Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 356
Filtrar
1.
PLoS One ; 19(10): e0311592, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39388479

RESUMO

The prevalence of hepatopancreatic diseases in cultured shrimp has increased in recent years. Decapod Hepanhamaparvovirus 1 (DHPV) infection was identified by histology in samples that could not be detected by PCR-based assay for this virus. Employing Laser Microdissection (LMD), we dissected cells containing intranuclear inclusion bodies pathognomonic for DHPV infection from histological sections. Whole Genome Amplification and NGS were used to generate five complete genomes of the novel DHPV isolate that showed identities ranging from 77% to 98% to previously reported isolates. Phylogenetic analyses revealed the DHPV isolate represents a novel genotype, Genotype V. We developed PCR and in situ hybridization methods tailored for the specific detection of this genotype. Our approach of combining LMD with NGS opens avenues for rapid identification of emerging viral pathogens and retrospective studies to understand origin and evolution of viruses showcasing the transformative potential of the innovative approach used in this study.


Assuntos
Genoma Viral , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Filogenia , Animais , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Penaeidae/virologia , Microdissecção/métodos , Microdissecção e Captura a Laser/métodos
2.
Anal Chem ; 96(32): 12973-12982, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39089681

RESUMO

There is increasing interest in developing in-depth proteomic approaches for mapping tissue heterogeneity in a cell-type-specific manner to better understand and predict the function of complex biological systems such as human organs. Existing spatially resolved proteomics technologies cannot provide deep proteome coverage due to limited sensitivity and poor sample recovery. Herein, we seamlessly combined laser capture microdissection with a low-volume sample processing technology that includes a microfluidic device named microPOTS (microdroplet processing in one pot for trace samples), multiplexed isobaric labeling, and a nanoflow peptide fractionation approach. The integrated workflow allowed us to maximize proteome coverage of laser-isolated tissue samples containing nanogram levels of proteins. We demonstrated that the deep spatial proteomics platform can quantify more than 5000 unique proteins from a small-sized human pancreatic tissue pixel (∼60,000 µm2) and differentiate unique protein abundance patterns in pancreas. Furthermore, the use of the microPOTS chip eliminated the requirement for advanced microfabrication capabilities and specialized nanoliter liquid handling equipment, making it more accessible to proteomic laboratories.


Assuntos
Peptídeos , Proteoma , Proteômica , Humanos , Proteoma/análise , Proteômica/métodos , Peptídeos/análise , Peptídeos/química , Pâncreas/metabolismo , Pâncreas/química , Nanotecnologia , Técnicas Analíticas Microfluídicas/instrumentação , Microdissecção e Captura a Laser/métodos
3.
PLoS One ; 19(8): e0308662, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39186704

RESUMO

Laser microdissection technology is favored by biomedical researchers for its ability to rapidly and accurately isolate target cells and tissues. However, the precision cutting capabilities of existing laser microdissection systems are hindered by limitations in overall mechanical movement accuracy, resulting in suboptimal cutting quality. Additionally, the use of current laser microdissection systems for target acquisition may lead to tissue burns and reduced acquisition rates due to inherent flaws in the capture methods. To address these challenges and achieve precise and efficient separation and capture of cellular tissues, we integrated a digital micromirror device (DMD) into the existing system optics to modulate spatial light. This allows the system to not only implement the traditional point scanning cutting method but also utilize the projection cutting method.We have successfully cut various patterns on commonly used laser microdissection materials such as PET films and mouse tissues. Under projection cutting mode, we were able to achieve precise cutting of special shapes with a diameter of 7.5 micrometers in a single pass, which improved cutting precision and efficiency. Furthermore, we employed a negative pressure adsorption method to efficiently collect target substances. This approach not only resulted in a single-pass capture rate exceeding 90% for targets of different sizes but also enabled simultaneous capture of multiple targets, overcoming the limitations of traditional single-target capture and enhancing target capture efficiency, and avoiding potential tissue damage from lasers.In summary, the integration of the digital micromirror device into laser microdissection systems significantly enhances cutting precision and efficiency, overcoming limitations of traditional systems. This advancement demonstrates the accuracy and effectiveness of laser microdissection systems in isolating and capturing biological tissues, highlighting their potential in medical applications.


Assuntos
Microdissecção e Captura a Laser , Animais , Camundongos , Microdissecção e Captura a Laser/métodos , Microdissecção e Captura a Laser/instrumentação , Lasers , Luz , Pressão , Adsorção
4.
Methods Mol Biol ; 2823: 55-75, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39052214

RESUMO

Combining proteogenomics with laser capture microdissection (LCM) in cancer research offers a targeted way to explore the intricate interactions between tumor cells and the different microenvironment components. This is especially important for immuno-oncology (IO) research where improvements in the predictability of IO-based drugs are sorely needed, and depends on a better understanding of the spatial relationships involving the tumor, blood supply, and immune cell interactions, in the context of their associated microenvironments. LCM is used to isolate and obtain distinct histological cell types, which may be routinely performed on complex and heterogeneous solid tumor specimens. Once cells have been captured, nucleic acids and proteins may be extracted for in-depth multimodality molecular profiling assays. Optimizing the minute tissue quantities from LCM captured cells is challenging. Following the isolation of nucleic acids, RNA-seq may be performed for gene expression and DNA sequencing performed for the discovery and analysis of actionable mutations, copy number variation, methylation profiles, etc. However, there remains a need for highly sensitive proteomic methods targeting small-sized samples. A significant part of this protocol is an enhanced liquid chromatography mass spectrometry (LC-MS) analysis of micro-scale and/or nano-scale tissue sections. This is achieved with a silver-stained one-dimensional sodium dodecyl sulfate polyacrylamide gel electrophoresis (1D-SDS-PAGE) approach developed for LC-MS analysis of fresh-frozen tissue specimens obtained via LCM. Included is a detailed in-gel digestion method adjusted and specifically designed to maximize the proteome coverage from amount-limited LCM samples to better facilitate in-depth molecular profiling. Described is a proteogenomic approach leveraged from microdissected fresh frozen tissue. The protocols may also be applicable to other types of specimens having limited nucleic acids, protein quantity, and/or sample volume.


Assuntos
Microdissecção e Captura a Laser , Proteogenômica , Proteogenômica/métodos , Humanos , Microdissecção e Captura a Laser/métodos , Cromatografia Líquida/métodos , Neoplasias/patologia , Neoplasias/genética , Descoberta de Drogas/métodos , Espectrometria de Massas/métodos , Proteômica/métodos , Microambiente Tumoral , Microdissecção/métodos
5.
Methods Mol Biol ; 2823: 193-223, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39052222

RESUMO

Archived tumor specimens are routinely preserved by formalin fixation and paraffin embedding. Despite the conventional wisdom that proteomics might be ineffective due to the cross-linking and pre-analytical variables, these samples have utility for both discovery and targeted proteomics. Building on this capability, proteomics approaches can be used to maximize our understanding of cancer biology and clinical relevance by studying preserved tumor tissues annotated with the patients' medical histories. Proteomics of formalin-fixed paraffin-embedded (FFPE) tissues also integrates with histological evaluation and molecular pathology strategies, so that additional collection of research biopsies or resected tumor aliquots is not needed. The acquisition of data from the same tumor sample also overcomes concerns about biological variation between samples due to intratumoral heterogeneity. However, the protein extraction and proteomics sample preparation from FFPE samples can be onerous, particularly for small (i.e., limited or precious) samples. Therefore, we provide a protocol for a recently introduced kit-based EasyPep method with benchmarking against a modified version of the well-established filter-aided sample preparation strategy using laser-capture microdissected lung adenocarcinoma tissues from a genetically engineered mouse model. This model system allows control over the tumor preparation and pre-analytical variables while also supporting the development of methods for spatial proteomics to examine intratumoral heterogeneity. Data are posted in ProteomeXchange (PXD045879).


Assuntos
Formaldeído , Inclusão em Parafina , Proteômica , Fixação de Tecidos , Proteômica/métodos , Inclusão em Parafina/métodos , Fixação de Tecidos/métodos , Formaldeído/química , Animais , Camundongos , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Microdissecção e Captura a Laser/métodos , Neoplasias/patologia , Neoplasias/metabolismo , Neoplasias/genética , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma de Pulmão/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-38996754

RESUMO

Diabetic nephropathy (DN) remains the primary cause of end-stage renal disease (ESRD), warranting equal attention and separate analysis of glomerular, tubular, and interstitial lesions in its diagnosis and intervention. This study aims to identify the specific proteomics characteristics of DN, and assess changes in the biological processes associated with DN. 5 patients with DN and 5 healthy kidney transplant donor control individuals were selected for analysis. The proteomic characteristics of glomeruli, renal tubules, and renal interstitial tissue obtained through laser capture microscopy (LCM) were studied using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Significantly, the expression of multiple heat shock proteins (HSPs), tubulins, and heterogeneous nuclear ribonucleoproteins (hnRNPs) in glomeruli and tubules was significantly reduced. Differentially expressed proteins (DEPs) in the glomerulus showed significant enrichment in pathways related to cell junctions and cell movement, including the regulation of actin cytoskeleton and tight junction. DEPs in renal tubules were significantly enriched in glucose metabolism-related pathways, such as glucose metabolism, glycolysis/gluconeogenesis, and the citric acid cycle. Moreover, the glycolysis/gluconeogenesis pathway was a co-enrichment pathway in both DN glomeruli and tubules. Notably, ACTB emerged as the most crucial protein in the protein-protein interaction (PPI) analysis of DEPs in both glomeruli and renal tubules. In this study, we delve into the unique proteomic characteristics of each sub-region of renal tissue. This enhances our understanding of the potential pathophysiological changes in DN, particularly the potential involvement of glycolysis metabolic disorder, glomerular cytoskeleton and cell junctions. These insights are crucial for further research into the identification of disease biomarkers and the pathogenesis of DN.


Assuntos
Nefropatias Diabéticas , Rim , Microdissecção e Captura a Laser , Proteoma , Proteômica , Espectrometria de Massas em Tandem , Humanos , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Proteômica/métodos , Microdissecção e Captura a Laser/métodos , Masculino , Pessoa de Meia-Idade , Feminino , Espectrometria de Massas em Tandem/métodos , Rim/química , Rim/metabolismo , Rim/patologia , Proteoma/análise , Proteoma/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Adulto , Idoso
7.
Exp Eye Res ; 246: 109989, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38969282

RESUMO

RNA sequencing (RNA-seq) coupled with laser capture microdissection (LCM) is a powerful tool for transcriptomic analysis in unfixed fresh-frozen tissues. Fixation of ocular tissues for immunohistochemistry commonly involves the use of paraformaldehyde (PFA) followed by embedding in Optimal Cutting Temperature (OCT) medium for long-term cryopreservation. However, the quality of RNA derived from such archival PFA-fixed/OCT-embedded samples is often compromised, limiting its suitability for transcriptomic studies. In this study, we aimed to develop a methodology to extract high-quality RNA from PFA-fixed canine eyes by utilizing LCM to isolate retinal tissue. We demonstrate the efficacy of an optimized LCM and RNA purification protocol for transcriptomic profiling of PFA-fixed retinal specimens. We compared four pairs of canine retinal tissues, where one eye was subjected to PFA-fixation prior to OCT embedding, while the contralateral eye was embedded fresh frozen (FF) in OCT without fixation. Since the RNA obtained from PFA-fixed retinas were contaminated with genomic DNA, we employed two rounds of DNase I treatment to obtain RNA suitable for RNA-seq. Notably, the quality of sequencing reads and gene sets identified from both PFA-fixed and FF tissues were nearly identical. In summary, our study introduces an optimized workflow for transcriptomic profiling from PFA-fixed archival retina. This refined methodology paves the way for improved transcriptomic analysis of preserved ocular tissue, bridging the gap between optimal sample preservation and high-quality RNA data acquisition.


Assuntos
Fixadores , Formaldeído , Perfilação da Expressão Gênica , Microdissecção e Captura a Laser , Retina , Fixação de Tecidos , Transcriptoma , Animais , Retina/metabolismo , Perfilação da Expressão Gênica/métodos , Microdissecção e Captura a Laser/métodos , Fixação de Tecidos/métodos , Cães , Fluxo de Trabalho , Criopreservação , RNA/genética , Polímeros
8.
STAR Protoc ; 5(2): 103050, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38703368

RESUMO

Single-cell RNA sequencing (scRNA-seq) combined with laser capture microdissection (LCM) offers a versatile framework for comprehensive transcriptomics from tissue sections. Here, we present a detailed protocol for DRaqL (direct RNA recovery and quenching for LCM) in combination with Smart-seq2 (DRaqL-Smart-seq2), which enables high-quality RNA sequencing for single cells obtained from alcohol-fixed murine ovarian sections. Additionally, we provide an optional procedure for scRNA-seq from formalin-fixed sections (DRaqL-Protease-Smart-seq2). We outline key steps for cell lysis, cDNA amplification, and sequencing library preparation. For complete details on the use and execution of this protocol, please refer to Ikeda et al.1.


Assuntos
Microdissecção e Captura a Laser , Análise de Célula Única , Análise de Célula Única/métodos , Animais , Camundongos , Feminino , Microdissecção e Captura a Laser/métodos , RNA-Seq/métodos , Análise de Sequência de RNA/métodos , Fixação de Tecidos/métodos , Ovário/citologia , Ovário/metabolismo , RNA/genética , RNA/análise , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Biblioteca Gênica , Análise da Expressão Gênica de Célula Única
9.
J Clin Pathol ; 77(6): 430-434, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38429092

RESUMO

We demonstrate a method for tissue microdissection using scanning laser ablation that is approximately two orders of magnitude faster than conventional laser capture microdissection. Our novel approach uses scanning laser optics and a slide coating under the tissue that can be excited by the laser to selectively eject regions of tissue for further processing. Tissue was dissected at 0.117 s/mm2 without reduction in yield, sequencing insert size or base quality compared with undissected tissue. From eight cases, 58-416 mm2 of tissue was obtained from one to four slides in 7-48 seconds total dissection time per case. These samples underwent exome sequencing and we found the variant allelic fraction increased in regions enriched for tumour as expected. This suggests that our ablation technique may be useful as a tool in both clinical and research labs.


Assuntos
Microdissecção e Captura a Laser , Humanos , Microdissecção e Captura a Laser/métodos , Terapia a Laser/métodos , Microdissecção/métodos , Sequenciamento do Exoma , Fatores de Tempo
10.
Mol Cell Proteomics ; 23(5): 100750, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38513891

RESUMO

Spatial tissue proteomics integrating whole-slide imaging, laser microdissection, and ultrasensitive mass spectrometry is a powerful approach to link cellular phenotypes to functional proteome states in (patho)physiology. To be applicable to large patient cohorts and low sample input amounts, including single-cell applications, loss-minimized and streamlined end-to-end workflows are key. We here introduce an automated sample preparation protocol for laser microdissected samples utilizing the cellenONE robotic system, which has the capacity to process 192 samples in 3 h. Following laser microdissection collection directly into the proteoCHIP LF 48 or EVO 96 chip, our optimized protocol facilitates lysis, formalin de-crosslinking, and tryptic digest of low-input archival tissue samples. The seamless integration with the Evosep ONE LC system by centrifugation allows 'on-the-fly' sample clean-up, particularly pertinent for laser microdissection workflows. We validate our method in human tonsil archival tissue, where we profile proteomes of spatially-defined B-cell, T-cell, and epithelial microregions of 4000 µm2 to a depth of ∼2000 proteins and with high cell type specificity. We finally provide detailed equipment templates and experimental guidelines for broad accessibility.


Assuntos
Microdissecção e Captura a Laser , Proteômica , Fluxo de Trabalho , Humanos , Proteômica/métodos , Microdissecção e Captura a Laser/métodos , Tonsila Palatina/citologia , Tonsila Palatina/metabolismo , Automação , Proteoma , Linfócitos B/metabolismo , Linfócitos B/citologia , Espectrometria de Massas/métodos , Linfócitos T/metabolismo , Linfócitos T/citologia
11.
PLoS One ; 19(3): e0297739, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38457477

RESUMO

In recent years, the importance of isolating single cells from blood circulation for several applications, such as non-invasive tumour diagnosis, the monitoring of minimal residual disease, and the analysis of circulating fetal cells for prenatal diagnosis, urged the need to set up innovative methods. For such applications, different methods were developed. All show some weaknesses, especially a limited sensitivity, and specificity. Here we present a new method for isolating a single or a limited number of cells adhered to SBS slides (Tethis S.p.a.) (a glass slide coated with Nanostructured Titanium Dioxide) by Laser Capture Microdissection (LCM) and subsequent Whole Genome Amplification. SBS slides have been shown to have an optimal performance in immobilizing circulating tumour cells (CTCs) from early breast cancer patients. In this work, we spiked cancer cells in blood samples to mimic CTCs. By defining laser parameters to cut intact samples, we were able to isolate genetically intact single cells. We demonstrate that SBS slides are optimally suited for isolating cells using LCM and that this method provides high-quality DNA, ideal for gene-specific assays such as PCR and Sanger sequencing for mutation analysis.


Assuntos
Células Neoplásicas Circulantes , Gravidez , Feminino , Humanos , Microdissecção e Captura a Laser/métodos , Células Neoplásicas Circulantes/patologia , DNA
12.
J Am Soc Nephrol ; 35(1): 117-128, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37749770

RESUMO

Laser capture microdissection and mass spectrometry (LCM/MS) is a technique that involves dissection of glomeruli from paraffin-embedded biopsy tissue, followed by digestion of the dissected glomerular proteins by trypsin, and subsequently mass spectrometry to identify and semiquantitate the glomerular proteins. LCM/MS has played a crucial role in the identification of novel types of amyloidosis, biomarker discovery in fibrillary GN, and more recently discovery of novel target antigens in membranous nephropathy (MN). In addition, LCM/MS has also confirmed the role for complement proteins in glomerular diseases, including C3 glomerulopathy. LCM/MS is now widely used as a clinical test and considered the gold standard for diagnosis and typing amyloidosis. For the remaining glomerular diseases, LCM/MS has remained a research tool. In this review, we discuss the usefulness of LCM/MS in other glomerular diseases, particularly MN, deposition diseases, and diseases of complement pathways, and advocate more routine use of LCM/MS at the present time in at least certain diseases, such as MN, for target antigen detection. We also discuss the limitations of LCM/MS, particularly the difficulties faced from moving from a research-based technique to a clinical test. Nonetheless, the role of LCM/MS in glomerular diseases is expanding. Currently, LCM/MS may be used to identify the etiology in certain glomerular diseases, but in the future, LCM/MS can play a valuable role in determining pathways of complement activation, inflammation, and fibrosis.


Assuntos
Amiloidose , Glomerulonefrite Membranosa , Nefropatias , Humanos , Nefropatias/patologia , Glomérulos Renais/patologia , Espectrometria de Massas , Microdissecção e Captura a Laser/métodos , Glomerulonefrite Membranosa/metabolismo
13.
Sci Rep ; 13(1): 18678, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37907539

RESUMO

The combination of MALDI mass spectrometry imaging, laser-capture microdissection, and quantitative proteomics allows the identification and characterization of molecularly distinct tissue compartments. Such workflows are typically performed using consecutive tissue sections, and so reliable sectioning and mounting of high-quality tissue sections is a prerequisite of such investigations. Embedding media facilitate the sectioning process but can introduce contaminants which may adversely affect either the mass spectrometry imaging or proteomics analyses. Seven low-temperature embedding media were tested in terms of embedding temperature and cutting performance. The two media that provided the best results (5% gelatin and 2% low-melting point agarose) were compared with non-embedded tissue by both MALDI mass spectrometry imaging of lipids and laser-capture microdissection followed by bottom-up proteomics. Two out of the seven tested media (5% gelatin and 2% low-melting point agarose) provided the best performances on terms of mechanical properties. These media allowed for low-temperature embedding and for the collection of high-quality consecutive sections. Comparisons with non-embedded tissues revealed that both embedding media had no discernable effect on proteomics analysis; 5% gelatin showed a light ion suppression effect in the MALDI mass spectrometry imaging experiments, 2% agarose performed similarly to the non-embedded tissue. 2% low-melting point agarose is proposed for tissue embedding in experiments involving MALDI mass spectrometry imaging of lipids and laser-capture microdissection, proteomics of consecutive tissue sections.


Assuntos
Gelatina , Proteômica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Sefarose , Proteômica/métodos , Gelatina/química , Microdissecção e Captura a Laser/métodos , Lasers , Lipídeos , Inclusão em Parafina
14.
Nat Methods ; 20(10): 1530-1536, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37783884

RESUMO

Single-cell proteomics by mass spectrometry is emerging as a powerful and unbiased method for the characterization of biological heterogeneity. So far, it has been limited to cultured cells, whereas an expansion of the method to complex tissues would greatly enhance biological insights. Here we describe single-cell Deep Visual Proteomics (scDVP), a technology that integrates high-content imaging, laser microdissection and multiplexed mass spectrometry. scDVP resolves the context-dependent, spatial proteome of murine hepatocytes at a current depth of 1,700 proteins from a cell slice. Half of the proteome was differentially regulated in a spatial manner, with protein levels changing dramatically in proximity to the central vein. We applied machine learning to proteome classes and images, which subsequently inferred the spatial proteome from imaging data alone. scDVP is applicable to healthy and diseased tissues and complements other spatial proteomics and spatial omics technologies.


Assuntos
Proteoma , Proteômica , Animais , Camundongos , Proteoma/análise , Espectrometria de Massas/métodos , Proteômica/métodos , Microdissecção e Captura a Laser/métodos
15.
J Genet Genomics ; 50(9): 641-651, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37544594

RESUMO

Spatial omics technologies have become powerful methods to provide valuable insights into cells and tissues within a complex context, significantly enhancing our understanding of the intricate and multifaceted biological system. With an increasing focus on spatial heterogeneity, there is a growing need for unbiased, spatially resolved omics technologies. Laser capture microdissection (LCM) is a cutting-edge method for acquiring spatial information that can quickly collect regions of interest (ROIs) from heterogeneous tissues, with resolutions ranging from single cells to cell populations. Thus, LCM has been widely used for studying the cellular and molecular mechanisms of diseases. This review focuses on the differences among four types of commonly used LCM technologies and their applications in omics and disease research. Key attributes of application cases are also highlighted, such as throughput and spatial resolution. In addition, we comprehensively discuss the existing challenges and the great potential of LCM in biomedical research, disease diagnosis, and targeted therapy from the perspective of high-throughput, multi-omics, and single-cell resolution.


Assuntos
Pesquisa Biomédica , Multiômica , Microdissecção e Captura a Laser/métodos
16.
Curr Protoc ; 3(7): e844, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37486164

RESUMO

To study the transcriptome of individual plant cells at specific points in time, we developed protocols for fixation, embedding, and sectioning of plant tissue followed by laser capture microdissection (LCM) and processing for RNA recovery. LCM allows the isolation of individual cell types from heterogeneous tissue sections and is particularly suited to plant processing because it does not require the breakdown of cell walls. This approach allows accurate separation of a small volume of cells that can be used to study gene expression profiles in different tissues or cell layers. The technique requires neither separation of cells by enzymatic digestion of any kind nor cell-specific reporter genes, and it allows storage of fixed and embedded tissue for months before capture. The methods for fixation, embedding, sectioning, and capturing of plant cells that we describe yield high-quality RNA suitable for making libraries for RNASeq. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Tissue Preparation for Laser Capture Microdissection Basic Protocol 2: Tissue Sectioning Basic Protocol 3: Laser Capture Microdissection of Embedded Tissue Basic Protocol 4: RNA Extraction from Laser Capture Microdissection Samples.


Assuntos
Plantas , RNA , Microdissecção e Captura a Laser/métodos , RNA/genética , Plantas/genética , Transcriptoma , Inclusão em Parafina
17.
Methods Mol Biol ; 2672: 163-175, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37335475

RESUMO

Laser microdissection (LM) is a powerful tool for various molecular analyses providing pure samples for genomic, transcriptomic, and proteomic studies. Cell subgroups, individual cells, or even chromosomes can be separated via laser beam from complex tissues, visualized under the microscope, and used for subsequent molecular analyses. This technique provides information on nucleic acids and proteins, keeping their spatiotemporal information intact. In short, the slide with tissue is placed under the microscope, imaged by a camera onto a computer screen, where the operator selects cells/chromosomes based on morphology or staining and commands the laser beam to cut the specimen following the selected path. Samples are then collected in a tube and subjected to downstream molecular analysis, such as RT-PCR, next-generation sequencing, or immunoassay.


Assuntos
Genoma , Proteômica , Microdissecção e Captura a Laser/métodos , Cromossomos , Análise de Célula Única
18.
FEBS Lett ; 597(3): 418-426, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36285639

RESUMO

Whole-organ transcriptomic analyses have emerged as a common method for characterizing developmental transitions in mammalian organs. However, it is unclear if all cell types in an organ follow the whole-organ defined developmental trajectory. Recently, a postnatal two-stage developmental process was described for the mouse stomach. Here, using laser capture microdissection to obtain in situ transcriptomic data, we show that mouse gastric pit cells exhibit four postnatal developmental stages. Interestingly, early stages are characterized by the up-regulation of genes associated with metabolism, a functionality not typically associated with pit cells. Hence, beyond revealing that not all constituent cells develop according to the whole-organ determined pathway, these results broaden our understanding of the pit cell phenotypic landscape during stomach development.


Assuntos
Perfilação da Expressão Gênica , Transcriptoma , Animais , Camundongos , Perfilação da Expressão Gênica/métodos , Mucosa Gástrica , Microdissecção e Captura a Laser/métodos , Mamíferos
19.
J Proteome Res ; 21(10): 2435-2442, 2022 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-36153828

RESUMO

Paneth cells are antimicrobial peptide-secreting cells located at the base of the crypts of the small intestine. The proteome of Paneth cells is not well defined because of their coexistence with stem cells, making it difficult to culture Paneth cells alone in vitro. Using a simplified toluidine blue O method for staining mouse intestinal tissue, laser capture microdissection (LCM) to isolate cells from the crypt region, and surfactant-assisted one-pot protein digestion, we identified more than 1300 proteins from crypts equivalent to 18,000 cells. Compared with the proteomes of villi and smooth muscle regions, the crypt proteome is highly enriched in defensins, lysozymes, and other antimicrobial peptides that are characteristic of Paneth cells. The sensitivity of the LCM-based proteomics approach was also assessed using a smaller number of cell equivalent tissues: a comparable proteomic coverage can be achieved with 3600 cells. This work is the first proteomics study of intestinal tissue enriched with Paneth cells. The simplified workflow enables profiling of Paneth cell-associated pathological changes at the proteome level directly from frozen intestinal tissue. It may also be useful for proteomics studies of other spatially resolved cell types from other tissues.


Assuntos
Celulas de Paneth , Proteoma , Animais , Defensinas/metabolismo , Microdissecção e Captura a Laser/métodos , Camundongos , Celulas de Paneth/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteômica/métodos , Tensoativos , Cloreto de Tolônio/metabolismo
20.
STAR Protoc ; 3(4): 101698, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36149794

RESUMO

We describe a pipeline for optimized and streamlined multiplexed immunofluorescence-guided laser capture microdissection allowing the harvest of individual cells based on their phenotype and tissue localization for transcriptomic analysis with next-generation RNA sequencing. Here, we analyze transcriptomes of CD3+ T cells, CD14+ monocytes/macrophages, and melanoma cells in non-dissociated metastatic melanoma tissue. While this protocol is described for melanoma tissues, we successfully applied it to human tonsil, skin, and breast cancer tissues as well as mouse lung tissues. For complete details on the use and execution of this protocol, please refer to Martinek et al. (2022).


Assuntos
Microdissecção e Captura a Laser , Melanoma , Animais , Humanos , Camundongos , Imunofluorescência , Perfilação da Expressão Gênica/métodos , Microdissecção e Captura a Laser/métodos , Melanoma/genética , Melanoma/cirurgia , Transcriptoma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA