1
|
van der Vloet L, Hilaire PBS, Bouillod C, Isin EM, Heeren RMA, Vandenbosch M. How can MSI enhance our understanding of ASO distribution? Drug Discov Today 2024; 30:104275. [PMID: 39701373 DOI: 10.1016/j.drudis.2024.104275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 12/05/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
In the dynamic field of drug discovery and development, a comprehensive understanding of drug absorption, distribution, metabolism, excretion, and toxicity is crucial. Mass spectrometry imaging (MSI) has become a key analytical tool in the pharmaceutical industry, allowing evaluation of drug biodistribution and molecular profiles. Antisense oligonucleotides (ASOs) are emerging drug candidates for treating neurologic diseases. This review explores the potential of MSI in investigating ASOs' spatial distribution within neurological disease models. Here, we focus on multimodal molecular imaging to gain insights into ASO distribution, simultaneously with a better understanding of the molecular pathways affected by ASOs. An improved understanding of therapeutic ASOs in tissue will potentially improve neurologic therapies, emphasizing their importance in patient care.
Collapse
Affiliation(s)
- Laura van der Vloet
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | | | - Christophe Bouillod
- Institut de Recherche et Développement Servier Paris-Saclay, Rue Francis Perrin, 91190 Gif-sur-Yvette, France
| | - Emre M Isin
- Institut de Recherche et Développement Servier Paris-Saclay, Rue Francis Perrin, 91190 Gif-sur-Yvette, France
| | - Ron M A Heeren
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.
| | - Michiel Vandenbosch
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
2
|
Lai H, Fan P, Wang H, Wang Z, Chen N. New perspective on central nervous system disorders: focus on mass spectrometry imaging. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:8080-8102. [PMID: 39508396 DOI: 10.1039/d4ay01205d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
An abnormally organized brain spatial network is linked to the development of various central nervous system (CNS) disorders, including neurodegenerative diseases and neuropsychiatric disorders. However, the complicated molecular mechanisms of these diseases remain unresolved, making the development of treatment strategies difficult. A novel molecular imaging technique, called mass spectrometry imaging (MSI), captures molecular information on the surface of samples in situ. With MSI, multiple compounds can be simultaneously visualized in a single experiment. The high spatial resolution enables the simultaneous visualization of the spatial distribution and relative content of various compounds. The wide application of MSI in biomedicine has facilitated extensive studies on CNS disorders in recent years. This review provides a concise overview of the processes, applications, advantages, and disadvantages, as well as mechanisms of the main types of MSI. Meanwhile, this review summarizes the main applications of MSI in studying CNS diseases, including Alzheimer's disease (AD), CNS tumors, stroke, depression, Huntington's disease (HD), and Parkinson's disease (PD). Finally, this review comprehensively discusses the synergistic application of MSI with other advanced imaging modalities, its utilization in organoid models, its integration with spatial omics techniques, and provides an outlook on its future potential in single-cell analysis.
Collapse
Affiliation(s)
- Huaqing Lai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Pinglong Fan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Huiqin Wang
- Hunan University of Chinese Medicine, Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha 410208, Hunan, China
| | - Zhenzhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Naihong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
3
|
Qiu T. Mass Spectrometry Imaging for Spatial Toxicology Research. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5104. [PMID: 39624029 PMCID: PMC11612705 DOI: 10.1002/jms.5104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 12/06/2024]
Abstract
The spatial information of xenobiotics distribution, metabolism, and toxicity mechanisms in situ has drawn increasing attention in both pharmaceutical and environmental toxicology research to aid drug development and environmental risk assessments. Mass spectrometry imaging (MSI) provides a label-free, multiplexed, and high-throughput tool to characterize xenobiotics, their metabolites, and endogenous molecules in situ with spatial resolution, providing knowledge on spatially resolved absorption, distribution, metabolism, excretion, and toxicity on the molecular level. In this perspective, we briefly summarize applications of MSI in toxicology on xenobiotic distribution and metabolism, quantification, toxicity mechanisms, and biomarker discovery. We identified several challenges regarding how we can fully harness the power of MSI in both fundamental toxicology research and regulatory practices. First, how can we increase the coverage, sensitivity, and specificity in detecting xenobiotics and their metabolites in complex biological matrices? Second, how can we link the spatial molecular information of xenobiotics to toxicity consequences to understand toxicity mechanisms, predict exposure outcomes, and aid biomarker discovery? Finally, how can we standardize the MSI experiment and data analysis workflow to provide robust conclusions for regulation and drug development? With these questions in mind, we provide our perspectives on the future directions of MSI as a promising tool in spatial toxicology research.
Collapse
Affiliation(s)
- Tian (Autumn) Qiu
- Department of ChemistryMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
4
|
Moore AM, Bowman A, Wali SN, Weigand MR, Wagner D, Yang J, Laskin J. Quantitative Analysis of Drugs in a Mimetic Tissue Model Using Nano-DESI on a Triple Quadrupole Mass Spectrometer. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024. [PMID: 39485739 DOI: 10.1021/jasms.4c00345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Mass spectrometry is a powerful analytical technique used at every stage of the pharmaceutical research process. A specialized branch of this method, mass spectrometry imaging (MSI), has emerged as an important tool for determining the spatial distribution of drugs in biological samples. Despite the importance of MSI, its quantitative capabilities are still limited due to the complexity of biological samples and the lack of separation prior to analysis. This makes the simultaneous quantification and visualization of analytes challenging. Several techniques have been developed to address this challenge and enable quantitative MSI. One such approach is the mimetic tissue model, which involves the incorporation of an analyte of interest into tissue homogenates at several concentrations. A calibration curve that accounts for signal suppression by the complex biological matrix is then created by measuring the signal of the analyte in the series of tissue homogenates. Herein, we use the mimetic tissue model on a triple quadrupole mass spectrometer (QqQ) in multiple reaction monitoring mode to demonstrate the quantitative abilities of nanospray desorption electrospray ionization (nano-DESI) and compare these results with those obtained using atmospheric pressure matrix-assisted laser desorption/ionization (AP-MALDI). For the tested compounds, our findings indicate that nano-DESI achieves lower standard deviations than AP-MALDI, resulting in superior limits of detection for the studied analytes. Additionally, we discuss the limitations of the mimetic tissue model in the quantification of certain analytes and the challenges involved with the implementation of the model.
Collapse
Affiliation(s)
- Alyssa M Moore
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Andrew Bowman
- AbbVie Incorporated, North Chicago, Illinois 60064, United States
| | - Syeda Nazifa Wali
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Miranda R Weigand
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - David Wagner
- AbbVie Incorporated, North Chicago, Illinois 60064, United States
| | - Junhai Yang
- AbbVie Incorporated, North Chicago, Illinois 60064, United States
| | - Julia Laskin
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
5
|
Xie F, Gales T, Ringenberg MA, Wolf AI, Groseclose MR. Characterizing the Distribution of a Stimulator of Interferon Genes Agonist and Its Metabolites in Mouse Liver by Matrix-Assisted Laser Desorption/Ionization Imaging Mass Spectrometry. Drug Metab Dispos 2024; 52:1181-1186. [PMID: 37884391 DOI: 10.1124/dmd.122.001076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/06/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
A STING (stimulator of interferon genes) agonist GSK3996915 under investigation in early discovery for hepatitis B was orally dosed to a mouse model for understanding the parent drug distribution in liver, the target organ. Matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) was used to quantify the distribution of GSK3996915 in liver collected from mice administered a single oral dose at 90 mg/kg. GSK3996915 was detected with a zonal distribution localized in the portal triad and highly concentrated in the main bile ducts, indicating clearance through biliary excretion. High spatial resolution imaging showed the distribution of the parent drug localized to the cellular populations in the sinusoids, including the Kupffer cells. Additionally, a series of drug-related metabolites were observed to be localized in the central zones of the liver. These results exemplify the potential of utilizing MALDI IMS for measuring not only quantitative drug distribution and target exposure but also drug metabolism and elimination in a single suite of experiments. SIGNIFICANCE STATEMENT: An integrated imaging approach utilizing matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) complemented with immunohistochemistry (IHC) and histology was used to address the question of target exposure at the cellular level. Localized quantification of the parent drug in the target organ and identification of potential metabolites in the context of tissue histology were also achieved in one experimental suite to support characterization of pharmacokinetic properties of the drug in the early discovery stage.
Collapse
Affiliation(s)
- Fang Xie
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - Tracy Gales
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - M A Ringenberg
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - Amaya I Wolf
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - M Reid Groseclose
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| |
Collapse
|
6
|
Singh M, Negi R, Alka, Vinayagam R, Kang SG, Shukla P. Age-Related Macular Degeneration (AMD): Pathophysiology, Drug Targeting Approaches, and Recent Developments in Nanotherapeutics. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1647. [PMID: 39459435 PMCID: PMC11509623 DOI: 10.3390/medicina60101647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/26/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024]
Abstract
The most prevalent reason for vision impairment in aging inhabitants is age-related macular degeneration (AMD), a posterior ocular disease with a poor understanding of the anatomic, genetic, and pathophysiological progression of the disease. Recently, new insights exploring the role of atrophic changes in the retinal pigment epithelium, extracellular drusen deposits, lysosomal lipofuscin, and various genes have been investigated in the progression of AMD. Hence, this review explores the incidence and risk factors for AMD, such as oxidative stress, inflammation, the complement system, and the involvement of bioactive lipids and their role in angiogenesis. In addition to intravitreal anti-vascular endothelial growth factor (VEGF) therapy and other therapeutic interventions such as oral kinase inhibitors, photodynamic, gene, and antioxidant therapy, as well as their benefits and drawbacks as AMD treatment options, strategic drug delivery methods, including drug delivery routes with a focus on intravitreal pharmacokinetics, are investigated. Further, the recent advancements in nanoformulations such as polymeric and lipid nanocarriers, liposomes, etc., intended for ocular drug delivery with pros and cons are too summarized. Therefore, the purpose of this review is to give new researchers an understanding of AMD pathophysiology, with an emphasis on angiogenesis, inflammation, the function of bioactive lipids, and therapy options. Additionally, drug delivery options that focus on the development of drug delivery system(s) via several routes of delivery can aid in the advancement of therapeutic choices.
Collapse
Affiliation(s)
- Mahendra Singh
- Department of Biotechnology, Institute of Biotechnology, School of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Riyakshi Negi
- Department of Pharmaceutical Sciences, School of Heath Sciences and Technology, UPES, Dehradun 246008, India; (R.N.); (A.)
| | - Alka
- Department of Pharmaceutical Sciences, School of Heath Sciences and Technology, UPES, Dehradun 246008, India; (R.N.); (A.)
| | - Ramachandran Vinayagam
- Department of Biotechnology, Institute of Biotechnology, School of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Sang Gu Kang
- Department of Biotechnology, Institute of Biotechnology, School of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Prashant Shukla
- Department of Pharmaceutical Sciences, School of Heath Sciences and Technology, UPES, Dehradun 246008, India; (R.N.); (A.)
| |
Collapse
|
7
|
Kibbe RR, Sohn AL, Muddiman DC. Leveraging Supervised Machine Learning Algorithms for System Suitability Testing of Mass Spectrometry Imaging Platforms. J Proteome Res 2024; 23:4384-4391. [PMID: 39226439 DOI: 10.1021/acs.jproteome.4c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Quality control and system suitability testing are vital protocols implemented to ensure the repeatability and reproducibility of data in mass spectrometry investigations. However, mass spectrometry imaging (MSI) analyses present added complexity since both chemical and spatial information are measured. Herein, we employ various machine learning algorithms and a novel quality control mixture to classify the working conditions of an MSI platform. Each algorithm was evaluated in terms of its performance on unseen data, validated with negative control data sets to rule out confounding variables or chance agreement, and utilized to determine the necessary sample size to achieve a high level of accurate classifications. In this work, a robust machine learning workflow was established where models could accurately classify the instrument condition as clean or compromised based on data metrics extracted from the analyzed quality control sample. This work highlights the power of machine learning to recognize complex patterns in MSI data and use those relationships to perform a system suitability test for MSI platforms.
Collapse
Affiliation(s)
- Russell R Kibbe
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Alexandria L Sohn
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - David C Muddiman
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
8
|
Mino T, Nonaka H, Hamachi I. Molecular anchoring and fluorescent labeling in animals compatible with tissue clearing for 3D imaging. Curr Opin Chem Biol 2024; 81:102474. [PMID: 38838505 DOI: 10.1016/j.cbpa.2024.102474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024]
Abstract
Analyzing the quantity and distribution of molecules throughout intact biological tissue is crucial for understanding various biological phenomena. Traditional methods involving destructive extraction result in the loss of spatial information. Conversely, tissue-clearing techniques combined with fluorescence imaging have recently emerged as a powerful tool for deep tissue imaging without sacrificing spatial coverage. Key to this approach is the anchoring and labeling of targets in intact tissue. In this review, methods for anchoring and labeling proteins, lipids, carbohydrates, and small molecules are presented. Future directions include the development of activity-based probes that work in vivo and mark transient events with spatial information to enable a deeper understanding of biological phenomena.
Collapse
Affiliation(s)
- Takeharu Mino
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Hiroshi Nonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan; ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan; ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan.
| |
Collapse
|
9
|
Zivko C, Hahm TH, Tressler C, Brown D, Glunde K, Mahairaki V. Mass Spectrometry Imaging of Organoids to Improve Preclinical Research. Adv Healthc Mater 2024; 13:e2302499. [PMID: 38247228 DOI: 10.1002/adhm.202302499] [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: 08/01/2023] [Revised: 12/18/2023] [Indexed: 01/23/2024]
Abstract
Preclinical models are essential research tools before novel therapeutic or diagnostic methods can be applied to humans. These range from in vitro cell monocultures to vastly more complex animal models, but clinical translation to humans often fails to deliver significant results. Three-dimensional (3D) organoid systems are being increasingly studied to establish physiologically relevant in vitro platforms in a trade-off between the complexity of the research question and the complexity of practical experimental setups. The sensitivity and precision of analytical tools are yet another limiting factors in what can be investigated, and mass spectrometry (MS) is one of the most powerful analytical techniques available to the scientific community. Its innovative use to spatially resolve biological samples has opened many research avenues in the field of MS imaging (MSI). Here, this work aims to explore the current scientific landscape in the application of MSI on organoids, with an emphasis on their combined potential to facilitate and improve preclinical studies.
Collapse
Affiliation(s)
- Cristina Zivko
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Tae-Hun Hahm
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Cay Tressler
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Dalton Brown
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Kristine Glunde
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Vasiliki Mahairaki
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
10
|
Zhang J, Mao Z, Zhang D, Guo L, Zhao H, Miao M. Mass spectrometry imaging as a promising analytical technique for herbal medicines: an updated review. Front Pharmacol 2024; 15:1442870. [PMID: 39148546 PMCID: PMC11324582 DOI: 10.3389/fphar.2024.1442870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/18/2024] [Indexed: 08/17/2024] Open
Abstract
Herbal medicines (HMs) have long played a pivotal role in preventing and treating various human diseases and have been studied widely. However, the complexities present in HM metabolites and their unclear mechanisms of action have posed significant challenges in the modernization of traditional Chinese medicine (TCM). Over the past two decades, mass spectrometry imaging (MSI) has garnered increasing attention as a robust analytical technique that enables the simultaneous execution of qualitative, quantitative, and localization analyses without complex sample pretreatment. With advances in technical solutions, MSI has been extensively applied in the field of HMs. MSI, a label-free ion imaging technique can comprehensively map the spatial distribution of HM metabolites in plant native tissues, thereby facilitating the effective quality control of HMs. Furthermore, the spatial dimension information of small molecule endogenous metabolites within animal tissues provided by MSI can also serve as a supplement to uncover pharmacological and toxicological mechanisms of HMs. In the review, we provide an overview of the three most common MSI techniques. In addition, representative applications in HM are highlighted. Finally, we discuss the current challenges and propose several potential solutions. We hope that the summary of recent findings will contribute to the application of MSI in exploring metabolites and mechanisms of action of HMs.
Collapse
Affiliation(s)
- Jinying Zhang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, China
| | - Zhiguo Mao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, China
| | - Ding Zhang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, China
| | - Lin Guo
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, China
| | - Hui Zhao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, China
| | - Mingsan Miao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Collaborative Innovation Center for Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, China
| |
Collapse
|
11
|
Rahman MF, Islam A, Islam MM, Mamun MA, Xu L, Sakamoto T, Sato T, Takahashi Y, Kahyo T, Aoyagi S, Kaibuchi K, Setou M. Mass Spectrometry Imaging Combined with Sparse Autoencoder Method Reveals Altered Phosphorylcholine Distribution in Imipramine Treated Wild-Type Mice Brains. Int J Mol Sci 2024; 25:7969. [PMID: 39063212 PMCID: PMC11276679 DOI: 10.3390/ijms25147969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Mass spectrometry imaging (MSI) is essential for visualizing drug distribution, metabolites, and significant biomolecules in pharmacokinetic studies. This study mainly focuses on imipramine, a tricyclic antidepressant that affects endogenous metabolite concentrations. The aim was to use atmospheric pressure matrix-assisted laser desorption/ionization (AP-MALDI)-MSI combined with different dimensionality reduction methods to examine the distribution and impact of imipramine on endogenous metabolites in the brains of treated wild-type mice. Brain sections from both control and imipramine-treated mice underwent AP-MALDI-MSI. Dimensionality reduction methods, including principal component analysis, multivariate curve resolution, and sparse autoencoder (SAE), were employed to extract valuable information from the MSI data. Only the SAE method identified phosphorylcholine (ChoP) as a potential marker distinguishing between the control and treated mice brains. Additionally, a significant decrease in ChoP accumulation was observed in the cerebellum, hypothalamus, thalamus, midbrain, caudate putamen, and striatum ventral regions of the treated mice brains. The application of dimensionality reduction methods, particularly the SAE method, to the AP-MALDI-MSI data is a novel approach for peak selection in AP-MALDI-MSI data analysis. This study revealed a significant decrease in ChoP in imipramine-treated mice brains.
Collapse
Affiliation(s)
- Md Foyzur Rahman
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Ariful Islam
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Md. Monirul Islam
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Md. Al Mamun
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- Preppers Co., Ltd., 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Lili Xu
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Takumi Sakamoto
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- Preppers Co., Ltd., 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Tomohito Sato
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Yutaka Takahashi
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- Preppers Co., Ltd., 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Tomoaki Kahyo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- Quantum Imaging Laboratory, Division of Research and Development in Photonics Technology/International Mass Imaging and Spatial Omics Center, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| | - Satoka Aoyagi
- Faculty of Science and Technology, Seikei University, 3-3-1 Kichijoji-kitamachi, Musashino-shi 180-8633, Tokyo, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
- International Mass Imaging and Spatial Omics Center, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Chuo-ku, Hamamatsu 431-3192, Shizuoka, Japan
| |
Collapse
|
12
|
Agrohia DK, Goswami R, Jantarat T, Çiçek YA, Thongsukh K, Jeon T, Bell JM, Rotello VM, Vachet RW. Suborgan Level Quantitation of Proteins in Tissues Delivered by Polymeric Nanocarriers. ACS NANO 2024; 18:16808-16818. [PMID: 38870478 PMCID: PMC11497159 DOI: 10.1021/acsnano.4c02344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Amidst the rapid growth of protein therapeutics as a drug class, there is an increased focus on designing systems to effectively deliver proteins to target organs. Quantitative monitoring of protein distributions in tissues is essential for optimal development of delivery systems; however, existing strategies can have limited accuracy, making it difficult to assess suborgan dosing. Here, we describe a quantitative imaging approach that utilizes metal-coded mass tags and laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) to quantify the suborgan distributions of proteins in tissues that have been delivered by polymeric nanocarriers. Using this approach, we measure nanomole per gram levels of proteins as delivered by guanidinium-functionalized poly(oxanorborneneimide) (PONI) polymers to various tissues, including the alveolar region of the lung. Due to the multiplexing capability of the LA-ICP-MS imaging, we are also able to simultaneously quantify protein and polymer distributions, obtaining valuable information about the relative excretion pathways of the protein cargo and carrier. This imaging approach will facilitate quantitative correlations between nanocarrier properties and protein cargo biodistributions.
Collapse
Affiliation(s)
- Dheeraj K. Agrohia
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Ritabrita Goswami
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Teerapong Jantarat
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Yağız Anil Çiçek
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Korndanai Thongsukh
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Taewon Jeon
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Jonathan M. Bell
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Richard W. Vachet
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
13
|
Tenebro CP, Marcial NBJM, Salcepuedes JJ, Torrecampo JC, Hernandez RD, Francisco JAP, Infante KMG, Belardo VJ, Paderes MC, Alvero RGY, Saludes JP, Dalisay DS. Visualization of renal rotenone accumulation after oral administration and in situ detection of kidney injury biomarkers via MALDI mass spectrometry imaging. Front Mol Biosci 2024; 11:1366278. [PMID: 39011141 PMCID: PMC11246995 DOI: 10.3389/fmolb.2024.1366278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 06/04/2024] [Indexed: 07/17/2024] Open
Abstract
The examination of drug accumulation within complex biological systems offers valuable insights into the molecular aspects of drug metabolism and toxicity. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) is an innovative methodology that enables the spatial visualization and quantification of biomolecules as well as drug and its metabolites in complex biological system. Hence, this method provides valuable insights into the metabolic profile and any molecular changes that may occur as a result of drug treatment. The renal system is particularly vulnerable to adverse effects of drug-induced harm and toxicity. In this study, MALDI MSI was utilized to examine the spatial distribution of drug and renal metabolites within kidney tissues subsequent to a single oral dosage of the anticancer compound rotenone. The integration of ion mobility spectrometry with MALDI MSI enhanced the data acquisition and analysis, resulting to improved mass resolution. Subsequently, the MS/MS fragment ions of rotenone reference drug were detected and characterized using MALDI HDMS/MS imaging. Notably, drug accumulation was observed in the cortical region of the representative kidney tissue sections treated with rotenone. The histological examination of treated kidney tissues did not reveal any observable changes. Differential ion intensity of renal endogenous metabolites was observed between untreated and rotenone-treated tissues. In the context of treated kidney tissues, the ion intensity level of sphingomyelin (D18:1/16:0), a sphingolipid indicator of glomerular cell injury and renal damage, was found to be elevated significantly compared to untreated kidney tissues. Conversely, the ion intensities of choline, glycero-3-phosphocholine (GPC), inosine, and a lysophosphatidylcholine LysoPC(18:0) exhibited a significant decrease. The results of this study demonstrate the potential of MALDI MSI as a novel technique for investigating the in situ spatial distribution of drugs and renal endogenous molecules while preserving the anatomical integrity of the kidney tissue. This technique can be used to study drug-induced metabolism and toxicity in a dynamic manner.
Collapse
Affiliation(s)
- Chuckcris P Tenebro
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
| | - Neaven Bon Joy M Marcial
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
| | - Janine J Salcepuedes
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
| | - Josie C Torrecampo
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
| | - Rajelle D Hernandez
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, Philippines
| | | | | | | | - Monissa C Paderes
- Institute of Chemistry, University of the Philippines Diliman, Quezon City, Philippines
| | | | - Jonel P Saludes
- Center for Natural Drug Discovery and Development, University of San Agustin, Iloilo City, Philippines
- Department of Chemistry, University of San Agustin, Iloilo City, Philippines
- Balik Scientist Program, Department of Science and Technology-Philippine Council for Health Research and Development, Taguig City, Philippines
| | - Doralyn S Dalisay
- Center for Chemical Biology and Biotechnology, University of San Agustin, Iloilo City, Philippines
- Balik Scientist Program, Department of Science and Technology-Philippine Council for Health Research and Development, Taguig City, Philippines
- Department of Biology, University of San Agustin, Iloilo City, Philippines
| |
Collapse
|
14
|
Khalil SM, Qin X, Hakenjos JM, Wang J, Hu Z, Liu X, Wang J, Maletic-Savatic M, MacKenzie KR, Matzuk MM, Li F. MALDI Imaging Mass Spectrometry Visualizes the Distribution of Antidepressant Duloxetine and Its Major Metabolites in Mouse Brain, Liver, Kidney, and Spleen Tissues. Drug Metab Dispos 2024; 52:673-680. [PMID: 38658163 PMCID: PMC11185819 DOI: 10.1124/dmd.124.001719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024] Open
Abstract
Imaging mass spectrometry (IMS) is a powerful tool for mapping the spatial distribution of unlabeled drugs and metabolites that may find application in assessing drug delivery, explaining drug efficacy, and identifying potential toxicity. This study focuses on determining the spatial distribution of the antidepressant duloxetine, which is widely prescribed despite common adverse effects (liver injury, constant headaches) whose mechanisms are not fully understood. We used high-resolution IMS with matrix-assisted laser desorption/ionization to examine the distribution of duloxetine and its major metabolites in four mouse organs where it may contribute to efficacy or toxicity: brain, liver, kidney, and spleen. In none of these tissues is duloxetine or its metabolites homogeneously distributed, which has implications for both efficacy and toxicity. We found duloxetine to be similarly distributed in spleen red pulp and white pulp but differentially distributed in different anatomic regions of the liver, kidney, and brain, with dose-dependent patterns. Comparison with hematoxylin and eosin staining of tissue sections reveals that the ion images of endogenous lipids help delineate anatomic regions in the brain and kidney, while heme ion images assist in differentiating regions within the spleen. These endogenous metabolites may serve as a valuable resource for examining the spatial distribution of other drugs in tissues when staining images are not available. These findings may facilitate future mechanistic studies of the therapeutic and adverse effects of duloxetine. In the current work, we did not perform absolute quantification of duloxetine, which will be reported in due course. SIGNIFICANCE STATEMENT: The study utilized imaging mass spectrometry to examine the spatial distribution of duloxetine and its primary metabolites in mouse brain, liver, kidney, and spleen. These results may pave the way for future investigations into the mechanisms behind duloxetine's therapeutic and adverse effects. Furthermore, the mass spectrometry images of specific endogenous metabolites such as heme could be valuable in analyzing the spatial distribution of other drugs within tissues in scenarios where histological staining images are unavailable.
Collapse
Affiliation(s)
- Saleh M Khalil
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Xuan Qin
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - John M Hakenjos
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Jian Wang
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Zhaoyong Hu
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Xinli Liu
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Jin Wang
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Mirjana Maletic-Savatic
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Kevin R MacKenzie
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Martin M Matzuk
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Feng Li
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| |
Collapse
|
15
|
El Allouche Y, Alaqarbeh M, El Aissouq A, El Rhabori S, Ech-Chahdi Y, Bouachrine M, Zaitan H, Khalil F. Chemoinformatics Study of Benzodiazepine-1, 2, 3-triazole Derivatives Targeting Butyrylcholinesterase. J Fluoresc 2024:10.1007/s10895-024-03812-8. [PMID: 38884828 DOI: 10.1007/s10895-024-03812-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
This study aims to assess the potential bioactivity of newly designed benzodiazepine-1,2,3-triazole derivatives using in-silico methodologies, with a primary focus on elucidating their inhibitory interactions with the butyrylcholinesterase (BuChE) enzyme, which is implicated in Alzheimer's disease. We employed multiple linear regression (MLR) methods to conduct a quantitative structure-activity relationship (QSAR) analysis on a collection of 31 benzodiazepine-1,2,3-triazole derivatives, with the goal of investigating, assessing, and predicting their activities, as well as designing novel compounds. This approach yielded highly accurate results, with coefficients of determination (R²) of 0.77 and 0.81 for the training and test datasets, respectively. Additionally, the optimized compounds were subjected to an Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) analysis, demonstrating their potential as non-hepatotoxic agents with enhanced absorption and blood-brain barrier permeability. To further validate these findings, the most favorable docking conformations were analyzed using molecular dynamics (MD) simulations with GROMACS software, predicting the stability of the formed complexes. These simulations underscored the critical role of hydrogen bonds in stabilizing the compounds at the BuChE receptor binding site. The results hold great promise for the development of innovative benzodiazepine-1,2,3-triazole derivatives as effective BuChE inhibitors, potentially leading to therapeutic interventions for Alzheimer's disease.
Collapse
Affiliation(s)
- Yassine El Allouche
- Laboratory of Processes, Materials, and Environment (LPME), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Fez, Morocco.
| | - Marwa Alaqarbeh
- Basic Science Department, Prince Al Hussein bin Abdullah II Academy for Civil Protection, Al-Balqa Applied University, Al-Salt, 19117, Jordan
| | - Abdellah El Aissouq
- Laboratory of Processes, Materials, and Environment (LPME), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Fez, Morocco.
| | - Said El Rhabori
- Laboratory of Processes, Materials, and Environment (LPME), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Youssra Ech-Chahdi
- Laboratory of Processes, Materials, and Environment (LPME), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mohammed Bouachrine
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University of Meknes, Meknes, Morocco
| | - Hicham Zaitan
- Laboratory of Processes, Materials, and Environment (LPME), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Fouad Khalil
- Laboratory of Processes, Materials, and Environment (LPME), Faculty of Science and Technology, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
16
|
Čužić S, Antolić M, Ognjenović A, Milutinović V, Iviš SV, Glojnarić I, Bosnar M, Požgaj L, Prenc E, Haber VE. Translational pathology in drug discovery. Front Pharmacol 2024; 15:1409092. [PMID: 38915468 PMCID: PMC11194691 DOI: 10.3389/fphar.2024.1409092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/23/2024] [Indexed: 06/26/2024] Open
Affiliation(s)
- Snježana Čužić
- In vivo Pharmacology and Toxicology, Selvita, Zagreb, Croatia
| | - Maja Antolić
- In vivo Pharmacology and Toxicology, Selvita, Zagreb, Croatia
| | - Anja Ognjenović
- In vivo Pharmacology and Toxicology, Selvita, Zagreb, Croatia
| | - Vuk Milutinović
- In vivo Pharmacology and Toxicology, Selvita, Zagreb, Croatia
| | | | - Ines Glojnarić
- In vivo Pharmacology and Toxicology, Selvita, Zagreb, Croatia
| | | | - Lidija Požgaj
- Pharmacology and Translational Research, Selvita, Zagreb, Croatia
| | - Ema Prenc
- Pharmacology and Translational Research, Selvita, Zagreb, Croatia
| | | |
Collapse
|
17
|
Sohn AL, Kibbe RR, Dioli OE, Hector EC, Bai H, Garrard KP, Muddiman DC. A statistical approach to system suitability testing for mass spectrometry imaging. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2024; 38:e9725. [PMID: 38456255 PMCID: PMC10926995 DOI: 10.1002/rcm.9725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 03/09/2024]
Abstract
RATIONALE Mass spectrometry imaging (MSI) elevates the power of conventional mass spectrometry (MS) to multidimensional space, elucidating both chemical composition and localization. However, the field lacks any robust quality control (QC) and/or system suitability testing (SST) protocols to monitor inconsistencies during data acquisition, both of which are integral to ensure the validity of experimental results. To satisfy this demand in the community, we propose an adaptable QC/SST approach with five analyte options amendable to various ionization MSI platforms (e.g., desorption electrospray ionization, matrix-assisted laser desorption/ionization [MALDI], MALDI-2, and infrared matrix-assisted laser desorption electrospray ionization [IR-MALDESI]). METHODS A novel QC mix was sprayed across glass slides to collect QC/SST regions-of-interest (ROIs). Data were collected under optimal conditions and on a compromised instrument to construct and refine the principal component analysis (PCA) model in R. Metrics, including mass measurement accuracy and spectral accuracy, were evaluated, yielding an individual suitability score for each compound. The average of these scores is utilized to inform if troubleshooting is necessary. RESULTS The PCA-based SST model was applied to data collected when the instrument was compromised. The resultant SST scores were used to determine a statistically significant threshold, which was defined as 0.93 for IR-MALDESI-MSI analyses. This minimizes the type-I error rate, where the QC/SST would report the platform to be in working condition when cleaning is actually necessary. Further, data scored after a partial cleaning demonstrate the importance of QC and frequent full instrument cleaning. CONCLUSIONS This study is the starting point for addressing an important issue and will undergo future development to improve the efficiency of the protocol. Ultimately, this work is the first of its kind and proposes this approach as a proof of concept to develop and implement universal QC/SST protocols for a variety of MSI platforms.
Collapse
Affiliation(s)
- Alexandria L. Sohn
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, NC 27695
| | - Russell R. Kibbe
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, NC 27695
| | - Olivia E. Dioli
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, NC 27695
| | - Emily C. Hector
- Department of Statistics, North Carolina State University, Raleigh, NC 27695
| | - Hongxia Bai
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, NC 27695
| | - Kenneth P. Garrard
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, NC 27695
| | - David C. Muddiman
- FTMS Laboratory for Human Health Research, Department of Chemistry, North Carolina State University, Raleigh, NC 27695
| |
Collapse
|
18
|
Kawaguchi M, Yonetani Y, Mizuguchi T, Spratt SJ, Asanuma M, Shimizu H, Sasaki M, Ozeki Y. Visualization of Modified Bisarylbutadiyne-Tagged Small Molecules in Live-Cell Nuclei by Stimulated Raman Scattering Microscopy. Anal Chem 2024; 96:6643-6651. [PMID: 38626411 DOI: 10.1021/acs.analchem.3c05946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Visualizing the distribution of small-molecule drugs in living cells is an important strategy for developing specific, effective, and minimally toxic drugs. As an alternative to fluorescence imaging using bulky fluorophores or cell fixation, stimulated Raman scattering (SRS) imaging combined with bisarylbutadiyne (BADY) tagging enables the observation of small molecules closer to their native intracellular state. However, there is evidence that the physicochemical properties of BADY-tagged analogues of small-molecule drugs differ significantly from those of their parent drugs, potentially affecting their intracellular distribution. Herein, we developed a modified BADY to reduce deviations in physicochemical properties (in particular, lipophilicity and membrane permeability) between tagged and parent drugs, while maintaining high Raman activity in live-cell SRS imaging. We highlight the practical application of this approach by revealing the nuclear distribution of a modified BADY-tagged analogue of JQ1, a bromodomain and extra-terminal motif inhibitor with applications in targeted cancer therapy, in living HeLa cells. The modified BADY, methoxypyridazyl pyrimidyl butadiyne (MPDY), revealed intranuclear JQ1, while BADY-tagged JQ1 did not show a clear nuclear signal. We anticipate that the present approach combining MPDY tagging with live-cell SRS imaging provides important insight into the behavior of intracellular drugs and represents a promising avenue for improving drug development.
Collapse
Affiliation(s)
| | - Yuki Yonetani
- Future Technology R&D Center, Canon Inc., Tokyo 146-8501, Japan
| | - Takaha Mizuguchi
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo 153-8904, Japan
| | - Spencer J Spratt
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo 153-8904, Japan
| | - Masato Asanuma
- Department of Electrical Engineering and Information Systems, The University of Tokyo, Tokyo 113-8656, Japan
| | - Hiroki Shimizu
- Organic & Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-8630, Japan
| | - Masato Sasaki
- Organic & Biomolecular Chemistry Department, Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-8630, Japan
| | - Yasuyuki Ozeki
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo 153-8904, Japan
- Department of Electrical Engineering and Information Systems, The University of Tokyo, Tokyo 113-8656, Japan
| |
Collapse
|
19
|
Chen M, Chen S, Wang X, Ye Z, Liu K, Qian Y, Tang M, Wu T. The discovery of regional neurotoxicity-associated metabolic alterations induced by carbon quantum dots in brain of mice using a spatial metabolomics analysis. Part Fibre Toxicol 2024; 21:19. [PMID: 38600504 PMCID: PMC11005155 DOI: 10.1186/s12989-024-00580-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/21/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Recently, carbon quantum dots (CQDs) have been widely used in various fields, especially in the diagnosis and therapy of neurological disorders, due to their excellent prospects. However, the associated inevitable exposure of CQDs to the environment and the public could have serious severe consequences limiting their safe application and sustainable development. RESULTS In this study, we found that intranasal treatment of 5 mg/kg BW (20 µL/nose of 0.5 mg/mL) CQDs affected the distribution of multiple metabolites and associated pathways in the brain of mice through the airflow-assisted desorption electrospray ionization mass spectrometry imaging (AFADESI-MSI) technique, which proved effective in discovery has proven to be significantly alerted and research into tissue-specific toxic biomarkers and molecular toxicity analysis. The neurotoxic biomarkers of CQDs identified by MSI analysis mainly contained aminos, lipids and lipid-like molecules which are involved in arginine and proline metabolism, biosynthesis of unsaturated fatty acids, and glutamine and glutamate metabolism, etc. as well as related metabolic enzymes. The levels or expressions of these metabolites and enzymes changed by CQDs in different brain regions would induce neuroinflammation, organelle damage, oxidative stress and multiple programmed cell deaths (PCDs), leading to neurodegeneration, such as Parkinson's disease-like symptoms. This study enlightened risk assessments and interventions of QD-type or carbon-based nanoparticles on the nervous system based on toxic biomarkers regarding region-specific profiling of altered metabolic signatures. CONCLUSION These findings provide information to advance knowledge of neurotoxic effects of CQDs and guide their further safety evaluation.
Collapse
Affiliation(s)
- Min Chen
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, 210009, Nanjing, P.R. China
| | - Siyuan Chen
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, 210009, Nanjing, P.R. China
| | - Xinyu Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, 210009, Nanjing, P.R. China
| | - Zongjian Ye
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, 210009, Nanjing, P.R. China
| | - Kehan Liu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, 210009, Nanjing, P.R. China
| | - Yijing Qian
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, 210009, Nanjing, P.R. China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, 210009, Nanjing, P.R. China
| | - Tianshu Wu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, 210009, Nanjing, P.R. China.
| |
Collapse
|
20
|
Gorman BL, Torti SV, Torti FM, Anderton CR. Mass spectrometry imaging of metals in tissues and cells: Methods and biological applications. Biochim Biophys Acta Gen Subj 2024; 1868:130329. [PMID: 36791830 PMCID: PMC10423302 DOI: 10.1016/j.bbagen.2023.130329] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Metals are pervasive throughout biological processes, where they play essential structural and catalytic roles. Metals can also exhibit deleterious effects on human health. Powerful analytical techniques, such as mass spectrometry imaging (MSI), are required to map metals due to their low concentrations within biological tissue. SCOPE OF REVIEW This Mini Review focuses on key MSI technology that can image metal distributions in situ, describing considerations for each technique (e.g., resolution, sensitivity, etc.). We highlight recent work using MSI for mapping trace metals in tissues, detecting metal-based drugs, and simultaneously imaging metals and biomolecules. MAJOR CONCLUSIONS MSI has enabled significant advances in locating bioactive metals at high spatial resolution and correlating their distributions with that of biomolecules. The use of metal-based immunochemistry has enabled simultaneous high-throughput protein and biomolecule imaging. GENERAL SIGNIFICANCE The techniques and examples described herein can be applied to many biological questions concerning the important biological roles of metals, metal toxicity, and localization of metal-based drugs.
Collapse
Affiliation(s)
- Brittney L Gorman
- Environmental Molecular Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, United States of America
| | - Suzy V Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Frank M Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, United States of America
| | - Christopher R Anderton
- Environmental Molecular Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, United States of America.
| |
Collapse
|
21
|
Gorman BL, Taylor MJ, Tesfay L, Lukowski JK, Hegde P, Eder JG, Bloodsworth KJ, Kyle JE, Torti S, Anderton CR. Applying Multimodal Mass Spectrometry to Image Tumors Undergoing Ferroptosis Following In Vivo Treatment with a Ferroptosis Inducer. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:5-12. [PMID: 38079508 DOI: 10.1021/jasms.3c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Epithelial ovarian cancer (EOC) is the most common form of ovarian cancer. The poor prognosis generally associated with this disease has led to the search for improved therapies such as ferroptosis-inducing agents. Ferroptosis is a form of regulated cell death that is dependent on iron and is characterized by lipid peroxidation. Precise mapping of lipids and iron within tumors exposed to ferroptosis-inducing agents may provide insight into processes of ferroptosis in vivo and ultimately assist in the optimal deployment of ferroptosis inducers in cancer therapy. In this work, we present a method for combining matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) with secondary ion mass spectrometry (SIMS) to analyze changes in spatial lipidomics and metal composition, respectively, in ovarian tumors following exposure to a ferroptosis inducer. Tumors were obtained by injecting human ovarian cancer tumor-initiating cells into mice, followed by treatment with the ferroptosis inducer erastin. SIMS imaging detected iron accumulation in the tumor tissue, and sequential MALDI-MS imaging of the same tissue section displayed two chemically distinct regions of lipids. One region was associated with the iron-rich area detected with SIMS, and the other region encompassed the remainder of the tissue section. Bulk lipidomics confirmed the lipid assignments putatively assigned from the MALDI-MS data. Overall, we demonstrate the ability of multimodal MSI to identify the spatial locations of iron and lipids in the same tissue section and associate these regions with clinical pathology.
Collapse
Affiliation(s)
- Brittney L Gorman
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Michael J Taylor
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Lia Tesfay
- Department of Molecular Biology and Biophysics, University of Connecticut Health, Farmington, Connecticut 06030, United States
| | - Jessica K Lukowski
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
- School of Medicine, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Poornima Hegde
- Department of Pathology and Laboratory Medicine, University of Connecticut Health, Farmington, 06030, Connecticut United States
| | - Josie G Eder
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Kent J Bloodsworth
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Jennifer E Kyle
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Suzy Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health, Farmington, Connecticut 06030, United States
| | - Christopher R Anderton
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| |
Collapse
|
22
|
Krestensen KK, Heeren RMA, Balluff B. State-of-the-art mass spectrometry imaging applications in biomedical research. Analyst 2023; 148:6161-6187. [PMID: 37947390 DOI: 10.1039/d3an01495a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Mass spectrometry imaging has advanced from a niche technique to a widely applied spatial biology tool operating at the forefront of numerous fields, most notably making a significant impact in biomedical pharmacological research. The growth of the field has gone hand in hand with an increase in publications and usage of the technique by new laboratories, and consequently this has led to a shift from general MSI reviews to topic-specific reviews. Given this development, we see the need to recapitulate the strengths of MSI by providing a more holistic overview of state-of-the-art MSI studies to provide the new generation of researchers with an up-to-date reference framework. Here we review scientific advances for the six largest biomedical fields of MSI application (oncology, pharmacology, neurology, cardiovascular diseases, endocrinology, and rheumatology). These publications thereby give examples for at least one of the following categories: they provide novel mechanistic insights, use an exceptionally large cohort size, establish a workflow that has the potential to become a high-impact methodology, or are highly cited in their field. We finally have a look into new emerging fields and trends in MSI (immunology, microbiology, infectious diseases, and aging), as applied MSI is continuously broadening as a result of technological breakthroughs.
Collapse
Affiliation(s)
- Kasper K Krestensen
- The Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Ron M A Heeren
- The Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, 6229 ER Maastricht, The Netherlands.
| | - Benjamin Balluff
- The Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
23
|
Djambazova KV, van Ardenne JM, Spraggins JM. Advances in Imaging Mass Spectrometry for Biomedical and Clinical Research. Trends Analyt Chem 2023; 169:117344. [PMID: 38045023 PMCID: PMC10688507 DOI: 10.1016/j.trac.2023.117344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Imaging mass spectrometry (IMS) allows for the untargeted mapping of biomolecules directly from tissue sections. This technology is increasingly integrated into biomedical and clinical research environments to supplement traditional microscopy and provide molecular context for tissue imaging. IMS has widespread clinical applicability in the fields of oncology, dermatology, microbiology, and others. This review summarizes the two most widely employed IMS technologies, matrix-assisted laser desorption/ionization (MALDI) and desorption electrospray ionization (DESI), and covers technological advancements, including efforts to increase spatial resolution, specificity, and throughput. We also highlight recent biomedical applications of IMS, primarily focusing on disease diagnosis, classification, and subtyping.
Collapse
Affiliation(s)
- Katerina V. Djambazova
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Jacqueline M. van Ardenne
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Jeffrey M. Spraggins
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
24
|
Chen Y, Liu Y, Li X, He Y, Li W, Peng Y, Zheng J. Recent Advances in Mass Spectrometry-Based Spatially Resolved Molecular Imaging of Drug Disposition and Metabolomics. Drug Metab Dispos 2023; 51:1273-1283. [PMID: 37295949 DOI: 10.1124/dmd.122.001069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Mass spectrometric imaging is a nontargeted, tag-free, high-throughput, and highly responsive analytical approach. The highly accurate molecular visualization detection technology enables qualitative and quantitative analyses of biologic tissues or cells scanned by mass spectrometry in situ, extracting known and unknown multiple compounds, and simultaneously assessing relative contents of targeting molecules by monitoring their molecular ions and pinpointing the spatial locations of those molecules distributed. Five mass spectrometric imaging techniques and their characteristics are introduced in the review, including matrix-assisted laser desorption ionization mass spectrometry, secondary ion mass spectrometry, desorption electrospray ionization mass spectrometry, laser ablation electrospray ionization mass spectrometry, and laser ablation inductively coupled plasma mass spectrometry. The mass spectrometry-based techniques provide the possibility for spatial metabolomics with the capability of high throughput and precision detection. The approaches have been widely employed to spatially image not only metabolome of endogenous amino acids, peptides, proteins, neurotransmitters, and lipids but also the disposition of exogenous chemicals, such as pharmaceutical agents, environmental pollutants, toxicants, natural products, and heavy metals. The techniques also provide us with spatial distribution imaging of analytes in single cells, tissue microregions, organs, and whole animals. SIGNIFICANCE STATEMENT: The review article includes an overview of five commonly used mass spectrometers for spatial imaging and describes the advantages and disadvantages of each. Examples of the technology applications cover drug disposition, diseases, and omics. Technical aspects of relative and absolute quantification by mass spectrometric imaging and challenges for future new applications are discussed as well. The reviewed knowledge may benefit the development of new drugs and provide a better understanding of biochemical processes related to physiology and diseases.
Collapse
Affiliation(s)
- Yu Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); School of Basic Medicine, School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); Division of Pain Management, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C.); and Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P.R. China (Y.P., J.Z.)
| | - Ying Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); School of Basic Medicine, School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); Division of Pain Management, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C.); and Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P.R. China (Y.P., J.Z.)
| | - Ximei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); School of Basic Medicine, School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); Division of Pain Management, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C.); and Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P.R. China (Y.P., J.Z.)
| | - Yan He
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); School of Basic Medicine, School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); Division of Pain Management, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C.); and Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P.R. China (Y.P., J.Z.)
| | - Weiwei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); School of Basic Medicine, School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); Division of Pain Management, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C.); and Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P.R. China (Y.P., J.Z.)
| | - Ying Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); School of Basic Medicine, School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); Division of Pain Management, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C.); and Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P.R. China (Y.P., J.Z.)
| | - Jiang Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); School of Basic Medicine, School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C., Y.L., X.L., Y.H., W.L.); Division of Pain Management, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, P.R. China (Y.C.); and Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, P.R. China (Y.P., J.Z.)
| |
Collapse
|
25
|
Herdiana Y, Husni P, Nurhasanah S, Shamsuddin S, Wathoni N. Chitosan-Based Nano Systems for Natural Antioxidants in Breast Cancer Therapy. Polymers (Basel) 2023; 15:2953. [PMID: 37447598 DOI: 10.3390/polym15132953] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer is a major cause of death globally, accounting for around 13% of all deaths. Chemotherapy, the common treatment for cancer, can have side effects that lead to the production of reactive oxygen species (ROS) and an increase in oxidative stress in the body. Antioxidants are important for maintaining the health of cells and helping the immune system function properly. They play a crucial role in balancing the body's internal environment. Using natural antioxidants is an alternative to mitigate the harmful effects of oxidative stress. However, around 80% of natural antioxidants have limited effectiveness when taken orally because they do not dissolve well in water or other solvents. This poor solubility affects their ability to be absorbed by the body and limits their bioavailability. One strategy that has been considered is to increase their water solubility to increase their oral bioavailability. Chitosan-based nanoparticle (CSNP) systems have been extensively explored due to their reliability and simpler synthesis routes. This review focuses on the various methods of chitosan-based nanoformulation for developing effective oral dosage forms for natural antioxidants based on the pharmacokinetics and pharmacodynamics properties. Chitosan (CS) could be a model, because of its wide use in polymeric NPs research, thus providing a better understanding of the role of vehicles that carry natural antioxidants in maintaining the stability and enhancing the performance of cancer drugs.
Collapse
Affiliation(s)
- Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Patihul Husni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Siti Nurhasanah
- Faculty of Agricultural Industrial Technology, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
- USM-RIKEN Interdisciplinary Collaboration on Advanced Sciences (URICAS), Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| |
Collapse
|
26
|
Fangma Y, Liu M, Liao J, Chen Z, Zheng Y. Dissecting the brain with spatially resolved multi-omics. J Pharm Anal 2023; 13:694-710. [PMID: 37577383 PMCID: PMC10422112 DOI: 10.1016/j.jpha.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 08/15/2023] Open
Abstract
Recent studies have highlighted spatially resolved multi-omics technologies, including spatial genomics, transcriptomics, proteomics, and metabolomics, as powerful tools to decipher the spatial heterogeneity of the brain. Here, we focus on two major approaches in spatial transcriptomics (next-generation sequencing-based technologies and image-based technologies), and mass spectrometry imaging technologies used in spatial proteomics and spatial metabolomics. Furthermore, we discuss their applications in neuroscience, including building the brain atlas, uncovering gene expression patterns of neurons for special behaviors, deciphering the molecular basis of neuronal communication, and providing a more comprehensive explanation of the molecular mechanisms underlying central nervous system disorders. However, further efforts are still needed toward the integrative application of multi-omics technologies, including the real-time spatial multi-omics analysis in living cells, the detailed gene profile in a whole-brain view, and the combination of functional verification.
Collapse
Affiliation(s)
- Yijia Fangma
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengting Liu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jie Liao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
27
|
Maciel LÍL, Bernardo RA, Martins RO, Batista Junior AC, Oliveira JVA, Chaves AR, Vaz BG. Desorption electrospray ionization and matrix-assisted laser desorption/ionization as imaging approaches for biological samples analysis. Anal Bioanal Chem 2023:10.1007/s00216-023-04783-8. [PMID: 37329466 DOI: 10.1007/s00216-023-04783-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/19/2023]
Abstract
The imaging of biological tissues can offer valuable information about the sample composition, which improves the understanding of analyte distribution in such complex samples. Different approaches using mass spectrometry imaging (MSI), also known as imaging mass spectrometry (IMS), enabled the visualization of the distribution of numerous metabolites, drugs, lipids, and glycans in biological samples. The high sensitivity and multiple analyte evaluation/visualization in a single sample provided by MSI methods lead to various advantages and overcome drawbacks of classical microscopy techniques. In this context, the application of MSI methods, such as desorption electrospray ionization-MSI (DESI-MSI) and matrix-assisted laser desorption/ionization-MSI (MALDI-MSI), has significantly contributed to this field. This review discusses the evaluation of exogenous and endogenous molecules in biological samples using DESI and MALDI imaging. It offers rare technical insights not commonly found in the literature (scanning speed and geometric parameters), making it a comprehensive guide for applying these techniques step-by-step. Furthermore, we provide an in-depth discussion of recent research findings on using these methods to study biological tissues.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Boniek Gontijo Vaz
- Instituto de Química, Universidade Federal de Goiás, Goiânia, GO, 74690-900, Brazil.
| |
Collapse
|
28
|
Cappuccio G, Khalil SM, Osenberg S, Li F, Maletic-Savatic M. Mass spectrometry imaging as an emerging tool for studying metabolism in human brain organoids. Front Mol Biosci 2023; 10:1181965. [PMID: 37304070 PMCID: PMC10251497 DOI: 10.3389/fmolb.2023.1181965] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/02/2023] [Indexed: 06/13/2023] Open
Abstract
Human brain organoids are emerging models to study human brain development and pathology as they recapitulate the development and characteristics of major neural cell types, and enable manipulation through an in vitro system. Over the past decade, with the advent of spatial technologies, mass spectrometry imaging (MSI) has become a prominent tool for metabolic microscopy, providing label-free, non-targeted molecular and spatial distribution information of the metabolites within tissue, including lipids. This technology has never been used for studies of brain organoids and here, we set out to develop a standardized protocol for preparation and mass spectrometry imaging of human brain organoids. We present an optimized and validated sample preparation protocol, including sample fixation, optimal embedding solution, homogenous deposition of matrices, data acquisition and processing to maximize the molecular information derived from mass spectrometry imaging. We focus on lipids in organoids, as they play critical roles during cellular and brain development. Using high spatial and mass resolution in positive- and negative-ion modes, we detected 260 lipids in the organoids. Seven of them were uniquely localized within the neurogenic niches or rosettes as confirmed by histology, suggesting their importance for neuroprogenitor proliferation. We observed a particularly striking distribution of ceramide-phosphoethanolamine CerPE 36:1; O2 which was restricted within rosettes and of phosphatidyl-ethanolamine PE 38:3, which was distributed throughout the organoid tissue but not in rosettes. This suggests that ceramide in this particular lipid species might be important for neuroprogenitor biology, while its removal may be important for terminal differentiation of their progeny. Overall, our study establishes the first optimized experimental pipeline and data processing strategy for mass spectrometry imaging of human brain organoids, allowing direct comparison of lipid signal intensities and distributions in these tissues. Further, our data shed new light on the complex processes that govern brain development by identifying specific lipid signatures that may play a role in cell fate trajectories. Mass spectrometry imaging thus has great potential in advancing our understanding of early brain development as well as disease modeling and drug discovery.
Collapse
Affiliation(s)
- Gerarda Cappuccio
- Department of Pediatrics–Neurology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Saleh M. Khalil
- Department of Pediatrics–Neurology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Sivan Osenberg
- Department of Pediatrics–Neurology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Feng Li
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Mirjana Maletic-Savatic
- Department of Pediatrics–Neurology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
29
|
Nonaka H, Mino T, Sakamoto S, Oh JH, Watanabe Y, Ishikawa M, Tsushima A, Amaike K, Kiyonaka S, Tamura T, Aricescu AR, Kakegawa W, Miura E, Yuzaki M, Hamachi I. Revisiting PFA-mediated tissue fixation chemistry: FixEL enables trapping of small molecules in the brain to visualize their distribution changes. Chem 2023; 9:523-540. [PMID: 38094901 PMCID: PMC7615374 DOI: 10.1016/j.chempr.2022.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Various small molecules have been used as functional probes for tissue imaging in medical diagnosis and pharmaceutical drugs for disease treatment. The spatial distribution, target selectivity, and diffusion/excretion kinetics of small molecules in structurally complicated specimens are critical for function. However, robust methods for precisely evaluating these parameters in the brain have been limited. Herein, we report a new method termed "fixation-driven chemical cross-linking of exogenous ligands (FixEL)," which traps and images exogenously administered molecules of interest (MOIs) in complex tissues. This method relies on protein-MOI interactions and chemical cross-linking of amine-tethered MOI with paraformaldehyde used for perfusion fixation. FixEL is used to obtain images of the distribution of the small molecules, which addresses selective/nonselective binding to proteins, time-dependent localization changes, and diffusion/retention kinetics of MOIs such as the scaffold of PET tracer derivatives or drug-like small molecules.
Collapse
Affiliation(s)
- Hiroshi Nonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
- ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan
| | - Takeharu Mino
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Seiji Sakamoto
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Jae Hoon Oh
- ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan
| | - Yu Watanabe
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Mamoru Ishikawa
- ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan
| | - Akihiro Tsushima
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Kazuma Amaike
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Shigeki Kiyonaka
- ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Tomonori Tamura
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
- ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan
| | - A. Radu Aricescu
- Division of Structural Biology, University of Oxford, Oxford OX3 7BN, UK
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Wataru Kakegawa
- ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan
- Department of Neurophysiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Eriko Miura
- Department of Neurophysiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Michisuke Yuzaki
- Department of Neurophysiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
- ERATO (Exploratory Research for Advanced Technology, JST), Tokyo 102-0075, Japan
| |
Collapse
|
30
|
Lim MJ, Yagnik G, Henkel C, Frost SF, Bien T, Rothschild KJ. MALDI HiPLEX-IHC: multiomic and multimodal imaging of targeted intact proteins in tissues. Front Chem 2023; 11:1182404. [PMID: 37201132 PMCID: PMC10187789 DOI: 10.3389/fchem.2023.1182404] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/14/2023] [Indexed: 05/20/2023] Open
Abstract
Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) is one of the most widely used methods for imaging the spatial distribution of unlabeled small molecules such as metabolites, lipids and drugs in tissues. Recent progress has enabled many improvements including the ability to achieve single cell spatial resolution, 3D-tissue image reconstruction, and the precise identification of different isomeric and isobaric molecules. However, MALDI-MSI of high molecular weight intact proteins in biospecimens has thus far been difficult to achieve. Conventional methods normally require in situ proteolysis and peptide mass fingerprinting, have low spatial resolution, and typically detect only the most highly abundant proteins in an untargeted manner. In addition, MSI-based multiomic and multimodal workflows are needed which can image both small molecules and intact proteins from the same tissue. Such a capability can provide a more comprehensive understanding of the vast complexity of biological systems at the organ, tissue, and cellular levels of both normal and pathological function. A recently introduced top-down spatial imaging approach known as MALDI HiPLEX-IHC (MALDI-IHC for short) provides a basis for achieving this high-information content imaging of tissues and even individual cells. Based on novel photocleavable mass-tags conjugated to antibody probes, high-plex, multimodal and multiomic MALDI-based workflows have been developed to image both small molecules and intact proteins on the same tissue sample. Dual-labeled antibody probes enable multimodal mass spectrometry and fluorescent imaging of targeted intact proteins. A similar approach using the same photocleavable mass-tags can be applied to lectin and other probes. We detail here several examples of MALDI-IHC workflows designed to enable high-plex, multiomic and multimodal imaging of tissues at a spatial resolution as low as 5 µm. This approach is compared to other existing high-plex methods such as imaging mass cytometry, MIBI-TOF, GeoMx and CODEX. Finally, future applications of MALDI-IHC are discussed.
Collapse
Affiliation(s)
- Mark J. Lim
- AmberGen, Inc., Billerica, MA, United States
- *Correspondence: Mark J. Lim, ; Kenneth J. Rothschild,
| | | | | | | | - Tanja Bien
- Bruker Daltonics GmbH & Co. KG, Bremen, Germany
| | - Kenneth J. Rothschild
- AmberGen, Inc., Billerica, MA, United States
- Department of Physics and Photonics Center, Boston University, Boston, MA, United States
- *Correspondence: Mark J. Lim, ; Kenneth J. Rothschild,
| |
Collapse
|
31
|
Bowman AP, Sawicki J, Talaty NN, Buck WR, Yang J, Wagner DS. Evaluation of Quantitative Platforms for Single Target Mass Spectrometry Imaging. Pharmaceuticals (Basel) 2022; 15:ph15101180. [PMID: 36297291 PMCID: PMC9609477 DOI: 10.3390/ph15101180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 12/05/2022] Open
Abstract
(1) Imaging of pharmaceutical compounds in tissue is an increasingly important subsection of Mass Spectrometry Imaging (MSI). Identifying proper target engagement requires MS platforms with high sensitivity and spatial resolution. Three prominent categories of drugs are small molecule drugs, antibody-drug conjugate payloads, and protein degraders. (2) We tested six common MSI platforms for their limit of detection (LoD) on a representative compound for each category: a Matrix-Assisted Laser Desorption/Ionization (MALDI) Fourier Transform Ion Cyclotron, a MALDI-2 Time-of-Flight (ToF), a MALDI-2 Trapped Ion Mobility Spectrometry ToF, a Desorption Electrospray Ionization Orbitrap, and 2 Atmospheric Pressure-MALDI Triple Quadrupoles. Samples were homogenized tissue mimetic models of rat liver spiked with known concentrations of analytes. (3) We found that the AP-MALDI-QQQ platform outperformed all 4 competing platforms by a minimum of 2- to 52-fold increase in LoD for representative compounds from each category of pharmaceutical. (4) AP-MALDI-QQQ platforms are effective, cost-efficient mass spectrometers for the identification of targeted analytes of interest.
Collapse
|
32
|
Nwabufo CK, Aigbogun OP. The Role of Mass Spectrometry Imaging in Pharmacokinetic Studies. Xenobiotica 2022; 52:811-827. [PMID: 36048000 DOI: 10.1080/00498254.2022.2119900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Although liquid chromatography-tandem mass spectrometry is the gold standard analytical platform for the quantification of drugs, metabolites, and biomarkers in biological samples, it cannot localize them in target tissues.The localization and quantification of drugs and/or their associated metabolites in target tissues is a more direct measure of bioavailability, biodistribution, efficacy, and regional toxicity compared to the traditional substitute studies using plasma.Therefore, combining high spatial resolution imaging functionality with the superior selectivity and sensitivity of mass spectrometry into one analytical technique will be a valuable tool for targeted localization and quantification of drugs, metabolites, and biomarkers.Mass spectrometry imaging (MSI) is a tagless analytical technique that allows for the direct localization and quantification of drugs, metabolites, and biomarkers in biological tissues, and has been used extensively in pharmaceutical research.The overall goal of this current review is to provide a detailed description of the working principle of MSI and its application in pharmacokinetic studies encompassing absorption, distribution, metabolism, excretion, and toxicity processes, followed by a discussion of the strategies for addressing the challenges associated with the functional utility of MSI in pharmacokinetic studies that support drug development.
Collapse
Affiliation(s)
- Chukwunonso K Nwabufo
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Omozojie P Aigbogun
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada.,Department of Chemistry, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|