Heldmann S, Becker M, Gottschalk M, Kobarg JH, Wirtz S, Schiffler S, Thiele H, Walch A, Maass P, Alexandrov T. MRI-compatible pipeline for three-dimensional MALDI imaging mass spectrometry using PAXgene fixation. J Proteomics 2013; 90:52-60. [DOI: 10.1016/j.jprot.2013.03.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 03/02/2013] [Accepted: 03/19/2013] [Indexed: 12/18/2022]
|
45
|
Moore JL, Becker KW, Nicklay JJ, Boyd KL, Skaar EP, Caprioli RM. Imaging mass spectrometry for assessing temporal proteomics: analysis of calprotectin in Acinetobacter baumannii pulmonary infection. Proteomics 2013; 14:820-828. [PMID: 23754577 DOI: 10.1002/pmic.201300046] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 04/23/2013] [Accepted: 05/11/2013] [Indexed: 12/13/2022]
Abstract
Imaging MS is routinely used to show spatial localization of proteins within a tissue sample and can also be employed to study temporal protein dynamics. The antimicrobial S100 protein calprotectin, a heterodimer of subunits S100A8 and S100A9, is an abundant cytosolic component of neutrophils. Using imaging MS, calprotectin can be detected as a marker of the inflammatory response to bacterial challenge. In a murine model of Acinetobacter baumannii pneumonia, protein images of S100A8 and S100A9 collected at different time points throughout infection aid in visualization of the innate immune response to this pathogen. Calprotectin is detectable within 6 h of infection as immune cells respond to the invading pathogen. As the bacterial burden decreases, signals from the inflammatory proteins decrease. Calprotectin is no longer detectable 96-144 h post infection, correlating to a lack of detectable bacterial burden in lungs. These experiments provide a label-free, multiplexed approach to study host response to a bacterial threat and eventual clearance of the pathogen over time.
Collapse
Affiliation(s)
- Jessica L Moore
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA.,Mass Spectrometry Research Center, Vanderbilt University, School of Medicine, Nashville, TN, USA
| | - Kyle W Becker
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joshua J Nicklay
- Mass Spectrometry Research Center, Vanderbilt University, School of Medicine, Nashville, TN, USA.,Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Richard M Caprioli
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA.,Mass Spectrometry Research Center, Vanderbilt University, School of Medicine, Nashville, TN, USA.,Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA.,Departments of Pharmacology and Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW There is still a fundamental lack of understanding of what protected vaccinee's in the moderately successful RV144 Thailand trial. It is clear that better tools are needed to identify and study correlates of protection and immune responses to vaccine challenge. Quantitative mass spectrometry (MS) has evolved considerably to become a useful tool in biomarker discovery; however, until recently it has been scarcely used to define host responses to HIV exposure and/or viral infection. In this review we discuss current quantitative MS techniques, their application in current HIV studies as well as novel approaches that could be used to better examine innate or adaptive immune responses in HIV vaccine or microbicide trials. RECENT FINDINGS Several recently published studies have allowed researchers to utilize quantitative MS as part of a systems biology approach to better understand the HIV-affected host's interaction with HIV and/or vaccine challenge. Proteomics has shown it can play a major role in studies to demonstrate insight into HIV replication, early stages of pathogenesis, and identify potential correlates of mucosal protection. SUMMARY Novel advances in quantitative proteomic techniques are allowing the opportunity to profile and evaluate HIV specific innate and adaptive immune responses, and will increase our understanding of HIV pathogenesis.
Collapse
|
47
|
MntABC and MntH contribute to systemic Staphylococcus aureus infection by competing with calprotectin for nutrient manganese. Infect Immun 2013; 81:3395-405. [PMID: 23817615 DOI: 10.1128/iai.00420-13] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
During infection, vertebrates limit access to manganese and zinc, starving invading pathogens, such as Staphylococcus aureus, of these essential metals in a process termed "nutritional immunity." The manganese and zinc binding protein calprotectin is a key component of the nutrient-withholding response, and mice lacking this protein do not sequester manganese from S. aureus liver abscesses. One potential mechanism utilized by S. aureus to minimize host-imposed manganese and zinc starvation is the expression of the metal transporters MntABC and MntH. We performed transcriptional analyses of both mntA and mntH, which revealed increased expression of both systems in response to calprotectin treatment. MntABC and MntH compete with calprotectin for manganese, which enables S. aureus growth and retention of manganese-dependent superoxide dismutase activity. Loss of MntABC and MntH results in reduced staphylococcal burdens in the livers of wild-type but not calprotectin-deficient mice, suggesting that these systems promote manganese acquisition during infection. During the course of these studies, we observed that metal content and the importance of calprotectin varies between murine organs, and infection leads to profound changes in the anatomical distribution of manganese and zinc. In total, these studies provide insight into the mechanisms utilized by bacteria to evade host-imposed nutrient metal starvation and the critical importance of restricting manganese availability during infection.
Collapse
|
48
|
Angel PM, Caprioli RM. Matrix-assisted laser desorption ionization imaging mass spectrometry: in situ molecular mapping. Biochemistry 2013; 52:3818-28. [PMID: 23259809 PMCID: PMC3864574 DOI: 10.1021/bi301519p] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Matrix-assisted laser desorption ionization imaging mass spectrometry (IMS) is a relatively new imaging modality that allows mapping of a wide range of biomolecules within a thin tissue section. The technology uses a laser beam to directly desorb and ionize molecules from discrete locations on the tissue that are subsequently recorded in a mass spectrometer. IMS is distinguished by the ability to directly measure molecules in situ ranging from small metabolites to proteins, reporting hundreds to thousands of expression patterns from a single imaging experiment. This article reviews recent advances in IMS technology, applications, and experimental strategies that allow it to significantly aid in the discovery and understanding of molecular processes in biological and clinical samples.
Collapse
Affiliation(s)
- Peggi M. Angel
- Mass Spectrometry Research Center and Department of Biochemistry, Vanderbilt University Medical Center, 465 21st Avenue South, MRB III Suite 9160, Nashville, Tennessee 37232, United States
| | - Richard M. Caprioli
- Mass Spectrometry Research Center and Department of Biochemistry, Medicine, Pharmacology, and Chemistry, Vanderbilt University Medical Center, 465 21st Avenue South, MRB III Suite 9160, Nashville, Tennessee 37232, United States
| |
Collapse
|
49
|
Norris JL, Caprioli RM. Analysis of tissue specimens by matrix-assisted laser desorption/ionization imaging mass spectrometry in biological and clinical research. Chem Rev 2013; 113:2309-42. [PMID: 23394164 PMCID: PMC3624074 DOI: 10.1021/cr3004295] [Citation(s) in RCA: 526] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jeremy L. Norris
- National Research Resource for Imaging Mass Spectrometry, Mass Spectrometry Research Center, and Department of Biochemistry, Vanderbilt University School of Medicine, 9160 Medical Research Building III, 465 21st Avenue South, Nashville, TN 37232-8575
| | - Richard M. Caprioli
- National Research Resource for Imaging Mass Spectrometry, Mass Spectrometry Research Center, and Department of Biochemistry, Vanderbilt University School of Medicine, 9160 Medical Research Building III, 465 21st Avenue South, Nashville, TN 37232-8575
| |
Collapse
|
50
|
A consideration of biomarkers to be used for evaluation of inflammation in human nutritional studies. Br J Nutr 2013; 109 Suppl 1:S1-34. [PMID: 23343744 DOI: 10.1017/s0007114512005119] [Citation(s) in RCA: 262] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
To monitor inflammation in a meaningful way, the markers used must be valid: they must reflect the inflammatory process under study and they must be predictive of future health status. In 2009, the Nutrition and Immunity Task Force of the International Life Sciences Institute, European Branch, organized an expert group to attempt to identify robust and predictive markers, or patterns or clusters of markers, which can be used to assess inflammation in human nutrition studies in the general population. Inflammation is a normal process and there are a number of cells and mediators involved. These markers are involved in, or are produced as a result of, the inflammatory process irrespective of its trigger and its location and are common to all inflammatory situations. Currently, there is no consensus as to which markers of inflammation best represent low-grade inflammation or differentiate between acute and chronic inflammation or between the various phases of inflammatory responses. There are a number of modifying factors that affect the concentration of an inflammatory marker at a given time, including age, diet and body fatness, among others. Measuring the concentration of inflammatory markers in the bloodstream under basal conditions is probably less informative compared with data related to the concentration change in response to a challenge. A number of inflammatory challenges have been described. However, many of these challenges are poorly standardised. Patterns and clusters may be important as robust biomarkers of inflammation. Therefore, it is likely that a combination of multiple inflammatory markers and integrated readouts based upon kinetic analysis following defined challenges will be the most informative biomarker of inflammation.
Collapse
|