101
|
Lai YH, Cai YH, Lee H, Ou YM, Hsiao CH, Tsao CW, Chang HT, Wang YS. Reducing Spatial Heterogeneity of MALDI Samples with Marangoni Flows During Sample Preparation. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2016; 27:1314-21. [PMID: 27126469 DOI: 10.1007/s13361-016-1406-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 04/02/2016] [Accepted: 04/05/2016] [Indexed: 05/20/2023]
Abstract
This work demonstrates a method to prepare homogeneous distributions of analytes to improve data reproducibility in matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MS). Natural-air drying processes normally result in unwanted heterogeneous spatial distributions of analytes in MALDI crystals and make quantitative analysis difficult. This study demonstrates that inducing Marangoni flows within drying droplets can significantly reduce the heterogeneity problem. The Marangoni flows are accelerated by changing substrate temperatures to create temperature gradients across droplets. Such hydrodynamic flows are analyzed semi-empirically. Using imaging mass spectrometry, changes of heterogeneity of molecules with the change of substrate temperature during drying processes are demonstrated. The observed heterogeneities of the biomolecules reduce as predicted Marangoni velocities increase. In comparison to conventional methods, drying droplets on a 5 °C substrate while keeping the surroundings at ambient conditions typically reduces the heterogeneity of biomolecular ions by 65%-80%. The observation suggests that decreasing substrate temperature during droplet drying processes is a simple and effective means to reduce analyte heterogeneity for quantitative applications. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Yin-Hung Lai
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, Republic of China
| | - Yi-Hong Cai
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, Republic of China
| | - Hsun Lee
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, Republic of China
| | - Yu-Meng Ou
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, Republic of China
- Chemistry Department, National Taiwan University, Taipei, 106, Taiwan, Republic of China
| | - Chih-Hao Hsiao
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, Republic of China
| | - Chien-Wei Tsao
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, Republic of China
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, 106, Taiwan, Republic of China
| | - Huan-Tsung Chang
- Chemistry Department, National Taiwan University, Taipei, 106, Taiwan, Republic of China
| | - Yi-Sheng Wang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan, Republic of China.
| |
Collapse
|
102
|
Eberlin LS, Margulis K, Planell-Mendez I, Zare RN, Tibshirani R, Longacre TA, Jalali M, Norton JA, Poultsides GA. Pancreatic Cancer Surgical Resection Margins: Molecular Assessment by Mass Spectrometry Imaging. PLoS Med 2016; 13:e1002108. [PMID: 27575375 PMCID: PMC5019340 DOI: 10.1371/journal.pmed.1002108] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 07/07/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Surgical resection with microscopically negative margins remains the main curative option for pancreatic cancer; however, in practice intraoperative delineation of resection margins is challenging. Ambient mass spectrometry imaging has emerged as a powerful technique for chemical imaging and real-time diagnosis of tissue samples. We applied an approach combining desorption electrospray ionization mass spectrometry imaging (DESI-MSI) with the least absolute shrinkage and selection operator (Lasso) statistical method to diagnose pancreatic tissue sections and prospectively evaluate surgical resection margins from pancreatic cancer surgery. METHODS AND FINDINGS Our methodology was developed and tested using 63 banked pancreatic cancer samples and 65 samples (tumor and specimen margins) collected prospectively during 32 pancreatectomies from February 27, 2013, to January 16, 2015. In total, mass spectra for 254,235 individual pixels were evaluated. When cross-validation was employed in the training set of samples, 98.1% agreement with histopathology was obtained. Using an independent set of samples, 98.6% agreement was achieved. We used a statistical approach to evaluate 177,727 mass spectra from samples with complex, mixed histology, achieving an agreement of 81%. The developed method showed agreement with frozen section evaluation of specimen margins in 24 of 32 surgical cases prospectively evaluated. In the remaining eight patients, margins were found to be positive by DESI-MSI/Lasso, but negative by frozen section analysis. The median overall survival after resection was only 10 mo for these eight patients as opposed to 26 mo for patients with negative margins by both techniques. This observation suggests that our method (as opposed to the standard method to date) was able to detect tumor involvement at the margin in patients who developed early recurrence. Nonetheless, a larger cohort of samples is needed to validate the findings described in this study. Careful evaluation of the long-term benefits to patients of the use of DESI-MSI for surgical margin evaluation is also needed to determine its value in clinical practice. CONCLUSIONS Our findings provide evidence that the molecular information obtained by DESI-MSI/Lasso from pancreatic tissue samples has the potential to transform the evaluation of surgical specimens. With further development, we believe the described methodology could be routinely used for intraoperative surgical margin assessment of pancreatic cancer.
Collapse
Affiliation(s)
- Livia S. Eberlin
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Katherine Margulis
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Ivette Planell-Mendez
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Richard N. Zare
- Department of Chemistry, Stanford University, Stanford, California, United States of America
- * E-mail:
| | - Robert Tibshirani
- Department of Biomedical Data Sciences, Stanford University, Stanford, California, United States of America
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Teri A. Longacre
- Department of Pathology, Stanford University, Stanford, California, United States of America
| | - Moe Jalali
- Department of Surgery, Stanford University, Stanford, California, United States of America
| | - Jeffrey A. Norton
- Department of Surgery, Stanford University, Stanford, California, United States of America
| | - George A. Poultsides
- Department of Surgery, Stanford University, Stanford, California, United States of America
| |
Collapse
|
103
|
Kertesz V, Weiskittel TM, Vavrek M, Freddo C, Van Berkel GJ. Extraction efficiency and implications for absolute quantitation of propranolol in mouse brain, liver and kidney tissue sections using droplet-based liquid microjunction surface sampling high-performance liquid chromatography/electrospray ionization tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2016; 30:1705-1712. [PMID: 28328034 DOI: 10.1002/rcm.7607] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 06/06/2023]
Abstract
RATIONALE Currently, the absolute quantitation aspects of droplet-based surface sampling for tissue analysis using a fully automated autosampler/high-performance liquid chromatography/electrospray ionization tandem mass spectrometry (HPLC/ESI-MS/MS) system have not been fully evaluated. Knowledge of extraction efficiency and its reproducibility is required to judge the potential of the method for absolute quantitation of analytes from tissue sections. METHODS Adjacent tissue sections of propranolol-dosed mouse brain (10-μm-thick), kidney (10-μm-thick) and liver (8-, 10-, 16- and 24-μm-thick) were obtained. The absolute concentration of propranolol was determined in tissue punches from serial sections using standard bulk tissue extraction protocols and subsequent HPLC separations and MS/MS analysis. These values were used to determine propranolol extraction efficiency from the tissues with the droplet-based surface sampling approach. RESULTS Extraction efficiency of propranolol using 10-μm-thick brain, kidney and liver tissues using droplet-based surface sampling varied between ~45 and 63%. The extraction efficiency decreased from ~65% to ~36% with liver thickness increasing from 8 μm to 24 μm. Selecting half of the samples as standards, the precision and accuracy of propranolol concentrations were determined for the other half of the samples that were employed as a quality control data set. The resulting precision (±15%) and accuracy (±3%) were within acceptable limits. CONCLUSIONS Quantitation of adjacent mouse tissue sections of different organs and of various thicknesses by droplet-based surface sampling in comparison with bulk extraction of tissue punches showed that extraction efficiency was incomplete using the former method, and that it depended on the organ and tissue thickness. However, once extraction efficiency was determined and applied, the droplet-based approach provided satisfactory quantitation accuracy and precision for assay validations. Thus, once the extraction efficiency was calibrated for a given tissue type, tissue thickness and drug, the droplet-based approach provides a non-labour-intensive and high-throughput means to acquire spatially resolved quantitative analysis of multiple samples of the same type. Published in 2016. This article is a U.S. Government work and is in the public domain in the USA.
Collapse
Affiliation(s)
- Vilmos Kertesz
- Mass Spectrometry and Laser Spectroscopy Group, Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831-6131, USA
| | - Taylor M Weiskittel
- Mass Spectrometry and Laser Spectroscopy Group, Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831-6131, USA
- ORISE HERE Intern, University of Tennessee, Knoxville, TN, 37996, USA
| | - Marissa Vavrek
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, West Point, PA, 19486, USA
| | - Carol Freddo
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, West Point, PA, 19486, USA
| | - Gary J Van Berkel
- Mass Spectrometry and Laser Spectroscopy Group, Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831-6131, USA
| |
Collapse
|
104
|
Abstract
The clinical development of molecularly targeted cancer therapies is enhanced by proof of mechanism of action as well as proof of concept, which relate molecular pharmacodynamics to efficacy via changes in cancer cell biology and physiology resulting from drug action on its intended target. Here, we present an introduction to the field of clinical pharmacodynamics, its medical and laboratory aspects, and its practical incorporation into clinical trials. We also describe key success factors that are useful for judging the quality of clinical pharmacodynamic studies, including biopsy quality and suitability, specimen handling, assay fitness-for-purpose, and reagent quality control. This introduction provides not only context for the following articles in this issue, but also an appreciation of the role of well-conducted clinical pharmacodynamic studies in oncology drug development.
Collapse
Affiliation(s)
- Ralph E Parchment
- Clinical Pharmacodynamics Program, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD.
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| |
Collapse
|
105
|
Lee JK, Jansson ET, Nam HG, Zare RN. High-Resolution Live-Cell Imaging and Analysis by Laser Desorption/Ionization Droplet Delivery Mass Spectrometry. Anal Chem 2016; 88:5453-61. [PMID: 27110027 PMCID: PMC5446058 DOI: 10.1021/acs.analchem.6b00881] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We have developed a new ambient-ionization mass spectrometric technique named laser desorption/ionization droplet delivery mass spectrometry (LDIDD-MS). LDIDD-MS permits high-resolution, high-sensitivity imaging of tissue samples as well as measurements of both single-cell apoptosis and live-cell exocytosis. A pulsed (15 Hz) UV laser beam (266 nm) is focused on a surface covered with target analytes to trigger their desorption and ionization. A spray of liquid droplets is simultaneously directed onto the laser-focused surface region to capture the ionized analytes and deliver them to a mass spectrometer. The approach of rapid and effective capturing of molecules after laser desorption/ionization allows the limit of detection for the amino acid lysine to be as low as 2 amol under ambient ionization conditions. Two-dimensional maps of the desorbed/ionized species are recorded by moving the sample on an XY translational stage. The spatial resolution for imaging with LDIDD-MS was determined to be 2.4 μm for an ink-printed pattern and 3 μm for mouse brain tissue. We applied LDIDD-MS to single-cell analysis of apoptotic HEK cells. Differences were observed in the profiles of fatty acids and lipids between healthy HEK cells and those undergoing apoptosis. We observed upregulation of phosphatidylcholine (PC) with a relatively shorter carbon chain length and downregulation of PC with a relatively longer carbon chain length. We also applied LDIDD-MS for a real-time direct measurements of live-cell exocytosis. The catecholamine dopamine and trace amines (phenethylamine and tyramine) were detected from live PC12 cells without damaging them.
Collapse
Affiliation(s)
- Jae Kyoo Lee
- Department of Chemistry, Stanford University, Stanford, California 94305 United States
| | - Erik T. Jansson
- Department of Chemistry, Stanford University, Stanford, California 94305 United States
- Department of Chemistry – BMC, Uppsala University, SE-75124 Uppsala, Sweden
| | - Hong Gil Nam
- Center for Plant Aging Research, Institute for Basic Science, Daegu 711-873, Republic of Korea
- Department of New Biology, DGIST, Daegu 711-873, Republic of Korea
| | - Richard N. Zare
- Department of Chemistry, Stanford University, Stanford, California 94305 United States
| |
Collapse
|
106
|
Shariatgorji M, Strittmatter N, Nilsson A, Källback P, Alvarsson A, Zhang X, Vallianatou T, Svenningsson P, Goodwin RJA, Andren PE. Simultaneous imaging of multiple neurotransmitters and neuroactive substances in the brain by desorption electrospray ionization mass spectrometry. Neuroimage 2016; 136:129-38. [PMID: 27155126 DOI: 10.1016/j.neuroimage.2016.05.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 02/07/2023] Open
Abstract
With neurological processes involving multiple neurotransmitters and neuromodulators, it is important to have the ability to directly map and quantify multiple signaling molecules simultaneously in a single analysis. By utilizing a molecular-specific approach, namely desorption electrospray ionization mass spectrometry imaging (DESI-MSI), we demonstrated that the technique can be used to image multiple neurotransmitters and their metabolites (dopamine, dihydroxyphenylacetic acid, 3-methoxytyramine, serotonin, glutamate, glutamine, aspartate, γ-aminobutyric acid, adenosine) as well as neuroactive drugs (amphetamine, sibutramine, fluvoxamine) and drug metabolites in situ directly in brain tissue sections. The use of both positive and negative ionization modes increased the number of identified molecular targets. Chemical derivatization by charge-tagging the primary amines of molecules significantly increased the sensitivity, enabling the detection of low abundant neurotransmitters and other neuroactive substances previously undetectable by MSI. The sensitivity of the imaging approach of neurochemicals has a great potential in many diverse applications in fields such as neuroscience, pharmacology, drug discovery, neurochemistry, and medicine.
Collapse
Affiliation(s)
- Mohammadreza Shariatgorji
- Biomolecular Imaging and Proteomics, National Resource for Mass Spectrometry Imaging, Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-75124 Uppsala, Sweden
| | | | - Anna Nilsson
- Biomolecular Imaging and Proteomics, National Resource for Mass Spectrometry Imaging, Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-75124 Uppsala, Sweden
| | - Patrik Källback
- Biomolecular Imaging and Proteomics, National Resource for Mass Spectrometry Imaging, Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-75124 Uppsala, Sweden
| | - Alexandra Alvarsson
- Center for Molecular Medicine, Department of Neurology and Clinical Neuroscience, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Xiaoqun Zhang
- Center for Molecular Medicine, Department of Neurology and Clinical Neuroscience, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Theodosia Vallianatou
- Biomolecular Imaging and Proteomics, National Resource for Mass Spectrometry Imaging, Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-75124 Uppsala, Sweden
| | - Per Svenningsson
- Center for Molecular Medicine, Department of Neurology and Clinical Neuroscience, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden
| | | | - Per E Andren
- Biomolecular Imaging and Proteomics, National Resource for Mass Spectrometry Imaging, Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-75124 Uppsala, Sweden.
| |
Collapse
|
107
|
Miyamoto S, Hsu CC, Hamm G, Darshi M, Diamond-Stanic M, Declèves AE, Slater L, Pennathur S, Stauber J, Dorrestein PC, Sharma K. Mass Spectrometry Imaging Reveals Elevated Glomerular ATP/AMP in Diabetes/obesity and Identifies Sphingomyelin as a Possible Mediator. EBioMedicine 2016; 7:121-34. [PMID: 27322466 PMCID: PMC4909366 DOI: 10.1016/j.ebiom.2016.03.033] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/11/2016] [Accepted: 03/21/2016] [Indexed: 01/01/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is suppressed in diabetes and may be due to a high ATP/AMP ratio, however the quantitation of nucleotides in vivo has been extremely difficult. Via matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to localize renal nucleotides we found that the diabetic kidney had a significant increase in glomerular ATP/AMP ratio. Untargeted MALDI-MSI analysis revealed that a specific sphingomyelin species (SM(d18:1/16:0)) accumulated in the glomeruli of diabetic and high-fat diet-fed mice compared with wild-type controls. In vitro studies in mesangial cells revealed that exogenous addition of SM(d18:1/16:0) significantly elevated ATP via increased glucose consumption and lactate production with a consequent reduction of AMPK and PGC1α. Furthermore, inhibition of sphingomyelin synthases reversed these effects. Our findings suggest that AMPK is reduced in the diabetic kidney due to an increase in the ATP/AMP ratio and that SM(d18:1/16:0) could be responsible for the enhanced ATP production via activation of the glycolytic pathway. MALDI-MSI revealed an increase in glomerular ATP/AMP ratio in the diabetic kidney. SM(d18:1/16:0) is increased in the glomeruli of diabetic and high-fat diet-fed mice. SM(d18:1/16:0) stimulated ATP production via enhanced aerobic glycolysis and reduced AMPK activity in mesangial cells. AMPK is known to be suppressed in states of high ATP/AMP ratio but the measurement of nucleotides in vivo has been difficult. Miyamoto et al. utilize matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to investigate the distribution of nucleotides and find an increase in glomerular ATP/AMP ratio in the diabetic kidney. Untargeted MALDI-MSI revealed that sphingomyelin(d18:1/16:0) is accumulated in the glomeruli of diabetic and high-fat diet-fed mice compared with controls. Sphingomyelin(d18:1/16:0) promotes ATP production in mesangial cells via activation of the glycolytic pathway. The inhibition of sphingomyelin(d18:1/16:0) synthesis may lead to novel therapeutic targets for the treatment of caloric-induced CKD.
Collapse
Affiliation(s)
- Satoshi Miyamoto
- Institute of Metabolomic Medicine, University of California San Diego, La Jolla, CA 92093, USA; Center for Renal Translational Medicine, Division of Nephrology-Hypertension, University of California San Diego, La Jolla, CA 92093, USA; Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, La Jolla, CA 92093, USA; Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Cheng-Chih Hsu
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA; Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Gregory Hamm
- ImaBiotech, MS Imaging Department, Lille 59120, France
| | - Manjula Darshi
- Institute of Metabolomic Medicine, University of California San Diego, La Jolla, CA 92093, USA; Center for Renal Translational Medicine, Division of Nephrology-Hypertension, University of California San Diego, La Jolla, CA 92093, USA
| | - Maggie Diamond-Stanic
- Institute of Metabolomic Medicine, University of California San Diego, La Jolla, CA 92093, USA; Center for Renal Translational Medicine, Division of Nephrology-Hypertension, University of California San Diego, La Jolla, CA 92093, USA; Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, La Jolla, CA 92093, USA
| | - Anne-Emilie Declèves
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension, University of California San Diego, La Jolla, CA 92093, USA
| | - Larkin Slater
- Institute of Metabolomic Medicine, University of California San Diego, La Jolla, CA 92093, USA; Center for Renal Translational Medicine, Division of Nephrology-Hypertension, University of California San Diego, La Jolla, CA 92093, USA; Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, La Jolla, CA 92093, USA
| | | | | | - Pieter C Dorrestein
- Institute of Metabolomic Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Kumar Sharma
- Institute of Metabolomic Medicine, University of California San Diego, La Jolla, CA 92093, USA; Center for Renal Translational Medicine, Division of Nephrology-Hypertension, University of California San Diego, La Jolla, CA 92093, USA; Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, La Jolla, CA 92093, USA.
| |
Collapse
|
108
|
Ogrinc Potočnik N, Porta T, Becker M, Heeren RMA, Ellis SR. Use of advantageous, volatile matrices enabled by next-generation high-speed matrix-assisted laser desorption/ionization time-of-flight imaging employing a scanning laser beam. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2015; 29:2195-203. [PMID: 26522310 DOI: 10.1002/rcm.7379] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 08/28/2015] [Accepted: 08/29/2015] [Indexed: 05/03/2023]
Abstract
RATIONALE In mass spectrometry imaging (MSI) it is often desirable to analyse the same sample in both polarities to extract the most information. However, many matrices that produce high-quality spectra in matrix-assisted laser desorption/ionization (MALDI) are volatile, greatly limiting their use in long imaging experiments. We demonstrate that using a new high speed MALDI-MSI instrument, volatile matrices, including those that produce intense lipid signals in both positive and negative ion mode, can now be effectively used in MSI. METHODS A prototype Bruker rapifleX MALDI Tissuetyper™ time-of-flight (TOF) instrument was used for high-speed imaging. This allows acquisition rates up to 50 pixels/s made possible by use of a 10 kHz laser and two rotating mirrors that allow the laser beam to be moved over, and synchronised with, the rapidly moving sample. MSI experiments were performed on mouse brain sections using non-vacuum stable dithranol and 2,6-dihydroxyacetophenone (DHA) matrices with pixel sizes ranging from 10 × 10 µm(2) to 50 × 50 µm(2). RESULTS Both DHA and dithranol produced rich, complementary lipid spectra in both positive and negative ion modes. Due to the rapid acquisition speed of the instrument, both matrices could be effectively used for MSI despite their volatility. For example, an entire mouse brain could be imaged consecutively in both positive and negative ion mode with 50 × 50 µm(2) pixels in ~35 min. We demonstrate that these speeds make possible both faster and higher resolution imaging of biological tissues on practical timescales. CONCLUSIONS These high acquisition speeds now make possible whole new classes of matrices that are unstable under high vacuum for MALDI-MSI studies. This provides researchers with far greater range and flexibility in choosing the best matrix for the given sample and analytes that they wish to detect. In addition, such instruments allow MSI to be performed at higher resolution across larger areas on practical time scales.
Collapse
Affiliation(s)
- Nina Ogrinc Potočnik
- M4I, The Maastricht Multimodal Molecular Imaging Institute, 6229 ER Maastricht, The Netherlands
| | - Tiffany Porta
- M4I, The Maastricht Multimodal Molecular Imaging Institute, 6229 ER Maastricht, The Netherlands
| | - Michael Becker
- Bruker Daltonik GmbH, Fahrenheitstrasse 4, 28359, Bremen, Germany
| | - Ron M A Heeren
- M4I, The Maastricht Multimodal Molecular Imaging Institute, 6229 ER Maastricht, The Netherlands
| | - Shane R Ellis
- M4I, The Maastricht Multimodal Molecular Imaging Institute, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
109
|
Pharmacokinetic and pharmacometabolomic study of pirfenidone in normal mouse tissues using high mass resolution MALDI-FTICR-mass spectrometry imaging. Histochem Cell Biol 2015; 145:201-11. [PMID: 26645566 DOI: 10.1007/s00418-015-1382-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2015] [Indexed: 10/22/2022]
Abstract
Given the importance of pirfenidone as the first worldwide-approved drug for idiopathic pulmonary fibrosis treatment, its pharmacodynamic properties and the metabolic response to pirfenidone treatment have not been fully elucidated. The aim of the present study was to get molecular insights of pirfenidone-related pharmacometabolomic response using MALDI-FTICR-MSI. Quantitative MALDI-FTICR-MSI was carried out for determining the pharmacokinetic properties of pirfenidone and its related metabolites 5-hydroxymethyl pirfenidone and 5-carboxy pirfenidone in lung, liver and kidney. To monitor the effect of pirfenidone administration on endogenous cell metabolism, additional in situ endogenous metabolite imaging was performed in lung tissue sections. While pirfenidone is highly abundant and delocalized across the whole micro-regions of lung, kidney and liver, 5-hydroxymethyl pirfenidone and 5-carboxy pirfenidone demonstrate heterogeneous distribution patterns in lung and kidney. In situ endogenous metabolite imaging study of lung tissue indicates no significant effects of pirfenidone on metabolic pathways. Remarkably, we found 129 discriminative m/z values which represent clear differences between control and treated lungs, the majority of which are currently unknown. PCA analysis and heatmap view can accurately distinguish control and treated groups. This is the first pharmacokinetic study to investigate the tissue distribution of orally administered pirfenidone and its related metabolites simultaneously in organs without labeling. The combination of pharmametabolome with histological features provides detailed mapping of drug effects on metabolism as response of healthy lung tissue to pirfenidone treatment.
Collapse
|
110
|
Ifa DR, Eberlin LS. Ambient Ionization Mass Spectrometry for Cancer Diagnosis and Surgical Margin Evaluation. Clin Chem 2015; 62:111-23. [PMID: 26555455 DOI: 10.1373/clinchem.2014.237172] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 09/28/2015] [Indexed: 01/12/2023]
Abstract
BACKGROUND There is a clinical need for new technologies that would enable rapid disease diagnosis based on diagnostic molecular signatures. Ambient ionization mass spectrometry has revolutionized the means by which molecular information can be obtained from tissue samples in real time and with minimal sample pretreatment. New developments in ambient ionization techniques applied to clinical research suggest that ambient ionization mass spectrometry will soon become a routine medical tool for tissue diagnosis. CONTENT This review summarizes the main developments in ambient ionization techniques applied to tissue analysis, with focus on desorption electrospray ionization mass spectrometry, probe electrospray ionization, touch spray, and rapid evaporative ionization mass spectrometry. We describe their applications to human cancer research and surgical margin evaluation, highlighting integrated approaches tested for ex vivo and in vivo human cancer tissue analysis. We also discuss the challenges for clinical implementation of these tools and offer perspectives on the future of the field. SUMMARY A variety of studies have showcased the value of ambient ionization mass spectrometry for rapid and accurate cancer diagnosis. Small molecules have been identified as potential diagnostic biomarkers, including metabolites, fatty acids, and glycerophospholipids. Statistical analysis allows tissue discrimination with high accuracy rates (>95%) being common. This young field has challenges to overcome before it is ready to be broadly accepted as a medical tool for cancer diagnosis. Growing research in new, integrated ambient ionization mass spectrometry technologies and the ongoing improvements in the existing tools make this field very promising for future translation into the clinic.
Collapse
Affiliation(s)
- Demian R Ifa
- Department of Chemistry, York University, Toronto, ON, Canada
| | - Livia S Eberlin
- Department of Chemistry, The University of Texas at Austin, Austin, TX.
| |
Collapse
|
111
|
Chen W, Wang L, Van Berkel GJ, Kertesz V, Gan J. Quantitation of repaglinide and metabolites in mouse whole-body thin tissue sections using droplet-based liquid microjunction surface sampling-high-performance liquid chromatography-electrospray ionization tandem mass spectrometry. J Chromatogr A 2015; 1439:137-143. [PMID: 26589943 DOI: 10.1016/j.chroma.2015.10.093] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 10/07/2015] [Accepted: 10/25/2015] [Indexed: 12/01/2022]
Abstract
Herein, quantitation aspects of a fully automated autosampler/HPLC-MS/MS system applied for unattended droplet-based surface sampling of repaglinide dosed thin tissue sections with subsequent HPLC separation and mass spectrometric analysis of parent drug and various drug metabolites were studied. Major organs (brain, lung, liver, kidney and muscle) from whole-body thin tissue sections and corresponding organ homogenates prepared from repaglinide dosed mice were sampled by surface sampling and by bulk extraction, respectively, and analyzed by HPLC-MS/MS. A semi-quantitative agreement between data obtained by surface sampling and that by employing organ homogenate extraction was observed. Drug concentrations obtained by the two methods followed the same patterns for post-dose time points (0.25, 0.5, 1 and 2 h). Drug amounts determined in the specific tissues was typically higher when analyzing extracts from the organ homogenates. In addition, relative comparison of the levels of individual metabolites between the two analytical methods also revealed good semi-quantitative agreement.
Collapse
Affiliation(s)
- Weiqi Chen
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, Princeton, NJ 08543, USA
| | - Lifei Wang
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, Princeton, NJ 08543, USA
| | - Gary J Van Berkel
- Organic and Biological Mass Spectrometry Group, Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
| | - Vilmos Kertesz
- Organic and Biological Mass Spectrometry Group, Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA.
| | - Jinping Gan
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, Princeton, NJ 08543, USA.
| |
Collapse
|
112
|
Kulkarni P, Kaftan F, Kynast P, Svatoš A, Böcker S. Correcting mass shifts: A lock mass-free recalibration procedure for mass spectrometry imaging data. Anal Bioanal Chem 2015; 407:7603-13. [DOI: 10.1007/s00216-015-8935-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 07/18/2015] [Accepted: 07/21/2015] [Indexed: 11/30/2022]
|
113
|
Weaver EM, Hummon AB, Keithley RB. Chemometric analysis of MALDI mass spectrometric images of three-dimensional cell culture systems. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2015; 7:7208-7219. [PMID: 26604989 PMCID: PMC4654961 DOI: 10.1039/c5ay00293a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
As imaging mass spectrometry (IMS) has grown in popularity in recent years, the applications of this technique have become increasingly diverse. Currently there is a need for sophisticated data processing strategies that maximize the information gained from large IMS data sets. Traditional two-dimensional heat maps of single ions generated in IMS experiments lack analytical detail, yet manual analysis of multiple peaks across hundreds of pixels within an entire image is time-consuming, tedious and subjective. Here, various chemometric methods were used to analyze data sets obtained by matrix-assisted laser desorption/ionization (MALDI) IMS of multicellular spheroids. HT-29 colon carcinoma multicellular spheroids are an excellent in vitro model system that mimic the three dimensional morphology of tumors in vivo. These data are especially challenging to process because, while different microenvironments exist, the cells are clonal which can result in strong similarities in the mass spectral profiles within the image. In this proof-of-concept study, a combination of principal component analysis (PCA), clustering methods, and linear discriminant analysis was used to identify unique spectral features present in spatially heterogeneous locations within the image. Overall, the application of these exploratory data analysis tools allowed for the isolation and detection of proteomic changes within IMS data sets in an easy, rapid, and unsupervised manner. Furthermore, a simplified, non-mathematical theoretical introduction to the techniques is provided in addition to full command routines within the MATLAB programming environment, allowing others to easily utilize and adapt this approach.
Collapse
Affiliation(s)
- Eric M. Weaver
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | - Amanda B. Hummon
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
- Corresponding Author:
| | | |
Collapse
|
114
|
Yalcin EB, de la Monte SM. Review of matrix-assisted laser desorption ionization-imaging mass spectrometry for lipid biochemical histopathology. J Histochem Cytochem 2015. [PMID: 26209083 DOI: 10.1369/0022155415596202] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Matrix-Assisted Laser Desorption Ionization-Imaging Mass Spectrometry (MALDI-IMS) is a rapidly evolving method used for the in situ visualization and localization of molecules such as drugs, lipids, peptides, and proteins in tissue sections. Therefore, molecules such as lipids, for which antibodies and other convenient detection reagents do not exist, can be detected, quantified, and correlated with histopathology and disease mechanisms. Furthermore, MALDI-IMS has the potential to enhance our understanding of disease pathogenesis through the use of "biochemical histopathology". Herein, we review the underlying concepts, basic methods, and practical applications of MALDI-IMS, including post-processing steps such as data analysis and identification of molecules. The potential utility of MALDI-IMS as a companion diagnostic aid for lipid-related pathological states is discussed.
Collapse
Affiliation(s)
- Emine B Yalcin
- Liver Research Center, Division of Gastroenterology and Department of Medicine (EBY, SMD)
| | - Suzanne M de la Monte
- Liver Research Center, Division of Gastroenterology and Department of Medicine (EBY, SMD),Departments of Neurology, Neurosurgery, and Pathology (SMD),Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI (EBY, SMD)
| |
Collapse
|
115
|
Liubchenko GA, Moriev RM, Kholodna LS. Modern fluorescent techniques to investigate the mechanisms of lymphocyte activation. UKRAINIAN BIOCHEMICAL JOURNAL 2015; 87:33-45. [PMID: 26036129 DOI: 10.15407/ubj87.01.033] [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: 11/17/2022] Open
Abstract
Fluorescent proteins are promising toolsfor studying intracellular signaling processes in lymphocytes. This brief review summarizes fluorescence-based imaging techniques developed in recent years and discusses new methodological advances, such as fluorescent photoswitches, fluorescence recovery after photobleaching (FRAP), fluorescent resonance energy transfer (FRET), fluorescence lifetime imaging microscopy (FLIM), photoactivated localization microscopy (PALM), stochastic optical reconstruction microscopy (STORM), stimulated emission depletion (STED), total internal reflection fluorescence (TIRF) and other techiques. This survey also highlights recent advances in vitro imaging of live tissues, novel applications of flow cytometry with genetically modifed fluorescent proteins, and future prospects for the development of new immunological test systems based on fluorescent protein technology.
Collapse
|
116
|
Cho YT, Su H, Wu WJ, Wu DC, Hou MF, Kuo CH, Shiea J. Biomarker Characterization by MALDI-TOF/MS. Adv Clin Chem 2015; 69:209-54. [PMID: 25934363 DOI: 10.1016/bs.acc.2015.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mass spectrometric techniques frequently used in clinical diagnosis, such as gas chromatography-mass spectrometry, liquid chromatography-mass spectrometry, ambient ionization mass spectrometry, and matrix-assisted laser desorption ionization/time-of-flight mass spectrometry (MALDI-TOF/MS), are discussed. Due to its ability to rapidly detect large biomolecules in trace amounts, MALDI-TOF/MS is an ideal tool for characterizing disease biomarkers in biologic samples. Clinical applications of MS for the identification and characterization of microorganisms, DNA fragments, tissues, and biofluids are introduced. Approaches for using MALDI-TOF/MS to detect various disease biomarkers including peptides, proteins, and lipids in biological fluids are further discussed. Finally, various sample pretreatment methods which improve the detection efficiency of disease biomarkers are introduced.
Collapse
Affiliation(s)
- Yi-Tzu Cho
- Department of Cosmetic Applications and Management, Yuh-Ing Junior College of Health Care & Management, Kaohsiung, Taiwan
| | - Hung Su
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Wen-Jeng Wu
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Feng Hou
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chao-Hung Kuo
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jentaie Shiea
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung, Taiwan; Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan; Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
117
|
Golf O, Muirhead LJ, Speller A, Balog J, Abbassi-Ghadi N, Kumar S, Mróz A, Veselkov K, Takáts Z. XMS: cross-platform normalization method for multimodal mass spectrometric tissue profiling. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:44-54. [PMID: 25380777 DOI: 10.1007/s13361-014-0997-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 06/04/2023]
Abstract
Here we present a proof of concept cross-platform normalization approach to convert raw mass spectra acquired by distinct desorption ionization methods and/or instrumental setups to cross-platform normalized analyte profiles. The initial step of the workflow is database driven peak annotation followed by summarization of peak intensities of different ions from the same molecule. The resulting compound-intensity spectra are adjusted to a method-independent intensity scale by using predetermined, compound-specific normalization factors. The method is based on the assumption that distinct MS-based platforms capture a similar set of chemical species in a biological sample, though these species may exhibit platform-specific molecular ion intensity distribution patterns. The method was validated on two sample sets of (1) porcine tissue analyzed by laser desorption ionization (LDI), desorption electrospray ionization (DESI), and rapid evaporative ionization mass spectrometric (REIMS) in combination with Fourier transformation-based mass spectrometry; and (2) healthy/cancerous colorectal tissue analyzed by DESI and REIMS with the latter being combined with time-of-flight mass spectrometry. We demonstrate the capacity of our method to reduce MS-platform specific variation resulting in (1) high inter-platform concordance coefficients of analyte intensities; (2) clear principal component based clustering of analyte profiles according to histological tissue types, irrespective of the used desorption ionization technique or mass spectrometer; and (3) accurate "blind" classification of histologic tissue types using cross-platform normalized analyte profiles.
Collapse
Affiliation(s)
- Ottmar Golf
- Institute for Inorganic and Analytical Chemistry, Justus Liebig University, Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Waki M, Sugiyama E, Kondo T, Sano K, Setou M. Nanoparticle-assisted laser desorption/ionization for metabolite imaging. Methods Mol Biol 2015; 1203:159-173. [PMID: 25361676 DOI: 10.1007/978-1-4939-1357-2_16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS) has enabled the spatial analysis of various molecules, including peptides, nucleic acids, lipids, and drug molecules. To expand the capabilities of MALDI-IMS, we have established an imaging technique using metal nanoparticles (NPs) to visualize metabolites, termed nanoparticle-assisted laser desorption/ionization imaging mass spectrometry (nano-PALDI-IMS). By utilizing Ag-, Fe-, Au-, and TiO2-derived NPs, we have succeeded in visualizing various metabolites, including fatty acid and glycosphingolipids, with higher sensitivity and spatial resolution than conventional techniques. Herein, we describe the practical experimental procedures and methods associated with nano-PALDI-IMS for the visualization of these molecules.
Collapse
Affiliation(s)
- Michihiko Waki
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu-shi, Shizuoka, Japan
| | | | | | | | | |
Collapse
|
119
|
Li H, Smith BK, Shrestha B, Márk L, Vertes A. Automated cell-by-cell tissue imaging and single-cell analysis for targeted morphologies by laser ablation electrospray ionization mass spectrometry. Methods Mol Biol 2015; 1203:117-127. [PMID: 25361672 DOI: 10.1007/978-1-4939-1357-2_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Mass spectrometry imaging (MSI) is an emerging technology for the mapping of molecular distributions in tissues. In most of the existing studies, imaging is performed by sampling on a predefined rectangular grid that does not reflect the natural cellular pattern of the tissue. Delivering laser pulses by a sharpened optical fiber in laser ablation electrospray ionization (LAESI) mass spectrometry (MS) has enabled the direct analysis of single cells and subcellular compartments. Cell-by-cell imaging had been demonstrated using LAESI-MS, where individual cells were manually selected to serve as natural pixels for tissue imaging. Here we describe a protocol for a novel cell-by-cell LAESI imaging approach that automates cell recognition and addressing for systematic ablation of individual cells. Cell types with particular morphologies can also be selected for analysis. First, the cells are recognized as objects in a microscope image. The coordinates of their centroids are used by a stage-control program to sequentially position the cells under the optical fiber tip for laser ablation. This approach increases the image acquisition efficiency and stability, and enables the investigation of extended or selected tissue areas. In the LAESI process, the ablation events result in mass spectra that represent the metabolite levels in the ablated cells. Peak intensities of selected ions are used to represent the metabolite distributions in the tissue with single-cell resolution.
Collapse
Affiliation(s)
- Hang Li
- Department of Chemistry, W. M. Keck Institute for Proteomics Technology and Applications, The George Washington University, 725 21-st Street, N.W., Washington, DC, 20052, USA
| | | | | | | | | |
Collapse
|
120
|
Mainini V, Lalowski M, Gotsopoulos A, Bitsika V, Baumann M, Magni F. MALDI-imaging mass spectrometry on tissues. Methods Mol Biol 2015; 1243:139-64. [PMID: 25384744 DOI: 10.1007/978-1-4939-1872-0_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF)-profiling and imaging mass spectrometry (MSI) are promising technologies for measuring hundreds of different molecules directly on tissues. For instance, small molecules, drugs and their metabolites, endogenous lipids, carbohydrates and complex peptides/proteins can be measured at the same time. In the most advanced instruments, it is achieved without significant disruption of sample integrity. MSI is a unique approach for assessing the spatial distribution of molecules using graphical multidimensional maps of their constituent analytes, which may for instance be correlated with histopathological alterations in patient tissues. MALDI-TOF-MSI technology has been implemented in hospitals of several countries, where it is routinely used for quick pathogen(s) identification, a task formerly accomplished by laborious and expensive DNA/RNA-based PCR (polymerase chain reaction) screening.In this chapter, we describe how MSI is performed, what is required from the researcher, the instrument vendors and finally what can be achieved with MSI. We restrict our descriptions only to MALDI-MSI although several other MS techniques of ionization can easily be linked to MSI.
Collapse
Affiliation(s)
- Veronica Mainini
- Department of Health Sciences, University Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | | | | | | | | | | |
Collapse
|
121
|
Ghorai S, Seneviratne CA, Murray KK. Tip-enhanced laser ablation sample transfer for biomolecule mass spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:63-70. [PMID: 25287125 PMCID: PMC4276512 DOI: 10.1007/s13361-014-1005-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/10/2014] [Accepted: 09/11/2014] [Indexed: 06/03/2023]
Abstract
Atomic force microscope (AFM) tip-enhanced laser ablation was used to transfer molecules from thin films to a suspended silver wire for off-line mass spectrometry using laser desorption ionization (LDI) and matrix-assisted laser desorption ionization (MALDI). An AFM with a 30 nm radius gold-coated silicon tip was used to image the sample and to hold the tip 15 nm from the surface for material removal using a 355 nm Nd:YAG laser. The ablated material was captured on a silver wire that was held 300 μm vertically and 100 μm horizontally from the tip. For the small molecules anthracene and rhodamine 6G, the wire was cut and affixed to a metal target using double-sided conductive tape and analyzed by LDI using a commercial laser desorption time-of-flight mass spectrometer. Approximately 100 fg of material was ablated from each of the 1 μm ablation spots and transferred with approximately 3% efficiency. For larger polypeptide molecules angiotensin II and bovine insulin, the captured material was dissolved in saturated matrix solution and deposited on a target for MALDI analysis.
Collapse
Affiliation(s)
- Suman Ghorai
- Louisiana State University, Louisiana State University, Baton Rouge, Louisiana, 70803, USA
| | | | - Kermit K. Murray
- Louisiana State University, Louisiana State University, Baton Rouge, Louisiana, 70803, USA
| |
Collapse
|
122
|
Kriegsmann J, Kriegsmann M, Casadonte R. MALDI TOF imaging mass spectrometry in clinical pathology: a valuable tool for cancer diagnostics (review). Int J Oncol 2014; 46:893-906. [PMID: 25482502 DOI: 10.3892/ijo.2014.2788] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/04/2014] [Indexed: 11/06/2022] Open
Abstract
Matrix-assisted laser desorption/ionization (MALDI) time-of-flight (TOF) imaging mass spectrometry (IMS) is an evolving technique in cancer diagnostics and combines the advantages of mass spectrometry (proteomics), detection of numerous molecules, and spatial resolution in histological tissue sections and cytological preparations. This method allows the detection of proteins, peptides, lipids, carbohydrates or glycoconjugates and small molecules.Formalin-fixed paraffin-embedded tissue can also be investigated by IMS, thus, this method seems to be an ideal tool for cancer diagnostics and biomarker discovery. It may add information to the identification of tumor margins and tumor heterogeneity. The technique allows tumor typing, especially identification of the tumor of origin in metastatic tissue, as well as grading and may provide prognostic information. IMS is a valuable method for the identification of biomarkers and can complement histology, immunohistology and molecular pathology in various fields of histopathological diagnostics, especially with regard to identification and grading of tumors.
Collapse
Affiliation(s)
- Jörg Kriegsmann
- MVZ for Histology, Cytology and Molecular Diagnostics, Trier, Germany
| | - Mark Kriegsmann
- Institute for Pathology, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
123
|
Waki M, Ide Y, Ishizaki I, Nagata Y, Masaki N, Sugiyama E, Kurabe N, Nicolaescu D, Yamazaki F, Hayasaka T, Ikegami K, Kondo T, Shibata K, Hiraide T, Taki Y, Ogura H, Shiiya N, Sanada N, Setou M. Single-cell time-of-flight secondary ion mass spectrometry reveals that human breast cancer stem cells have significantly lower content of palmitoleic acid compared to their counterpart non-stem cancer cells. Biochimie 2014; 107 Pt A:73-7. [DOI: 10.1016/j.biochi.2014.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 10/02/2014] [Indexed: 12/13/2022]
|
124
|
McDonnell LA, Römpp A, Balluff B, Heeren RMA, Albar JP, Andrén PE, Corthals GL, Walch A, Stoeckli M. Discussion point: reporting guidelines for mass spectrometry imaging. Anal Bioanal Chem 2014; 407:2035-45. [DOI: 10.1007/s00216-014-8322-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
125
|
Three-dimensional imaging of lipids and metabolites in tissues by nanospray desorption electrospray ionization mass spectrometry. Anal Bioanal Chem 2014; 407:2063-71. [PMID: 25395201 DOI: 10.1007/s00216-014-8174-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/03/2014] [Accepted: 09/08/2014] [Indexed: 10/24/2022]
Abstract
Three-dimensional (3D) imaging of tissue sections is a new frontier in mass spectrometry imaging (MSI). Here, we report on fast 3D imaging of lipids and metabolites associated with mouse uterine decidual cells and embryo at the implantation site on day 6 of pregnancy. 2D imaging of 16-20 serial tissue sections deposited on the same glass slide was performed using nanospray desorption electrospray ionization (nano-DESI)-an ambient ionization technique that enables sensitive localized analysis of analytes on surfaces without special sample pretreatment. In this proof-of-principle study, nano-DESI was coupled to a high-resolution Q-Exactive instrument operated at high repetition rate of >5 Hz with moderate mass resolution of 35,000 (m/Δm at m/z 200), which enabled acquisition of the entire 3D image with a spatial resolution of ∼150 μm in less than 4.5 h. The results demonstrate localization of acetylcholine in the primary decidual zone (PDZ) of the implantation site throughout the depth of the tissue examined, indicating an important role of this signaling molecule in decidualization. Choline and phosphocholine-metabolites associated with cell growth-are enhanced in the PDZ and abundant in other cellular regions of the implantation site. Very different 3D distributions were obtained for fatty acids (FA), oleic acid and linoleic acid (FA 18:1 and FA 18:2), differing only by one double bond. Localization of FA 18:2 in the PDZ indicates its important role in decidualization while FA 18:1 is distributed more evenly throughout the tissue. In contrast, several lysophosphatidylcholines (LPC) observed in this study show donut-like distributions with localization around the PDZ. Complementary distributions with minimal overlap were observed for LPC 18:0 and FA 18:2 while the 3D image of the potential precursor phosphatidylcholine 36:2 (PC 36:2) showed a significant overlap with both LPC 18:0 and FA 18:2.
Collapse
|
126
|
Balluff B, Frese CK, Maier SK, Schöne C, Kuster B, Schmitt M, Aubele M, Höfler H, Deelder AM, Heck A, Hogendoorn PCW, Morreau J, Maarten Altelaar AF, Walch A, McDonnell LA. De novo discovery of phenotypic intratumour heterogeneity using imaging mass spectrometry. J Pathol 2014; 235:3-13. [PMID: 25201776 DOI: 10.1002/path.4436] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/04/2014] [Accepted: 09/03/2014] [Indexed: 12/31/2022]
Abstract
An essential and so far unresolved factor influencing the evolution of cancer and the clinical management of patients is intratumour clonal and phenotypic heterogeneity. However, the de novo identification of tumour subpopulations is so far both a challenging and an unresolved task. Here we present the first systematic approach for the de novo discovery of clinically detrimental molecular tumour subpopulations. In this proof-of-principle study, spatially resolved, tumour-specific mass spectra were acquired, using matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry, from tissues of 63 gastric carcinoma and 32 breast carcinoma patients. The mass spectra, representing the proteomic heterogeneity within tumour areas, were grouped by a corroborated statistical clustering algorithm in order to obtain segmentation maps of molecularly distinct regions. These regions were presumed to represent different phenotypic tumour subpopulations. This was confirmed by linking the presence of these tumour subpopulations to the patients' clinical data. This revealed several of the detected tumour subpopulations to be associated with a different overall survival of the gastric cancer patients (p = 0.025) and the presence of locoregional metastases in patients with breast cancer (p = 0.036). The procedure presented is generic and opens novel options in cancer research, as it reveals microscopically indistinct tumour subpopulations that have an adverse impact on clinical outcome. This enables their further molecular characterization for deeper insights into the biological processes of cancer, which may finally lead to new targeted therapies.
Collapse
Affiliation(s)
- Benjamin Balluff
- Centre for Proteomics and Metabolomics, Leiden University Medical Centre, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Aichler M, Luber B, Lordick F, Walch A. Proteomic and metabolic prediction of response to therapy in gastric cancer. World J Gastroenterol 2014; 20:13648-13657. [PMID: 25320503 PMCID: PMC4194549 DOI: 10.3748/wjg.v20.i38.13648] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 06/13/2014] [Indexed: 02/06/2023] Open
Abstract
Several new treatment options for gastric cancer have been introduced but the prognosis of patients diagnosed with gastric cancer is still poor. Disease prognosis could be improved for high-risk individuals by implementing earlier screenings. Because many patients are asymptomatic during the early stages of gastric cancer, the diagnosis is often delayed and patients present with unresectable locally advanced or metastatic disease. Cytotoxic treatment has been shown to prolong survival in general, but not all patients are responders. The application of targeted therapies and multimodal treatment has improved prognosis for those with advanced disease. However, these new therapeutic strategies do not uniformly benefit all patients. Predicting whether patients will respond to specific therapies would be of particular value and would allow for stratifying patients for personalized treatment strategies. Metabolic imaging by positron emission tomography was the first technique with the potential to predict the response of esophago-gastric cancer to neoadjuvant therapy. Exploring and validating tissue-based biomarkers are ongoing processes. In this review, we discuss the status of several targeted therapies for gastric cancer, as well as proteomic and metabolic methods for investigating biomarkers for therapy response prediction in gastric cancer.
Collapse
|
128
|
Wang Y, Zhang X, Zhai Y, Jiang Y, Fang X, Zhou M, Deng Y, Xu W. Mass Selective Ion Transfer and Accumulation in Ion Trap Arrays. Anal Chem 2014; 86:10164-70. [DOI: 10.1021/ac502583b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Yuzhuo Wang
- School
of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaohua Zhang
- Department
of Chemistry, FUDAN University, Shanghai 200433, China
| | - Yanbing Zhai
- School
of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - You Jiang
- National Institute
of Metrology, Beijing 100013, China
| | - Xiang Fang
- National Institute
of Metrology, Beijing 100013, China
| | - Mingfei Zhou
- Department
of Chemistry, FUDAN University, Shanghai 200433, China
| | - Yulin Deng
- School
of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Wei Xu
- School
of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
129
|
Liquid microjunction surface sampling of acetaminophen, terfenadine and their metabolites in thin tissue sections. Bioanalysis 2014; 6:2599-606. [DOI: 10.4155/bio.14.130] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: The aim of this work was to evaluate the analytical performance of a fully automated droplet-based surface-sampling system for determining the distribution of the drugs acetaminophen and terfenadine, and their metabolites, in rat thin tissue sections. Results: The rank order of acetaminophen concentration observed in tissues was stomach > small intestine > liver, while the concentrations of its glucuronide and sulfate metabolites were greatest in the liver and small intestine. Terfenadine was most concentrated in the liver and kidney, while its major metabolite, fexofenadine, was found in the liver and small intestine. Conclusion: The spatial distributions of both drugs and their respective metabolites observed in this work were consistent with previous studies using radiolabeled drugs.
Collapse
|
130
|
Application of desorption electrospray ionization mass spectrometry imaging in breast cancer margin analysis. Proc Natl Acad Sci U S A 2014; 111:15184-9. [PMID: 25246570 DOI: 10.1073/pnas.1408129111] [Citation(s) in RCA: 189] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Distinguishing tumor from normal glandular breast tissue is an important step in breast-conserving surgery. Because this distinction can be challenging in the operative setting, up to 40% of patients require an additional operation when traditional approaches are used. Here, we present a proof-of-concept study to determine the feasibility of using desorption electrospray ionization mass spectrometry imaging (DESI-MSI) for identifying and differentiating tumor from normal breast tissue. We show that tumor margins can be identified using the spatial distributions and varying intensities of different lipids. Several fatty acids, including oleic acid, were more abundant in the cancerous tissue than in normal tissues. The cancer margins delineated by the molecular images from DESI-MSI were consistent with those margins obtained from histological staining. Our findings prove the feasibility of classifying cancerous and normal breast tissues using ambient ionization MSI. The results suggest that an MS-based method could be developed for the rapid intraoperative detection of residual cancer tissue during breast-conserving surgery.
Collapse
|
131
|
Verbeeck N, Yang J, De Moor B, Caprioli R, Waelkens E, Van de Plas R. Automated anatomical interpretation of ion distributions in tissue: linking imaging mass spectrometry to curated atlases. Anal Chem 2014; 86:8974-82. [PMID: 25153352 PMCID: PMC4165455 DOI: 10.1021/ac502838t] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 07/30/2014] [Indexed: 12/17/2022]
Abstract
Imaging mass spectrometry (IMS) has become a prime tool for studying the distribution of biomolecules in tissue. Although IMS data sets can become very large, computational methods have made it practically feasible to search these experiments for relevant findings. However, these methods lack access to an important source of information that many human interpretations rely upon: anatomical insight. In this work, we address this need by (1) integrating a curated anatomical data source with an empirically acquired IMS data source, establishing an algorithm-accessible link between them and (2) demonstrating the potential of such an IMS-anatomical atlas link by applying it toward automated anatomical interpretation of ion distributions in tissue. The concept is demonstrated in mouse brain tissue, using the Allen Mouse Brain Atlas as the curated anatomical data source that is linked to MALDI-based IMS experiments. We first develop a method to spatially map the anatomical atlas to the IMS data sets using nonrigid registration techniques. Once a mapping is established, a second computational method, called correlation-based querying, gives an elementary demonstration of the link by delivering basic insight into relationships between ion images and anatomical structures. Finally, a third algorithm moves further beyond both registration and correlation by providing automated anatomical interpretation of ion images. This task is approached as an optimization problem that deconstructs ion distributions as combinations of known anatomical structures. We demonstrate that establishing a link between an IMS experiment and an anatomical atlas enables automated anatomical annotation, which can serve as an important accelerator both for human and machine-guided exploration of IMS experiments.
Collapse
Affiliation(s)
- Nico Verbeeck
- Department
of Electrical Engineering (ESAT), STADIUS Center for
Dynamical Systems, Signal Processing, and Data Analytics, KU Leuven, Kasteelpark
Arenberg 10, Box 2446, 3001 Leuven, Belgium
- iMinds
Medical IT, Kasteelpark
Arenberg 10, Box 2446, 3001 Leuven, Belgium
| | - Junhai Yang
- Mass
Spectrometry Research Center and Departments of Biochemistry, Chemistry,
Pharmacology, and Medicine, Vanderbilt University, 465 21st Avenue South, MRB III Suite
9160, Nashville, Tennessee 37232, United States
| | - Bart De Moor
- Department
of Electrical Engineering (ESAT), STADIUS Center for
Dynamical Systems, Signal Processing, and Data Analytics, KU Leuven, Kasteelpark
Arenberg 10, Box 2446, 3001 Leuven, Belgium
- iMinds
Medical IT, Kasteelpark
Arenberg 10, Box 2446, 3001 Leuven, Belgium
| | - Richard
M. Caprioli
- Mass
Spectrometry Research Center and Departments of Biochemistry, Chemistry,
Pharmacology, and Medicine, Vanderbilt University, 465 21st Avenue South, MRB III Suite
9160, Nashville, Tennessee 37232, United States
| | - Etienne Waelkens
- Sybioma, KU Leuven, Campus Gasthuisberg O&N 2, Herestraat 49, Box
802, 3000 Leuven, Belgium
- Department
of Cellular and Molecular Medicine, KU Leuven, Campus Gasthuisberg O&N 1, Herestraat
49, Box 901, 3000 Leuven, Belgium
| | - Raf Van de Plas
- Mass
Spectrometry Research Center and Departments of Biochemistry, Chemistry,
Pharmacology, and Medicine, Vanderbilt University, 465 21st Avenue South, MRB III Suite
9160, Nashville, Tennessee 37232, United States
| |
Collapse
|
132
|
Klein O, Strohschein K, Nebrich G, Oetjen J, Trede D, Thiele H, Alexandrov T, Giavalisco P, Duda GN, von Roth P, Geissler S, Klose J, Winkler T. MALDI imaging mass spectrometry: discrimination of pathophysiological regions in traumatized skeletal muscle by characteristic peptide signatures. Proteomics 2014; 14:2249-60. [PMID: 25056804 DOI: 10.1002/pmic.201400088] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/24/2014] [Accepted: 07/21/2014] [Indexed: 01/06/2023]
Abstract
Due to formation of fibrosis and the loss of contractile muscle tissue, severe muscle injuries often result in insufficient healing marked by a significant reduction of muscle force and motor activity. Our previous studies demonstrated that the local transplantation of mesenchymal stromal cells into an injured skeletal muscle of the rat improves the functional outcome of the healing process. Since, due to the lack of sufficient markers, the accurate discrimination of pathophysiological regions in injured skeletal muscle is inadequate, underlying mechanisms of the beneficial effects of mesenchymal stromal cell transplantation on primary trauma and trauma adjacent muscle area remain elusive. For discrimination of these pathophysiological regions, formalin-fixed injured skeletal muscle tissue was analyzed by MALDI imaging MS. By using two computational evaluation strategies, a supervised approach (ClinProTools) and unsupervised segmentation (SCiLS Lab), characteristic m/z species could be assigned to primary trauma and trauma adjacent muscle regions. Using "bottom-up" MS for protein identification and validation of results by immunohistochemistry, we could identify two proteins, skeletal muscle alpha actin and carbonic anhydrase III, which discriminate between the secondary damage on adjacent tissue and the primary traumatized muscle area. Our results underscore the high potential of MALDI imaging MS to describe the spatial characteristics of pathophysiological changes in muscle.
Collapse
Affiliation(s)
- Oliver Klein
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany; Core Unit Proteomics, Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Min KW, Bang JY, Kim KP, Kim WS, Lee SH, Shanta SR, Lee JH, Hong JH, Lim SD, Yoo YB, Na CH. Imaging mass spectrometry in papillary thyroid carcinoma for the identification and validation of biomarker proteins. J Korean Med Sci 2014; 29:934-40. [PMID: 25045225 PMCID: PMC4101781 DOI: 10.3346/jkms.2014.29.7.934] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/24/2014] [Indexed: 01/09/2023] Open
Abstract
Direct tissue imaging mass spectrometry (IMS) by matrix-assisted laser desorption ionization and time-of-flight (MALDI-TOF) mass spectrometry has become increasingly important in biology and medicine, because this technology can detect the relative abundance and spatial distribution of interesting proteins in tissues. Five thyroid cancer samples, along with normal tissue, were sliced and transferred onto conductive glass slides. After laser scanning by MALDI-TOF equipped with a smart beam laser, images were created for individual masses and proteins were classified at 200-µm spatial resolution. Based on the spatial distribution, region-specific proteins on a tumor lesion could be identified by protein extraction from tumor tissue and analysis using liquid chromatography with tandem mass spectrometry (LC-MS/MS). Using all the spectral data at each spot, various intensities of a specific peak were detected in the tumor and normal regions of the thyroid. Differences in the molecular weights of expressed proteins between tumor and normal regions were analyzed using unsupervised and supervised clustering. To verify the presence of discovered proteins through IMS, we identified ribosomal protein P2, which is specific for cancer. We have demonstrated the feasibility of IMS as a useful tool for the analysis of tissue sections, and identified the tumor-specific protein ribosomal protein P2.
Collapse
Affiliation(s)
- Kyueng-Whan Min
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
| | - Joo-Young Bang
- Department of Molecular Biotechnology, WCU Program, Konkuk University, Seoul, Korea
- SMART Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Kwang Pyo Kim
- Department of Molecular Biotechnology, WCU Program, Konkuk University, Seoul, Korea
- SMART Institute of Advanced Biomedical Science, Konkuk University, Seoul, Korea
| | - Wan-Seop Kim
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
- The Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - Sang Hwa Lee
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
| | - Selina Rahman Shanta
- Department of Molecular Biotechnology, WCU Program, Konkuk University, Seoul, Korea
| | - Jeong Hwa Lee
- Department of Molecular Biotechnology, WCU Program, Konkuk University, Seoul, Korea
| | - Ji Hye Hong
- Department of Molecular Biotechnology, WCU Program, Konkuk University, Seoul, Korea
| | - So Dug Lim
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
| | - Young-Bum Yoo
- Department of Surgery, Konkuk University School of Medicine, Seoul, Korea
| | - Chan-Hyun Na
- Department of Molecular Biotechnology, WCU Program, Konkuk University, Seoul, Korea
| |
Collapse
|
134
|
Ogrinc Potočnik N, Škrášková K, Flinders B, Pelicon P, Heeren RMA. Gold sputtered fiducial markers for combined secondary ion mass spectrometry and MALDI imaging of tissue samples. Anal Chem 2014; 86:6781-5. [PMID: 24918500 DOI: 10.1021/ac500308s] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mass spectrometry imaging (MSI) is a label free technique capable of providing simultaneous identification and localization of biomolecules. A multimodal approach is required that allows for the study of the complexity of biological tissue samples to overcome the limitations of a single MSI technique. Secondary ion mass spectrometry (SIMS) allows for high spatial resolution imaging while matrix-assisted laser desorption (MALDI) offers a significantly wider mass range. The combination of coregistered SIMS and MALDI images results in detailed and unique biomolecular information. In this Technical Note, we describe how gold sputtered/implanted fiducial markers (FM) are created and can be used to ensure a proper overlay and coregistration of the two-dimensional images provided by the two MSI modalities.
Collapse
|
135
|
Ostermann KM, Luf A, Lutsch NM, Dieplinger R, Mechtler TP, Metz TF, Schmid R, Kasper DC. MALDI Orbitrap mass spectrometry for fast and simplified analysis of novel street and designer drugs. Clin Chim Acta 2014; 433:254-8. [DOI: 10.1016/j.cca.2014.03.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 03/13/2014] [Accepted: 03/13/2014] [Indexed: 11/26/2022]
|
136
|
Horn PJ, Chapman KD. Lipidomics in situ: Insights into plant lipid metabolism from high resolution spatial maps of metabolites. Prog Lipid Res 2014; 54:32-52. [DOI: 10.1016/j.plipres.2014.01.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/14/2014] [Accepted: 01/14/2014] [Indexed: 12/31/2022]
|
137
|
Acquadro E, Caron I, Tortarolo M, Bucci EM, Bendotti C, Corpillo D. Human SOD1-G93A specific distribution evidenced in murine brain of a transgenic model for amyotrophic lateral sclerosis by MALDI imaging mass spectrometry. J Proteome Res 2014; 13:1800-9. [PMID: 24579824 DOI: 10.1021/pr400942n] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, fatal neurodegenerative disease caused by the degeneration of motor neurons. The transgenic mouse model carrying the human SOD1G93A mutant gene (hSOD1G93A mouse) represents one of the most reliable and widely used model of this pathology. In the present work, the innovative technique of matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) was applied in the study of pathological alterations at the level of small brain regions such as facial and trigeminal nuclei, which in rodents are extremely small and would be difficult to analyze with classical proteomics approaches. Comparing slices from three mice groups (transgenic hSOD1G93A, transgenic hSOD1WT, and nontransgenic, Ntg), this technique allowed us to evidence the accumulation of hSOD1G93A in the facial and trigeminal nuclei, where it generates aggregates. This phenomenon is likely to be correlated to the degeneration observed in these regions. Moreover, a statistical analysis allowed us to highlight other proteins as differentially expressed among the three mice groups analyzed. Some of them were identified by reverse-phase HPLC fractionation of extracted proteins and mass spectrometric analysis before and after trypsin digestion. In particular, the 40S ribosomal protein S19 (RPS19) was upregulated in the parenkyma and reactive glial cells in facial nuclei of hSOD1G93A mice when compared to transgenic hSOD1WT and nontransgenic ones.
Collapse
Affiliation(s)
- Elena Acquadro
- ABLE Bioscences, BioIndustry Park Silvano Fumero S.p.A., Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy
| | | | | | | | | | | |
Collapse
|
138
|
Automated liquid microjunction surface sampling-HPLC-MS/MS analysis of drugs and metabolites in whole-body thin tissue sections. Bioanalysis 2014; 5:819-26. [PMID: 23534426 DOI: 10.4155/bio.13.42] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The aim of this work was to develop a fully automated liquid extraction-based surface sampling system utilizing a commercially available autosampler coupled with HPLC-MS/MS detection. RESULTS Discrete spots selected for droplet-based sampling and automated sample queue generation, for both the autosampler and MS, were enabled by using in-house developed software. In addition, co-registration of spatially resolved sampling positions and HPLC-MS information to generate heat maps of compounds monitored for subsequent data analysis was also available in the software. The system was evaluated with whole-body thin tissue sections from propranolol-dosed rats. CONCLUSION The spatial distributions of both the drug and its hydroxypropranolol glucuronide metabolites were consistent with previous studies employing other liquid extraction-based surface sampling methodologies.
Collapse
|
139
|
Neubert P, Walch A. Current frontiers in clinical research application of MALDI imaging mass spectrometry. Expert Rev Proteomics 2014; 10:259-73. [DOI: 10.1586/epr.13.19] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
140
|
Cillero-Pastor B, Heeren RMA. Matrix-Assisted Laser Desorption Ionization Mass Spectrometry Imaging for Peptide and Protein Analyses: A Critical Review of On-Tissue Digestion. J Proteome Res 2013; 13:325-35. [DOI: 10.1021/pr400743a] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Berta Cillero-Pastor
- FOM Institute AMOLF, Biomolecular Imaging Mass Spectrometry (BIMS), AMOLF Science Park 104, 1098 XG Amsterdam, The Netherlands
| | - Ron M. A. Heeren
- FOM Institute AMOLF, Biomolecular Imaging Mass Spectrometry (BIMS), AMOLF Science Park 104, 1098 XG Amsterdam, The Netherlands
| |
Collapse
|
141
|
A critical evaluation of the current state-of-the-art in quantitative imaging mass spectrometry. Anal Bioanal Chem 2013; 406:1275-89. [DOI: 10.1007/s00216-013-7478-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/28/2013] [Accepted: 10/31/2013] [Indexed: 01/29/2023]
|
142
|
Le CH, Han J, Borchers CH. Dithranol as a matrix for matrix assisted laser desorption/ionization imaging on a fourier transform ion cyclotron resonance mass spectrometer. J Vis Exp 2013:e50733. [PMID: 24300588 DOI: 10.3791/50733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mass spectrometry imaging (MSI) determines the spatial localization and distribution patterns of compounds on the surface of a tissue section, mainly using MALDI (matrix assisted laser desorption/ionization)-based analytical techniques. New matrices for small-molecule MSI, which can improve the analysis of low-molecular weight (MW) compounds, are needed. These matrices should provide increased analyte signals while decreasing MALDI background signals. In addition, the use of ultrahigh-resolution instruments, such as Fourier transform ion cyclotron resonance (FTICR) mass spectrometers, has the ability to resolve analyte signals from matrix signals, and this can partially overcome many problems associated with the background originating from the MALDI matrix. The reduction in the intensities of the metastable matrix clusters by FTICR MS can also help to overcome some of the interferences associated with matrix peaks on other instruments. High-resolution instruments such as the FTICR mass spectrometers are advantageous as they can produce distribution patterns of many compounds simultaneously while still providing confidence in chemical identifications. Dithranol (DT; 1,8-dihydroxy-9,10-dihydroanthracen-9-one) has previously been reported as a MALDI matrix for tissue imaging. In this work, a protocol for the use of DT for MALDI imaging of endogenous lipids from the surfaces of mammalian tissue sections, by positive-ion MALDI-MS, on an ultrahigh-resolution hybrid quadrupole FTICR instrument has been provided.
Collapse
Affiliation(s)
- Cuong H Le
- University of Victoria-Genome BC Proteomics Centre, University of Victoria
| | | | | |
Collapse
|
143
|
Rapid estimation of the energy charge from cell lysates using matrix-assisted laser desorption/ionization mass spectrometry: role of in-source fragmentation. Anal Biochem 2013; 447:107-13. [PMID: 24269891 DOI: 10.1016/j.ab.2013.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/04/2013] [Accepted: 11/08/2013] [Indexed: 11/21/2022]
Abstract
Nucleotides are key players in the central energy metabolism of cells. Here we show how to estimate the energy charge from cell lysates by direct negative ion matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) using 9-aminoacridine as matrix. We found a high level of in-source decay of all the phosphorylated nucleotides, with some of them producing considerable amounts of adenosine-5'-diphosphate (ADP) fragment ions. We investigated the behavior of adenosine-5'-monophosphate (AMP), ADP, and adenosine-5'-triphosphate (ATP) as well as the cofactors coenzyme A (CoA) and acetyl-coenzyme A (ACoA) and nicotinamide adenine dinucleotides (NAD⁺ and NADH) in detail. In-source decay of these compounds depends strongly on the applied laser power and on the extraction pulse delay. At standard instrument settings, the 9-aminoacridine (9-AA) matrix resulted in a much higher in-source decay compared with 2,4,6-trihydroxyacetophenone (2,4,6-THAP). By adding ¹³C-labeled ATP to a cell lysate, we were able to determine the degree of in-source decay during an experiment. Analyzing a cell extract of the monocytic cell line THP-1 with [¹³C]ATP as internal standard, we were able to obtain values for the energy charge that were similar to those determined by a reference liquid chromatography electrospray ionization coupled to mass spectrometry (LC-ESI-MS) method.
Collapse
|
144
|
Lanekoff I, Burnum-Johnson K, Thomas M, Short J, Carson JP, Cha J, Dey SK, Yang P, Conaway MCP, Laskin J. High-speed tandem mass spectrometric in situ imaging by nanospray desorption electrospray ionization mass spectrometry. Anal Chem 2013; 85:9596-603. [PMID: 24040919 PMCID: PMC3867692 DOI: 10.1021/ac401760s] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nanospray desorption electrospray ionization (nano-DESI) combined with tandem mass spectrometry (MS/MS), high-resolution mass analysis of the fragment ions (m/Δm = 17 500 at m/z 200), and rapid spectral acquisition enabled simultaneous imaging and identification of a large number of metabolites and lipids from 92 selected m/z windows (±1 Da) with a spatial resolution of better than 150 μm. Mouse uterine sections of implantation sites on day 6 of pregnancy were analyzed in the ambient environment without any sample pretreatment. MS/MS imaging was performed by scanning the sample under the nano-DESI probe at 10 μm/s, while higher-energy collision-induced dissociation (HCD) spectra were acquired for a targeted inclusion list of 92 m/z values at a rate of ∼6.3 spectra/s. Molecular ions and their corresponding fragments, separated by high-resolution mass analysis, were assigned on the basis of accurate mass measurement. Using this approach, we were able to identify and image both abundant and low-abundance isobaric and isomeric species within each m/z window. MS/MS analysis enabled efficient separation and identification of isomeric and isobaric phospholipids that are difficult to separate in full-scan mode. Furthermore, we identified several metabolites associated with early pregnancy and obtained the first 2D images of these molecules.
Collapse
Affiliation(s)
- Ingela Lanekoff
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352
| | - Kristin Burnum-Johnson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352
| | - Mathew Thomas
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352
| | - Joshua Short
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352
| | - James P. Carson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352
| | - Jeeyeon Cha
- Division of Reproductive Sciences, The Perinatal Institute, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Sudhansu K. Dey
- Division of Reproductive Sciences, The Perinatal Institute, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | | | | | - Julia Laskin
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352
| |
Collapse
|
145
|
MALDI-MS tissue imaging identification of biliverdin reductase B overexpression in prostate cancer. J Proteomics 2013; 91:500-14. [DOI: 10.1016/j.jprot.2013.08.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 07/30/2013] [Accepted: 08/03/2013] [Indexed: 01/18/2023]
|
146
|
Oetjen J, Aichler M, Trede D, Strehlow J, Berger J, Heldmann S, Becker M, Gottschalk M, Kobarg JH, Wirtz S, Schiffler S, Thiele H, Walch A, Maass P, Alexandrov T. MRI-compatible pipeline for three-dimensional MALDI imaging mass spectrometry using PAXgene fixation. J Proteomics 2013; 90:52-60. [DOI: 10.1016/j.jprot.2013.03.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 03/02/2013] [Accepted: 03/19/2013] [Indexed: 12/18/2022]
|
147
|
Passarelli MK, Ewing AG. Single-cell imaging mass spectrometry. Curr Opin Chem Biol 2013; 17:854-9. [PMID: 23948695 DOI: 10.1016/j.cbpa.2013.07.017] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/03/2013] [Accepted: 07/03/2013] [Indexed: 10/26/2022]
Abstract
Single-cell imaging mass spectrometry (IMS) is a powerful technique used to map the distributions of endogenous biomolecules with subcellular resolution. Currently, secondary ion mass spectrometry is the predominant technique for single-cell IMS, thanks to its submicron lateral resolution and surface sensitivity. However, recent methodological and technological developments aimed at improving the spatial resolution of matrix assisted laser desorption ionization (MALDI) have made this technique a potential platform of single-cell IMS. MALDI opens the field of single-cell IMS to new possibilities, including single cell proteomic imaging and atmospheric pressure analyses; however, sensitivity is a challenge. In this report, we estimate the availability of proteins and lipids in a single cell and discuss strategies employed to improve sensitivity at the single-cell level.
Collapse
|
148
|
Nakamoto A, Sato T, Hirosawa N, Nakamoto N, Enoki Y, Chida D, Usui M, Takeda S, Nagai T, Sasaki A, Sakamoto Y, Yoda T. Proteomics-based identification of novel proteins in temporal tendons of patients with masticatory muscle tendon--aponeurosis hyperplasia. Int J Oral Maxillofac Surg 2013; 43:113-9. [PMID: 23870541 DOI: 10.1016/j.ijom.2013.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 06/02/2013] [Accepted: 06/06/2013] [Indexed: 11/26/2022]
Abstract
Masticatory muscle tendon-aponeurosis hyperplasia (MMTAH) is a new disease associated with limited mouth opening that is often misdiagnosed as a temporomandibular disorder; subsequently, patients are mistakenly treated with irreversible operations. Due to the poor presentation and characterization of symptoms, the underlying pathological conditions remain unclear. We have previously conducted a proteomic analysis of tendons derived from one MMTAH subject and one facial deformity subject using two-dimensional fluorescence difference gel electrophoresis and liquid chromatography coupled with tandem mass spectrometry. However, the results were obtained for only one subject. The aim of the present study was to confirm the expression of specific molecules in tendon tissues from multiple subjects with MMTAH by applying two-dimensional polyacrylamide gel electrophoresis with matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Of the 19 proteins identified in tendons from both MMTAH and facial deformity patients, fibrinogen fragment D and beta-crystallin A4 were up-regulated, whereas myosin light chain 4 was down-regulated in MMTAH. We also found fibrinogen to be expressed robustly in tendon tissues of MMTAH patients. Our data provide the possibility that the distinctive expression of these novel proteins is associated with the pathology of MMTAH.
Collapse
Affiliation(s)
- A Nakamoto
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| | - T Sato
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan.
| | - N Hirosawa
- Department of Biomedical Research Center, Division of Analytical Science, Saitama Medical University, Saitama, Japan
| | - N Nakamoto
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| | - Y Enoki
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| | - D Chida
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| | - M Usui
- Department of Periodontology, Kyushu Dental College, Kitakyushu City, Fukuoka, Japan
| | - S Takeda
- Division of Endocrinology, Metabolism and Nephrology, Keio University, Tokyo, Japan
| | - T Nagai
- Department of Pathology, Saitama Medical University, Saitama, Japan
| | - A Sasaki
- Department of Pathology, Saitama Medical University, Saitama, Japan
| | - Y Sakamoto
- Department of Biomedical Research Center, Division of Analytical Science, Saitama Medical University, Saitama, Japan
| | - T Yoda
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| |
Collapse
|
149
|
Griffiths RL, Sarsby J, Guggenheim EJ, Race AM, Steven RT, Fear J, Lalor PF, Bunch J. Formal lithium fixation improves direct analysis of lipids in tissue by mass spectrometry. Anal Chem 2013; 85:7146-53. [PMID: 23879734 DOI: 10.1021/ac400737z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mass spectrometry imaging is a powerful method for imaging and in situ characterization of lipids in thin tissue sections. Structural elucidation of lipids is often achieved via collision induced dissociation, and lithium-lipid adducts have been widely reported as providing the most structurally informative fragment ions. We present a method for the incorporation of lithium salts into tissue imaging experiments via fixation of samples in formal lithium solutions. The method is suitable for preparation of single tissue sections, or as an immersion fixation method for whole tissue blocks or organs prior to sectioning. We compare lithium adduct detection and MALDI-MSI of murine brain from analysis of tissues prepared in different ways. Tissues prepared in formal solutions containing lithium or sodium salts before coating in matrix via air-spray deposition are compared with fresh samples coated in lithium-doped matrix preparations by either dry-coating or air-spray deposition. Sample preparation via fixation in formal lithium is shown to yield the highest quality images of lithium adducts, resulting in acquisition of more informative product ion spectra in MALDI MS/MS profiling and imaging experiments. Finally, the compatibility of formal lithium solutions with standard histological staining protocols (hemotoxylin and eosin, Van Giessen and Oil Red O) is demonstrated in a study of human liver tissue.
Collapse
Affiliation(s)
- Rian L Griffiths
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Liu X, Weaver EM, Hummon AB. Evaluation of therapeutics in three-dimensional cell culture systems by MALDI imaging mass spectrometry. Anal Chem 2013; 85:6295-302. [PMID: 23724927 PMCID: PMC4118837 DOI: 10.1021/ac400519c] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug penetration into solid tumors is critical for the effectiveness of clinical chemotherapy. Failing to consider the efficiency of drug penetration can lead to fatal recurrence in many cancers. Three-dimensional (3D) cell cultures have served as an important model system and have contributed to valuable assays in drug discovery studies. However, limited methodologies result in incomplete evaluation of the distribution of many anticancer drugs. As a proof-of-concept study, we have applied matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) in HCT 116 colon carcinoma multicellular spheroids to assess the distribution of the anticancer drug, irinotecan. The time-dependent penetration of irinotecan was visualized and the localization of three metabolites as well as the parent drug in treated spheroids was mapped. To validate the identities of the metabolites, we analyzed extracts from drug-treated spheroids using nanoflow liquid chromatography-tandem mass spectrometry (nLC-MS/MS). Ten metabolites were identified with nLC-MS/MS, including those detected by MALDI IMS. This novel approach allows the measurement of drug penetration and distribution in 3D culture mimics and provides a more cost and time-effective approach for the testing of new pharmaceuticals compared to animal models.
Collapse
Affiliation(s)
- Xin Liu
- University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Eric M. Weaver
- University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Amanda B. Hummon
- University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|