Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 44(3): 741-754, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38299357

RESUMO

BACKGROUND: The metabolic alterations occurring within the arterial architecture during atherosclerosis development remain poorly understood, let alone those particular to each arterial tunica. We aimed first to identify, in a spatially resolved manner, the specific metabolic changes in plaque, media, adventitia, and cardiac tissue between control and atherosclerotic murine aortas. Second, we assessed their translatability to human tissue and plasma for cardiovascular risk estimation. METHODS: In this observational study, mass spectrometry imaging (MSI) was applied to identify region-specific metabolic differences between atherosclerotic (n=11) and control (n=11) aortas from low-density lipoprotein receptor-deficient mice, via histology-guided virtual microdissection. Early and advanced plaques were compared within the same atherosclerotic animals. Progression metabolites were further analyzed by MSI in 9 human atherosclerotic carotids and by targeted mass spectrometry in human plasma from subjects with elective coronary artery bypass grafting (cardiovascular risk group, n=27) and a control group (n=27). RESULTS: MSI identified 362 local metabolic alterations in atherosclerotic mice (log2 fold-change ≥1.5; P≤0.05). The lipid composition of cardiac tissue is altered during atherosclerosis development and presents a generalized accumulation of glycerophospholipids, except for lysolipids. Lysolipids (among other glycerophospholipids) were found at elevated levels in all 3 arterial layers of atherosclerotic aortas. LPC(18:0) (lysophosphatidylcholine; P=0.024) and LPA(18:1) (lysophosphatidic acid; P=0.025) were found to be significantly elevated in advanced plaques as compared with mouse-matched early plaques. Higher levels of both lipid species were also observed in fibrosis-rich areas of advanced- versus early-stage human samples. They were found to be significantly reduced in human plasma from subjects with elective coronary artery bypass grafting (P<0.001 and P=0.031, respectively), with LPC(18:0) showing significant association with cardiovascular risk (odds ratio, 0.479 [95% CI, 0.225-0.883]; P=0.032) and diagnostic potential (area under the curve, 0.778 [95% CI, 0.638-0.917]). CONCLUSIONS: An altered phospholipid metabolism occurs in atherosclerosis, affecting both the aorta and the adjacent heart tissue. Plaque-progression lipids LPC(18:0) and LPA(18:1), as identified by MSI on tissue, reflect cardiovascular risk in human plasma.


Assuntos
Doenças da Aorta , Aterosclerose , Doenças Cardiovasculares , Placa Aterosclerótica , Humanos , Animais , Camundongos , Placa Aterosclerótica/metabolismo , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/metabolismo , Fatores de Risco , Aterosclerose/diagnóstico , Aterosclerose/metabolismo , Aorta/diagnóstico por imagem , Aorta/metabolismo , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Glicerofosfolipídeos/metabolismo , Fatores de Risco de Doenças Cardíacas
2.
Analyst ; 148(24): 6161-6187, 2023 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-37947390

RESUMO

Mass spectrometry imaging has advanced from a niche technique to a widely applied spatial biology tool operating at the forefront of numerous fields, most notably making a significant impact in biomedical pharmacological research. The growth of the field has gone hand in hand with an increase in publications and usage of the technique by new laboratories, and consequently this has led to a shift from general MSI reviews to topic-specific reviews. Given this development, we see the need to recapitulate the strengths of MSI by providing a more holistic overview of state-of-the-art MSI studies to provide the new generation of researchers with an up-to-date reference framework. Here we review scientific advances for the six largest biomedical fields of MSI application (oncology, pharmacology, neurology, cardiovascular diseases, endocrinology, and rheumatology). These publications thereby give examples for at least one of the following categories: they provide novel mechanistic insights, use an exceptionally large cohort size, establish a workflow that has the potential to become a high-impact methodology, or are highly cited in their field. We finally have a look into new emerging fields and trends in MSI (immunology, microbiology, infectious diseases, and aging), as applied MSI is continuously broadening as a result of technological breakthroughs.


Assuntos
Pesquisa Biomédica , Diagnóstico por Imagem , Humanos , Espectrometria de Massas/métodos , Diagnóstico por Imagem/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos
3.
J Pathol ; 256(2): 202-213, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34719782

RESUMO

The response to neoadjuvant therapy can vary widely between individual patients. Histopathological tumor regression grading (TRG) is a strong factor for treatment response and survival prognosis of esophageal adenocarcinoma (EAC) patients following neoadjuvant treatment and surgery. However, TRG systems are usually based on the estimation of residual tumor but do not consider stromal or metabolic changes after treatment. Spatial metabolomics analysis is a powerful tool for molecular tissue phenotyping but has not been used so far in the context of neoadjuvant treatment of esophageal cancer. We used imaging mass spectrometry to assess the potential of spatial metabolomics on tumor and stroma tissue for evaluating therapy response of neoadjuvant-treated EAC patients. With an accuracy of 89.7%, the binary classifier trained on spatial tumor metabolite data proved to be superior for stratifying patients when compared with histopathological response assessment, which had an accuracy of 70.5%. Sensitivities and specificities for the poor and favorable survival patient groups ranged from 84.9% to 93.3% using the metabolic classifier and from 62.2% to 78.1% using TRG. The tumor classifier was the only significant prognostic factor (HR 3.38, 95% CI 1.40-8.12, p = 0.007) when adjusted for clinicopathological parameters such as TRG (HR 1.01, 95% CI 0.67-1.53, p = 0.968) or stromal classifier (HR 1.86, 95% CI 0.81-4.25, p = 0.143). The classifier even allowed us to further stratify patients within the TRG1-3 categories. The underlying mechanisms of response to treatment have been figured out through network analysis. In summary, metabolic response evaluation outperformed histopathological response evaluation in our study with regard to prognostic stratification. This finding indicates that the metabolic constitution of the tumor may have a greater impact on patient survival than the quantity of residual tumor cells or the stroma. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Adenocarcinoma/tratamento farmacológico , Biomarcadores Tumorais/metabolismo , Metabolismo Energético , Neoplasias Esofágicas/tratamento farmacológico , Metaboloma , Metabolômica , Terapia Neoadjuvante , Gradação de Tumores , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Quimiorradioterapia Adjuvante , Quimioterapia Adjuvante , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Esofagectomia , Alemanha , Humanos , Aprendizado de Máquina , Terapia Neoadjuvante/efeitos adversos , Terapia Neoadjuvante/mortalidade , Valor Preditivo dos Testes , Reprodutibilidade dos Testes , Estudos Retrospectivos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Suíça , Fatores de Tempo , Resultado do Tratamento
4.
Proteomics ; 22(10): e2100223, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35170848

RESUMO

MALDI MS imaging (MSI) is a powerful analytical tool for spatial peptide detection in heterogeneous tissues. Proper sample preparation is crucial to achieve high quality, reproducible measurements. Here we developed an optimized protocol for spatially resolved proteolytic peptide detection with MALDI time-of-flight MSI of fresh frozen prostate tissue sections. The parameters tested included four different tissue washes, four methods of protein denaturation, four methods of trypsin digestion (different trypsin densities, sprayers, and incubation times), and five matrix deposition methods (different sprayers, settings, and matrix concentrations). Evaluation criteria were the number of detected and excluded peaks, percentage of high mass peaks, signal-to-noise ratio, spatial localization, and average intensities of identified peptides, all of which were integrated into a weighted quality evaluation scoring system. Based on these scores, the optimized protocol included an ice-cold EtOH+H2 O wash, a 5 min heating step at 95°C, tryptic digestion incubated for 17h at 37°C and CHCA matrix deposited at a final amount of 1.8 µg/mm2 . Including a heat-induced protein denaturation step after tissue wash is a new methodological approach that could be useful also for other tissue types. This optimized protocol for spatial peptide detection using MALDI MSI facilitates future biomarker discovery in prostate cancer and may be useful in studies of other tissue types.


Assuntos
Peptídeos , Próstata , Humanos , Masculino , Próstata/metabolismo , Proteínas , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Tripsina/metabolismo
5.
J Proteome Res ; 21(1): 49-66, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34874173

RESUMO

Intestinal ischemia-reperfusion (IR) injury is a severe clinical condition, and unraveling its pathophysiology is crucial to improve therapeutic strategies and reduce the high morbidity and mortality rates. Here, we studied the dynamic proteome and phosphoproteome in the human intestine during ischemia and reperfusion, using liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis to gain quantitative information of thousands of proteins and phosphorylation sites, as well as mass spectrometry imaging (MSI) to obtain spatial information. We identified a significant decrease in abundance of proteins related to intestinal absorption, microvillus, and cell junction, whereas proteins involved in innate immunity, in particular the complement cascade, and extracellular matrix organization increased in abundance after IR. Differentially phosphorylated proteins were involved in RNA splicing events and cytoskeletal and cell junction organization. In addition, our analysis points to mitogen-activated protein kinase (MAPK) and cyclin-dependent kinase (CDK) families to be active kinases during IR. Finally, matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) MSI presented peptide alterations in abundance and distribution, which resulted, in combination with Fourier-transform ion cyclotron resonance (FTICR) MSI and LC-MS/MS, in the annotation of proteins related to RNA splicing, the complement cascade, and extracellular matrix organization. This study expanded our understanding of the molecular changes that occur during IR in the human intestine and highlights the value of the complementary use of different MS-based methodologies.


Assuntos
Proteômica , Espectrometria de Massas em Tandem , Cromatografia Líquida/métodos , Humanos , Proteoma , Proteômica/métodos , Reperfusão , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos
6.
Anal Chem ; 93(3): 1393-1400, 2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33373197

RESUMO

In quantitative mass spectrometry imaging (MSI), the gold standard adds a single structural homologue of the target compound at a known concentration to the sample. This internal standard enables to map the detected intensity of the target molecule against an external calibration curve. This approach, however, ignores local noise levels and disproportional ion suppression effects, which might depend on the concentration of the target compound. To overcome these issues, we propose a novel approach that applies several isotopically labeled versions, each at a different concentration, to the sample. This allows creating individual internal calibration curves for every MSI pixel. As proof of principle, we have quantified an endogenous peptide of histone H4 by matrix-assisted laser desorption/ionization-Q-MSI (MALDI-Q-MSI), using a mixture of three isotopically labeled versions. The usage of a fourth label allowed us to compare the gold standard to our multilabel approach. We observed substantial heterogeneity in ion suppression across the tissue, which disclosed itself as varying slopes in the per-pixel regression analyses. These slopes were histology-dependent and differed from each other by up to a factor of 4. The results were validated by liquid chromatography-mass spectrometry (LC-MS), exhibiting a high agreement between LC-MS and MALDI-Q-MSI (Pearson correlation r = 0.87). A comparison between the multilabel and single-label approaches revealed a higher accuracy for the multilabel method when the local target compound concentration differed too much from the concentration of the single label. In conclusion, we show that the multilabel approach provides superior quantitation compared to a single-label approach, in case the target compound is inhomogeneously distributed at a wide concentration range in the tissue.


Assuntos
Histonas/química , Peptídeos/análise , Animais , Colo/química , Colo/metabolismo , Espectrometria de Massas , Suínos
7.
Proteomics ; 20(23): e1900369, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32767647

RESUMO

Mass spectrometry imaging (MSI) allows investigating the spatial distribution of chemical compounds directly in biological tissues. As the analytical depth of MSI is limited, MSI needs to be coupled to more sensitive local extraction-based omics approaches to achieve a comprehensive molecular characterization. For this, it is important to retain the spatial information provided by MSI for follow-up omics studies. It has been shown that regiospecific MSI data can be used to guide a laser microdissection system for ultra-sensitive liquid chromatography-mass spectrometry (LC-MS) analyses. So far, this combination has required separate and specialized mass spectrometry (MS) instrumentation. Recent advances in dual-source instrumentation, harboring both matrix assisted laser/desorption ionization (MALDI) and electrospray ionization (ESI) sources, promise state-of-the-art MSI and liquid-based proteomic capabilities on the same MS instrument. This study demonstrates that such an instrument can offer both fast lipid-based MSI at high mass and high lateral resolution and sensitive LC-MS on local protein extracts from the exact same tissue section.


Assuntos
Lipídeos , Proteômica , Cromatografia Líquida , Microdissecção e Captura a Laser , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
8.
Anal Chem ; 92(4): 3171-3179, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-31944670

RESUMO

Levels of zinc, along with its mechanistically related metabolites citrate and aspartate, are widely reported as reduced in prostate cancer compared to healthy tissue and are therefore pointed out as potential cancer biomarkers. Previously, it has only been possible to analyze zinc and metabolites by separate detection methods. Through matrix-assisted laser desorption/ionization mass spectrometry imaging (MSI), we were for the first time able to demonstrate, in two different sample sets (n = 45 and n = 4), the simultaneous spatial detection of zinc, in the form of ZnCl3-, together with citrate, aspartate, and N-acetylaspartate on human prostate cancer tissues. The reliability of the ZnCl3- detection was validated by total zinc determination using laser ablation inductively coupled plasma MSI on adjacent serial tissue sections. Zinc, citrate, and aspartate were correlated with each other (range r = 0.46 to 0.74) and showed a significant reduction in cancer compared to non-cancer epithelium (p < 0.05, log2 fold change range: -0.423 to -0.987), while no significant difference between cancer and stroma tissue was found. Simultaneous spatial detection of zinc and its metabolites is not only a valuable tool for analyzing the role of zinc in prostate metabolism but might also provide a fast and simple method to detect zinc, citrate, and aspartate levels as a biomarker signature for prostate cancer diagnostics and prognostics.


Assuntos
Próstata/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Zinco/metabolismo , Ácido Aspártico/metabolismo , Citratos/metabolismo , Humanos , Masculino , Próstata/citologia , Próstata/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Fatores de Tempo
9.
Clin Chem Lab Med ; 58(6): 914-929, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31665113

RESUMO

Mass spectrometry (MS) is the workhorse of metabolomics, proteomics and lipidomics. Mass spectrometry imaging (MSI), its extension to spatially resolved analysis of tissues, is a powerful tool for visualizing molecular information within the histological context of tissue. This review summarizes recent developments in MSI and highlights current challenges that remain to achieve molecular imaging at the cellular level of clinical specimens. We focus on matrix-assisted laser desorption/ionization (MALDI)-MSI. We discuss the current status of each of the analysis steps and remaining challenges to reach the desired level of cellular imaging. Currently, analyte delocalization and degradation, matrix crystal size, laser focus restrictions and detector sensitivity are factors that are limiting spatial resolution. New sample preparation devices and laser optic systems are being developed to push the boundaries of these limitations. Furthermore, we review the processing of cellular MSI data and images, and the systematic integration of these data in the light of available algorithms and databases. We discuss roadblocks in the data analysis pipeline and show how technology from other fields can be used to overcome these. Finally, we conclude with curative and community efforts that are needed to enable contextualization of the information obtained.


Assuntos
Análise de Célula Única/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Animais , Humanos
10.
Angew Chem Int Ed Engl ; 59(40): 17447-17450, 2020 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-32668069

RESUMO

The large-scale and label-free molecular characterization of single cells in their natural tissue habitat remains a major challenge in molecular biology. We present a method that integrates morphometric image analysis to delineate and classify individual cells with their single-cell-specific molecular profiles. This approach provides a new means to study spatial biological processes such as cancer field effects and the relationship between morphometric and molecular features.


Assuntos
Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Animais , Colo/citologia , Colo/patologia , Modelos Animais de Doenças , Lipídeos/química , Análise de Célula Única , Neoplasias Gástricas/patologia , Suínos
11.
Anal Bioanal Chem ; 411(22): 5647-5653, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31263919

RESUMO

Mass spectrometry imaging (MSI) is an analytical technique for the unlabeled and multiplex imaging of molecules in biological tissue sections. It therefore enables the spatial and molecular annotations of tissues complementary to histology. It has already been shown that MSI can guide subsequent material isolation technologies such as laser microdissection (LMD) to enable a more in-depth molecular characterization of MSI-highlighted tissue regions. However, with MSI now reaching spatial resolutions at the single-cell scale, there is a need for a precise co-registration between MSI and the LMD. As proof-of-principle, MSI of lipids was performed on a breast cancer tissue followed by a segmentation of the data to detect molecularly distinct segments within its tumor areas. After image processing of the segmentation results, the coordinates of the MSI-detected segments were passed to the LMD system by three co-registration steps. The errors of each co-registration step were quantified and the total error was found to be less than 13 µm. With this link established, MSI data can now accurately guide LMD to excise MSI-defined regions of interest for subsequent extract-based analyses. In our example, the excised tissue material was then subjected to ultrasensitive microproteomics in order to determine predominant molecular mechanisms in each of the MSI-highlighted intratumor segments. This work shows how the strengths of MSI, histology, and extract-based omics can be combined to enable a more comprehensive molecular characterization of in situ biological processes.


Assuntos
Neoplasias da Mama/metabolismo , Espectrometria de Massas/métodos , Proteínas de Neoplasias/metabolismo , Proteômica , Neoplasias da Mama/patologia , Feminino , Humanos , Lasers , Espectrometria de Massas/normas
12.
Proc Natl Acad Sci U S A ; 113(43): 12244-12249, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27791011

RESUMO

The identification of tumor subpopulations that adversely affect patient outcomes is essential for a more targeted investigation into how tumors develop detrimental phenotypes, as well as for personalized therapy. Mass spectrometry imaging has demonstrated the ability to uncover molecular intratumor heterogeneity. The challenge has been to conduct an objective analysis of the resulting data to identify those tumor subpopulations that affect patient outcome. Here we introduce spatially mapped t-distributed stochastic neighbor embedding (t-SNE), a nonlinear visualization of the data that is able to better resolve the biomolecular intratumor heterogeneity. In an unbiased manner, t-SNE can uncover tumor subpopulations that are statistically linked to patient survival in gastric cancer and metastasis status in primary tumors of breast cancer.


Assuntos
Neoplasias da Mama/patologia , Variação Genética , Prognóstico , Neoplasias Gástricas/patologia , Idoso , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Linhagem da Célula/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Medicina de Precisão , Neoplasias Gástricas/genética , Análise de Sobrevida
13.
Anal Bioanal Chem ; 410(23): 5969-5980, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29968108

RESUMO

Mass spectrometry imaging (MSI) has provided many results with translational character, which still have to be proven robust in large patient cohorts and across different centers. Although formalin-fixed paraffin-embedded (FFPE) specimens are most common in clinical practice, no MSI multicenter study has been reported for FFPE samples. Here, we report the results of the first round robin MSI study on FFPE tissues with the goal to investigate the consequences of inter- and intracenter technical variation on masking biological effects. A total of four centers were involved with similar MSI instrumentation and sample preparation equipment. A FFPE multi-organ tissue microarray containing eight different types of tissue was analyzed on a peptide and metabolite level, which enabled investigating different molecular and biological differences. Statistical analyses revealed that peptide intercenter variation was significantly lower and metabolite intercenter variation was significantly higher than the respective intracenter variations. When looking at relative univariate effects of mass signals with statistical discriminatory power, the metabolite data was more reproducible across centers compared to the peptide data. With respect to absolute effects (cross-center common intensity scale), multivariate classifiers were able to reach on average > 90% accuracy for peptides and > 80% for metabolites if trained with sufficient amount of cross-center data. Overall, our study showed that MSI data from FFPE samples could be reproduced to a high degree across centers. While metabolite data exhibited more reproducibility with respect to relative effects, peptide data-based classifiers were more directly transferable between centers and therefore more robust than expected. Graphical abstract ᅟ.


Assuntos
Espectrometria de Massas , Metabolômica , Inclusão em Parafina , Peptídeos/análise , Análise Serial de Tecidos , Fixação de Tecidos , Animais , Formaldeído/química , Espectrometria de Massas/métodos , Metabolômica/métodos , Camundongos , Inclusão em Parafina/métodos , Proteômica/métodos , Reprodutibilidade dos Testes , Análise Serial de Tecidos/métodos , Fixação de Tecidos/métodos
14.
Biochim Biophys Acta Proteins Proteom ; 1865(7): 957-966, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27725306

RESUMO

Mass spectrometry imaging (MSI) has been widely used for the direct molecular assessment of tissue samples and has demonstrated great potential to complement current histopathological methods in cancer research. It is now well established that tissue preparation is key to a successful MSI experiment; for histologically heterogeneous tumor tissues, other parts of the workflow are equally important to the experiment's success. To demonstrate these facets here we describe a matrix-assisted laser desorption/ionization MSI biomarker discovery investigation of high-grade, complex karyotype sarcomas, which often have histological overlap and moderate response to chemo-/radio-therapy. Multiple aspects of the workflow had to be optimized, ranging from the tissue preparation and data acquisition protocols, to the post-MSI histological staining method, data quality control, histology-defined data selection, data processing and statistical analysis. Only as a result of developing every step of the biomarker discovery workflow was it possible to identify a panel of protein signatures that could distinguish between different subtypes of sarcomas or could predict patient survival outcome. This article is part of a Special Issue entitled: MALDI Imaging, edited by Dr. Corinna Henkel and Prof. Peter Hoffmann.


Assuntos
Neoplasias/diagnóstico , Neoplasias/patologia , Biomarcadores/metabolismo , Humanos , Neoplasias/metabolismo , Sarcoma/diagnóstico , Sarcoma/metabolismo , Sarcoma/patologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos
15.
Analyst ; 142(15): 2690-2712, 2017 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-28642940

RESUMO

Mass spectrometry is being used in many clinical research areas ranging from toxicology to personalized medicine. Of all the mass spectrometry techniques, mass spectrometry imaging (MSI), in particular, has continuously grown towards clinical acceptance. Significant technological and methodological improvements have contributed to enhance the performance of MSI recently, pushing the limits of throughput, spatial resolution, and sensitivity. This has stimulated the spread of MSI usage across various biomedical research areas such as oncology, neurological disorders, cardiology, and rheumatology, just to name a few. After highlighting the latest major developments and applications touching all aspects of translational research (i.e. from early pre-clinical to clinical research), we will discuss the present challenges in translational research performed with MSI: data management and analysis, molecular coverage and identification capabilities, and finally, reproducibility across multiple research centers, which is the largest remaining obstacle in moving MSI towards clinical routine.


Assuntos
Pesquisa Biomédica , Diagnóstico por Imagem , Espectrometria de Massas , Humanos , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
16.
Angew Chem Int Ed Engl ; 56(25): 7146-7150, 2017 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-28493648

RESUMO

Mass spectrometry imaging (MSI) simultaneously detects and identifies the spatial distribution of numerous molecules throughout tissues. Currently, MSI is limited to providing a static and ex vivo snapshot of highly dynamic systems in which molecules are constantly synthesized and consumed. Herein, we demonstrate an innovative MSI methodology to study dynamic molecular changes of amino acids within biological tissues by measuring the dilution and conversion of stable isotopes in a mouse model. We evaluate the method specifically on hepatocellular metabolism of the essential amino acid l-phenylalanine, associated with liver diseases. Crucially, the method reveals the localized dynamics of l-phenylalanine metabolism, including its in vivo hydroxylation to l-tyrosine and co-localization with other liver metabolites in a time course of samples from different animals. This method thus enables the dynamics of localized biochemical synthesis to be studied directly from biological tissues.


Assuntos
Isótopos de Carbono/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Espectrometria de Massas/métodos , Fenilalanina/metabolismo , Tirosina/metabolismo , Animais , Modelos Animais de Doenças , Cromatografia Gasosa-Espectrometria de Massas/métodos , Xenoenxertos , Hidroxilação , Cinética , Camundongos , Camundongos Nus , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Espectroscopia de Infravermelho com Transformada de Fourier , Espectrometria de Massas em Tandem/métodos
17.
Proteomics ; 16(11-12): 1802-13, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27174013

RESUMO

The combination of high heterogeneity, both intratumoral and intertumoral, with their rarity has made diagnosis, prognosis of high-grade sarcomas difficult. There is an urgent need for more objective molecular biomarkers, to differentiate between the many different subtypes, and to also provide new treatment targets. Mass spectrometry imaging (MSI) has amply demonstrated its ability to identify potential new markers for patient diagnosis, survival, metastasis and response to therapy in cancer research. In this study, we investigated the ability of MALDI-MSI of proteins to distinguish between high-grade osteosarcoma (OS), leiomyosarcoma (LMS), myxofibrosarcoma (MFS) and undifferentiated pleomorphic sarcoma (UPS) (Ntotal = 53). We also investigated if there are individual proteins or protein signatures that are statistically associated with patient survival. Twenty diagnostic protein signals were found characteristic for specific tumors (p ≤ 0.05), amongst them acyl-CoA-binding protein (m/z 11 162), macrophage migration inhibitory factor (m/z 12 350), thioredoxin (m/z 11 608) and galectin-1 (m/z 14 633) were assigned. Another nine protein signals were found to be associated with overall survival (p ≤ 0.05), including proteasome activator complex subunit 1 (m/z 9753), indicative for non-OS patients with poor survival; and two histone H4 variants (m/z 11 314 and 11 355), indicative of poor survival for LMS patients.


Assuntos
Biomarcadores Tumorais/genética , Fibrossarcoma/diagnóstico por imagem , Leiomiossarcoma/diagnóstico por imagem , Osteossarcoma/diagnóstico por imagem , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Adulto , Idoso , Intervalo Livre de Doença , Feminino , Fibrossarcoma/diagnóstico , Fibrossarcoma/genética , Fibrossarcoma/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Leiomiossarcoma/diagnóstico , Leiomiossarcoma/genética , Leiomiossarcoma/patologia , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Osteossarcoma/diagnóstico , Osteossarcoma/genética , Osteossarcoma/patologia , Prognóstico , Proteômica/métodos
18.
Anal Chem ; 88(10): 5281-9, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-27065343

RESUMO

In research and clinical settings, formalin-fixed and paraffin-embedded (FFPE) tissue specimens are collected routinely and therefore this material constitutes a highly valuable source to gather insight in metabolic changes of diseases. Among mass spectrometry techniques to examine the molecular content of FFPE tissue, mass spectrometry imaging (MSI) is the most appropriate when morphological and histological features are to be related to metabolic information. Currently, high-resolution mass spectrometers are widely used for metabolomics studies. However, with regards to matrix-assisted laser desorption/ionization (MALDI) MSI, no study has so far addressed the necessity of instrumental mass resolving power in terms of clinical diagnosis and prognosis using archived FFPE tissue. For this matter we performed for the first time a comprehensive comparison between a high mass resolution Fourier-transform ion cyclotron resonance (FTICR) mass spectrometer and a time-of-flight (TOF) instrument with lower mass resolving power. Spectra analysis revealed that about one-third of the detected peaks remained unresolved by MALDI-TOF, which led to a 3-5 times lower number of m/z features compared to FTICR measurements. Overlaid peak information and background noise in TOF images made a precise assignment of molecular attributes to morphological features more difficult and limited classification approaches. This clearly demonstrates the need for high-mass resolution capabilities for metabolite imaging. Nevertheless, MALDI-TOF allowed reproducing and verifying individual markers identified previously by MALDI-FTICR MSI. The systematic comparison gives rise to a synergistic combination of the different MSI platforms for high-throughput discovery and validation of biomarkers.


Assuntos
Neoplasias do Colo/patologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Biomarcadores/análise , Neoplasias do Colo/mortalidade , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Formaldeído/química , Análise de Fourier , Humanos , Processamento de Imagem Assistida por Computador , Metabolômica , Inclusão em Parafina , Taxa de Sobrevida
19.
Anal Chem ; 88(11): 5871-8, 2016 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-27180608

RESUMO

Mass spectrometry imaging (MSI) is a powerful molecular imaging technique. In microprobe MSI, images are created through a grid-wise interrogation of individual spots by mass spectrometry across a surface. Classical statistical tests for within-sample comparisons fail as close-by measurement spots violate the assumption of independence of these tests, which can lead to an increased false-discovery rate. For spatial data, this effect is referred to as spatial autocorrelation. In this study, we investigated spatial autocorrelation in three different matrix-assisted laser desorption/ionization MSI data sets. These data sets cover different molecular classes (metabolites/drugs, lipids, and proteins) and different spatial resolutions ranging from 20 to 100 µm. Significant spatial autocorrelation was detected in all three data sets and found to increase with decreasing pixel size. To enable statistical testing for differences in mass signal intensities between regions of interest within MSI data sets, we propose the use of Conditional Autoregressive (CAR) models. We show that, by accounting for spatial autocorrelation, discovery rates (i.e., the ratio between the features identified and the total number of features) could be reduced between 21% and 69%. The reliability of this approach was validated by control mass signals based on prior knowledge. In light of the advent of larger MSI data sets based on either an increased spatial resolution or 3D data sets, accounting for effects due to spatial autocorrelation becomes even more indispensable. Here, we propose a generic and easily applicable workflow to enable within-sample statistical comparisons.

20.
Analyst ; 141(12): 3832-41, 2016 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-27041214

RESUMO

The high ion signals produced by many lipids in mass spectrometry imaging (MSI) make them an ideal molecular class to study compositional changes throughout tissue sections and their relationship with disease. However, the large extent of structural diversity observed in the lipidome means optimal ion signal for different lipid classes is often obtained in opposite polarities. In this work we demonstrate how new high speed MALDI-MSI technologies combined with precise laser position control enables the acquisition of positive and negative ion mode lipid data from the same tissue section much faster than is possible with other MSI instruments. Critically, using this approach we explicitly demonstrate how such dual polarity acquisitions provide more information regarding molecular composition and spatial distributions throughout biological tissues. For example, in applying this approach to the zebra finch songbird brain we reveal the high abundance of DHA containing phospholipids (PC in positive mode and PE, PS in negative ion mode) in the nuclei that control song learning behaviour. To make the most of dual polarity data from single tissues we have also developed a pLSA-based multivariate analysis technique that includes both positive and negative ion data in the classification approach. In doing so the correlation amongst different lipid classes that ionise best in opposite polarities and contribute to certain spatial patterns within the tissue can be directly revealed. To demonstrate we apply this approach to studying the lipidomic changes throughout the tumor microenvironment within xenografts from a lung cancer model.


Assuntos
Química Encefálica , Encéfalo/fisiologia , Tentilhões/fisiologia , Neoplasias Experimentais/química , Fosfolipídeos/análise , Animais , Humanos , Lasers , Lipídeos , Camundongos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA